1
|
Francis J, Baxter M, Giza D, Cheung KL, Parks R. Patient-Reported Outcomes in Geriatric Oncology-Balancing Quality of Life and Therapeutic Effectiveness Using Primary Breast Cancer as a Model. Drugs Aging 2025; 42:1-7. [PMID: 39671154 DOI: 10.1007/s40266-024-01167-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2024] [Indexed: 12/14/2024]
Abstract
Breast cancer remains the most prevalent cancer worldwide, necessitating advancements in its management. Surgery remains the recommended primary treatment although neoadjuvant or adjuvant treatments, such as chemotherapy, may also be indicated. However, such medications confer a risk of toxicity, often resulting in dose reductions and hospitalisations. This morbidity is particularly pertinent within older patients, for whom their experience of breast cancer is already faced through the lens of unique challenges often including comorbidity, socioeconomic decline and limited support networks. Quality of life (QoL) assessments acknowledge the impact of diagnosis and treatment on patients' psychological, emotional and physical well-being. Multiple tools exist (each with their own strengths and weaknesses) ranging from the more comprehensive [such as the European Organization for the Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30)] to the more broadly focused [including the General Functional Assessment of Cancer Therapy (FACT-G)]. However, while such tools have existed for some time, there remains a gap in clinical guidance as to their integration, particularly within older patient cohorts. This article seeks to address these complexities in breast cancer decision-making by exploring how QoL assessment can best be utilised inform efficacy-tolerability trade-offs, and subsequently facilitate optimal patient-centred care.
Collapse
Affiliation(s)
- James Francis
- Nottingham Breast Cancer Research Centre, School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Uttoxeter Road, Derby, DE22 3DT, UK
| | - Mark Baxter
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland
| | - Dana Giza
- Division of Geriatric and Palliative Medicine, University of Texas Health Science Centre, McGovern Medical School, Houston, TX, USA
| | - Kwok-Leung Cheung
- Nottingham Breast Cancer Research Centre, School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Uttoxeter Road, Derby, DE22 3DT, UK
| | - Ruth Parks
- Nottingham Breast Cancer Research Centre, School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Uttoxeter Road, Derby, DE22 3DT, UK.
| |
Collapse
|
2
|
Kidane RD, Ruddy KJ, Lin G, Sandhu NP. Cardiovascular Health Considerations for Primary Care Physicians Treating Breast Cancer Survivors. Mayo Clin Proc 2025; 100:124-140. [PMID: 39641716 DOI: 10.1016/j.mayocp.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/09/2024] [Accepted: 08/13/2024] [Indexed: 12/07/2024]
Abstract
Breast cancer (BC) survivors are at increased risk for cardiovascular disease (CVD) and require their primary care physicians to manage their long-term general medical care, including cardiovascular (CV) health. Yet, evidence exists that some primary care physicians possess insufficient knowledge about survivorship care. With the goal of bridging these knowledge gaps, a PubMed review was conducted from July 7, 2020, through October 2, 2020, with an updated PubMed review from January 3, 2024, through April 28, 2024, focusing on CV health considerations in the primary care of BC survivors. Search terms included variations of "breast cancer survivors" and "cardiovascular." In total, 152 publications were included. Breasts cancer survivors may have increased CVD risk because some anticancer therapies are cardiotoxic and risk factors for BC often also increase the risk for CVD. Multiple risk factors overlap for BC and CVD such as older age, Western diet, early menarche, physical inactivity, high body mass index, and smoking. In this review, results are summarized from studies that report the presence of CV risk factors and CVD in BC survivors. Also described are the CV effects of BC therapies (chemotherapy, hormonal agents, targeted therapies, and radiotherapy) and the type of CV evaluation (cardiac imaging and measurement of biomarkers) that these patients may need. Primary care physicians have an important role in managing the CV health of BC survivors from preventing, assessing, and managing CV risk factors to referring patients to appropriate specialists when needed.
Collapse
Affiliation(s)
- Redet D Kidane
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Kathryn J Ruddy
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Grace Lin
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Nicole P Sandhu
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Ayub MA, Tyagi AR, Srivastava SK, Singh P. Quantum DFT analysis and molecular docking investigation of various potential breast cancer drugs. J Mater Chem B 2024; 13:218-238. [PMID: 39545283 DOI: 10.1039/d4tb01803f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Breast cancer is among the deadliest cancers worldwide, highlighting the urgent need for effective treatments. This study employs density functional theory (DFT) and molecular docking analyses to evaluate the anti-cancer efficacy and specificity of drug molecules lapatinib, tucatinib, neratinib, anastrozole, and letrozole. DFT analysis provides comprehensive insights into the structural, electronic, optical, and vibrational properties of these drugs, helping to elucidate their molecular stability and reactivity through global reactivity descriptors. Additionally, molecular docking simulations reveal the binding conformations and interaction profiles of these drugs with key breast cancer targets, underscoring their therapeutic potential. Docking results indicate that lapatinib, tucatinib, and neratinib have high binding affinities for HER2, with lapatinib exhibiting the strongest overall binding, particularly with PDK1 (PDB ID: 1UU7), PAK4 (PDB ID: 2X4Z), GSK3 (PDB ID: 1GNG), and HER2 (PDB ID: 2IOK). The stable hydrogen bonding and other interactions observed with lapatinib support its effectiveness in treating HER2-positive breast cancers, tucatinib's selective HER2 binding reduces off-target effects, while neratinib's irreversible binding provides prolonged inhibition, making it useful for overcoming resistance in HER2-positive cases. In contrast, anastrozole and letrozole show lower binding affinities for HER2 and EGFR due to their simpler structures but are potent aromatase inhibitors, making them effective in treating estrogen receptor-positive (ER-positive) breast cancers. In conclusion, DFT and molecular docking studies affirm the suitability of lapatinib, tucatinib, and neratinib for HER2-positive cancers, while anastrozole and letrozole are effective in ER-positive cancers, emphasizing the role of molecular structure and binding affinity in optimizing cancer treatment strategies.
Collapse
Affiliation(s)
- Md Ashraf Ayub
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari-845401, Bihar, India.
| | - Ankit Raj Tyagi
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari-845401, Bihar, India.
| | - Sunil Kumar Srivastava
- Department of Physics, School of Physical Sciences, Mahatma Gandhi Central University, Motihari-845401, Bihar, India.
| | - Pranveer Singh
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari-845401, Bihar, India.
| |
Collapse
|
4
|
Mehta K, Hegde M, Girisa S, Vishwa R, Alqahtani MS, Abbas M, Shakibaei M, Sethi G, Kunnumakkara AB. Targeting RTKs/nRTKs as promising therapeutic strategies for the treatment of triple-negative breast cancer: evidence from clinical trials. Mil Med Res 2024; 11:76. [PMID: 39668367 PMCID: PMC11636053 DOI: 10.1186/s40779-024-00582-z] [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: 10/19/2023] [Accepted: 11/08/2024] [Indexed: 12/14/2024] Open
Abstract
The extensive heterogeneity and the limited availability of effective targeted therapies contribute to the challenging prognosis and restricted survival observed in triple-negative breast cancer (TNBC). Recent research indicates the aberrant expression of diverse tyrosine kinases (TKs) within this cancer, contributing significantly to tumor cell proliferation, survival, invasion, and migration. The contemporary paradigm shift towards precision medicine has highlighted TKs and their receptors as promising targets for pharmacotherapy against a range of malignancies, given their pivotal roles in tumor initiation, progression, and advancement. Intensive investigations have focused on various monoclonal antibodies (mAbs) and small molecule inhibitors that specifically target proteins such as epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor (VEGFR), cellular mesenchymal-epithelial transition factor (c-MET), human epidermal growth factor receptor 2 (HER2), among others, for combating TNBC. These agents have been studied both in monotherapy and in combination with other chemotherapeutic agents. Despite these advances, a substantial terrain of unexplored potential lies within the realm of TK targeted therapeutics, which hold promise in reshaping the therapeutic landscape. This review summarizes the various TK targeted therapeutics that have undergone scrutiny as potential therapeutic interventions for TNBC, dissecting the outcomes and revelations stemming from diverse clinical investigations. A key conclusion from the umbrella clinical trials evidences the necessity for in-depth molecular characterization of TNBCs for the maximum efficiency of TK targeted therapeutics, either as standalone treatments or a combination. Moreover, our observation highlights that the outcomes of TK targeted therapeutics in TNBC are substantially influenced by the diversity of the patient cohort, emphasizing the prioritization of individual patient genetic/molecular profiles for precise TNBC patient stratification for clinical studies.
