1
|
Saeed BA, Ahmed WU, Alshekh AA, Wu L, Lu Y, Mirgany TO, Kadi AA, Alwabli RI, Wang N, Rahman AFMM. Potent Anti-Cancer Activity of Benzo[d]Thiazole-Isatin Conjugates: Induces S Phase Arrest and Cell Necrosis in A549 and HepG2 Cells. Chem Biodivers 2025:e202500575. [PMID: 40304277 DOI: 10.1002/cbdv.202500575] [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: 02/17/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/02/2025]
Abstract
The search for new molecules in cancer therapeutics is essential for overcoming treatment limitations, targeting evolving cancer characteristics, minimizing side effects, and advancing scientific knowledge. In this context, a series of benzo[d]thiazole-isatin conjugates (6a-6m) has been synthesized successfully in three steps with excellent yields (>90%). The preliminary antiproliferative results demonstrate that these synthetic derivatives are comparable to the well-known tyrosine kinase inhibitor (TKI) sunitinib. Moreover, the evaluation of their cytotoxicity has revealed outstanding activity against various cancer cell lines, including A549, HePG2, MDA-MB-231, and SGC7901. This significant efficacy highlights their potential as effective anti-cancer agents. Mechanism of action (MoA) studies revealed that compound 6g arrests the A549 and HepG2 cells at S phase, and the compound 6g on HepG2 cells results in a large number of cell necroses. Binding affinity of 6g with EGFR and cyclin-dependent kinase 2 (CDK2) kinase gives better results than reference drugs gefitinib and roscovitine, respectively.
Collapse
Affiliation(s)
- Bashayer A Saeed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Wahid U Ahmed
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Aljawharah A Alshekh
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Lingjie Wu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Yang Lu
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, Zhejiang, China
| | - Tebyan O Mirgany
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Adnan A Kadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Reem I Alwabli
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ning Wang
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, Zhejiang, China
| | - A F M Motiur Rahman
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Ashok Kumar P, Sandhu M, Desai D, Sravanthi Venkata M, Kumar VS, Benjamin S. A Meta-Analysis Studying the Difference in Response to Trastuzumab-Deruxtecan Based on HER2 Immunohistochemistry Staining in HER2 Low Metastatic Breast Cancer Patients. Am J Ther 2025:00045391-990000000-00298. [PMID: 40298948 DOI: 10.1097/mjt.0000000000001894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Affiliation(s)
- Prashanth Ashok Kumar
- Division of Hematology-Oncology, Upstate Cancer Center, Upstate University Hospital, Syracuse, NY
- George Washington University Hospital and Cancer Canter, Washington, DC
| | - Michael Sandhu
- Department of Medicine, Upstate University Hospital, Syracuse, NY
| | - Devashish Desai
- Department of Medicine, Upstate University Hospital, Syracuse, NY
| | | | | | - Sam Benjamin
- Division of Hematology-Oncology, Upstate Cancer Center, Upstate University Hospital, Syracuse, NY
| |
Collapse
|
3
|
Dabiri M, Tehrani M, Rafiei A, Valadan R. Production and functional analysis of a phage displayed scFv recombinant antibody targeting EGFR/HER2 dimerization domain. Protein Expr Purif 2025; 228:106649. [PMID: 39722421 DOI: 10.1016/j.pep.2024.106649] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Tumor cells exploit epidermal growth factor receptor (EGFR) family to develop resistance against therapeutic antibodies, such as Herceptin. Upon ligand binding, dimerization between EGFR and HER2 is one of the most important causes of treatment failure in breast cancer and other cancers expressing EGFR and HER2. The aim of this study was to develop and evaluate the function of a human recombinant single-chain variable fragment (scFv) antibody against the dimerization domain of EGFR to inhibit its interaction with other members of the epidermal growth factor receptor family, especially HER2. METHODS scFv against EGFR was expressed and purified. Cell-ELISA, MTT assay, inhibition of STAT3 phosphorylation, quantitative RT-PCR, and dimerization inhibition were performed on EGFR and HER2 expressing cell lines to characterize functional properties of the produced scFv. The conformational structure of the produced scFv and its binding ability to EGFR was computationally investigated. RESULTS In vitro binding analysis by cell-ELISA revealed the EGFR binding ability of the purified antibodies and confirmed by immunoblotting. ScFvs preferentially reduced the proliferation and survival of MCF7, MDA-MB-468, and SKOV3 cell lines with no effect on the VERO line. More considerably, MCF7 cells treated with the scFv antibody showed reduced STAT3 phosphorylation, decreased Bcl-2 expression, and increased Bax expression. Finally, the scFvs hindered EGFR and HER2 dimerization. CONCLUSION The produced scFv antibody showed to be functional in a simultaneous blockade of EGFR and HER2, suggesting its potential as a promising candidate for targeted therapy against various EGFR overexpressing tumors.
Collapse
Affiliation(s)
- Mina Dabiri
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Department of Biomedical and Biological Sciences, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ, 08028, United States.
| | - Mohsen Tehrani
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Alireza Rafiei
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Reza Valadan
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
4
|
Al-Ghamdi AR, Ahmed WU, Al-Wabli RI, Al-Mutairi MS, Rahman AFMM. Synthesis and Anticancer Evaluation of O-Alkylated ( E)-Chalcone Derivatives: A Focus on Estrogen Receptor Inhibition. Int J Mol Sci 2025; 26:833. [PMID: 39859546 PMCID: PMC11766267 DOI: 10.3390/ijms26020833] [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: 11/13/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer remains a leading cause of morbidity and mortality worldwide, highlighting the urgent need for novel therapeutic agents. This study investigated the synthesis and biological evaluation of O-alkyl (E)-chalcone derivatives (4a-4v) as potential anticancer agents. The compounds were synthesized via aldol condensation of substituted aldehydes and acetophenones, with structures confirmed by IR, NMR, and mass spectrometry. In vitro cytotoxicity assays revealed varying effectiveness, with compounds 4a, 4b, 4q, and 4v exhibiting potent activity against MDA-MB-231 and MCF-7, showing IC50 values between 2.08 and 13.58 µM, besides HCT-116 and HeLa cancer cell lines (IC50 values between 6.59 and 22.64 µM). Notably, compound 4b displayed remarkable selectivity, with an IC50 of 54.59 µM against the non-cancerous WI-38 cell line. Additionally, protein kinase inhibition assays indicated that compounds 4b and 4q effectively inhibited EGFR and VEGFR-2, with 4b outperforming the standard inhibitor erlotinib. Molecular docking studies of compound 4q showed strong binding affinities in the ATP-binding pockets of EGFR, HER2, VEGFR2, and CDK2. In silico analyses further highlighted the favorable pharmacokinetic properties of compound 4q, underscoring its potential as a selective tyrosine kinase inhibitor. These findings suggest the therapeutic promise of O-alkyl (E)-chalcone derivatives in cancer treatment.
Collapse
Affiliation(s)
- Alwah R. Al-Ghamdi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.R.A.-G.); (R.I.A.-W.)
| | - Wahid U. Ahmed
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China;
| | - Reem I. Al-Wabli
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.R.A.-G.); (R.I.A.-W.)
| | - Maha S. Al-Mutairi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.R.A.-G.); (R.I.A.-W.)
| | - A. F. M. Motiur Rahman
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.R.A.-G.); (R.I.A.-W.)
| |
Collapse
|
5
|
Mussa A, Ismail NH, Hamid M, Al-Hatamleh MAI, Bragoli A, Hajissa K, Mokhtar NF, Mohamud R, Uskoković V, Hassan R. Understanding the role of TNFR2 signaling in the tumor microenvironment of breast cancer. J Exp Clin Cancer Res 2024; 43:312. [PMID: 39609700 PMCID: PMC11603874 DOI: 10.1186/s13046-024-03218-1] [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: 08/21/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed malignancy among women. It is characterized by a high level of heterogeneity that emerges from the interaction of several cellular and soluble components in the tumor microenvironment (TME), such as cytokines, tumor cells and tumor-associated immune cells. Tumor necrosis factor (TNF) receptor 2 (TNFR2) appears to play a significant role in microenvironmental regulation, tumor progression, immune evasion, drug resistance, and metastasis of many types of cancer, including BC. However, the significance of TNFR2 in BC biology is not fully understood. This review provides an overview of TNFR2 biology, detailing its activation and its interactions with important signaling pathways in the TME (e.g., NF-κB, MAPK, and PI3K/Akt pathways). We discuss potential therapeutic strategies targeting TNFR2, with the aim of enhancing the antitumor immune response to BC. This review provides insights into role of TNFR2 as a major immune checkpoint for the future treatment of patients with BC.
Collapse
Affiliation(s)
- Ali Mussa
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
- Department of Biology, Faculty of Education, Omdurman Islamic University, P.O. Box 382, Omdurman, Sudan
| | - Nor Hayati Ismail
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
| | - Mahasin Hamid
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Hunan Province, Changsha, 410013, China
- Department of Zoology, Faculty of Sciences and Information Technology, University of Nyala, Nyala, 63311, Sudan
| | - Mohammad A I Al-Hatamleh
- Division of Hematology and Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Anthony Bragoli
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Khalid Hajissa
- Department of Zoology, Faculty of Science and Technology, Omdurman Islamic University, P.O. Box 382, Omdurman, Sudan
| | - Noor Fatmawati Mokhtar
- Institute for Research in Molecular Medicine (iNFORMM), Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia.
| | - Vuk Uskoković
- TardigradeNano LLC, Irvine, CA, 92604, USA
- Division of Natural Sciences, Fullerton College, Fullerton, CA, 92832, USA
| | - Rosline Hassan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia.
| |
Collapse
|
6
|
Saadan N, Ahmed WU, Kadi AA, Al-Mutairi MS, Al-Wabli RI, Rahman AFMM. Synthesis and Evaluation of Thiazolyl-indole-2-carboxamide Derivatives as Potent Multitarget Anticancer Agents. ACS OMEGA 2024; 9:41944-41967. [PMID: 39398118 PMCID: PMC11465279 DOI: 10.1021/acsomega.4c06889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024]
Abstract
Cancer is a complex disease driven by the dysregulation of multiple signaling pathways and cellular processes. The development of compounds capable of exerting multitarget actions against these key pathways involved in cancer progression is a promising therapeutic approach. Here, a series of novel (E/Z)-N-(4-(2-(2-(substituted)hydrazineyl)-2-oxoethyl)thiazol-2-yl)-1H-indole-2-carboxamide derivatives (6a-6z) were designed, synthesized, and evaluated for their biological activity. Compounds 6e, 6i, 6q, 6v, 7a, and 7b exhibited exceptional cytotoxicity against various cancer cell lines, particularly 6i (IC50 = 6.10 ± 0.4 μM against MCF-7 cell lines) and 6v (IC50 = 6.49 ± 0.3 μM against MCF-7 cell lines). These potent compounds inhibited key protein kinases like EGFR, HER2, VEGFR-2, and CDK2, induced cell cycle arrest at the G2/M phase, and promoted apoptosis. Docking studies revealed improved binding affinity of 6i and 6v with target proteins compared to reference drugs. These findings highlight the promising potential of 6i and 6v as multitarget cancer therapeutics deserving further development.
Collapse
Affiliation(s)
- Njood
M. Saadan
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Wahid U. Ahmed
- School
of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Adnan A. Kadi
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Maha S. Al-Mutairi
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Reem I. Al-Wabli
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - A. F. M. Motiur Rahman
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
7
|
Mahtani R, Harpalani N, Yan F, Phiel K, Kovalenko I. Expanding treatment options for patients with HER2+ metastatic breast cancer with margetuximab plus chemotherapy: a case report series. Front Oncol 2024; 14:1419246. [PMID: 39220641 PMCID: PMC11362812 DOI: 10.3389/fonc.2024.1419246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Background Human epidermal growth factor receptor 2 protein (HER2)-positive (+) metastatic breast cancer (MBC) is an aggressive disease and patients often undergo multiple lines of therapy following HER2 targeted therapies. The most recent National Comprehensive Cancer Network (NCCN) guidelines recommend margetuximab plus chemotherapy as fourth-line or later therapy for HER2+/hormone receptor (HR) + or negative (-) MBC. The aim of this case series is to provide information regarding margetuximab utilization in clinical practice as later-line therapy in women with HER2+ MBC. Case summaries Margetuximab plus chemotherapy was used as fourth- or later-line treatment in patients who had received multiple HER2-targeted agents, including trastuzumab, pertuzumab, ado-trastuzumab emtansine, trastuzumab deruxtecan, tucatinib, and neratinib. Patients responded to margetuximab plus chemotherapy with real-world progression-free survival (PFS) of 3, 4, and 7 months. Conclusion Clinical outcomes from three heavily pretreated patients with metastatic HER2+/HR+ MBC demonstrated that margetuximab plus chemotherapy resulted in real-world PFS comparable to that reported in the controlled pivotal clinical trial and support use of this targeted therapy option in appropriately identified patients.