Collapse
Affiliation(s)
- Kasshish Mehta
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Ravichandran Vishwa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, 61421, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, 61421, Abha, Saudi Arabia
| | - Mehdi Shakibaei
- Department of Human-Anatomy, Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Ludwig-Maximilian-University, 80336, Munich, Germany
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India.
| |
Collapse
|
5
|
Gao X, Guo X, Yuan W, Jiang S, Lu Z, Luo Q, Zha Y, Wang L, Li S, Wang K, Zhu X, Yao Y. Pyrotinib induces cell death in HER2-positive breast cancer via triggering HSP90-dependent HER2 degradation and ROS/HSF-1-dependent oxidative DNA damage. Cell Stress Chaperones 2024; 29:777-791. [PMID: 39566595 DOI: 10.1016/j.cstres.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024] Open
Abstract
HER2-positive breast cancer (HER2+ BC) is distinguished by its poor prognosis, propensity for early onset, and high risk of recurrence and metastasis. Consequently, anti-HER2-targeted therapy has emerged as a principal strategy in the treatment of this form of breast cancer. Pyrotinib, a novel irreversible pan-HER2 tyrosine kinase inhibitor, has brought fresh hope to patients with advanced HER2+ breast cancer. In this study, we conducted a comprehensive exploration of pyrotinib's antitumor mechanism. The in vitro results showed that pyrotinib significantly inhibited SKBR3 cells viability and induced apoptosis by promoting HER2 endocytosis and ubiquitylation, leading to HER2 degradation through the displacement of HSP90 from HER2. Beyond targeting the HER2 signaling pathway, pyrotinib also induced DNA damage, which was mediated by the activation of the reactive oxygen species/heat shock factor 1 signaling pathway and the downregulation of proliferating cell nuclear antigen expression. Furthermore, the in vivo results demonstrated a pronounced anticancer effect of pyrotinib in the SKBR3 xenograft mouse model, concomitant with a reduction in HER2 expression. In summary, our findings provide novel insights into the mechanism of pyrotinib in the treatment of HER2+ BC.
Collapse
Affiliation(s)
- Xiaomin Gao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, China; Department of Pharmacy, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu Province, China
| | - Xu Guo
- Department of Pharmacy, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu Province, China; Department of Pharmacy, Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Wenbo Yuan
- Department of Pharmacy, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu Province, China
| | - Sunmin Jiang
- Department of Pharmacy, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu Province, China
| | - Zihong Lu
- Department of Pharmacy, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu Province, China
| | - Qing Luo
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Yuan Zha
- Department of Pharmacy, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu Province, China; Nanjing Medical University, Wuxi Medical Center, Wuxi, Jiangsu Province, China
| | - Ling Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shu Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Ke Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xue Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu Province, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Ying Yao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, China; Department of Pharmacy, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu Province, China.
| |
Collapse
|
6
|
Šermukšnytė A, Stasevych M, Komarovska-Porokhnyavets O, Zvarych V, Jakubauskienė E, Kantminienė K, Tumosienė I. Novel Antimicrobial and Antitumor Agents Bearing Pyridine-1,2,4-triazole-3-thione-hydrazone Scaffold: Synthesis, Biological Evaluation, and Molecular Docking Investigation. Biomolecules 2024; 14:1529. [PMID: 39766236 PMCID: PMC11673677 DOI: 10.3390/biom14121529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
A series of target 4-substituted-5-(2-(pyridine-2-ylamino)ethyl)-2,4-dihydro-3H-1,2,4-triazole-3-thiones and their chloro analogs 7-21 were synthesized in a reaction of the selected aldehydes with the corresponding 4-amino-1,2,4-triazole-3-thiones 5 and 6, which were obtained from 3-(pyridin-2-ylamino)propanoic acid (3) or 3-((5-chloropyridin-2-yl)amino)propanoic acid (4), respectively, with thioacetohydrazide. The antibacterial and antifungal activities of the synthesized hydrazones were screened against the bacteria Escherichia coli, Staphylococcus aureus, and Mycobacterium luteum and the fungi Candida tenuis and Aspergillus niger by agar diffusion and serial dilution methods. 4-Amino-5-(2-((5-chloropyridin-2-yl)amino)ethyl)-2,4-dihydro-3H-1,2,4-triazole-3-thione (6) and 4-(benzylideneamino)-5-(2-(pyridin-2-ylamino)ethyl)-2,4-dihydro-3H-1,2,4-triazole-3-thione (7) were identified as exceptionally active (MIC 0.9 µg/mL) against the fungus C. tenuis. 5-Chloropyridine derivatives bearing 4-benzylidene 8, 2-nitrobenzylidene 10, pyridinylmethylene 16, and 4-methylthiobenzylidene 21 moieties showed very high antibacterial activity (MIC 3.9 µg/mL) against the M. luteum strain. The cell viability screening of the synthesized compounds using triple-negative breast cancer MDA-MB-231 and glioblastoma U-87 cell lines by MTT assay identified three active hydrazones, of which 5-(2-(pyridin-2-ylamino)ethyl)-4-((pyridin-3-ylmethylene)amino)-2,4-dihydro-3H-1,2,4-triazole-3-thione (15) had the highest effect on the viability of cells (IC50 value 39.2 ± 1.7 μM against MDA-MD-231). The in silico molecular modeling results suggested that these three most active hydrazones might have influenced the mitogen-activated protein kinase pathway through the inhibition of BRAF and MEK serine-threonine protein kinases. 5-(2-((5-Chloropyridin-2-yl)amino)ethyl)-4-((4-(methylthio)benzylidene)amino)-2,4-dihydro-3H-1,2,4-triazole-3-thione (21) demonstrated the highest affinity among them.
Collapse
Affiliation(s)
- Aida Šermukšnytė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų pl. 19, 50254 Kaunas, Lithuania; (A.Š.); (I.T.)
| | - Maryna Stasevych
- Department of Technology of Biologically Active Substances, Pharmacy, and Biotechnology, Lviv Polytechnic National University, S. Bandera Str. 12, 79013 Lviv, Ukraine; (M.S.); (O.K.-P.)
| | - Olena Komarovska-Porokhnyavets
- Department of Technology of Biologically Active Substances, Pharmacy, and Biotechnology, Lviv Polytechnic National University, S. Bandera Str. 12, 79013 Lviv, Ukraine; (M.S.); (O.K.-P.)
| | - Viktor Zvarych
- Department of Automated Control Systems, Lviv Polytechnic National University, S. Bandera Str. 12, 79013 Lviv, Ukraine;
| | - Eglė Jakubauskienė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, 10257 Vilnius, Lithuania;
| | - Kristina Kantminienė
- Department of Physical and Inorganic Chemistry, Kaunas University of Technology, Radvilėnų pl. 19, 50254 Kaunas, Lithuania
| | - Ingrida Tumosienė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų pl. 19, 50254 Kaunas, Lithuania; (A.Š.); (I.T.)
| |
Collapse
|
7
|
Sankarapandian V, Rajendran RL, Miruka CO, Sivamani P, Maran BAV, Krishnamoorthy R, Gangadaran P, Ahn BC. A review on tyrosine kinase inhibitors for targeted breast cancer therapy. Pathol Res Pract 2024; 263:155607. [PMID: 39326367 DOI: 10.1016/j.prp.2024.155607] [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: 06/26/2024] [Revised: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024]
Abstract
Breast cancer is a heterogeneous disease with complex molecular pathogenesis. Overexpression of several tyrosine kinase receptors is associated with poor prognosis, therefore, they can be key targets in breast cancer therapy. Tyrosine kinase inhibitors (TKIs) have emerged as leading agents in targeted cancer therapy due to their effectiveness in disrupting key molecular pathways involved in tumor growth. TKIs target various tyrosine kinases, including the human epidermal growth factor receptor 2 (HER2), epidermal growth factor receptor (EGFR), Vascular endothelial growth factor receptor (VEGFR), anaplastic lymphoma kinase (ALK), vascular endothelial growth factor receptor (VEGFR)-associated multi-targets, rearranged during transfection (RET), fibroblast growth factor receptor (FGFR), receptor tyrosine kinase-like orphan signal 1 (ROS1), Mitogen-activated protein kinase (MAPK), and tropomyosin receptor kinase (TRK). These drugs target the tyrosine kinase domain of receptor tyrosine kinases and play a vital role in proliferation and migration of breast cancer cells. Several TKIs, including lapatinib, neratinib, and tucatinib, have been developed and are currently used in clinical settings, often in combination with chemotherapy, endocrine therapy, or other targeted agents. TKIs have demonstrated remarkable benefits in enhancing progression-free and overall survival in patients with breast cancer and have become a standard of care for this population. This review provides an overview of TKIs currently being examined in preclinical studies and clinical trials, especially in combination with drugs approved for breast cancer treatment. TKIs have emerged as a promising therapeutic option for patients with breast cancer and hold potential for treating other breast cancer subtypes. The development of new TKIs and their integration into personalized treatment strategies will continue to shape the future of breast cancer therapy.
Collapse
Affiliation(s)
- Vidya Sankarapandian
- Department of Microbiology and Immunology, Kampala International University, Western Campus, Box 20000, Uganda
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Conrad Ondieki Miruka
- Department of Biochemistry, Kampala International University, Western Campus, Box 20000, Uganda
| | - Poornima Sivamani
- Department of Pharmacology and Clinical pharmacology, Christian Medical College, Vellore 632004, India
| | - Balu Alagar Venmathi Maran
- Graduate School of Integrated Science and Technology, Nagasaki University, 1-14 Bunkyomachi, Nagasaki 852-8521, Japan
| | - Rajapandiyan Krishnamoorthy
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea..
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea..
| |
Collapse
|
8
|
Addisu S, Bekele A, Seifu D, Assefa M, Gemechu T, Hoenerhoff MJ, Merajver SD. Epidermal growth factor receptor (EGFR) and vascular endothelial growth factor A (VEGF-A) expressions in Ethiopian female breast cancer and their association with histopathologic features. PLoS One 2024; 19:e0308411. [PMID: 39405290 PMCID: PMC11478813 DOI: 10.1371/journal.pone.0308411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 07/22/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGF) play important role in breast tumor growth, invasion, metastasis, patient survival and drug resistance. The aim of this study was to evaluate the protein expression status of EGFR and VEGF-A, as well as their association with hormone receptor status and histopathological characteristics in the invasive type of female breast cancer among Ethiopians. METHOD The primary breast tumor tissues were obtained from 85 Ethiopian invasive breast cancer cases that underwent modified radical mastectomy (MRM) from June 2014 to June 2015. Their FFPE blocks were analyzed for EGFR and VEGF protein expressions using immunohistochemical techniques. The expressions were also correlated with histopathologic features. RESULT Epidermal growth factor receptor over-expression was observed in 22% of the tumor samples. VEGF-A expression was negative in 13.41%, low in 63.41%, moderate in 20.73%, and high in 2.44%. EGFR expression, but not VEGF-A, showed a significant inverse correlation with both estrogen receptor (ER) (P = 0.01) and progesterone receptor (PR) statuses (P = 0.04). EGFR and VEGF expressions did not show significant association with tumor size, grade, lymph node status or age at diagnosis. CONCLUSION Epidermal growth factor receptor expression was most likely associated with ER and PR negative tumors. Assessments of multiple molecular markers aid to understand the biological behavior of the disease in Ethiopian population. It might also help to predict which group of patients might get more benefit from the selected treatment strategies and which are not.
Collapse
Affiliation(s)
- Sisay Addisu
- Department of Biochemistry, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Abebe Bekele
- Department of Surgery, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Daniel Seifu
- Department of Biochemistry, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Mathewos Assefa
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Tufa Gemechu
- Department of Pathology, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Mark J. Hoenerhoff
- In Vivo Animal Core, Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Sofia D. Merajver
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, United States of America
| |
Collapse
|
9
|
Raj A, Chandran C S, Dua K, Kamath V, Alex AT. Targeting overexpressed surface proteins: A new strategy to manage the recalcitrant triple-negative breast cancer. Eur J Pharmacol 2024; 981:176914. [PMID: 39154820 DOI: 10.1016/j.ejphar.2024.176914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and heterogeneous cancer that lacks all three molecular markers, Estrogen, Progesterone, and Human Epidermal Growth Factor Receptor 2 (HER2). This unique characteristic of TNBC makes it more resistant to hormonal therapy; hence, chemotherapy and surgery are preferred. Active targeting with nanoparticles is more effective in managing TNBC than a passive approach. The surface of TNBC cells overexpresses several cell-specific proteins, which can be explored for diagnostic and therapeutic purposes. Immunohistochemical analysis has revealed that TNBC cells overexpress αVβ3 integrin, Intercellular Adhesion Molecule 1 (ICAM-1), Glucose Transporter 5 (GLUT5), Transmembrane Glycoprotein Mucin 1 (MUC-1), and Epidermal Growth Factor Receptor (EGFR). These surface proteins can be targeted using ligands, such as aptamers, antibodies, and sugar molecules. Targeting the surface proteins of TNBC with ligands helps harmonize treatment and improve patient compliance. In this review, we discuss the proteins expressed, which are limited to αVβ3 integrin proteins, ICAM-1, GLUT-5, MUC1, and EGFR, on the surface of TNBC, the challenges associated with the preclinical setup of breast cancer for targeted nanoformulations, internalization techniques and their challenges, suggestions to overcome the limitations of successful translation of nanoparticles, and the possibility of ligand-conjugated nanoparticles targeting these surface receptors for a better therapeutic outcome.
Collapse
Affiliation(s)
- Alan Raj
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka state, India, 576104.
| | - Sarath Chandran C
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Government Medical College Kannur, Pariyaram, Kerala, India, 670 503; Kerala University of Health Sciences, Thrissur, Kerala, India - 680 596.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, Faculty of Health, University of Technology Sydney, Sydney, Australia-2007; Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney, Australia-2007.
| | - Venkatesh Kamath
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka state, India, 576104.
| | - Angel Treasa Alex
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka state, India, 576104.
| |
Collapse
|
10
|
Nevins S, McLoughlin CD, Oliveros A, Stein JB, Rashid MA, Hou Y, Jang MH, Lee KB. Nanotechnology Approaches for Prevention and Treatment of Chemotherapy-Induced Neurotoxicity, Neuropathy, and Cardiomyopathy in Breast and Ovarian Cancer Survivors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300744. [PMID: 37058079 PMCID: PMC10576016 DOI: 10.1002/smll.202300744] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/05/2023] [Indexed: 06/19/2023]
Abstract
Nanotechnology has emerged as a promising approach for the targeted delivery of therapeutic agents while improving their efficacy and safety. As a result, nanomaterial development for the selective targeting of cancers, with the possibility of treating off-target, detrimental sequelae caused by chemotherapy, is an important area of research. Breast and ovarian cancer are among the most common cancer types in women, and chemotherapy is an essential treatment modality for these diseases. However, chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy are common side effects that can affect breast and ovarian cancer survivors quality of life. Therefore, there is an urgent need to develop effective prevention and treatment strategies for these adverse effects. Nanoparticles (NPs) have extreme potential for enhancing therapeutic efficacy but require continued research to elucidate beneficial interventions for women cancer survivors. In short, nanotechnology-based approaches have emerged as promising strategies for preventing and treating chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy. NP-based drug delivery systems and therapeutics have shown potential for reducing the side effects of chemotherapeutics while improving drug efficacy. In this article, the latest nanotechnology approaches and their potential for the prevention and treatment of chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy in breast and ovarian cancer survivors are discussed.
Collapse
Affiliation(s)
- Sarah Nevins
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Callan D. McLoughlin
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Alfredo Oliveros
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Joshua B. Stein
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Mohammad Abdur Rashid
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| |
Collapse
|
11
|
Yu C, Richly M, Hoang TT, El Beheiry M, Türkcan S, Masson JB, Alexandrou A, Bouzigues CI. Confinement energy landscape classification reveals membrane receptor nano-organization mechanisms. Biophys J 2024; 123:1882-1895. [PMID: 38845200 PMCID: PMC11267427 DOI: 10.1016/j.bpj.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 03/01/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024] Open
Abstract
The cell membrane organization has an essential functional role through the control of membrane receptor confinement in micro- or nanodomains. Several mechanisms have been proposed to account for these properties, although some features have remained controversial, notably the nature, size, and stability of cholesterol- and sphingolipid-rich domains or lipid rafts. Here, we probed the effective energy landscape acting on single-nanoparticle-labeled membrane receptors confined in raft nanodomains- epidermal growth factor receptor (EGFR), Clostridium perfringens ε-toxin receptor (CPεTR), and Clostridium septicum α-toxin receptor (CSαTR)-and compared it with hop-diffusing transferrin receptors. By establishing a new analysis pipeline combining Bayesian inference, decision trees, and clustering approaches, we systematically classified single-protein trajectories according to the type of effective confining energy landscape. This revealed the existence of only two distinct organization modalities: confinement in a quadratic energy landscape for EGFR, CPεTR, and CSαTR (A), and free diffusion in confinement domains resulting from the steric hindrance due to F-actin barriers for transferrin receptor (B). The further characterization of effective confinement energy landscapes by Bayesian inference revealed the role of interactions with the domain environment in cholesterol- and sphingolipid-rich domains with (EGFR) or without (CPεTR and CSαTR) interactions with F-actin to regulate the confinement energy depth. These two distinct mechanisms result in the same organization type (A). We revealed that the apparent domain sizes for these receptor trajectories resulted from Brownian exploration of the energy landscape in a steady-state-like regime at a common effective temperature, independently of the underlying molecular mechanisms. These results highlight that confinement domains may be adequately described as interaction hotspots rather than rafts with abrupt domain boundaries. Altogether, these results support a new model for functional receptor confinement in membrane nanodomains and pave the way to the constitution of an atlas of membrane protein organization.