Collapse
Affiliation(s)
- Reshma Mahtani
- Miami Cancer Institute – Medical Oncology, Baptist Health South Florida, Miami, FL, United States
- Miami Cancer Institute, Baptist Hospital of Miami, Miami, FL, United States
- Memorial Sloan Kettering Cancer Alliance, Miami Cancer Institute, Miami, FL, United States
| | - Natasha Harpalani
- Miami Cancer Institute, Baptist Hospital of Miami, Miami, FL, United States
| | - Fengting Yan
- Breast Medical Oncology, Swedish Cancer Institute, Seattle, WA, United States
- First Hill - True Family Women’s Cancer Center, Swedish Health Services, Seattle, WA, United States
| | | | - Iuliia Kovalenko
- Internal Medicine, University of Pittsburgh Medical Center (UPMC) Harrisburg, Harrisburg, PA, United States
| |
Collapse
|
8
|
Yang H, Wang X, Blanco-Gómez A, He L, García-Sancha N, Corchado-Cobos R, Pérez-Baena MJ, Jiménez-Navas A, Wang P, Inman JL, Snijders AM, Threadgill DW, Balmain A, Chang H, Perez-Losada J, Mao JH. A susceptibility gene signature for ERBB2-driven mammary tumour development and metastasis in collaborative cross mice. EBioMedicine 2024; 106:105260. [PMID: 39067134 PMCID: PMC11338061 DOI: 10.1016/j.ebiom.2024.105260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Deeper insights into ERBB2-driven cancers are essential to develop new treatment approaches for ERBB2+ breast cancers (BCs). We employed the Collaborative Cross (CC) mouse model to unearth genetic factors underpinning Erbb2-driven mammary tumour development and metastasis. METHODS 732 F1 hybrid female mice between FVB/N MMTV-Erbb2 and 30 CC strains were monitored for mammary tumour phenotypes. GWAS pinpointed SNPs that influence various tumour phenotypes. Multivariate analyses and models were used to construct the polygenic score and to develop a mouse tumour susceptibility gene signature (mTSGS), where the corresponding human ortholog was identified and designated as hTSGS. The importance and clinical value of hTSGS in human BC was evaluated using public datasets, encompassing TCGA, METABRIC, GSE96058, and I-SPY2 cohorts. The predictive power of mTSGS for response to chemotherapy was validated in vivo using genetically diverse MMTV-Erbb2 mice. FINDINGS Distinct variances in tumour onset, multiplicity, and metastatic patterns were observed in F1-hybrid female mice between FVB/N MMTV-Erbb2 and 30 CC strains. Besides lung metastasis, liver and kidney metastases emerged in specific CC strains. GWAS identified specific SNPs significantly associated with tumour onset, multiplicity, lung metastasis, and liver metastasis. Multivariate analyses flagged SNPs in 20 genes (Stx6, Ramp1, Traf3ip1, Nckap5, Pfkfb2, Trmt1l, Rprd1b, Rer1, Sepsecs, Rhobtb1, Tsen15, Abcc3, Arid5b, Tnr, Dock2, Tti1, Fam81a, Oxr1, Plxna2, and Tbc1d31) independently tied to various tumour characteristics, designated as a mTSGS. hTSGS scores (hTSGSS) based on their transcriptional level showed prognostic values, superseding clinical factors and PAM50 subtype across multiple human BC cohorts, and predicted pathological complete response independent of and superior to MammaPrint score in I-SPY2 study. The power of mTSGS score for predicting chemotherapy response was further validated in an in vivo mouse MMTV-Erbb2 model, showing that, like findings in human patients, mouse tumours with low mTSGS scores were most likely to respond to treatment. INTERPRETATION Our investigation has unveiled many new genes predisposing individuals to ERBB2-driven cancer. Translational findings indicate that hTSGS holds promise as a biomarker for refining treatment strategies for patients with BC. FUNDING The U.S. Department of Defense (DoD) Breast Cancer Research Program (BCRP) (BC190820), United States; MCIN/AEI/10.13039/501100011039 (PID2020-118527RB-I00, PDC2021-121735-I00), the "European Union Next Generation EU/PRTR," the Regional Government of Castile and León (CSI144P20), European Union.
Collapse
Affiliation(s)
- Hui Yang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Xinzhi Wang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Adrián Blanco-Gómez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain; Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | - Li He
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA; Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430079, China
| | - Natalia García-Sancha
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain; Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | - Roberto Corchado-Cobos
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain; Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | - Manuel Jesús Pérez-Baena
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain; Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | - Alejandro Jiménez-Navas
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain; Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | - Pin Wang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA; Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Jamie L Inman
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Antoine M Snijders
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA; Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - David W Threadgill
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA; Department of Molecular and Cellular Medicine and Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843, USA
| | - Allan Balmain
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Hang Chang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA; Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| | - Jesus Perez-Losada
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain; Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain.
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA; Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| |
Collapse
|
9
|
Hicken EJ, Brown K, Dwulet NC, Gaudino JJ, Hansen EP, Hartley DP, Kowalski JP, Laird ER, Lazzara NC, Li B, Mou TC, Mutryn MF, Oko L, Pajk S, Pipal RW, Rosen RZ, Shelp R, Singh A, Wang J, Wise CE, Wong C, Wong JY. Discovery of Potent and Selective Covalent Inhibitors of HER2 WT and HER2 YVMA. J Med Chem 2024; 67:9759-9771. [PMID: 38820338 DOI: 10.1021/acs.jmedchem.4c00978] [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: 06/02/2024]
Abstract
HER2 overexpression and amplification have been identified as oncogenic drivers, and the development of therapies to treat tumors harboring these markers has received considerable attention. Activation of HER2 signaling and subsequent cell growth can also be induced by HER2 mutations, including the common YVMA insertion in exon 20 within the kinase domain. Enhertu is currently the only approved treatment for HER2 mutant tumors in NSCLC. TKIs tested in this space have suffered from off-target activity, primarily due to EGFRWT inhibition or attenuated activity against HER2 mutants. The goal of this work was to identify a TKI that would provide robust inhibition of oncogenic HER2WT and HER2 mutants while sparing EGFRWT activity. Herein, we describe the development of a potent, covalent inhibitor of HER2WT and the YVMA insertion mutant while providing oral bioavailability and avoiding the inhibition of EGFRWT.
Collapse
Affiliation(s)
- Erik J Hicken
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Karin Brown
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Natalie C Dwulet
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - John J Gaudino
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Erik P Hansen
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Dylan P Hartley
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - John P Kowalski
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Ellen R Laird
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Nicholas C Lazzara
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Bin Li
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Tung-Chung Mou
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Marie F Mutryn
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Lauren Oko
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Spencer Pajk
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Robert W Pipal
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Rachel Z Rosen
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Russell Shelp
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Anurag Singh
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Jing Wang
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Courtney E Wise
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Christina Wong
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| | - Jim Y Wong
- Pfizer Boulder Research and Development, Boulder, Colorado 80301, United States
| |
Collapse
|
10
|
Jia HY, Zhang XY, Ye BC, Yin BC. An Orthogonal CRISPR/dCas12a System for RNA Imaging in Live Cells. Anal Chem 2024; 96:5913-5921. [PMID: 38563119 DOI: 10.1021/acs.analchem.3c05975] [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: 04/04/2024]
Abstract
CRISPR/Cas technology has made great progress in the field of live-cell imaging beyond genome editing. However, effective and easy-to-use CRISPR systems for labeling multiple RNAs of interest are still needed. Here, we engineered a CRISPR/dCas12a system that enables the specific recognition of the target RNA under the guidance of a PAM-presenting oligonucleotide (PAMmer) to mimic the PAM recognition mechanism for DNA substrates. We demonstrated the feasibility and specificity of this system for specifically visualizing endogenous mRNA. By leveraging dCas12a-mediated precursor CRISPR RNA (pre-crRNA) processing and the orthogonality of dCas12a from different bacteria, we further demonstrated the proposed system as a simple and versatile molecular toolkit for multiplexed imaging of different types of RNA transcripts in live cells with high specificity. This programmable dCas12a system not only broadens the RNA imaging toolbox but also facilitates diverse applications for RNA manipulation.
Collapse
Affiliation(s)
- Hai-Yan Jia
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
| | - Xin-Yue Zhang
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
| | - Bang-Ce Ye
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
- School of Chemistry and Chemical Engineering, Shihezi University, Xinjiang 832000, China
| | - Bin-Cheng Yin
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
- School of Chemistry and Chemical Engineering, Shihezi University, Xinjiang 832000, China
| |
Collapse
|
11
|
Narwadkar YS, Parghane RV, Sahu S, Lad S, Deep K, Wanage G, Suralkar T, Banerjee S, Gupta S, Basu S, Badwe RA. Clinical Internal Dosimetry and Biodistribution of 177 Lu-DOTA-Trastuzumab in HER2-Positive Metastatic and Locally Advanced Breast Carcinoma. Clin Nucl Med 2024; 49:e149-e155. [PMID: 38350067 DOI: 10.1097/rlu.0000000000005067] [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: 02/15/2024]
Abstract
OBJECTIVE The aim of this study was to assess the biodistribution and dosimetry of 177 Lu-DOTA-trastuzumab in patients with HER2-positive breast carcinoma using whole-body (WB) planar imaging at multiple time points. PATIENTS AND METHODS This study was a prospective evaluation of HER2-positive metastatic/locally advanced breast carcinoma patients who underwent gamma camera imaging for dosimetry and biodistribution studies by using 177 Lu-DOTA-trastuzumab. The standard diagnostic dosimetry protocol was followed, which included cold trastuzumab injection followed by in-house produced 177 Lu-DOTA-trastuzumab. Serial WB planar images (anterior and posterior) were obtained on gamma camera after the infusion of 177 Lu-DOTA-trastuzumab at multiple time points. Whole-body and organ regions of interest were drawn, and the numbers of disintegrations were obtained. The mean absorbed doses for the liver, spleen, kidneys, heart, red marrow, and tumor were obtained from OLINDA EXM v2.1.1 and ORIGIN software. RESULTS The study included a cohort of 21 female breast carcinoma patients. Tracer activity ( 177 Lu-DOTA-trastuzumab) was noted in the physiological organs such as the liver, spleen, kidneys, heart, as well as in the tumors. On visual analysis of 177 Lu-DOTA-trastuzumab biodistribution, the liver activity showed gradual clearance over time, and although spleen was comparatively faintly visualized than liver and similarly, kidneys were faintly visualized suggestive of the alternate route of tracer excretion. The maximum number of patients (n = 12) showed 2 components of clearance, namely, fast and slow. The average effective half-life of all the patients (including single and 2 components of clearance) was 106.25 ± 22.14 hours (84.11-128.39 hours). The mean absorbed dose for the liver, spleen, kidneys, heart, whole body, and red marrow was 1.0702 ± 0.731, 1.4114 ± 0.462, 1.4232 ± 0.364, 1.4719 ± 0.602, 0.2412 ± 0.0295, and 0.1485 ± 0.0213 mGy/MBq, respectively, by OLINDA EXM and 0.5741 ± 0.333, 0.8096 ± 0.224, 0.7943 ± 0.235, 1.8971 ± 0.713, and 0.09619 ± 0.0144 for liver, spleen, kidneys, heart and whole body respectively by ORIGIN. The absorbed radiation dose for tumor was 1.94E+2 by OLINDA EXM software and 1.78E+2 by ORIGIN software. In this study, during and after infusion of 177 Lu-DOTA-trastuzumab, no major adverse effects were noted in any patient except 1 patient who had grade 1 nausea and managed conservatively by antiemetic drug. CONCLUSIONS The results of our study demonstrated expected and favorable biodistribution and dosimetry with 177 Lu-DOTA-trastuzumab in HER2-positive breast carcinoma patients. We noticed the mean absorbed dose to the normal organs within the limits of maximum tolerable dose, and also tumor dose was higher than the normal liver dose. Therefore, we conclude that 177 Lu-DOTA-trastuzumab radioimmunotherapy is feasible and a safe treatment option for treating HER2-positive breast carcinoma patients.
Collapse
Affiliation(s)
| | | | - Sudeep Sahu
- From the Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe
| | - Sangita Lad
- From the Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe
| | | | | | - Tejal Suralkar
- Department of Radiation Oncology, The Cama and Albless Hospital
| | - Sharmila Banerjee
- Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre
| | - Sudeep Gupta
- Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre
| | | | | |
Collapse
|
12
|
Ebrahimnejad P, Mohammadi Z, Babaei A, Ahmadi M, Amirkhanloo S, Asare-Addo K, Nokhodchid A. Novel Strategies Using Sagacious Targeting for Site-Specific Drug Delivery in Breast Cancer Treatment: Clinical Potential and Applications. Crit Rev Ther Drug Carrier Syst 2024; 41:35-84. [PMID: 37824418 DOI: 10.1615/critrevtherdrugcarriersyst.v41.i1.20] [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: 10/14/2023]
Abstract
For more than a decade, researchers have been working to achieve new strategies and smart targeting drug delivery techniques and technologies to treat breast cancer (BC). Nanotechnology presents a hopeful strategy for targeted drug delivery into the building of new therapeutics using the properties of nanomaterials. Nanoparticles are of high regard in the field of diagnosis and the treatment of cancer. The use of these nanoparticles as an encouraging approach in the treatment of various cancers has drawn the interest of researchers in recent years. In order to achieve the maximum therapeutic effectiveness in the treatment of BC, combination therapy has also been adopted, leading to minimal side effects and thus an enhancement in the quality of life for patients. This review article compares, discusses and criticizes the approaches to treat BC using novel design strategies and smart targeting of site-specific drug delivery systems.