Collapse
Affiliation(s)
- Chao Yu
- Laboratoire Optique et Biosciences, CNRS UMR74645, Inserm U1182, Ecole Polytechnique, Institut Polytechnique Paris, Palaiseau, France
| | - Maximilian Richly
- Laboratoire Optique et Biosciences, CNRS UMR74645, Inserm U1182, Ecole Polytechnique, Institut Polytechnique Paris, Palaiseau, France
| | - Thi Thuy Hoang
- Laboratoire Optique et Biosciences, CNRS UMR74645, Inserm U1182, Ecole Polytechnique, Institut Polytechnique Paris, Palaiseau, France
| | - Mohammed El Beheiry
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Decision and Bayesian Computation, Paris, France; Épiméthée, INRIA, Paris, France
| | - Silvan Türkcan
- Laboratoire Optique et Biosciences, CNRS UMR74645, Inserm U1182, Ecole Polytechnique, Institut Polytechnique Paris, Palaiseau, France
| | - Jean-Baptiste Masson
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Decision and Bayesian Computation, Paris, France; Épiméthée, INRIA, Paris, France
| | - Antigoni Alexandrou
- Laboratoire Optique et Biosciences, CNRS UMR74645, Inserm U1182, Ecole Polytechnique, Institut Polytechnique Paris, Palaiseau, France
| | - Cedric I Bouzigues
- Laboratoire Optique et Biosciences, CNRS UMR74645, Inserm U1182, Ecole Polytechnique, Institut Polytechnique Paris, Palaiseau, France.
| |
Collapse
|
12
|
Du J, Liu X, Sun J, Wu Q, Hu Y, Shi H, Zheng L, Liu Y, Wu C, Gao Y. Trastuzumab-functionalized bionic pyrotinib liposomes for targeted therapy of HER2-positive breast cancer. Breast Cancer Res 2024; 26:99. [PMID: 38867302 PMCID: PMC11167944 DOI: 10.1186/s13058-024-01853-2] [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: 09/19/2023] [Accepted: 06/01/2024] [Indexed: 06/14/2024] Open
Abstract
In this study, we prepared a bionic nanosystem of trastuzumab-functionalized SK-BR-3 cell membrane hybrid liposome-coated pyrotinib (Ptb-M-Lip-Her) for the treatment of HER2-positive breast cancer. Transmission electron microscopy, dynamic light scattering, polyacrylamide gel electrophoresis (SDS-PAGE) and western blotting were used to verify the successful preparation of Ptb-M-Lip-Her. In vitro drug release experiments proved that Ptb-M-Lip-Her had a sustained release effect. Cell uptake experiments and in vivo imaging experiments proved that Ptb-M-Lip-Her had good targeting ability to homologous tumor cells (SK-BR-3). The results of cell experiments such as MTT, flow cytometry, immunofluorescence staining and in vivo antitumor experiments showed that Ptb-M-Lip-Her could significantly promote apoptosis and inhibit the proliferation of SK-BR-3 cells. These results clearly indicated that Ptb-M-Lip-Her may be a promising biomimetic nanosystem for targeted therapy of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Jiaqun Du
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, 121001, Liaoning, China
| | - Xiaobang Liu
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, 121001, Liaoning, China
| | - Junpeng Sun
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, 121001, Liaoning, China
| | - Qian Wu
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, 121001, Liaoning, China
| | - Yu Hu
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, 121001, Liaoning, China
| | - Huan Shi
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, 121001, Liaoning, China
| | - Li Zheng
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, 121001, Liaoning, China
| | - Ying Liu
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, 121001, Liaoning, China.
| | - Chao Wu
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, 121001, Liaoning, China.
| | - Yu Gao
- Department of Medical Oncology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, the Fifth Section of Renmin Street, Guta District, Jinzhou, 121001, Liaoning Province, China.
| |
Collapse
|
13
|
Joo EH, Kim S, Park D, Lee T, Park WY, Han KY, Lee JE. Migratory Tumor Cells Cooperate with Cancer Associated Fibroblasts in Hormone Receptor-Positive and HER2-Negative Breast Cancer. Int J Mol Sci 2024; 25:5876. [PMID: 38892065 PMCID: PMC11172245 DOI: 10.3390/ijms25115876] [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: 03/20/2024] [Revised: 05/17/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Hormone receptor-positive and HER2-negative breast cancer (HR+/HER2-BC) is the most common type with a favorable prognosis under endocrine therapy. However, it still demonstrates unpredictable progression and recurrences influenced by high tumoral diversity and microenvironmental status. To address these heterogeneous molecular characteristics of HR+/HER2-BC, we aimed to simultaneously characterize its transcriptomic landscape and genetic architecture at the same resolution. Using advanced single-cell RNA and DNA sequencing techniques together, we defined four distinct tumor subtypes. Notably, the migratory tumor subtype was closely linked to genomic alterations of EGFR, related to the tumor-promoting behavior of IL6-positive inflammatory tumor-associated fibroblast, and contributing to poor prognosis. Our study comprehensively utilizes integrated analysis to uncover the complex dynamics of this breast cancer subtype, highlighting the pivotal role of the migratory tumor subtype in influencing surrounding cells. This sheds light on potential therapeutic targets by offering enhanced insights for HR+/HER2-BC treatment.
Collapse
Affiliation(s)
- Eun Hye Joo
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea; (E.H.J.); (W.-Y.P.)
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06355, Republic of Korea
| | - Sangmin Kim
- Department of Breast Cancer Center, Samsung Medical Center, Seoul 06351, Republic of Korea;
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Donghyun Park
- Planit Healthcare Inc., Seoul 06235, Republic of Korea;
| | - Taeseob Lee
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06355, Republic of Korea;
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea; (E.H.J.); (W.-Y.P.)
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06355, Republic of Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Kyung Yeon Han
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea; (E.H.J.); (W.-Y.P.)
| | - Jeong Eon Lee
- Department of Breast Cancer Center, Samsung Medical Center, Seoul 06351, Republic of Korea;
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| |
Collapse
|
14
|
Kheraldine H, Hassan AF, Alhussain H, Al-Thawadi H, Vranic S, Al Moustafa AE. Effects of neratinib on angiogenesis and the early stage of the embryo using chicken embryo as a model. BIOMOLECULES & BIOMEDICINE 2024; 24:575-581. [PMID: 38158791 PMCID: PMC11088882 DOI: 10.17305/bb.2023.9869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/17/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Angiogenesis is the process of forming new blood capillaries from pre-existing vessels. Even though it is essential during normal development, it plays a major role in cancer progression. Neratinib is a pan-human epidermal growth factor receptor (HER) inhibitor that has recently been approved for the treatment of HER2-positive breast cancer. However, its effects on angiogenesis and embryogenesis remain unknown. This study examined the antiangiogenic effects of neratinib using the chorioallantoic membrane (CAM) of chicken embryos. We also evaluated neratinib's toxicity during the early stages of normal development using the chicken embryos, primary embryonic fibroblasts (EFBs), and human umbilical vein endothelial cells (HUVEC). Our findings revealed that neratinib significantly inhibited the CAM angiogenesis compared to controls by reducing vessel percentage area and the average vessel length. Furthermore, neratinib downregulated vascular endothelial growth factor (VEGF), a key mediator of angiogenesis. At lower concentrations, neratinib was well-tolerated during early stages of normal development. Additionally, EFBs treated with neratinib showed no morphological or viability changes when compared to controls. However, at the highest concentration tested, neratinib treatment reduced HUVEC cell viability. This effect may be associated with the dysregulation of key apoptotic genes, including caspase-3, caspase-8, caspase-9, and the B-cell lymphoma 2 (Bcl2) gene. Our findings indicate a novel potential application of neratinib as an antiangiogenic agent, exhibiting tolerable toxicity in the early stages of embryogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Semir Vranic
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, Doha, Qatar
- Biomedical Research Center, Qatar University, Doha, Qatar
- Oncology Department, Faculty of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
15
|
Khaparkhuntikar K, Maji I, Gupta SK, Mahajan S, Aalhate M, Sriram A, Gupta U, Guru SK, Kulkarni P, Singh PK. Acalabrutinib as a novel hope for the treatment of breast and lung cancer: an in-silico proof of concept. J Biomol Struct Dyn 2024; 42:1469-1484. [PMID: 37272883 DOI: 10.1080/07391102.2023.2217923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/01/2023] [Indexed: 06/06/2023]
Abstract
Drug repurposing is proved to be a groundbreaking concept in the field of cancer research, accelerating the pace of de novo drug discovery by investigating the anti-cancer activity of the already approved drugs. On the other hand, it got highly benefitted from the advancement in the in-silico tools and techniques, which are used to build up the initial "proof of concept" based on the drug-target interaction. Acalabrutinib (ACL) is a well-known drug for the treatment of hematological malignancies. But, the therapeutic ability of ACL against solid tumors is still unexplored. Thereby, the activity of ACL on breast cancer and lung cancer was evaluated utilizing different computational methods. A series of proteins such as VEGFR1, ALK, BCL2, CXCR-4, mTOR, AKT, PI3K, HER-2, and Estrogen receptors were selected based on their involvement in the progression of the breast as well as lung cancer. A multi-level computational study starting from protein-ligand docking to molecular dynamic (MD) simulations were performed to detect the binding potential of ACL towards the selected proteins. Results of the study led to the identification of ACL as a ligand that showed a high docking score and binding energy with HER-2, mTOR, and VEGFR-1 successively. Whereas, the MD simulations study has also shown good docked complex stability of ACL with HER2 and VEGFR1. Our findings suggest that interaction with those receptors can lead to preventive action on both breast and lung cancer, thus it can be concluded that ACL could be a potential molecule for the same purpose.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Kedar Khaparkhuntikar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Indrani Maji
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Sunil Kumar Gupta
- Department of Bioinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Anitha Sriram
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Ujala Gupta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Santosh Kumar Guru
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Prachi Kulkarni
- Department of Physiology, Shri B. M. Patil Medical College, Hospital & Research Centre BLDE (Deemed to be University), Vijayapura, Karnataka, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
16
|
Limsakul P, Choochuen P, Jungrungrueang T, Charupanit K. Prognostic Markers in Tyrosine Kinases Specific to Basal-like 2 Subtype of Triple-Negative Breast Cancer. Int J Mol Sci 2024; 25:1405. [PMID: 38338684 PMCID: PMC10855431 DOI: 10.3390/ijms25031405] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Triple-negative breast cancer (TNBC), a heterogeneous and therapeutically challenging subtype, comprises over 50% of patients categorized into basal-like 1 (BL1) and basal-like 2 (BL2) intrinsic molecular subtypes. Despite their shared basal-like classification, BL2 is associated with a poor response to neoadjuvant chemotherapy and reduced relapse-free survival compared to BL1. Here, the study focused on identifying subtype-specific markers for BL2 through transcriptomic analysis of TNBC patients using RNA-seq and clinical integration. Six receptor tyrosine kinase (TK) genes, including EGFR, EPHA4, EPHB2, PDGFRA, PDGFRB, and ROR1, were identified as potential differentiators for BL2. Correlations between TK mRNA expression and TNBC prognosis, particularly EGFR, PDGFRA, and PDGFRB, revealed potential synergistic interactions in pathways related to cell survival and proliferation. Our findings also suggest promising dual markers for predicting disease prognosis. Furthermore, RT-qPCR validation demonstrated that identified BL2-specific TKs were expressed at a higher level in BL2 than in BL1 cell lines, providing insights into unique characteristics. This study advances the understanding of TNBC heterogeneity within the basal-like subtypes, which could lead to novel clinical treatment approaches and the development of targeted therapies.