Collapse
Affiliation(s)
- Pedram Ebrahimnejad
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran; Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Mohammadi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amirhossein Babaei
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Melika Ahmadi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shervin Amirkhanloo
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kofi Asare-Addo
- Department of Pharmacy, University of Huddersfield, Huddersfield, UK
| | - Ali Nokhodchid
- Lupin Pharmaceutical Research Center, Coral Springs, Florida, USA; Pharmaceutics Research Lab, Arundel Building, School of Life Sciences, University of Sussex, Brighton, UK
| |
Collapse
|
13
|
Angelico G, Broggi G, Tinnirello G, Puzzo L, Vecchio GM, Salvatorelli L, Memeo L, Santoro A, Farina J, Mulé A, Magro G, Caltabiano R. Tumor Infiltrating Lymphocytes (TILS) and PD-L1 Expression in Breast Cancer: A Review of Current Evidence and Prognostic Implications from Pathologist's Perspective. Cancers (Basel) 2023; 15:4479. [PMID: 37760449 PMCID: PMC10526828 DOI: 10.3390/cancers15184479] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
With the rise of novel immunotherapies able to stimulate the antitumor immune response, increasing literature concerning the immunogenicity of breast cancer has been published in recent years. Numerous clinical studies have been conducted in order to identify novel biomarkers that could reflect the immunogenicity of BC and predict response to immunotherapy. In this regard, TILs have emerged as an important immunological biomarker related to the antitumor immune response in BC. TILs are more frequently observed in triple-negative breast cancer and HER2+ subtypes, where increased TIL levels have been linked to a better response to neoadjuvant chemotherapy and improved survival. PD-L1 is a type 1 transmembrane protein ligand expressed on T lymphocytes, B lymphocytes, and antigen-presenting cells and is considered a key inhibitory checkpoint involved in cancer immune regulation. PD-L1 immunohistochemical expression in breast cancer is observed in about 10-30% of cases and is extremely variable based on tumor stage and molecular subtypes. Briefly, TNBC shows the highest percentage of PD-L1 positivity, followed by HER2+ tumors. On the other hand, PD-L1 is rarely expressed (0-10% of cases) in hormone-receptor-positive BC. The prognostic role of PD-L1 expression in BC is still controversial since different immunohistochemistry (IHC) clones, cut-off points, and scoring systems have been utilized across published studies. In the present paper, an extensive review of the current knowledge of the immune landscape of BC is provided. TILS and PD-L1 expression across different BC subtypes are discussed, providing a guide for their pathological assessment and reporting.
Collapse
Affiliation(s)
- Giuseppe Angelico
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.B.); (G.T.); (L.P.); (G.M.V.); (L.S.); (J.F.); (G.M.); (R.C.)
| | - Giuseppe Broggi
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.B.); (G.T.); (L.P.); (G.M.V.); (L.S.); (J.F.); (G.M.); (R.C.)
| | - Giordana Tinnirello
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.B.); (G.T.); (L.P.); (G.M.V.); (L.S.); (J.F.); (G.M.); (R.C.)
| | - Lidia Puzzo
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.B.); (G.T.); (L.P.); (G.M.V.); (L.S.); (J.F.); (G.M.); (R.C.)
| | - Giada Maria Vecchio
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.B.); (G.T.); (L.P.); (G.M.V.); (L.S.); (J.F.); (G.M.); (R.C.)
| | - Lucia Salvatorelli
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.B.); (G.T.); (L.P.); (G.M.V.); (L.S.); (J.F.); (G.M.); (R.C.)
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, 95029 Catania, Italy;
| | - Angela Santoro
- Pathology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (A.S.); (A.M.)
| | - Jessica Farina
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.B.); (G.T.); (L.P.); (G.M.V.); (L.S.); (J.F.); (G.M.); (R.C.)
| | - Antonino Mulé
- Pathology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (A.S.); (A.M.)
| | - Gaetano Magro
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.B.); (G.T.); (L.P.); (G.M.V.); (L.S.); (J.F.); (G.M.); (R.C.)
| | - Rosario Caltabiano
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.B.); (G.T.); (L.P.); (G.M.V.); (L.S.); (J.F.); (G.M.); (R.C.)
| |
Collapse
|
14
|
Qian J, Xu Y, Ling X, Wang F. Overview on Current Trends and Emerging Therapies in the Chemotherapy of Patients with Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer. Galen Med J 2023; 12:e3021. [PMID: 39464538 PMCID: PMC11512434 DOI: 10.31661/gmj.v12i0.3021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 04/08/2023] [Accepted: 04/15/2023] [Indexed: 10/29/2024] Open
Abstract
Over the past few decades, significant progress has been made in the management of human epidermal growth factor receptor 2 (HER2)-positive breast cancer (BC) due to the development of targeted therapies. However, HER2-positive BC is an aggressive subtype, posing significant challenges, including treatment resistance and disease recurrence. Current standard treatment options for HER2-positive BC include combinations of chemotherapy drugs, targeted therapies such as trastuzumab and pertuzumab, and hormone therapies. However, some important limitations of these treatments, such as resistance and adverse effects, are reported. Also, we showed emerging therapeutic options, such as novel chemotherapy agents, antibody-drug conjugates, and immune checkpoint inhibitors, and discussed their mechanisms of action, potential benefits, and potential future directions in the field.
Collapse
Affiliation(s)
- Jian Qian
- Thyroid and Breast Surgery, Hangzhou Linping District Maternal and Child Health Care
Hospital, Hangzhou 311199, Zhejiang, China
| | - Yunxia Xu
- Thyroid and Breast Surgery, Hangzhou Linping District Maternal and Child Health Care
Hospital, Hangzhou 311199, Zhejiang, China
| | - Xiaokai Ling
- Thyroid and Breast Surgery, Hangzhou Linping District Maternal and Child Health Care
Hospital, Hangzhou 311199, Zhejiang, China
| | - Fenhua Wang
- Thyroid and Breast Surgery, Hangzhou Linping District Maternal and Child Health Care
Hospital, Hangzhou 311199, Zhejiang, China
| |
Collapse
|
15
|
Valadan R, Dabiri M, Tehrani M, Hashemi Tabar G, Rafiei A. A cell-based subtractive panning strategy for selection of conformation-specific single-chain variable-fragment (scFv) against dimerization domain of EGFR. J Immunol Methods 2023; 515:113456. [PMID: 36898519 DOI: 10.1016/j.jim.2023.113456] [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: 12/28/2022] [Revised: 03/06/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023]
Abstract
BACKGROUND AND OBJECTIVE Overexpression of EGFR, a member of the ErbB receptor family, has been observed in several cancers and causes resistance to therapeutic antibodies, such as Herceptin. In this study, we produced a recombinant single-chain variable fragment (scFv) antibody against the EGFR dimerization domain. METHODS The recombinant scFv was generated using a cell-based subtractive panning strategy. Subtractive panning was performed on a genetically engineered, VERO/EGFR, cells as well as a triple-negative breast cancer, MDA-MB-468, cells. Phage cell-ELISA was used to monitor the binding of the selected scFvs to the dimerization domain of EGFR. Inhibition of EGFR and HER2 dimerization by the produced scFvs were finally evaluated using the dimerization inhibition test and the expression of apoptosis-related genes were measured using the quantitative RT-PCR. RESULTS PCR fingerprinting results showed a uniform digestion pattern following the third round of panning that confirmed the success of subtractive panning. Moreover, cell-ELISA validated the reactivity of the produced scFvs to EGFR following stimulation with EGF. Dimerization inhibition test showed the capacity of the scFvs to inhibit EGFR and HER2 dimerization. Investigation of apoptosis-related genes showed that treatment with the scFv antibody caused increased Bax and decreased Bcl2 expression. CONCLUSIONS Directed HER2 targeting was shown to be effective enough to block the functional domain of the cell receptor and its intracellular signaling pathway. The subtractive panning strategy used in this study could control the process of directed selection of specific antibodies against the dimerization domain of EGFR. Selected antibodies might then be functionally tested for antitumor effects in both in vitro and in vivo studies.
Collapse
Affiliation(s)
- Reza Valadan
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mina Dabiri
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohsen Tehrani
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Gholamreza Hashemi Tabar
- Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Department of Pathobiology, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Alireza Rafiei
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran..
| |
Collapse
|
16
|
Shagufta, Ahmad I. Therapeutic significance of molecular hybrids for breast cancer research and treatment. RSC Med Chem 2023; 14:218-238. [PMID: 36846377 PMCID: PMC9945856 DOI: 10.1039/d2md00356b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Worldwide, breast cancer is still a leading cause of cancer death in women. Indeed, over the years, several anti-breast cancer drugs have been developed; however, the complex heterogeneous nature of breast cancer disease reduces the applicability of conventional targeted therapies with the upsurge in side effects and multi-drug resistance. Molecular hybrids generated by a combination of two or more active pharmacophores emerged as a promising approach in recent years for the design and synthesis of anti-breast cancer drugs. The hybrid anti-breast cancer molecules are well known for their several advantages compared to the parent moiety. These hybrid forms of anti-breast cancer molecules demonstrated remarkable effects in blocking different pathways contributing to the pathogenies of breast cancer and improved specificity. In addition, these hybrids are patient compliant with reduced side effects and multi-drug resistance. The literature revealed that molecular hybrids are applied to discover and develop novel hybrids for various complex diseases. This review article highlights the recent progress (∼2018-2022) in developing molecular hybrids, including linked, merged, and fused hybrids, as promising anti-breast cancer agents. Furthermore, their design principles, biological potential, and future perspective are discussed. The provided information will lead to the development of novel anti-breast cancer hybrids with excellent pharmacological profiles in the future.
Collapse
Affiliation(s)
- Shagufta
- Department of Mathematics and Natural Sciences, School of Arts and Sciences, American University of Ras Al Khaimah Ras Al Khaimah United Arab Emirates
| | - Irshad Ahmad
- Department of Mathematics and Natural Sciences, School of Arts and Sciences, American University of Ras Al Khaimah Ras Al Khaimah United Arab Emirates
| |
Collapse
|
17
|
Phosphohistidine signaling promotes FAK-RB1 interaction and growth factor-independent proliferation of esophageal squamous cell carcinoma. Oncogene 2023; 42:449-460. [PMID: 36513743 DOI: 10.1038/s41388-022-02568-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 11/08/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Current clinical therapies targeting receptor tyrosine kinases including focal adhesion kinase (FAK) have had limited or no effect on esophageal squamous cell carcinoma (ESCC). Unlike esophageal adenocarcinomas, ESCC acquire glucose in excess of their anabolic need. We recently reported that glucose-induced growth factor-independent proliferation requires the phosphorylation of FAKHis58. Here, we confirm His58 phosphorylation in FAK immunoprecipitates of glucose-stimulated, serum-starved ESCC cells using antibodies specific for 3-phosphohistidine and mass spectrometry. We also confirm a role for the histidine kinase, NME1, in glucose-induced FAKpoHis58 and ESCC cell proliferation, correlating with increased levels of NME1 in ESCC tumors versus normal esophageal tissues. Unbiased screening identified glucose-induced retinoblastoma transcriptional corepressor 1 (RB1) binding to FAK, mediated through a "LxCxE" RB1-binding motif in FAK's FERM domain. Importantly, in the absence of growth factors, glucose increased FAK scaffolding of RB1 in the cytoplasm, correlating with increased ESCC G1→S phase transition. Our data strongly suggest that this glucose-mediated mitogenic pathway is novel and represents a unique targetable opportunity in ESCC.
Collapse
|
18
|
The Role of Radiolabeled Monoclonal Antibodies in Cancer Imaging and ADC Treatment. Cancer J 2022; 28:446-453. [DOI: 10.1097/ppo.0000000000000625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
19
|
Pattanayak B, Lameirinhas A, Torres-Ruiz S, Burgués O, Rovira A, Martínez MT, Tapia M, Zazo S, Albanell J, Rojo F, Bermejo B, Eroles P. Role of SALL4 in HER2+ Breast Cancer Progression: Regulating PI3K/AKT Pathway. Int J Mol Sci 2022; 23:13292. [PMID: 36362083 PMCID: PMC9655635 DOI: 10.3390/ijms232113292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/16/2022] [Accepted: 10/25/2022] [Indexed: 08/30/2023] Open
Abstract
Treatment for the HER2+ breast cancer subtype is still unsatisfactory, despite breakthroughs in research. The discovery of various new molecular mechanisms of transcription factors may help to make treatment regimens more effective. The transcription factor SALL4 has been related to aggressiveness and resistance therapy in cancer. Its molecular mechanisms and involvement in various signaling pathways are unknown in the HER2+ breast cancer subtype. In this study, we have evaluated the implication of SALL4 in the HER2+ subtype through its expression in patients' samples and gain and loss of function in HER2+ cell lines. We found higher SALL4 expression in breast cancer tissues compared to healthy tissue. Interestingly, high SALL4 expression was associated with disease relapse and poor patient survival. In HER2+ cell lines, transient overexpression of SALL4 modulates PI3K/AKT signaling through regulating PTEN expression and BCL2, which increases cell survival and proliferation while reducing the efficacy of trastuzumab. SALL4 has also been observed to regulate the epithelial-mesenchymal transition and stemness features. SALL4 overexpression significantly reduced the epithelial markers E-cadherin, while it increased the mesenchymal markers β-catenin, vimentin and fibronectin. Furthermore, it has been also observed an increased expression of MYC, an essential transcription factor for regulating epithelial-mesenchymal transition and/or cancer stem cells. Our study demonstrates, for the first time, the importance of SALL4 in the HER2+ subtype and partial regulation of trastuzumab sensitivity. It provides a viable molecular mechanism-driven therapeutic strategy for an important subset of HER2-overexpressing patients whose malignancies are mediated by SALL4 expression.
Collapse
Affiliation(s)
| | - Ana Lameirinhas
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
| | | | - Octavio Burgués
- Department of Pathology, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
| | - Ana Rovira
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
| | - María Teresa Martínez
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Marta Tapia
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Sandra Zazo
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
- Department of Pathology, Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Joan Albanell
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
- Department of Medical Oncology, Hospital del Mar, 08003 Barcelona, Spain
| | - Federico Rojo
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
- Department of Pathology, Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Begoña Bermejo
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Pilar Eroles
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
- Department of Physiology, Universidad de Valencia, 46010 Valencia, Spain
- Department of Biotechnology, Universidad Politécnica de Valencia, 46022 Valencia, Spain
| |
Collapse
|
20
|
Koronkiewicz M, Kazimierczuk Z, Orzeszko A. Antitumor activity of the protein kinase inhibitor 1-(β-D-2'-deoxyribofuranosyl)-4,5,6,7-tetrabromo- 1H-benzimidazole in breast cancer cell lines. BMC Cancer 2022; 22:1069. [PMID: 36243702 PMCID: PMC9571492 DOI: 10.1186/s12885-022-10156-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Background The protein kinases CK2 and PIM-1 are involved in cell proliferation and survival, the cell cycle, and drug resistance, and they are found overexpressed in virtually all types of human cancer, including breast cancer. In this study, we investigated the antitumor activity of a deoxynucleoside derivative, the protein kinase inhibitor compound 1-(β-D-2′-deoxyribofuranosyl)-4,5,6,7-tetrabromo-1H-benzimidazole (K164, also termed TDB), inter alia CK2 and PIM-1, on breast cancer cell lines (MDA-MB-231, MCF-7, and SK-BR-3). Methods An evaluation of the cytotoxic and proapoptotic effects, mitochondrial membrane potential (ΔΨm), and cell cycle progression was performed using an MTT assay, flow cytometry, and microscopic analysis. The Western blotting method was used to analyze the level of proteins important for the survival of breast cancer cells and proteins phosphorylated by the CK2 and PIM-1 kinases. Results The examined compound demonstrated the inhibition of cell viability in all the tested cell lines and apoptotic activity, especially in the MCF-7 and SK-BR-3 cells. Changes in the mitochondrial membrane potential (ΔΨm), cell cycle progression, and the level of the proteins studied were also observed. Conclusions The investigated CK2 and PIM-1 kinase inhibitor K164 is a promising compound that can be considered a potential agent in targeted therapy in selected types of breast cancer; therefore, further research is necessary. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10156-8.
Collapse
Affiliation(s)
- Mirosława Koronkiewicz
- Department of Biomedical Research, National Medicines Institute, Chełmska St. 30/34, 00-725, Warsaw, Poland.
| | - Zygmunt Kazimierczuk
- Institute of Chemistry, Warsaw University of Life Sciences, Nowoursynowska St. 159C, 02-787, Warsaw, Poland
| | - Andrzej Orzeszko
- Institute of Chemistry, Warsaw University of Life Sciences, Nowoursynowska St. 159C, 02-787, Warsaw, Poland
| |
Collapse
|
21
|
Uchimiak K, Badowska-Kozakiewicz AM, Sobiborowicz-Sadowska A, Deptała A. Current State of Knowledge on the Immune Checkpoint Inhibitors in Triple-Negative Breast Cancer Treatment: Approaches, Efficacy, and Challenges. Clin Med Insights Oncol 2022; 16:11795549221099869. [PMID: 35721387 PMCID: PMC9201309 DOI: 10.1177/11795549221099869] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype with limited treatment options. Recently, there has been a growing interest in immunotherapy with immune checkpoint inhibitors (ICIs) in TNBC, leading to extensive preclinical and clinical research. This review summarizes the current state of knowledge on ICIs efficacy and their predictive markers in TNBC and highlights the areas where the data are still limited. Currently, the only approved ICI-based regimen for TNBC is pembrolizumab with chemotherapy. Its advantage over chemotherapy alone was confirmed for non-metastatic TNBC regardless of programmed death-ligand 1 (PD-L1) expression (KEYNOTE-522) and for metastatic, PD-L1-positive TNBC (KEYNOTE-355). Pembrolizumab's efficacy was also evaluated in monotherapy, or in combination with niraparib and radiation therapy, showing potential efficacy and acceptable safety profile in phase 2 clinical trials. Atezolizumab + nab-paclitaxel increased the overall survival (OS) over placebo + nab-paclitaxel in early TNBC, regardless of PD-L1 status (IMpassion031). In IMpassion130 (untreated, advanced TNBC), the OS improvement was not statistically significant in the intention-to-treat population but clinically meaningful in the PD-L1 positive cohort. The durvalumab-anthracycline combination showed an increased response durability over placebo anthracycline in early TNBC (GeparNuevo). Several phase 1 clinical trials also showed a potential efficacy of atezolizumab and avelumab monotherapy in metastatic TNBC. ICIs appear to be applicable in both neoadjuvant and adjuvant settings, and are both pretreated and previously untreated patients. Further research is necessary to determine the most beneficial drug combinations and optimize patient selection. It is essential to identify the predictive markers for ICIs and factors affecting their expression.
Collapse
Affiliation(s)
- Katarzyna Uchimiak
- Students’ Scientific Organization of
Cancer Cell Biology, Department of Cancer Prevention, Medical University of Warsaw,
Warsaw, Poland
| | | | - Aleksandra Sobiborowicz-Sadowska
- Students’ Scientific Organization of
Cancer Cell Biology, Department of Cancer Prevention, Medical University of Warsaw,
Warsaw, Poland
| | - Andrzej Deptała
- Department of Cancer Prevention,
Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
22
|
Purrahman D, Mahmoudian-Sani MR, Saki N, Wojdasiewicz P, Kurkowska-Jastrzębska I, Poniatowski ŁA. Involvement of progranulin (PGRN) in the pathogenesis and prognosis of breast cancer. Cytokine 2022; 151:155803. [DOI: 10.1016/j.cyto.2022.155803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/26/2021] [Accepted: 01/09/2022] [Indexed: 12/19/2022]
|
23
|
Al Rabadi LS, Cook MM, Kaempf AJ, Saraceni MM, Savin MA, Mitri ZI. Ado-trastuzumab for the treatment of metastatic HER2-positive breast cancer in patients previously treated with Pertuzumab. BMC Cancer 2021; 21:1150. [PMID: 34706686 PMCID: PMC8549287 DOI: 10.1186/s12885-021-08894-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 10/13/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Docetaxel in combination with two HER2-directed therapies, trastuzumab and pertuzumab, is the current standard frontline therapy for patients with metastatic HER2-positive breast cancer. Ado-trastuzumab (T-DM1), an antibody-drug conjugate of trastuzumab and a cytotoxic microtubule-inhibitory agent, emtansine, is approved in patients that have progressed with prior trastuzumab-based therapy. However, the benefit of T-DM1 in patients previously treated with pertuzumab therapy for metastatic breast cancer remains unclear. METHODS We identified thirty-three adults with metastatic HER2-positive breast cancer treated between March 2013 and July 2018 with T-DM1 either as subsequent therapy after progression on a pertuzumab-based regimen (i.e., "pertuzumab-pretreated") or without prior exposure to pertuzumab (i.e., "pertuzumab-naïve"). Collected data included patient demographics, treatment history, adverse events, and clinical outcomes. For both cohorts receiving T-DM1, the primary endpoint was PFS and secondary endpoints were overall survival (OS), overall response rate (ORR), clinical benefit rate (CBR), and T-DM1-related toxicity rate. RESULTS Pertuzumab-pretreated patients (n = 23, with 21 evaluable for T-DM1 efficacy) had a median PFS of 9.5 months (95% CI: 2.9-NA), 1-year OS rate of 67.4% (95% CI: 50.0-90.9%) with an unreached median, ORR of 14.3% (95% CI: 3.0-36.3%), and CBR of 52.4% (95% CI: 29.8-74.3%), with none of these measures being statistically different than those estimated for the pertuzumab-naïve group (n = 10). Treatment with T-DM1 after prior pertuzumab exposure (median T-DM1 duration 2.9 months) resulted in no grade ≥ 3 adverse events. CONCLUSIONS In our cohort, prior exposure to pertuzumab did not significantly impact T-DM1's clinical efficacy or safety profile as second- or later-line therapy in patients with metastatic HER2-positive breast cancer.
Collapse
Affiliation(s)
- Luai S Al Rabadi
- Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Road, OC14HO, Portland, OR, 97239, USA
| | - Madeline M Cook
- School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Andy J Kaempf
- Knight Cancer Institute, Biostatistics Shared Resource, Oregon Health & Science University, Portland, OR, USA
| | - Megan M Saraceni
- Department of Pharmacy, Oregon Health & Science University, Portland, OR, USA
| | - Michael A Savin
- Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Road, OC14HO, Portland, OR, 97239, USA
| | - Zahi I Mitri
- Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Road, OC14HO, Portland, OR, 97239, USA.
| |
Collapse
|
24
|
Monoclonal antibody based radiopharmaceuticals for imaging and therapy. Curr Probl Cancer 2021; 45:100796. [PMID: 34657748 DOI: 10.1016/j.currproblcancer.2021.100796] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/03/2021] [Accepted: 09/02/2021] [Indexed: 11/23/2022]
Abstract
The concept of personalized medicine has been steadily growing for the past decades. Monoclonal antibodies (mAbs) are undoubtedly playing an important role in the transition away from conventional medical practice to a more tailored approach to deliver the best therapy with the highest safety margin to a specific patient. In certain instances, mAbs and antibody drug conjugates (ADCs) may represent the preferred therapeutic option for several types of cancers due to their high specificity and affinity to the antigen. Monoclonal antibodies can be labeled with specific radionuclides well-suited for PET (Positron Emission Tomography) or gamma camera scintigraphy. The use of radiolabeled mAbs allows the interrogation of specific biomarkers and assessment of tumor heterogeneity in vivo by a single diagnostic imaging scan that includes the whole-body in the field-of-view. Moreover, the same mAb can then be radiolabeled with an analogous radionuclide for the delivery of beta-minus radiation or alpha-particles as part of a radioimmunotherapy (RIT) approach. However, the path to develop, validate, and implement mAb-based radiopharmaceuticals from bench-to-bedside is complex due to the extensive pre-clinical experiments and toxicological studies required, and the necessity of labor-intensive clinical trials that often require multi-time-point imaging and blood draws for internal radiation dosimetry and pharmacokinetics. As more mAb-based radiopharmaceuticals have been developed and evaluated, the opportunities and limitations offered by mAbs have become better defined. Our aim with this manuscript is therefore to provide an overview of the recent advances in the development of mAb-based radiopharmaceuticals and their clinical applications in Oncology.
Collapse
|
25
|
Pellegrini M. Accurate prediction of breast cancer survival through coherent voting networks with gene expression profiling. Sci Rep 2021; 11:14645. [PMID: 34282236 PMCID: PMC8289832 DOI: 10.1038/s41598-021-94243-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
For a patient affected by breast cancer, after tumor removal, it is necessary to decide which adjuvant therapy is able to prevent tumor relapse and formation of metastases. A prediction of the outcome of adjuvant therapy tailored for the patient is hard, due to the heterogeneous nature of the disease. We devised a methodology for predicting 5-years survival based on the new machine learning paradigm of coherent voting networks, with improved accuracy over state-of-the-art prediction methods. The 'coherent voting communities' metaphor provides a certificate justifying the survival prediction for an individual patient, thus facilitating its acceptability in practice, in the vein of explainable Artificial Intelligence. The method we propose is quite flexible and applicable to other types of cancer.
Collapse
Affiliation(s)
- Marco Pellegrini
- Institute of Informatics and Telematics (IIT), CNR, 56124, Pisa, Italy.
| |
Collapse
|
26
|
Peeters M, Planchard D, Pegram M, Gonçalves J, Bocquet F, Jang H. Biosimilars in an era of rising oncology treatment options. Future Oncol 2021; 17:3881-3892. [PMID: 34189937 DOI: 10.2217/fon-2021-0546] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
New diagnostic technologies, including molecular profiling, have enabled advances in treatments of various cancers; this has significantly improved clinical outcomes, including overall survival. However, the high cost of biologic drugs may prevent patients from having access to optimal treatment. Introduction of lower priced biosimilar agents into the therapeutic armamentarium brings the potential to ease the burden on healthcare expenditure and facilitate better access to effective cancer treatments. Oncology biosimilars have shown comparable efficacy and safety based on clinical evidence and physicochemical quality data as well as in real-world settings. This paper aims to review changes in the management of oncology treatment and their implication with respect to biosimilars.