Collapse
Affiliation(s)
- Praopim Limsakul
- Division of Physical Science, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand;
- Center of Excellence for Trace Analysis and Biosensor (TAB-CoE), Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
| | - Pongsakorn Choochuen
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.C.); (T.J.)
| | - Thawirasm Jungrungrueang
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.C.); (T.J.)
| | - Krit Charupanit
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.C.); (T.J.)
| |
Collapse
|
17
|
Ozer LY, Fayed HS, Ericsson J, Al Haj Zen A. Development of a cancer metastasis-on-chip assay for high throughput drug screening. Front Oncol 2024; 13:1269376. [PMID: 38239643 PMCID: PMC10794518 DOI: 10.3389/fonc.2023.1269376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Metastasis is the cause of most triple-negative breast cancer deaths, yet anti-metastatic therapeutics remain limited. To develop new therapeutics to prevent metastasis, pathophysiologically relevant assays that recapitulate tumor microenvironment is essential for disease modeling and drug discovery. Here, we have developed a microfluidic metastasis-on-chip assay of the early stages of cancer metastasis integrated with the triple-negative breast cancer cell line (MDA-MB-231), stromal fibroblasts and a perfused microvessel. High-content imaging with automated quantification methods was optimized to assess the tumor cell invasion and intravasation within the model. Cell invasion and intravasation were enhanced when fibroblasts co-cultured with a breast cancer cell line (MDA-MB-231). However, the non-invasive breast cancer cell line, MCF7, remained non-invasive in our model, even in the presence of fibroblasts. High-content screening of a targeted anti-cancer therapy drug library was conducted to evaluate the drug response sensitivity of the optimized model. Through this screening, we identified 30 compounds that reduced the tumor intravasation by 60% compared to controls. Multi-parametric phenotypic analysis was applied by combining the data from the metastasis-on-chip, cell proliferation and 2D cell migration screens, revealing that the drug library was clustered into eight distinct groups with similar drug responses. Notably, MEK inhibitors were enriched in cluster cell invasion and intravasation. In contrast, drugs with molecular targets: ABL, KIT, PDGF, SRC, and VEGFR were enriched in the drug clusters showing a strong effect on tumor cell intravasation with less impact on cell invasion or cell proliferation, of which, Imatinib, a multi-kinase inhibitor targeting BCR-ABL/PDGFR/KIT. Further experimental analysis showed that Imatinib enhanced endothelial barrier stability as measured by trans-endothelial electrical resistance and significantly reduced the trans-endothelial invasion activity of tumor cells. Our findings demonstrate the potential of our metastasis-on-chip assay as a powerful tool for studying cancer metastasis biology, drug discovery aims, and assessing drug responses, offering prospects for personalized anti-metastatic therapies for triple-negative breast cancer patients.
Collapse
Affiliation(s)
| | | | | | - Ayman Al Haj Zen
- College of Health and Life Sciences, Hamad bin Khalifa University, Doha, Qatar
| |
Collapse
|
18
|
Sun K, Wang X, Zhang H, Lin G, Jiang R. Management and Mechanisms of Diarrhea Induced by Tyrosine Kinase Inhibitors in Human Epidermal Growth Factor Receptor-2-Positive Breast Cancer. Cancer Control 2024; 31:10732748241278039. [PMID: 39159918 PMCID: PMC11334140 DOI: 10.1177/10732748241278039] [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: 06/04/2024] [Revised: 07/20/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024] Open
Abstract
Breast cancer has the highest incidence among female malignancies, significantly impacting women's health. Recently, numerous HER2-targeted therapies have achieved excellent clinical outcomes. Currently, anti-HER2 drugs are divided into three main categories: monoclonal antibodies, small-molecule tyrosine kinase inhibitors, and antibody-coupled drugs (ADCs). The main toxic side effects of small molecule TKI-based therapy are diarrhea, hand-foot syndrome, rash, nausea, and vomiting. Diarrhea is a potential predictor of tumor response, affecting up to 95% of cancer patients treated with TKIs. Severe gastrointestinal toxicity can result in the need for dose reductions and treatment interruptions. This not only compromises the efficacy of TKIs but also deteriorates human nutrition and quality of life. The majority of individuals develop diarrhea within 7 days of starting treatment, with approximately 30% developing grade 3 or higher diarrhea within 2-3 days of starting treatment. The severity of diarrhea typically correlates with the dosage of most TKIs. Current prevention and management strategies are primarily empirical, focusing on symptom alleviation rather than addressing the toxicological mechanisms underlying TKI-induced diarrhea. Consequently, anti-diarrheal drugs are often less effective in managing this condition in cancer patients receiving TKIs. Moreover, our understanding of the toxicological mechanisms responsible for such diarrhea remains limited, underscoring the urgent need to identify these mechanisms in order to develop effective anti-diarrheal medications tailored to this specific context. This review aims to elucidate management approaches and mechanisms for diarrhea induced by TKIs during HER2-positive breast cance.
Collapse
Affiliation(s)
- Kena Sun
- Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Xiaojia Wang
- Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Huanping Zhang
- Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Guang Lin
- Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Ruiyuan Jiang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
19
|
Wang S, Gu Z, Zhu L, Han Y, Yu H, Fang W, Han B. Genetic insights into thymic carcinomas and thymic neuroendocrine neoplasms denote prognosis signatures and pathways. Chin Med J (Engl) 2023; 136:2712-2721. [PMID: 37749819 PMCID: PMC10684125 DOI: 10.1097/cm9.0000000000002852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Thymic carcinomas (TCs) and thymic neuroendocrine neoplasms (TNENs) are two aggressive subtypes of thymic malignancy. Traditional therapy for advanced TCs and TNENs has limited outcome. New genomic profiling of TCs and TNENs might provide insights that contribute to the development of new treatment approaches. METHODS We used gene panel sequencing technologies to investigate the genetic aberrations of 32 TC patients and 15 TNEN patients who underwent surgery at Shanghai Chest Hospital between 2015 and 2017. Patient samples were sequenced using a 324-gene platform with licensed technologies. In this study, we focused on clinically relevant genomic alterations (CRGAs), which are previously proven to be pathogenic alterations, to identify the pathology-specific mutational patterns, prognostic signatures of TCs and TNENs. RESULTS The mutational profiles between TCs and TNENs were diverse. The genetic alterations that ranked highest in TCs were in CDKN2A, TP53, ASXL1, CDKN2B, PIK3C2G, PTCH1, and ROS1 , while those in TNENs were in MEN1, MLL2, APC, RB1 , and TSC2 . Prognostic analysis showed that mutations of ROS1, CDKN2A, CDKN2B, BRAF, and BAP1 were significantly associated with worse outcomes in TC patients, and that mutation of ERBB2 indicated shortened disease-free survival (DFS) and overall survival (OS) in TNEN patients. Further investigation found that the prognosis-related genes were focused on signal pathways of cell cycle control, chromatin remodeling/DNA methylation, phosphoinositide 3-kinases (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR), and receptor tyrosine kinase (RTK)/RAS/mitogen-activated protein kinase (MAPK) signaling. CONCLUSION We profiled the mutational features of 47 Chinese patients with thymic malignancy of diverse pathologic phenotypes to uncover the integrated genomic landscape of these rare tumors, and identified the pathology-specific mutational patterns, prognostic signatures, and potential therapeutic targets for TCs and TNENs.