Collapse
Affiliation(s)
- Marc Peeters
- Department of Oncology, Antwerp University Hospital, Belgium
| | - David Planchard
- Department of Medical Oncology, Thoracic Unit, Institut Gustave Roussy, Villejuif, France
| | - Mark Pegram
- Stanford Comprehensive Cancer Institute, Stanford University School of Medicine Stanford, CA, USA
| | - João Gonçalves
- iMed-Research Institute for Medicines, Faculty of Pharmacy at University of Lisbon, Lisbon, Portugal
| | - François Bocquet
- Oncology Data Factory & Analytics Department, Institut de Cancérologie de l'Ouest, Nantes-Angers, France
| | - Harah Jang
- Samsung Bioepis Co., Ltd, Incheon, Republic of Korea
| |
Collapse
|
27
|
Schettini F, Conte B, Buono G, De Placido P, Parola S, Griguolo G, Fabi A, Bighin C, Riccardi F, Cianniello D, De Laurentiis M, Puglisi F, Pelizzari G, Bonotto M, Russo S, Frassoldati A, Pazzola A, Montemurro F, Lambertini M, Guarneri V, Cognetti F, Locci M, Generali D, Conte P, De Placido S, Giuliano M, Arpino G, Del Mastro L. T-DM1 versus pertuzumab, trastuzumab and a taxane as first-line therapy of early-relapsed HER2-positive metastatic breast cancer: an Italian multicenter observational study. ESMO Open 2021; 6:100099. [PMID: 33819752 PMCID: PMC8047485 DOI: 10.1016/j.esmoop.2021.100099] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/06/2021] [Accepted: 02/27/2021] [Indexed: 02/06/2023] Open
Abstract
Background The current standard first-line treatment of human epidermal growth factor receptor 2 (HER2)-positive (+) metastatic breast cancer is the combination of pertuzumab, trastuzumab and a taxane (P + T + taxane), while standard second-line is ado-trastuzumab-emtansine (T-DM1). The registration trial of pertuzumab, however, did not include early-relapsing patients, defined as patients experiencing tumor relapse ≤12 months from the end of (neo)adjuvant anti-HER2 therapy. Conversely, the pivotal trial of T-DM1 included some patients relapsing ≤6 months after the end of (neo)adjuvant trastuzumab. Thus, a proportion of early-relapsing patients are currently eligible to receive T-DM1 as first-line treatment. Nevertheless, no direct comparison exists between the two regimens in this clinical setting. Patients and methods We retrospectively compared T-DM1 versus P + T + taxane as first-line treatment in two cohorts of early-relapsing patients in an Italian ‘real-world’ setting, involving 14 public health care institutions. The primary endpoint was progression-free survival. Secondary endpoints included patients' characterization, overall survival and post-progression survival. Univariate and multivariate analyses were carried out. All tests were two-sided and a P ≤ 0.05 was considered statistically significant. Results Among 1252 screened patients, 75 met the inclusion criteria. Forty-four (58.7%) received P + T + taxane and 31 (41.3%) received T-DM1. The two cohorts showed similar characteristics of aggressiveness and no significant differences in treatment history. T-DM1, compared with P + T + taxane was associated with worse progression-free survival (adjusted hazard ratio: 2.26, 95% confidence interval: 1.13-4.52, P = 0.021) and overall survival (adjusted hazard ratio: 3.95, 95% confidence interval: 1.38-11.32, P = 0.010), irrespective of previous (neo)adjuvant treatment, age, hormone receptors status, time-to-relapse (≤6 months or within 6-12 months) and presence of visceral/brain metastases. No differences were observed in post-progression survival (P = 0.095). Conclusions Our study suggests superiority for P + T + taxane over T-DM1 as up-front treatment of early-relapsing HER2+ metastatic breast cancer, which merits further assessment in larger and prospective trials. This is the first study comparing pertuzumab + trastuzumab + taxane (P + T + taxane) with T-DM1 in early-relapsing HER2+ MBC. The majority of early-relapsing HER2+ MBC have high-grade, node-positive, large primary tumors. First-line T-DM1 compared with P + T + taxane is associated with worse progression-free survival. First-line T-DM1 compared with P + T + taxane is associated with worse overall survival. Post-progression survival does not differ between the two treatments cohorts.
Collapse
Affiliation(s)
- F Schettini
- Department of Clinical Medicine and Surgery, University of Naples 'Federico II', Naples, Italy; Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; SOLTI Breast Cancer Research Group, Barcelona, Spain.
| | - B Conte
- SOLTI Breast Cancer Research Group, Barcelona, Spain; Medical Oncology Unit 2, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - G Buono
- Oncology Unit, San Rocco Hospital, Sessa Aurunca, Italy
| | - P De Placido
- Department of Clinical Medicine and Surgery, University of Naples 'Federico II', Naples, Italy
| | - S Parola
- Department of Clinical Medicine and Surgery, University of Naples 'Federico II', Naples, Italy
| | - G Griguolo
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - A Fabi
- Division of Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - C Bighin
- Medical Oncology Unit 2, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - F Riccardi
- Medical Oncology, 'A. Cardarelli' Hospital, Naples, Italy
| | - D Cianniello
- National Cancer Institute Fondazione 'G. Pascale', Naples, Italy
| | - M De Laurentiis
- National Cancer Institute Fondazione 'G. Pascale', Naples, Italy
| | - F Puglisi
- Department of Medicine (DAME), University of Udine, Udine, Italy; Department of Clinical Oncology, CRO Aviano National Cancer Institute, Aviano, Italy
| | - G Pelizzari
- Department of Clinical Oncology, CRO Aviano National Cancer Institute, Aviano, Italy
| | - M Bonotto
- Department of Oncology, ASUFC University Hospital, Udine, Italy
| | - S Russo
- Department of Oncology, ASUFC University Hospital, Udine, Italy
| | - A Frassoldati
- Oncology Unit, University Hospital St. Anna, Ferrara, Italy
| | - A Pazzola
- Division of Medical Oncology, AOU Sassari, Sassari, Italy
| | - F Montemurro
- Depertment of Medical Oncology, Candiolo Cancer Institute, Candiolo, Italy
| | - M Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy; Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - V Guarneri
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - F Cognetti
- Department of Clinic and Molecular Medicine, 'La Sapienza' University of Rome, Rome, Italy
| | - M Locci
- Department of Neuroscience, Reproductive Medicine, Odontostomatology, University of Naples 'Federico II', Naples, Italy
| | - D Generali
- Breast Cancer Unit, Azienda Socio Sanitaria Territoriale di Cremona, Cremona, Italy; Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - P Conte
- Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - S De Placido
- Department of Clinical Medicine and Surgery, University of Naples 'Federico II', Naples, Italy
| | - M Giuliano
- Department of Clinical Medicine and Surgery, University of Naples 'Federico II', Naples, Italy
| | - G Arpino
- Department of Clinical Medicine and Surgery, University of Naples 'Federico II', Naples, Italy
| | - L Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy; U.O.S.D. Breast Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
28
|
El-Chami D, Al Haddad M, Abi-Habib R, El-Sibai M. Recombinant anthrax lethal toxin inhibits cell motility and invasion in breast cancer cells through the dysregulation of Rho GTPases. Oncol Lett 2020; 21:163. [PMID: 33552281 DOI: 10.3892/ol.2020.12424] [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: 08/17/2020] [Accepted: 11/27/2020] [Indexed: 11/05/2022] Open
Abstract
Breast cancer is the leading cause of cancer-associated death among women worldwide. Targeting breast cancer cell metastasis is an important therapeutic approach. The MAPK pathway is a key cell signaling pathway that plays a pivotal role in cellular invasion and migration. Numerous studies have identified the MAPK pathway as a way to target cell survival and motility. The present study treated MBA-MD-231 breast cancer cells with anthrax lethal toxin (LeTx), a potent MAPK inhibitor that selectively cleaves and inactivates all MEKs, as a potential therapeutic method to inhibit breast cancer cell migration. LeTx has been demonstrated to affect breast cancer cell migration. Cells treated with LeTx showed a significant decrease in motility, as observed using wound healing and random 2D motility assays. Additionally, cells treated with LeTx showed an increase in adhesion, which would explain the decrease in migration. Pull-down assays examining the activation status of the members of the Rho family of GTPases revealed an increase in RhoA activation accompanied by a decrease in Cdc42 activation following LeTx treatment. Finally, LeTx mediated a decrease in invasion using a Boyden chamber assay, which could be a result of the decrease in Cdc42 activation. The present study reported the effect of LeTx treatment on the migration, adhesion and invasion of breast cancer cells, demonstrating that this effect was associated with the dysregulation of the Rho GTPases, RhoA and Cdc42.
Collapse
Affiliation(s)
- Dana El-Chami
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Maria Al Haddad
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Ralph Abi-Habib
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut 1102 2801, Lebanon
| |
Collapse
|
29
|
Thanopoulou E, Khader L, Caira M, Wardley A, Ettl J, Miglietta F, Neven P, Guarneri V. Therapeutic Strategies for the Management of Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Positive (HR+/HER2+) Breast Cancer: A Review of the Current Literature. Cancers (Basel) 2020; 12:E3317. [PMID: 33182657 PMCID: PMC7696181 DOI: 10.3390/cancers12113317] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/22/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
Enormous advances have been made in the understanding and treatment of human epidermal growth factor receptor 2-positive breast cancer (HER2+ BC) in the last 30 years that have resulted in survival gains for affected patients. A growing body of evidence suggests that hormone receptor-positive (HR+)/HER2+ BC and HR-negative (HR-)/HER2+ BC are biologically different, with complex molecular bidirectional crosstalk between the estrogen receptor and HER2 pathway potentially affecting sensitivity to both HER2-targeted and endocrine therapy in patients with HR+/HER2+ BC. Subgroup analyses from trials enrolling patients with HER2+ BC and the results of clinical trials specifically designed to evaluate therapy in patients with HR+/HER2+ BC are helping to guide treatment decisions. In this context, encouraging results with strategies aimed at delaying or reversing drug resistance, including extended adjuvant therapy and the addition of drugs targeting alternative pathways, such as cyclin-dependent kinase (CDK) 4 and 6 inhibitors, have recently emerged. We have reached the point where tailoring the treatment according to risk and biology has become the paradigm in early BC. However, further clinical trials are needed that integrate translational research principles and identify and consider specific patient subgroups and biomarkers.
Collapse
Affiliation(s)
- Eirini Thanopoulou
- Eli Lilly and Company Limited, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK;
| | - Leila Khader
- Eli Lilly Italia S.p.A., 50019 Comune di Sesto Fiorentino, Florence, Italy; (L.K.); (M.C.)
| | - Morena Caira
- Eli Lilly Italia S.p.A., 50019 Comune di Sesto Fiorentino, Florence, Italy; (L.K.); (M.C.)
| | - Andrew Wardley
- The NIHR Manchester Clinical Research Facility at The Christie NHS Foundation Trust, School of Medical Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester M204BX, UK;
| | - Johannes Ettl
- Department of Obstetrics and Gynecology, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany;
| | - Federica Miglietta
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, 35128 Padua, Italy;
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy
| | - Patrick Neven
- Multidisciplinary Breast Center and Department of Gynecology and Obstetrics, UZ Leuven, 3000 Leuven, Belgium;
- Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Valentina Guarneri
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, 35128 Padua, Italy;
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy
| |
Collapse
|
30
|
Elumalai S, Managó S, De Luca AC. Raman Microscopy: Progress in Research on Cancer Cell Sensing. SENSORS (BASEL, SWITZERLAND) 2020; 20:E5525. [PMID: 32992464 PMCID: PMC7582629 DOI: 10.3390/s20195525] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023]
Abstract
In the last decade, Raman Spectroscopy (RS) was demonstrated to be a label-free, non-invasive and non-destructive optical spectroscopy allowing the improvement in diagnostic accuracy in cancer and analytical assessment for cell sensing. This review discusses how Raman spectra can lead to a deeper molecular understanding of the biochemical changes in cancer cells in comparison to non-cancer cells, analyzing two key examples, leukemia and breast cancer. The reported Raman results provide information on cancer progression and allow the identification, classification, and follow-up after chemotherapy treatments of the cancer cells from the liquid biopsy. The key obstacles for RS applications in cancer cell diagnosis, including quality, objectivity, number of cells and velocity of the analysis, are considered. The use of multivariant analysis, such as principal component analysis (PCA) and linear discriminate analysis (LDA), for an automatic and objective assessment without any specialized knowledge of spectroscopy is presented. Raman imaging for cancer cell mapping is shown and its advantages for routine clinical pathology practice and live cell imaging, compared to single-point spectral analysis, are debated. Additionally, the combination of RS with microfluidic devices and high-throughput screening for improving the velocity and the number of cells analyzed are also discussed. Finally, the combination of the Raman microscopy (RM) with other imaging modalities, for complete visualization and characterization of the cells, is described.