Collapse
Affiliation(s)
- Shuyuan Wang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhitao Gu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Lei Zhu
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yuchen Han
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Hong Yu
- Department of Radiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Wentao Fang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Baohui Han
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| |
Collapse
|
20
|
Guo L, Shao W, Zhou C, Yang H, Yang L, Cai Q, Wang J, Shi Y, Huang L, Zhang J. Neratinib for HER2-positive breast cancer with an overlooked option. Mol Med 2023; 29:134. [PMID: 37803271 PMCID: PMC10559443 DOI: 10.1186/s10020-023-00736-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 09/28/2023] [Indexed: 10/08/2023] Open
Abstract
Positive human epidermal growth factor receptor 2 (HER2) expression is associated with an increased risk of metastases especially those to the brain in patients with advanced breast cancer (BC). Neratinib as a tyrosine kinase inhibitor can prevent the transduction of HER1, HER2 and HER4 signaling pathways thus playing an anticancer effect. Moreover, neratinib has a certain efficacy to reverse drug resistance in patients with BC with previous HER2 monoclonal antibody or targeted drug resistance. Neratinib, as monotherapy and in combination with other therapies, has been tested in the neoadjuvant, adjuvant, and metastatic settings. Neratinib with high anticancer activity is indicated for the prolonged adjuvant treatment of HER2-positive early BC, or in combination with other drugs including trastuzumab, capecitabine, and paclitaxel for the treatment of advanced HER2-positive BC especially cancers with central nervous system (CNS) metastasis to reduce the risk of BC recurrence. This article reviewed the pharmacological profiles, efficacy, safety, tolerability, and current clinical trials pertaining to neratinib, with a particular focus on the use of neratinib in patients with metastatic breast cancer (MBC) involving the CNS. We further discussed the use of neratinib for HER2-negative and HER2-mutant breast cancers, and mechanisms of resistance to neratinib. The current evidence suggests that neratinib has promising efficacy in patients with BC which is at least non-inferior compared to previous therapeutic regimens. The most common AE was diarrhea, and the incidence, severity and duration of neratinib-related grade 3 diarrhea can be reduced with loperamide. Of note, neratinib has the potential to effectively control and prevent brain metastasis in patients with advanced BC, providing a therapeutic strategy for HER2-positive BC.
Collapse
Affiliation(s)
- Liting Guo
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| | - Weiwei Shao
- Department of Pathology, The First People's Hospital of Yancheng City, Yancheng, China
| | - Chenfei Zhou
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Hui Yang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Liu Yang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Qu Cai
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Junqing Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| | - Yan Shi
- Department of General Surgery, Shanghai Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, 358 Datong Road, Gaoqiao Town, Shanghai, 200137, China.
| | - Lei Huang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
- Medical Center on Aging of Ruijin Hospital, MCARJH, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| |
Collapse
|
21
|
Guiselin T, Lecoutey C, Rochais C, Dallemagne P. Conceptual Framework of the Design of Pleiotropic Drugs against Alzheimer's Disease. Pharmaceutics 2023; 15:2382. [PMID: 37896142 PMCID: PMC10610275 DOI: 10.3390/pharmaceutics15102382] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023] Open
Abstract
The multifactorial nature of some diseases, particularly neurodegenerative diseases such as Alzheimer's disease, frequently requires the use of several drugs. These drug cocktails are not without drawbacks in terms of increased adverse effects, drug-drug interactions or low adherence to treatment. The use of pleiotropic drugs, which combine, within a single molecule, several activities directed against distinct therapeutic targets, makes it possible to overcome some of these problems. In addition, these pleiotropic drugs generally lead to the expression of a synergy of effects, sometimes greater than that observed with a combination of drugs. This article will review, through recent examples, the different kinds of pleiotropic drugs being studied or already present on the market of medicines, with a focus on the structural aspect of such drug design.
Collapse
Affiliation(s)
| | | | | | - Patrick Dallemagne
- Normandie University, Unicaen, Centre d’Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France; (T.G.); (C.L.); (C.R.)
| |
Collapse
|
22
|
Bansal I, Pandey AK, Ruwali M. Small-molecule inhibitors of kinases in breast cancer therapy: recent advances, opportunities, and challenges. Front Pharmacol 2023; 14:1244597. [PMID: 37711177 PMCID: PMC10498465 DOI: 10.3389/fphar.2023.1244597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023] Open
Abstract
Breast cancer is the most common malignancy in women worldwide and despite significant advancements in detection, treatment, and management of cancer, it is still the leading cause of malignancy related deaths in women. Understanding the fundamental biology of breast cancer and creating fresh diagnostic and therapeutic strategies have gained renewed focus in recent studies. In the onset and spread of breast cancer, a group of enzymes known as kinases are extremely important. Small-molecule kinase inhibitors have become a promising class of medications for the treatment of breast cancer owing to their capacity to specifically target kinases involved in the growth and progression of cancer. The creation of targeted treatments that block these kinases and the signalling pathways that they activate has completely changed how breast cancer is treated. Many of these targeted treatments have been approved for the treatment of breast cancer as clinical trials have demonstrated their great efficacy. CDK4/6 inhibitors, like palbociclib, abemaciclib, and ribociclib, EGFR inhibitors such as gefitinib and erlotinib and HER2-targeting small-molecule kinases like neratinib and tucatinib are some examples that have shown potential in treating breast cancer. Yet, there are still difficulties in the development of targeted medicines for breast cancer, such as figuring out which patient subgroups may benefit from these therapies and dealing with drug resistance problems. Notwithstanding these difficulties, kinase-targeted treatments for breast cancer still have a lot of potential. The development of tailored medicines will continue to be fuelled by the identification of novel targets and biomarkers for breast cancer as a result of advancements in genomic and proteomic technology.
Collapse
Affiliation(s)
- Isha Bansal
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram, Haryana, India
| | - Amit Kumar Pandey
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER-Ahmedabad), Gandhinagar, Gujarat, India
| | - Munindra Ruwali
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram, Haryana, India
| |
Collapse
|
23
|
Bouabdallah S, Al-Maktoum A, Amin A. Steroidal Saponins: Naturally Occurring Compounds as Inhibitors of the Hallmarks of Cancer. Cancers (Basel) 2023; 15:3900. [PMID: 37568716 PMCID: PMC10417465 DOI: 10.3390/cancers15153900] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Cancer is a global health burden responsible for an exponentially growing number of incidences and mortalities, regardless of the significant advances in its treatment. The identification of the hallmarks of cancer is a major milestone in understanding the mechanisms that drive cancer initiation, development, and progression. In the past, the hallmarks of cancer have been targeted to effectively treat various types of cancers. These conventional cancer drugs have shown significant therapeutic efficacy but continue to impose unfavorable side effects on patients. Naturally derived compounds are being tested in the search for alternative anti-cancer drugs. Steroidal saponins are a group of naturally occurring compounds that primarily exist as secondary metabolites in plant species. Recent studies have suggested that steroidal saponins possess significant anti-cancer capabilities. This review aims to summarize the recent findings on steroidal saponins as inhibitors of the hallmarks of cancer and covers key studies published between the years 2014 and 2024. It is reported that steroidal saponins effectively inhibit the hallmarks of cancer, but poor bioavailability and insufficient preclinical studies limit their utilization.
Collapse
Affiliation(s)
- Salwa Bouabdallah
- Theranostic Biomarkers, LR23ES02, Faculty of Medicine of Tunis, Université Tunis El Manar, Tunis 1006, Tunisia
| | - Amna Al-Maktoum
- Biology Department, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| | - Amr Amin
- Biology Department, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| |
Collapse
|
24
|
Hu ZY, Yan M, Xiong H, Ran L, Zhong J, Luo T, Sun T, Xie N, Liu L, Yang X, Xiao H, Li J, Liu B, Ouyang Q. Pyrotinib in combination with letrozole for hormone receptor-positive, human epidermal growth factor receptor 2-positive metastatic breast cancer (PLEHERM): a multicenter, single-arm, phase II trial. BMC Med 2023; 21:226. [PMID: 37365596 DOI: 10.1186/s12916-023-02943-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Human epidermal growth factor receptor 2 (HER2) targeted therapy combined with endocrine therapy has been recommended as an alternative treatment strategy for patients with hormone receptor (HR)-positive, HER2-positive metastatic breast cancer (MBC). This study aimed to evaluate the role of pyrotinib, an oral pan-HER irreversible tyrosine kinase inhibitor, in combination with letrozole for patients with HR-positive, HER2-positive MBC. METHODS In this multi-center, phase II trial, HR-positive and HER2-positive MBC patients who were not previously treated for metastasis disease were enrolled. Patients received daily oral pyrotinib 400 mg and letrozole 2.5 mg until disease progression, unacceptable toxicity, or withdrawal of consent. The primary endpoint was the clinical benefit rate (CBR) assessed by an investigator according to the Response Evaluation Criteria in Solid Tumors version 1.1. RESULTS From November 2019 to December 2021, 53 patients were enrolled and received pyrotinib plus letrozole. As of August 2022, the median follow-up duration was 11.6 months (95% confidence interval [CI], 8.7-14.0 months). The CBR was 71.7% (95% CI, 57.7-83.2%), and the objective response rate was 64.2% (95% CI, 49.8-76.9%). The median progression-free survival was 13.7 months (95% CI, 10.7-18.7 months). The most common treatment-related adverse event of grade 3 or higher was diarrhea (18.9%). No treatment-related deaths were reported, and one patient experienced treatment discontinuation due to adverse event. CONCLUSIONS Our preliminary results suggested that pyrotinib plus letrozole is feasible for the first-line treatment of patients with HR-positive and HER2-positive MBC, with manageable toxicities. TRIAL REGISTRATION ClinicalTrials.gov, NCT04407988.
Collapse
Affiliation(s)
- Zhe-Yu Hu
- Medical Department of Breast Cancer, Hunan Cancer Hospital, No. 283, Tongzipo Road, Changsha, 410013, China
- Medical Department of Breast Cancer, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Min Yan
- Department of Breast Cancer, Henan Cancer Hospital, Zhengzhou, China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Li Ran
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jincai Zhong
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ting Luo
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Sun
- Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, China
| | - Ning Xie
- Medical Department of Breast Cancer, Hunan Cancer Hospital, No. 283, Tongzipo Road, Changsha, 410013, China
- Medical Department of Breast Cancer, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Liping Liu
- Medical Department of Breast Cancer, Hunan Cancer Hospital, No. 283, Tongzipo Road, Changsha, 410013, China
- Medical Department of Breast Cancer, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiaohong Yang
- Medical Department of Breast Cancer, Hunan Cancer Hospital, No. 283, Tongzipo Road, Changsha, 410013, China
- Medical Department of Breast Cancer, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Huawu Xiao
- Medical Department of Breast Cancer, Hunan Cancer Hospital, No. 283, Tongzipo Road, Changsha, 410013, China
- Medical Department of Breast Cancer, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jing Li
- Medical Department of Breast Cancer, Hunan Cancer Hospital, No. 283, Tongzipo Road, Changsha, 410013, China
- Medical Department of Breast Cancer, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Binliang Liu
- Medical Department of Breast Cancer, Hunan Cancer Hospital, No. 283, Tongzipo Road, Changsha, 410013, China
- Medical Department of Breast Cancer, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Quchang Ouyang
- Medical Department of Breast Cancer, Hunan Cancer Hospital, No. 283, Tongzipo Road, Changsha, 410013, China.
- Medical Department of Breast Cancer, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
| |
Collapse
|
25
|
He S, Hou Y, Hou L, Chen N, Yang X, Wang H, Han P, Fan Y, Zhao J, Zhang J, Geng J. Targeted RASSF1A expression inhibits proliferation of HER2‑positive breast cancer cells in vitro. Exp Ther Med 2023; 25:245. [PMID: 37153885 PMCID: PMC10160914 DOI: 10.3892/etm.2023.11944] [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: 11/16/2022] [Accepted: 03/15/2023] [Indexed: 05/10/2023] Open
Abstract
Human epidermal growth factor receptor 2-positive (HER2+) breast cancer, which accounts for 15-20% of all breast cancer, is associated with tumor recurrence and poor prognosis. RAS association domain family protein 1 subtype A (RASSF1A) is a tumor suppressor that is silenced in a variety of human cancers. The present study aimed to investigate the role of RASSF1A in HER2+ breast cancer and the therapeutic potential of RASSF1A-based targeted gene therapy for this malignancy. RASSF1A expression in human HER2+ breast cancer tissues and cell lines was evaluated by reverse transcription PCR and western blot analysis. The associations between tumorous RASSF1A level and tumor grade, TNM stage, tumor size, lymph node metastasis and five-year survival were examined. HER2+ and HER2-negative (HER2-) breast cancer cells were transfected with a lentiviral vector (LV-5HH-RASSF1A) that could express RASSF1A under the control of five copies of the hypoxia-responsive element (5HRE) and one copy of the HER2 promoter (HER2p). Cell proliferation was evaluated by the MTT and colony formation assays. It was found that tumorous RASSF1A level was negatively associated with tumor grade (P=0.014), TNM stage (P=0.0056), tumor size (P=0.014) and lymph node metastasis (P=0.029) and positively associated with five-year survival (P=0.038) in HER2+ breast cancer patients. Lentiviral transfection of HER2+ breast cancer cells resulted in increased RASSF1A expression and decreased cell proliferation, especially under hypoxic conditions. However, lentiviral transfection of HER2-breast cancer cells did not affect RASSF1A expression. In conclusion, these findings verified the clinical significance of RASSF1A as a tumor suppressor in HER2+ breast cancer and supported LV-5HH-RASSF1A as a potential targeted gene therapy for this malignancy.
Collapse
Affiliation(s)
- Sai He
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Yanni Hou
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Leina Hou
- Department of Anesthesiology, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Nan Chen
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaomin Yang
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Huxia Wang
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Pihua Han
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Yongguo Fan
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Jing Zhao
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Jingyuan Zhang
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Jie Geng
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
- Correspondence to: Dr Jie Geng, Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
26
|
Lisiak N, Dzikowska P, Wisniewska U, Kaczmarek M, Bednarczyk-Cwynar B, Zaprutko L, Rubis B. Biological Activity of Oleanolic Acid Derivatives HIMOXOL and Br-HIMOLID in Breast Cancer Cells Is Mediated by ER and EGFR. Int J Mol Sci 2023; 24:5099. [PMID: 36982173 PMCID: PMC10048893 DOI: 10.3390/ijms24065099] [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: 02/01/2023] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/30/2023] Open
Abstract
Breast cancer is one of the most frequently observed malignancies worldwide and represents a heterogeneous group of cancers. For this reason, it is crucial to properly diagnose every single case so a specific and efficient therapy can be adjusted. One of the most critical diagnostic parameters evaluated in cancer tissue is the status of the estrogen receptor (ER) and epidermal growth factor receptor (EGFR). Interestingly, the expression of the indicated receptors may be used in a personalized therapy approach. Importantly, the promising role of phytochemicals in the modulation of pathways controlled by ER and EGFR was also demonstrated in several types of cancer. One such biologically active compound is oleanolic acid, but due to poor water solubility and cell membrane permeability that limits its use, alternative derivative compounds were developed. These are HIMOXOL and Br-HIMOLID, which were demonstrated to be capable of inducing apoptosis and autophagy or diminishing the migratory and invasive potential of breast cancer cells in vitro. In our study, we revealed that proliferation, cell cycle, apoptosis, autophagy, and also the migratory potential of HIMOXOL and Br-HIMOLID in breast cancer cells are mediated by ER (MCF7) and EGFR (MDA-MB-231) receptors. These observations make the studied compounds interesting in the context of anticancer strategies.
Collapse
Affiliation(s)
- Natalia Lisiak
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806 Poznan, Poland
| | - Patrycja Dzikowska
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806 Poznan, Poland
| | - Urszula Wisniewska
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806 Poznan, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Garbary 15 St., 61-866 Poznan, Poland
| | - Barbara Bednarczyk-Cwynar
- Department of Organic Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6 St., 60-780 Poznan, Poland
| | - Lucjusz Zaprutko
- Department of Organic Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6 St., 60-780 Poznan, Poland
| | - Blazej Rubis
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806 Poznan, Poland
| |
Collapse
|
27
|
UPLC-MS/MS Method for Simultaneous Estimation of Neratinib and Naringenin in Rat Plasma: Greenness Assessment and Application to Therapeutic Drug Monitoring. SEPARATIONS 2023. [DOI: 10.3390/separations10030167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
Tyrosine kinase inhibitors have often been reported to treat early-stage hormone-receptor-positive breast cancers. In particular, neratinib has shown positive responses in stage I and II cases in women with HER2-positive breast cancers with trastuzumab. In order to augment the biopharmaceutical attributes of the drug, the work designed endeavors to explore the therapeutic benefits of neratinib in combination with naringenin, a phytoconstituent with reported uses in breast cancer. A UPLC-MS/MS method was developed for the simultaneous estimation of neratinib and naringenin in rat plasma, while imatinib was selected as the internal standard (IS). Acetonitrile was used as the liquid extractant. The reversed-phase separation was achieved on a C18 column (100 mm × 2.1 mm, 1.7 µm) with the isocratic flow of mobile phase-containing acetonitrile (0.1% formic acid) and 0.002 M ammonium acetate (50:50, % v/v) at flow rate 0.5 mL·min−1. The mass spectra were recorded by multiple reaction monitoring of the precursor-to-product ion transitions for neratinib (m/z 557.138→111.927), naringenin (m/z 273.115→152.954), and the IS (m/z 494.24→394.11). The method was validated for selectivity, trueness, precision, matrix effect, recovery, and stability over a concentration range of 10–1280 ng·mL−1 for both targets and was acceptable. The method was also assessed for greenness profile by an integrative qualitative and quantitative approach; the results corroborated the eco-friendly nature of the method. Therefore, the developed method has implications for its applicability in clinical sample analysis from pharmacokinetic studies in human studies to support the therapeutic drug monitoring (TDM) of combination drugs.