Collapse
Affiliation(s)
| | | | - Anna Chiara De Luca
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Via P. Castellino 111, 80131 Naples, Italy; (S.E.); (S.M.)
| |
Collapse
|
31
|
Lin Y, Zhang W, Cao H, Li G, Du W. Classifying Breast Cancer Subtypes Using Deep Neural Networks Based on Multi-Omics Data. Genes (Basel) 2020; 11:E888. [PMID: 32759821 PMCID: PMC7464481 DOI: 10.3390/genes11080888] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
With the high prevalence of breast cancer, it is urgent to find out the intrinsic difference between various subtypes, so as to infer the underlying mechanisms. Given the available multi-omics data, their proper integration can improve the accuracy of breast cancer subtype recognition. In this study, DeepMO, a model using deep neural networks based on multi-omics data, was employed for classifying breast cancer subtypes. Three types of omics data including mRNA data, DNA methylation data, and copy number variation (CNV) data were collected from The Cancer Genome Atlas (TCGA). After data preprocessing and feature selection, each type of omics data was input into the deep neural network, which consists of an encoding subnetwork and a classification subnetwork. The results of DeepMO based on multi-omics on binary classification are better than other methods in terms of accuracy and area under the curve (AUC). Moreover, compared with other methods using single omics data and multi-omics data, DeepMO also had a higher prediction accuracy on multi-classification. We also validated the effect of feature selection on DeepMO. Finally, we analyzed the enrichment gene ontology (GO) terms and biological pathways of these significant genes, which were discovered during the feature selection process. We believe that the proposed model is useful for multi-omics data analysis.
Collapse
Affiliation(s)
- Yuqi Lin
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (Y.L.); (W.Z.)
| | - Wen Zhang
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (Y.L.); (W.Z.)
| | - Huanshen Cao
- Center for Fundamental and Applied Microbiomics, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA;
| | - Gaoyang Li
- School of Life Science and Technology, Tongji University, Shanghai 200092, China;
| | - Wei Du
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (Y.L.); (W.Z.)
| |
Collapse
|
32
|
Qi WX, Zhao S, Chen J. Risk factors for developing cardiac toxicities in cancer patients treated with panitumumab combination therapy. Future Oncol 2020; 16:1359-1370. [PMID: 32422068 DOI: 10.2217/fon-2020-0050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: To evaluate the incidence and risk of cardiac toxicities associated with panitumumab in advanced cancer of Caucasian patients. Materials & methods: The incidence of cardiac toxicity was assessed by simple incidence rates and rates per 100 person-years. Univariate and multivariate Cox regression was conducted. Results: Panitumumab-containing therapy significantly increased the risk of developing cardiac arrhythmias (p = 0.036), but not for any cardiac event (p = 0.24) or ischemic event (p = 0.087). The absolute rate of developing cardiac arrhythmia was 10.0 events versus 7.5 events per 100 person-years. Pre-existing hypertension (p = 0.033), history of cardiac disease (p = 0.055) or panitumumab usage (p = 0.046) were risk factors for cardiac arrhythmias. Conclusion: The addition of panitumumab to chemotherapy increases the risk of developing cardiac arrhythmia, but not for any cardiac toxicity or ischemic events.
Collapse
Affiliation(s)
- Wei-Xiang Qi
- Department of Radiation Oncology, Rui Jin Hospital Affiliated Medicine School of Shanghai Jiao Tong University, Shanghai, PR China
| | - Shengguang Zhao
- Department of Radiation Oncology, Rui Jin Hospital Affiliated Medicine School of Shanghai Jiao Tong University, Shanghai, PR China
| | - Jiayi Chen
- Department of Radiation Oncology, Rui Jin Hospital Affiliated Medicine School of Shanghai Jiao Tong University, Shanghai, PR China
| |
Collapse
|
33
|
Hasham K, Ahmed N, Zeshan B. Circulating microRNAs in oncogenic viral infections: potential diagnostic biomarkers. SN APPLIED SCIENCES 2020. [DOI: 10.1007/s42452-020-2251-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
34
|
Zalloum H, AbuThiab T, Hameduh T, AlBayyari S, Zalloum W, Abu-Irmaileh B, Mubarak MS, Zihlif M. Comparative anti-proliferative effects of potential HER2 inhibitors on a panel of breast cancer cell lines. Breast Cancer 2019; 27:213-224. [PMID: 31559601 DOI: 10.1007/s12282-019-01011-z] [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: 03/27/2019] [Accepted: 09/14/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Breast cancer is one of the most lethal types of cancer in women worldwide. The human epidermal growth factor receptor 2 (HER2) is considered as a validated target in breast cancer therapy. Previously, we have used quantitative structure activity relationship QSAR equations and their associated pharmacophore models to screen for new promising HER2 structurally diverse inhibitory leads which were tested against HER2-overexpressing SKOV3 ovarian cancer cell line. OBJECTIVE In this study, we sought to explore the effect of most active ligands against different normal and breast cancer cell lines that represent different breast cancer subtypes with distinguished expression levels in HER2 and HER1. METHODS We have tested the promising compounds against SKBR3, MDA-MB-231, MCF7, human fibroblast, and MCF10 cell lines. To understand the inhibitory effects of the active ligands against HER2 over expressed breast cancer cell lines, all inhibitors and the control compound, lapatinib, were docked into the active site of HER2 enzyme performed using Ligand Fit docking engine and PMF scoring function. RESULTS Five ligands exhibited promising results with relatively low IC50 values on cells that amplify HER2 and high IC50 on those that do not express such a receptor. The most potent compound (compound 13) showed an IC50 of 0.046 µM. To test their toxicity against normal cells, the active compounds were tested against both normal fibroblast and normal breast cancer cell MCF-10 and relatively high IC50 values were scored. The IC50 values on HER2 over-expressed breast cancer and normal fibroblast cells provided a promising safety index. Docking results showed the highest similarity in the binding site between the most active ligand and the lapatinib. CONCLUSION Our pharmacophore model resulted in a high potent ligand that shows high potency against HER2 positive breast cancer and relatively low toxicity towards the normal human cells.
Collapse
Affiliation(s)
- Hiba Zalloum
- Hamdi Mango Research Center for Scientific Research, The University of Jordan, Amman, 11942, Jordan.
| | - Tuka AbuThiab
- Hamdi Mango Research Center for Scientific Research, The University of Jordan, Amman, 11942, Jordan
| | - Tareq Hameduh
- Hamdi Mango Research Center for Scientific Research, The University of Jordan, Amman, 11942, Jordan
| | - Sara AlBayyari
- Hamdi Mango Research Center for Scientific Research, The University of Jordan, Amman, 11942, Jordan
| | - Waleed Zalloum
- Department of Pharmacy, Faculty of Health Science, American University of Madaba, P.O. Box 2882, Amman, 11821, Jordan
| | - Basha'er Abu-Irmaileh
- Hamdi Mango Research Center for Scientific Research, The University of Jordan, Amman, 11942, Jordan
| | - Mohammad S Mubarak
- Department of Chemistry, Faculty of Science, The University of Jordan, Amman, 11942, Jordan
| | - Malek Zihlif
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
35
|
Aldossary MY, Alquraish F, Alazhri J. A Case of Locally Advanced Breast Cancer in a 59-Year-Old Man Requiring a Modified Approach to Management. AMERICAN JOURNAL OF CASE REPORTS 2019; 20:531-536. [PMID: 30992422 PMCID: PMC6485044 DOI: 10.12659/ajcr.915377] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Patient: Male, 59 Final Diagnosis: Invasive ductal carcinoma Symptoms: Foul-smelling discharge • painful breast mass Medication: — Clinical Procedure: Modified radical mastectomy Specialty: Surgery
Collapse
Affiliation(s)
- Mohammed Yousef Aldossary
- Department of General Surgery, Surgical Oncology Unit, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Fatimah Alquraish
- Department of General Surgery, King Abdulaziz Air Base Hospital, Dhahran, Saudi Arabia
| | - Jamila Alazhri
- Department of General Surgery, Surgical Oncology Unit, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| |
Collapse
|
36
|
Tao M, Song T, Du W, Han S, Zuo C, Li Y, Wang Y, Yang Z. Classifying Breast Cancer Subtypes Using Multiple Kernel Learning Based on Omics Data. Genes (Basel) 2019; 10:E200. [PMID: 30866472 PMCID: PMC6471546 DOI: 10.3390/genes10030200] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/25/2019] [Accepted: 03/02/2019] [Indexed: 12/31/2022] Open
Abstract
It is very significant to explore the intrinsic differences in breast cancer subtypes. These intrinsic differences are closely related to clinical diagnosis and designation of treatment plans. With the accumulation of biological and medicine datasets, there are many different omics data that can be viewed in different aspects. Combining these multiple omics data can improve the accuracy of prediction. Meanwhile; there are also many different databases available for us to download different types of omics data. In this article, we use estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) to define breast cancer subtypes and classify any two breast cancer subtypes using SMO-MKL algorithm. We collected mRNA data, methylation data and copy number variation (CNV) data from TCGA to classify breast cancer subtypes. Multiple Kernel Learning (MKL) is employed to use these omics data distinctly. The result of using three omics data with multiple kernels is better than that of using single omics data with multiple kernels. Furthermore; these significant genes and pathways discovered in the feature selection process are also analyzed. In experiments; the proposed method outperforms other state-of-the-art methods and has abundant biological interpretations.
Collapse
Affiliation(s)
- Mingxin Tao
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China.
- Computational System Biology Laboratory, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA.
| | - Tianci Song
- Computer Science & Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Wei Du
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China.
| | - Siyu Han
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China.
| | - Chunman Zuo
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China.
| | - Ying Li
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China.
| | - Yan Wang
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China.
| | - Zekun Yang
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China.
| |
Collapse
|
37
|
Siddique AB, Ebrahim HY, Akl MR, Ayoub NM, Goda AA, Mohyeldin MM, Nagumalli SK, Hananeh WM, Liu YY, Meyer SA, El Sayed KA. (-)-Oleocanthal Combined with Lapatinib Treatment Synergized against HER-2 Positive Breast Cancer In Vitro and In Vivo. Nutrients 2019; 11:nu11020412. [PMID: 30781364 PMCID: PMC6412724 DOI: 10.3390/nu11020412] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/30/2019] [Accepted: 02/11/2019] [Indexed: 12/11/2022] Open
Abstract
Dysregulation of epidermal growth factor receptor (EGFR)/human epidermal growth factor-2 (HER2) family is a hallmark of aggressive breast cancer. Small-molecule tyrosine kinase inhibitors are among the most effective cancer targeted treatments. (−)-Oleocanthal (OC) is a naturally occurring phenolic secoiridoid lead from extra-virgin olive oil with documented anti-cancer activities via targeting mesenchymal epithelial transition factor (c-Met). Dysregulation of c-Met promotes aggressiveness to breast cancer-targeted therapies. Lapatinib (LP) is an FDA-approved dual EGFR/HER2 inhibitor for HER2-amplified breast cancer. HER2-Positive tumor cells can escape targeted therapies like LP effects by overexpressing c-Met. Combined OC-LP treatment is hypothesized to be mechanistically synergistic against HER2-overexpressing breast cancer. Combined sub-effective treatments of OC-LP resulted in synergistic anti-proliferative effects against the HER2-positive BT-474 and SK-BR-3 breast cancer cell lines, compared to OC or LP monotherapy. Antibody array and Western blot analysis showed that combined OC-LP treatment significantly inhibited EGFR, HER2, and c-Met receptor activation, as well as multiple downstream signaling proteins, compared to individual OC or LP treatment. OC-LP Combination significantly inhibited invasion and migration of breast cancer cells through reduced activation of focal adhesion kinase (FAK) and paxillin. Combined treatment of OC-10 mg/kg with LP-12.5 mg/kg suppressed more than 90% of BT-474 tumor cells growth in a nude mouse xenograft model, compared to individual OC or LP treatment. Activated c-Met, EGFR, HER2, and protein kinase B (AKT) were significantly suppressed in combination-treated mice tumors, compared to OC or LP monotherapy. This study reveals the OC future potential as combination therapy to sensitize HER2-overexpressing breast cancers and significantly reduce required doses of targeted HER family therapeutics.
Collapse
Affiliation(s)
- Abu Bakar Siddique
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA.
| | - Hassan Y Ebrahim
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA.
| | - Mohamed R Akl
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA.
| | - Nehad M Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan.
| | - Amira A Goda
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA.
| | - Mohamed M Mohyeldin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA.
| | - Suresh K Nagumalli
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA.
| | - Wael M Hananeh
- Department of Pathology and Public Health, Faculty of Veterinary Medicine, Jordan University of Science and Technology (JUST), Irbid 22110, Jordan.
| | - Yong-Yu Liu
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA.
| | - Sharon A Meyer
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA.
| | - Khalid A El Sayed
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA.
| |
Collapse
|
38
|
Martínez-Pérez C, Turnbull AK, Dixon JM. The evolving role of receptors as predictive biomarkers for metastatic breast cancer. Expert Rev Anticancer Ther 2018; 19:121-138. [PMID: 30501540 DOI: 10.1080/14737140.2019.1552138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION In breast cancer, estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2) are essential biomarkers to predict response to endocrine and anti-HER2 therapies, respectively. In metastatic breast cancer, the use of these receptors and targeted therapies present additional challenges: temporal heterogeneity, together with limited sampling methodologies, hinders receptor status assessment, and the constant evolution of the disease invariably leads to resistance to treatment. Areas covered: This review summarizes the genomic abnormalities in ER and HER2, such as mutations, amplifications, translocations, and alternative splicing, emerging as novel biomarkers that provide an insight into underlying mechanisms of resistance and hold potential predictive value to inform treatment selection. We also describe how liquid biopsies for sampling of circulating markers and ultrasensitive detection technologies have emerged which complement ongoing efforts for biomarker discovery and analysis. Expert commentary: While evidence suggests that genomic aberrations in ER and HER2 could contribute to meeting the pressing need for better predictive biomarkers, efforts need to be made to standardize assessment methods and better understand the resistance mechanisms these markers denote. Taking advantage of emerging technologies, research in upcoming years should include prospective trials incorporating these predictors into the study design to validate their potential clinical value.
Collapse
Affiliation(s)
- Carlos Martínez-Pérez
- a Breast Cancer Now Edinburgh Team, Institute of Genetics and Molecular Medicine , University of Edinburgh, Western General Hospital , Edinburgh , UK
| | - Arran K Turnbull
- a Breast Cancer Now Edinburgh Team, Institute of Genetics and Molecular Medicine , University of Edinburgh, Western General Hospital , Edinburgh , UK
| | - J Michael Dixon
- a Breast Cancer Now Edinburgh Team, Institute of Genetics and Molecular Medicine , University of Edinburgh, Western General Hospital , Edinburgh , UK.,b Edinburgh Breast Unit , Western General Hospital , Edinburgh , UK
| |
Collapse
|
39
|
Predicting the Efficacy of HER2-Targeted Therapies: A Look at the Host. DISEASE MARKERS 2017; 2017:7849108. [PMID: 29403144 PMCID: PMC5748305 DOI: 10.1155/2017/7849108] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/21/2017] [Indexed: 12/29/2022]
Abstract
HER2 is overexpressed in 20% of invasive breast cancers (BCs) and correlates with a more aggressive disease. Until the advent of targeted agents, HER2 was associated with worse outcomes. Rationally designed HER2-targeted agents have been developed and introduced into clinical practice for women with HER2-amplified BC, improving disease-free and overall survival for primary and metastatic tumors. Trastuzumab, a recombinant humanized anti-HER2 monoclonal antibody, combined with chemotherapy, remains the standard of care for patients with HER2-positive BCs. However, many patients do not respond to this agent, whereas newer drugs have proven to be efficacious in clinical trials. The identification of biomarkers that select sensitive tumors and patients who will benefit from these new agents would help the incorporation of these therapies, limiting the risk of side effects and overtreatment and improving the outcomes of all patients with early-stage HER2-positive BC. We review the mechanisms of action of HER2-targeting agents, focusing on the involvement of the immune system and related predictive biomarkers.
Collapse
|
40
|
Chellappan DK, Chellian J, Ng ZY, Sim YJ, Theng CW, Ling J, Wong M, Foo JH, Yang GJ, Hang LY, Nathan S, Singh Y, Gupta G. The role of pazopanib on tumour angiogenesis and in the management of cancers: A review. Biomed Pharmacother 2017; 96:768-781. [PMID: 29054093 DOI: 10.1016/j.biopha.2017.10.058] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/05/2017] [Accepted: 10/10/2017] [Indexed: 01/03/2023] Open
Abstract
Pazopanib is a relatively new compound to be introduced into the chemotherapy field. It is thought to have decent anti-angiogenic properties, which gives an additional hope for the treatment of certain types of cancers. A systematic review solely discussing about pazopanib and its anti-angiogenic effect is yet to be published to date, despite several relevant clinical trials being conducted over the recent years. In this review, we aim to investigate the mechanism of pazopanib's anti-angiogenic effect and its effectiveness in treating several cancers. We have included, in this study, findings from electronically searchable data from randomized clinical trials, clinical studies, cohort studies and other relevant articles. A total of 352 studies were included in this review. From the studies, the effect of pazopanib in various cancers or models was observed and recorded. Study quality is indefinite, with a few decent quality articles. The most elaborately studied cancers include renal cell carcinoma, solid tumors, advanced solid tumors, soft tissue sarcoma, breast cancer and gynecological cancers. In addition, several less commonly studied cancers are included in the studies as well. Pazopanib had demonstrated its anti-angiogenic effect based on favorable results observed in cancers, which are caused by angiogenesis-related mechanisms, such as renal cell carcinoma, solid tumors, advanced solid tumors and soft tissue sarcoma. This review was conducted to study, analyze and review the anti-angiogenic properties of pazopanib in various cancers. The results obtained can provide a decent reference when considering treatment options for angiogenesis-related malignancies. Furthermore, the definite observations of the anti-angiogenic effects of pazopanib could provide newer insights leading to the future development of drugs of the same mechanism with increased efficiency and reduced adverse effects.
Collapse
Affiliation(s)
- Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Jestin Chellian
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Zhao Yin Ng
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia; School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, Jaipur, 302017, India
| | - Yan Jinn Sim
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Chiu Wei Theng
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Joyce Ling
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Mei Wong
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Jia Hui Foo
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Goh Jun Yang
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Li Yu Hang
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Saranyah Nathan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Yogendra Singh
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, Jaipur, 302017, India
| | - Gaurav Gupta
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, Jaipur, 302017, India.
| |
Collapse
|
41
|
Huang JP, Ling K. EZH2 and histone deacetylase inhibitors induce apoptosis in triple negative breast cancer cells by differentially increasing H3 Lys 27 acetylation in the BIM gene promoter and enhancers. Oncol Lett 2017; 14:5735-5742. [PMID: 29113202 PMCID: PMC5661363 DOI: 10.3892/ol.2017.6912] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 03/30/2017] [Indexed: 11/21/2022] Open
Abstract
Enhancer of zeste homolog 2 (EZH2), a subunit of polycomb repressive complex 2, is a histone methyl-transferase and is considered to work cooperatively with histone deacetylases (HDACs) in the same protein complex to mediate gene transcription repression by increasing histone H3 Lys27 trimethylation (H3K27me3), in particular in the nucleosome (s). EZH2 is overexpressed in numerous types of cancer, including triple negative breast cancer (TNBC), a subtype of breast cancer, which there are no effective treatment options for. Thus, inhibition of EZH2 may be harnessed for targeted therapy of this disease. The present study demonstrated that co-treatment with an EZH2 inhibitor and a HDAC inhibitor additively induced apoptosis in two TNBC cell lines, namely MDA-MB-231 and MDA-MB-436. The increased rate of cell death was associated with an elevation of B cell lymphoma-2 like 11 (BIM) expression level, a pro-apoptotic protein at the protein and mRNA expression levels in these two cell lines. The expression of forkhead box O1 (FOXO1), a known upstream transcriptional activator of BIM, was upregulated in both cell lines by the HDAC inhibitor, and the effect was more pronounced in MDA-MB-436 cells with higher phosphorylation levels of protein kinase B, a negative regulator of FOXO1, compared with MDA-MB-231 cells. Conversely, FOXO1 expression was inhibited following treatment with the EZH2 inhibitor, suggesting that EZH2 and HDAC inhibitors induced BIM expression via a FOXO1-independent mechanism. The present study further revealed that the EZH2 inhibitor, but not the HDAC inhibitor, induced high levels of H3K27 acetylation (H3K27ac) in the BIM promoter. By contrast, compared with the effect of the EZH2 inhibitor, HDAC inhibitor treatment resulted in an increase in H3K27ac at two BIM enhancers. Collectively, the results of the present study indicated that EZH2 and HDACs act differentially on H3K27ac levels in the nucleosome at the promoter and enhancer regions of the BIM gene. Through the upregulation of BIM, co-treatment with EZH2 and HDAC inhibitors had a pronounced therapeutic effect on TNBC cells, suggesting that co-targeting EZH2 and HDAC proteins represents a viable therapeutic option for the treatment of TNBC.
Collapse
Affiliation(s)
- Julia P Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
42
|
Kuninaka K, Takahashi R, Nakagawa Y, Nishimaki T. A case of HER2-positive male occult breast carcinoma with skin and lymph node metastases that exhibited complete response to trastuzumab monotherapy. Clin Case Rep 2017; 5:591-593. [PMID: 28469855 PMCID: PMC5412765 DOI: 10.1002/ccr3.884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 10/06/2016] [Accepted: 10/21/2016] [Indexed: 11/08/2022] Open
Abstract
Metastatic male occult HER2‐positive breast cancer can be successfully treated with trastuzumab monotherapy.
Collapse
Affiliation(s)
- Kouichi Kuninaka
- Division of Digestive and General Surgery Ryukyu University Graduate School of Medicine Okinawa Japan
| | - Ryo Takahashi
- Division of Digestive and General Surgery Ryukyu University Graduate School of Medicine Okinawa Japan
| | - Yutaka Nakagawa
- Division of Digestive and General Surgery Ryukyu University Graduate School of Medicine Okinawa Japan
| | - Tadashi Nishimaki
- Division of Digestive and General Surgery Ryukyu University Graduate School of Medicine Okinawa Japan
| |
Collapse
|
43
|
Santos JC, Ribeiro ML, Sarian LO, Ortega MM, Derchain SF. Exosomes-mediate microRNAs transfer in breast cancer chemoresistance regulation. Am J Cancer Res 2016; 6:2129-2139. [PMID: 27822407 PMCID: PMC5088281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/09/2016] [Indexed: 06/06/2023] Open
Abstract
Breast cancer is the most common and fatal type of cancer in women worldwide due to the metastatic process and resistance to treatment. Despite advances in molecular knowledge, little is known regarding resistance to chemotherapy. One highlighted aspect is the DNA damage response (DDR) pathway that is activated upon genotoxic damage, controlling the cell cycle arrest or DNA repair activation. Recently, studies have showed that cancer stem cells (CSCs) could promote chemoresistance through DDR pathway. Furthermore, it is known that the epithelial-mesenchymal transition (EMT) can generate cells with CSCs characteristics and therefore regulate the chemoresistance process. The exosomes are microvesicles filled with RNAs, proteins and microRNAs (miRNAs) that can be released by many cell types, including tumor cells and CSCs. The exosomes content may be cell-to-cell transferable and it could control a wide range of pathways during tumor development and metastasis. A big challenge for modern medicine is to determine the reasons why patients do not respond to chemotherapy treatments and also guide the most appropriate therapy for each one. Considering that the CSCs are able to stimulate the formation of a more aggressive tumor phenotype with migration and metastasis ability, resistance to treatment and disease recurrence, as well as few studies capable to determine clearly the interaction of breast CSCs with its microenvironment, the present review summarize the possibility that exosomes-mediate miRNAs transfer and regulate chemoresistance in breast tumor cells and CSCs, to clarify the complexity of breast cancer progression and therapy.
Collapse
Affiliation(s)
- Juliana Carvalho Santos
- Women’s Health Hospital “Prof Dr José Aristodemo Pinotti” (CAISM), State University of Campinas (UNICAMP)Campinas, SP, Brazil
| | - Marcelo Lima Ribeiro
- Clinical Pharmacology and Gastroenterology Unit, São Francisco University, São Francisco UniversityBragança Paulista, SP, Brazil
| | - Luis Otávio Sarian
- Women’s Health Hospital “Prof Dr José Aristodemo Pinotti” (CAISM), State University of Campinas (UNICAMP)Campinas, SP, Brazil
| | - Manoela Marques Ortega
- Clinical Pharmacology and Gastroenterology Unit, São Francisco University, São Francisco UniversityBragança Paulista, SP, Brazil
| | - Sophie Françoise Derchain
- Women’s Health Hospital “Prof Dr José Aristodemo Pinotti” (CAISM), State University of Campinas (UNICAMP)Campinas, SP, Brazil
| |
Collapse
|
44
|
Dash R, Uddin MMN, Hosen SZ, Rahim ZB, Dinar AM, Kabir MSH, Sultan RA, Islam A, Hossain MK. Molecular docking analysis of known flavonoids as duel COX-2 inhibitors in the context of cancer. Bioinformation 2015; 11:543-9. [PMID: 26770028 PMCID: PMC4702032 DOI: 10.6026/97320630011543] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 12/16/2022] Open
Abstract
Cyclooxygenase-2 (COX-2) catalyzed synthesis of prostaglandin E2 and it associates with tumor growth, infiltration, and metastasis in preclinical experiments. Known inhibitors against COX-2 exhibit toxicity. Therefore, it is of interest to screen natural compounds like flavanoids against COX-2. Molecular docking using 12 known flavanoids against COX-2 by FlexX and of ArgusLab were performed. All compounds showed a favourable binding energy of >-10 KJ/mol in FlexX and > -8 kcal/mol in ArgusLab. However, this data requires in vitro and in vivo verification for further consideration.