Collapse
|
28
|
Saha M, Ghosh SS. Engineered Hybrid Nanosystem for Homologous Targeting of EMT Induced Triple Negative Breast Cancer Cells. ACS APPLIED BIO MATERIALS 2023; 6:681-693. [PMID: 36662500 DOI: 10.1021/acsabm.2c00925] [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: 01/21/2023]
Abstract
The increased mortality rate due to metastatic breast cancer with poor prognosis has raised concern over its effective therapy. Though various therapies and anticancer drugs have been approved, there is still a lack in the targeting of metastatic triple negative breast cancer (TNBC). We have developed a hybrid nanosystem that was synthesized by fusing exosomes from MCF-7 cells and nanovesicles from the MDA MB-231 cells that would be targeted. The developed nanosystem was characterized by various techniques like Western blotting, AFM, FETEM, DLS, CD, and fluorescence spectroscopy. The hybrid system was used for the delivery of an HDAC inhibitor, Trichostatin A (TSA), in combination with lapatinib (a tyrosine kinase inhibitor) for cotherapy of epithelial to mesenchymal transition (EMT) induced TNBC. This targeted cotherapy module had higher efficiency and effectivity in the reduction of metastatic ability and proliferation of EMT induced MDA MB-231 cells as compared to free inhibitor treatment or untargeted cotherapy. Reduction in the expression of the Wnt/β-catenin signaling pathway molecules like β-catenin (by 0.7 fold), Gsk3β (by 0.6 fold), and pGsk-3β (0.3 fold) was observed upon treatment. This subsequently resulted in the suppression of EMT markers, thereby resulting in reversing EMT to MET and suppressing metastatic breast cancer.
Collapse
Affiliation(s)
- Muktashree Saha
- Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati781039, Assam, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati781039, Assam, India.,Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| |
Collapse
|
29
|
Transcriptomic Analysis of Subtype-Specific Tyrosine Kinases as Triple Negative Breast Cancer Biomarkers. Cancers (Basel) 2023; 15:cancers15020403. [PMID: 36672350 PMCID: PMC9856281 DOI: 10.3390/cancers15020403] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/22/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Triple negative breast cancer (TNBC) shows impediment to the development of targeted therapies due to the absence of specific molecular targets. The high heterogeneity across TNBC subtypes, which can be classified to be at least four subtypes, including two basal-like (BL1, BL2), a mesenchymal (M), and a luminal androgen receptor (LAR) subtype, limits the response to cancer therapies. Despite many attempts to identify TNBC biomarkers, there are currently no effective targeted therapies against this malignancy. In this study, thus, we identified the potential tyrosine kinase (TK) genes that are uniquely expressed in each TNBC subtype, since TKs have been typically used as drug targets. Differentially expressed TK genes were analyzed from The Cancer Genome Atlas (TCGA) database and were confirmed with the other datasets of both TNBC patients and cell lines. The results revealed that each TNBC subtype expressed distinct TK genes that were specific to the TNBC subtype. The identified subtype-specific TK genes of BL1, BL2, M, and LAR are LYN, CSF1R, FGRF2, and SRMS, respectively. These findings could serve as a potential biomarker of specific TNBC subtypes, which could lead to an effective treatment for TNBC patients.
Collapse
|
30
|
Singh D, Piplani M, Kharkwal H, Murugesan S, Singh Y, Aggarwal A, Chander S. Anticancer Potential of Compounds Bearing Thiazolidin-4-one Scaffold: Comprehensive Review. PHARMACOPHORE 2023. [DOI: 10.51847/ohzuia1yg6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
|
31
|
Abrams JN. Design, Synthesis, and In Vitro Mitotic Evaluation of 3‐Amino‐Isoquinolinones as Anticancer Agents. ChemistrySelect 2022. [DOI: 10.1002/slct.202202861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jason N. Abrams
- Department of Chemistry Texas A&M University Kingsville 700 University Blvd Kingsville TX 78363
| |
Collapse
|
32
|
Wang L, Wu F, Xu J, Wang Y, Fei W, Jiang H, Geng P, Zhou Q, Wang S, Zheng Y, Deng H. Differential effects of ketoconazole, fluconazole, and itraconazole on the pharmacokinetics of pyrotinib in vitro and in vivo. Front Pharmacol 2022; 13:962731. [PMID: 36160438 PMCID: PMC9490176 DOI: 10.3389/fphar.2022.962731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022] Open
Abstract
It has been reported that drug-drug interactions (DDIs) can affect the pharmacokinetics and pharmacodynamics of various oral drugs. To better understand the effects of azole antifungal drugs (ketoconazole, fluconazole, and itraconazole) on pyrotinib’s pharmacokinetics, DDIs between pyrotinib and three azoles were studied with Sprague-Dawley (SD) rat liver microsomes in vitro. Additionally, in vivo pyrotinib metabolic experiment was also performed. Twenty-four male SD rats were randomly divided into four groups: the ketoconazole (40 mg/kg), fluconazole (40 mg/kg), itraconazole (40 mg/kg), and the control group. UPLC-MS/MS was used for the determination of Pyrotinib’s plasma concentration in rats. In vitro experiments showed that IC50 values of ketoconazole, fluconazole and itraconazole were 0.06, 11.55, and 0.27 μM, respectively, indicating that these drugs might reduce the clearance rate of pyrotinib at different degrees. In rat studies, coadministration of pyrotinib with ketoconazole or fluconazole could dramatically increase the Cmax and AUC(0-t) values and decrease the clearance rate of pyrotinib, especially for ketoconazole. However, coadministration with itraconazole had no impact on the pharmacokinetic characters of pyrotinib. These data indicated that ketoconazole and fluconazole could significantly decrease the metabolism of pyrotinib both in vitro and in vivo. More attentions should be paid when pyrotinib is combined with azole antifungal drugs in clinic although further investigation is still required in future.
Collapse
Affiliation(s)
- Li Wang
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fan Wu
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jia Xu
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
| | - Yu Wang
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
| | - Weidong Fei
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Jiang
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
| | - Peiwu Geng
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
| | - Quan Zhou
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
| | - Shuanghu Wang
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
| | - Yongquan Zheng
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Huadong Deng, ; Yongquan Zheng,
| | - Huadong Deng
- Department of Ultrasonography, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
- *Correspondence: Huadong Deng, ; Yongquan Zheng,
| |
Collapse
|
33
|
Calaf GM, Crispin LA, Muñoz JP, Aguayo F, Bleak TC. Muscarinic Receptors Associated with Cancer. Cancers (Basel) 2022; 14:cancers14092322. [PMID: 35565451 PMCID: PMC9100020 DOI: 10.3390/cancers14092322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Recently, cancer research has described the presence of the cholinergic machinery, specifically muscarinic receptors, in a wide variety of cancers due to their activation and signaling pathways associated with tumor progression and metastasis, providing a wide overview of their contribution to different cancer formation and development for new antitumor targets. This review focused on determining the molecular signatures associated with muscarinic receptors in breast and other cancers and the need for pharmacological, molecular, biochemical, technological, and clinical approaches to improve new therapeutic targets. Abstract Cancer has been considered the pathology of the century and factors such as the environment may play an important etiological role. The ability of muscarinic agonists to stimulate growth and muscarinic receptor antagonists to inhibit tumor growth has been demonstrated for breast, melanoma, lung, gastric, colon, pancreatic, ovarian, prostate, and brain cancer. This work aimed to study the correlation between epidermal growth factor receptors and cholinergic muscarinic receptors, the survival differences adjusted by the stage clinical factor, and the association between gene expression and immune infiltration level in breast, lung, stomach, colon, liver, prostate, and glioblastoma human cancers. Thus, targeting cholinergic muscarinic receptors appears to be an attractive therapeutic alternative due to the complex signaling pathways involved.
Collapse
Affiliation(s)
- Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.); (J.P.M.); (T.C.B.)
- Correspondence:
| | - Leodan A. Crispin
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.); (J.P.M.); (T.C.B.)
| | - Juan P. Muñoz
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.); (J.P.M.); (T.C.B.)
| | - Francisco Aguayo
- Laboratorio de Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile;
| | - Tammy C. Bleak
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (L.A.C.); (J.P.M.); (T.C.B.)
| |
Collapse
|
34
|
Yang R, Li Y, Wang H, Qin T, Yin X, Ma X. Therapeutic progress and challenges for triple negative breast cancer: targeted therapy and immunotherapy. MOLECULAR BIOMEDICINE 2022; 3:8. [PMID: 35243562 PMCID: PMC8894518 DOI: 10.1186/s43556-022-00071-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/09/2022] [Indexed: 02/08/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a subtype of breast cancer, with estrogen receptor, human epidermal growth factor receptor 2 and progesterone receptor negative. TNBC is characterized by high heterogeneity, high rates of metastasis, poor prognosis, and lack of therapeutic targets. Now the treatment of TNBC is still based on surgery and chemotherapy, which is effective only in initial stage but almost useless in advanced stage. And due to the lack of hormone target, hormonal therapies have little beneficial effects. In recent years, signaling pathways and receptor-specific targets have been reported to be effective in TNBC patients under specific clinical conditions. Now targeted therapies have been approved for many other cancers and even other subtypes of breast cancer, but treatment options for TNBC are still limited. Most of TNBC patients showed no response, which may be related to the heterogeneity of TNBC, therefore more effective treatments and predictive biomarkers are needed. In the present review, we summarize potential treatment opinions for TNBC based on the dysregulated receptors and signaling pathways, which play a significant role in multiple stages of TNBC development. We also focus on the application of immunotherapy in TNBC, and summarize the preclinical and clinical trials of therapy for patients with TNBC. We hope to accelerate the research and development of new drugs for TNBC by understanding the relevant mechanisms, and to improve survival.
Collapse
Affiliation(s)
- Ruoning Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China.,Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yueyi Li
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China
| | - Hang Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China
| | - Taolin Qin
- West China Hospital, West China Medical School Sichuan University, Chengdu, PR, China
| | - Xiaomeng Yin
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China.
| |
Collapse
|