Collapse
Affiliation(s)
- Raju Dash
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong-4000, Bangladesh
| | | | - S.M. Zahid Hosen
- Molecular Modeling & Drug Design Laboratory (MMDDL), Pharmacology Research
Division, Bangladesh Council of Scientific & Industrial Research (BCSIR), Chittagong-4220, Bangladesh
| | - Zahed Bin Rahim
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong-4000, Bangladesh
| | - Abu Mansur Dinar
- Quality Control Operations, Square Pharmaceutical Ltd, Bangladesh
| | | | - Ramiz Ahmed Sultan
- Department of Pharmacy, University of Chittagong, Chittagong-4331, Bangladesh
| | - Ashekul Islam
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong-4331, Bangladesh
| | - Md Kamrul Hossain
- Department of Pharmacy, University of Chittagong, Chittagong-4331, Bangladesh
| |
Collapse
|
45
|
Nounou MI, ElAmrawy F, Ahmed N, Abdelraouf K, Goda S, Syed-Sha-Qhattal H. Breast Cancer: Conventional Diagnosis and Treatment Modalities and Recent Patents and Technologies. Breast Cancer (Auckl) 2015; 9:17-34. [PMID: 26462242 PMCID: PMC4589089 DOI: 10.4137/bcbcr.s29420] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/08/2015] [Accepted: 09/09/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Breast cancer is the most prevalent cancer among women worldwide. However, increased survival is due to the dramatic advances in the screening methods, early diagnosis, and breakthroughs in treatments. Over the course of the last decade, many acquisitions have taken place in this critical field of research in the pharmaceutical industry. Advances in molecular biology and pharmacology aided in better understanding of breast cancer, enabling the design of smarter therapeutics able to target cancer and respond to its microenvironment efficiently. Patents and research papers investigating diagnosis and treatment strategies for breast cancer using novel technologies have been surveyed for the past 15 years. Various nanocarriers have been introduced to improve the therapeutic efficacy of anticancer drugs, including liposomes, polymeric micelles, quantum dots, nanoparticles, and dendrimers. This review provides an overview of breast cancer, conventional therapy, novel technologies in the management of breast cancer, and rational approaches for targeting breast cancer. HIGHLIGHTS Breast cancer is the most common cancer in women worldwide. However, survival rates vary widely, optimistically heading toward a positive trend. Increased survival is due to the drastic shift in the screening methods, early diagnosis, and breakthroughs in treatments.Different strategies of breast cancer classification and staging have evolved over the years. Intrinsic (molecular) subtyping is essential in clinical trials and well understanding of the disease.Many novel technologies are being developed to detect distant metastases and recurrent disease as well as to assess response to breast cancer management.Intensive research efforts are actively ongoing to take novel breast cancer therapeutics to potential clinical application.Most of the recent research papers and patents discuss one of the following strategies: the development of new drug entities that specifically target the breast tumor cells; tailor designing a novel carrier system that can multitask and multifunction as a drug carrier, targeting vehicle and even as a diagnostic tool, direct conjugation of a therapeutic drug moiety with a targeting moiety, diagnostic moiety or pharmacokinetics altering moiety; or the use of innovative nontraditional approaches such as genetic engineering, stem cells, or vaccinations.
Collapse
Affiliation(s)
- Mohamed I. Nounou
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Fatema ElAmrawy
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Nada Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Kamilia Abdelraouf
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | | | | |
Collapse
|
46
|
Breast cancer with neoductgenesis: histopathological criteria and its correlation with mammographic and tumour features. Int J Breast Cancer 2014; 2014:581706. [PMID: 25400950 PMCID: PMC4220584 DOI: 10.1155/2014/581706] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 09/09/2014] [Indexed: 11/30/2022] Open
Abstract
Introduction. Breast cancer with mammographic casting type calcifications, high grade DCIS with an abnormal number of ducts, periductal desmoplastic reaction, lymphocyte infiltration, and tenascin-C (TN-C) overexpression has been proposed to represent a more aggressive form of breast cancer and has been denominated as breast cancer with neoductgenesis. We developed histopathological criteria for neoductgenesis in order to study reproducibility and correlation with other tumour markers. Methods. 74 cases of grades 2 and 3 DCIS, with or without an invasive component, were selected. A combined score of the degree(s) of concentration of ducts, lymphocyte infiltration, and periductal fibrosis was used to classify cases as showing neoductgenesis, or not. Diagnostic reproducibility, correlation with tumour markers, and mammographic features were studied. Results. Twenty-three of 74 cases were diagnosed with neoductgenesis. The kappa value between pathologists showed moderate reproducibility (0.50) (95% CI; 0.41–0.60). Neoductgenesis correlated significantly with malignant type microcalcifications and TN-C expression (P = 0.008 and 0.04) and with ER, PR, and HER2 status (P < 0.00001 for all three markers). Conclusions. We developed histological criteria for breast cancer with neoductgenesis. Neoductgenesis, by our applied histopathological definition was related to more aggressive tumour biology and malignant mammographic calcifications.
Collapse
|
47
|
Li G, Wang X, Hibshoosh H, Jin C, Halmos B. Modulation of ErbB2 blockade in ErbB2-positive cancers: the role of ErbB2 Mutations and PHLDA1. PLoS One 2014; 9:e106349. [PMID: 25238247 PMCID: PMC4169529 DOI: 10.1371/journal.pone.0106349] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 08/06/2014] [Indexed: 12/20/2022] Open
Abstract
We set out to study the key effectors of resistance and sensitivity to ErbB2 tyrosine kinase inhibitors, such as lapatinib in ErbB2-positive breast and lung cancers. A cell-based in vitro site-directed mutagenesis lapatinib resistance model identified several mutations, including the gatekeeper ErbB2 mutation ErbB2-T798I, as mediating resistance. ErbB2-T798I engineered cell models indeed show resistance to lapatinib but remain sensitive to the irreversible EGFR/ErbB2 inhibitor, PD168393, suggestive of potential alternative treatment strategies to overcome resistance. Gene expression profiling studies identified a select group of downstream targets regulated by ErbB2 signaling and define PHLDA1 as an immediately downregulated gene upon oncogenic ErbB2 signaling inhibition. We find significant down-regulation of PHLDA1 in primary breast cancer and PHLDA1 is statistically significantly less expressed in ErbB2 negative compared with ErbB2 positive tumors consistent with its regulation by ErbB2. Lastly, PHLDA1 overexpression blocks AKT signaling, inhibits cell growth and enhances lapatinib sensitivity further supporting an important negative growth regulator function. Our findings suggest that PHLDA1 might have key inhibitory functions in ErbB2 driven lung and breast cancer cells and a better understanding of its functions might point at novel therapeutic options. In summary, our studies define novel ways of modulating sensitivity and resistance to ErbB2 inhibition in ErbB2-dependent cancers.
Collapse
Affiliation(s)
- Guangyuan Li
- Department of Pathology, University Hospitals of Case Medical Center, Case Western Reserve University, Cleveland, Ohio, United States of America
- Division of Hematology/Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, United States of America
| | - Xiaoqi Wang
- Division of Hematology/Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, United States of America
| | - Hanina Hibshoosh
- Department of Pathology, Columbia University Medical Center, New York, New York, United States of America
| | - Cheng Jin
- Division of Hematology/Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, United States of America
| | - Balazs Halmos
- Division of Hematology/Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
48
|
Wållberg H, Ståhl S. Design and evaluation of radiolabeled tracers for tumor imaging. Biotechnol Appl Biochem 2014; 60:365-83. [PMID: 24033592 DOI: 10.1002/bab.1111] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 02/20/2013] [Indexed: 12/22/2022]
Abstract
The growing understanding of tumor biology and the identification of tumor-specific genetic and molecular alterations, such as the overexpression of membrane receptors and other proteins, allows for personalization of patient management using targeted therapies. However, this puts stringent demands on the diagnostic tools used to identify patients who are likely to respond to a particular treatment. Radionuclide molecular imaging is a promising noninvasive method to visualize and characterize the expression of such targets. A number of different proteins, from full-length antibodies and their derivatives to small scaffold proteins and peptide receptor-ligands, have been applied to molecular imaging, each demonstrating strengths and weaknesses. Here, we discuss the concept of molecular targeting and, in particular, molecular imaging of cancer-associated targets. Additionally, we describe important biotechnological considerations and desired features when designing and developing tracers for radionuclide molecular imaging.
Collapse
Affiliation(s)
- Helena Wållberg
- Division of Molecular Biotechnology, School of Biotechnology, AlbaNova University Center, KTH Royal Institute of Technology, Stockholm, Sweden
| | | |
Collapse
|
49
|
Yang CR, Liao WS, Wu YH, Murugan K, Chen C, Chao JI. CR108, a novel vitamin K3 derivative induces apoptosis and breast tumor inhibition by reactive oxygen species and mitochondrial dysfunction. Toxicol Appl Pharmacol 2013; 273:611-22. [PMID: 24128853 DOI: 10.1016/j.taap.2013.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/27/2013] [Accepted: 10/05/2013] [Indexed: 11/26/2022]
Abstract
Vitamin K3 derivatives have been shown to exert anticancer activities. Here we show a novel vitamin K3 derivative (S)-2-(2-hydroxy-3-methylbutylthio)naphthalene-1,4-dione, which is named as CR108 that induces apoptosis and tumor inhibition through reactive oxygen species (ROS) and mitochondrial dysfunction in human breast cancer. CR108 is more effective on the breast cancer cell death than other vitamin K3 derivatives. Moreover, CR108 induced apoptosis in both the non-HER-2-overexpressed MCF-7 and HER-2-overexpressed BT-474 breast cancer cells. CR108 caused the loss of mitochondrial membrane potential, cytochrome c released from mitochondria to cytosol, and cleaved PARP proteins for apoptosis induction. CR108 markedly increased ROS levels in breast cancer cells. N-acetylcysteine (NAC), a general ROS scavenger, completely blocked the CR108-induced ROS levels, mitochondrial dysfunction and apoptosis. Interestingly, CR108 increased the phosphorylation of p38 MAP kinase but conversely inhibited the survivin protein expression. NAC treatment prevented the activation of p38 MAP kinase and rescued the survivin protein levels. SB202190, a specific p38 MAP kinase inhibitor, recovered the survivin protein levels and attenuated the cytotoxicity of CR108-treated cells. Furthermore, CR108 inhibited the xenografted human breast tumor growth in nude mice. Together, we demonstrate that CR108 is a novel vitamin K3 derivative that induces apoptosis and tumor inhibition by ROS production and mitochondrial dysfunction and associates with the phosphorylation of p38 MAP kinase and the inhibition of survivin in the human breast cancer.
Collapse
Affiliation(s)
- Chun-Ru Yang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30068, Taiwan
| | | | | | | | | | | |
Collapse
|
50
|
Hurrell T, Outhoff K. The in vitro influences of epidermal growth factor and heregulin-β1 on the efficacy of trastuzumab used in Her-2 positive breast adenocarcinoma. Cancer Cell Int 2013; 13:97. [PMID: 24119761 PMCID: PMC3852844 DOI: 10.1186/1475-2867-13-97] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 10/10/2013] [Indexed: 11/15/2022] Open
Abstract
Background Human epidermal growth factor receptor-2 (Her-2) is over expressed in approximately 25-30% of all primary breast tumors resulting in a distinctive breast cancer subtype associated with a poor prognosis and a decrease in overall survival. Trastuzumab (Herceptin®), an anti-Her-2 monoclonal antibody, has dramatically altered the prognosis of Her-2 positive breast cancer. Trastuzumab is, however, associated with primary and acquired resistance. Aim and methods To investigate the in-vitro effects of trastuzumab on cell viability (tetrazolium conversion assay), cell cycling (propidium iodide staining), apoptosis (executioner caspases and annexin-V) and relative surface Her-2 receptor expression (anti-Her-2 affibody molecule) in Her-2-positive (SK-Br-3) and oestrogen receptor positive (MCF-7) breast adenocarcinoma cells and to determine potential augmentation of these effects by two endogenous ligands, epidermal growth factor (EGF) and heregulin-β1 (HRG- β1). Results Cell viability was decreased in SK-Br-3 cells by exposure to trastuzumab. This was associated with G1 accumulation and decreased relative surface Her-2 receptor density, supporting the cytostatic nature of trastuzumab in vitro. SK-Br-3 cells exposed to EGF and heregulin-β1 produced differential cell responses alone and in combination with trastuzumab, in some instances augmenting cell viability and cell cycling. Relative surface Her-2 receptor density was reduced substantially by trastuzumab, EGF and heregulin-β1. These reductions were amplified when ligands were used in combination with trastuzumab. Conclusion Cell type specific interactions of endogenous ligands appear to be dependent on absolute Her-receptor expression and cross activation of signaling pathways. This supports the notion that receptor density of Her-family members and multiplicity of growth ligands are of mutual importance in breast cancer cell proliferation and therefore also in resistance associated with trastuzumab.
Collapse
Affiliation(s)
- Tracey Hurrell
- Department of Pharmacology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Private Bag X323, Pretoria 0007, South Africa.
| | | |
Collapse
|