1
|
Cardoso VDO, Bistaffa MJ, Sterman RG, Lima LLD, Toldo GS, Cancino-Bernardi J, Zucolotto V. Nanomedicine Innovations for Lung Cancer Diagnosis and Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13197-13220. [PMID: 40045524 PMCID: PMC11891907 DOI: 10.1021/acsami.4c16840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 03/12/2025]
Abstract
Lung cancer remains a challenge within the realm of oncology. Characterized by late-stage diagnosis and resistance to conventional treatments, the currently available therapeutic strategies encompass surgery, radiotherapy, chemotherapy, immunotherapy, and biological therapy; however, overall patient survival remains suboptimal. Nanotechnology has ushered in a new era by offering innovative nanomaterials with the potential to precisely target cancer cells while sparing healthy tissues. It holds the potential to reshape the landscape of cancer management, offering hope for patients and clinicians. The assessment of these nanotechnologies follows a rigorous evaluation process similar to that applied to chemical drugs, which includes considerations of their pharmacokinetics, pharmacodynamics, toxicology, and clinical effectiveness. However, because of the characteristics of nanoparticles, standard toxicological tests require modifications to accommodate their unique characteristics. Effective therapeutic strategies demand a profound understanding of the disease and consideration of clinical outcomes, physicochemical attributes of nanomaterials, nanobiointeractions, nanotoxicity, and regulatory compliance to ensure patient safety. This review explores the promise of nanomedicine in lung cancer treatment by capitalizing on its unique physicochemical properties. We address the multifaceted challenges of lung cancer and its tumor microenvironment and provide an overview of recent developments in nanoplatforms for early diagnosis and treatment that can enhance patient outcomes and overall quality of life.
Collapse
Affiliation(s)
- Valéria
Maria de Oliveira Cardoso
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, 13560-970 São Carlos, São Paulo, Brazil
| | - Maria Julia Bistaffa
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, 13560-970 São Carlos, São Paulo, Brazil
| | - Raquel González Sterman
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, 13560-970 São Carlos, São Paulo, Brazil
| | - Lorena Leticia
Peixoto de Lima
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, 13560-970 São Carlos, São Paulo, Brazil
| | - Gustavo Silveira Toldo
- Chemistry
Department, Laboratory in Bioanalytical of Nanosystems, Faculty of
Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, 14040-901 Ribeirão Preto, São Paulo, Brazil
| | - Juliana Cancino-Bernardi
- Chemistry
Department, Laboratory in Bioanalytical of Nanosystems, Faculty of
Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, 14040-901 Ribeirão Preto, São Paulo, Brazil
| | - Valtencir Zucolotto
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, 13560-970 São Carlos, São Paulo, Brazil
- Comprehensive
Center for Precision Oncology, C2PO, University of São Paulo, São Paulo 01246-000, Brazil
| |
Collapse
|
2
|
Wang X, Qin Z, Qiu W, Xu K, Bai Y, Zeng B, Ma Y, Yang S, Shi Y, Fan Y. Novel EGFR inhibitors against resistant L858R/T790M/C797S mutant for intervention of non-small cell lung cancer. Eur J Med Chem 2024; 277:116711. [PMID: 39094277 DOI: 10.1016/j.ejmech.2024.116711] [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: 06/17/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
To overcome C797S mutation, the latest and most common resistance mechanism in the clinical treatment of third-generation EGFR inhibitor, a novel series of substituted 6-(2-aminopyrimidine)-indole derivatives were designed and synthesized. Through the structure-activity relationship (SAR) study, compound 11eg was identified as a novel and potent EGFR L858R/T790M/C797S inhibitor (IC50 = 0.053 μM) but had a weak effect on EGFRWT (IC50 = 1.05 μM). 11eg significantly inhibited the proliferation of the non-small cell lung cancer (NSCLC) cells harboring EGFRL858R/T790M/C797S with an IC50 of 0.052 μM. 11eg also showed potent inhibitory activity against other NSCLC cell lines harboring main EGFR mutants. Furthermore, 11eg exhibited much superior activity in arresting cell cycle and inducing apoptosis of NSCLC cells with mutant EGFRC797S. It blocked cellular EGFR signaling. Importantly, 11eg markedly suppressed the tumor growth in in vivo xenograft mouse model with good safety. Additionally, 11eg displayed good microsomal stability. These results demonstrated the potential of 11eg with novel scaffold as a promising lead compound targeting EGFRC797S to guide in-depth structural optimization.
Collapse
Affiliation(s)
- Xiaoxue Wang
- Eye Institute, Nankai University, 94 Weijin Road, Tianjin, 300071, China; School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Zhongxiang Qin
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Wenrui Qiu
- Tianjin Normal University, No.393, Extension of Bin Shui West Road, Xi Qing District, Tianjin, 300387, China
| | - Kejia Xu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Yuting Bai
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Beilei Zeng
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Yakun Ma
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Shuang Yang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| | - Yi Shi
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| | - Yan Fan
- Eye Institute, Nankai University, 94 Weijin Road, Tianjin, 300071, China; School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
3
|
Wang C, Wang X, Wang X, Tian B, Zhang S, Wang T, Ma Y, Fan Y. Design, synthesis and biological evaluation of potent epidermal growth factor receptor tyrosine kinase (EGFR-TK) inhibitors against resistance mutation for lung cancer treatment. Bioorg Chem 2024; 143:107004. [PMID: 38086238 DOI: 10.1016/j.bioorg.2023.107004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/05/2023] [Accepted: 11/23/2023] [Indexed: 01/24/2024]
Abstract
In this study, we identified a newly synthesized compound 7o with potent inhibition on EGFR primary mutants (L858R, Del19) and drug-resistant mutant T790M with nanomolar IC50 values. 7o showed strong antiproliferative effects against EGFR mutant-driven non-small cell lung cancer (NSCLC) cells such as H1975, PC-9 and HCC827, over cells expressing EGFRWT. Molecular docking was performed to investigate the possible binding modes of 7o inside the binding site of EGFRL858R/T790M and EGFRWT. Analysis of cell cycle evidenced that 7o induced cell cycle arrest in G1 phases in the EGFR mutant cells, H1975 and PC-9, which resulted in decreased S-phase populations. Moreover, compound 7o induced cancer cell apoptosis in in vitro assays. In addition, 7o inhibited cellular phosphorylation of EGFR. In vivo, oral administration of 7o caused rapid tumor regression in H1975 xenograft model. Therefore, 7o might deserve further optimization as cancer treatment agent for EGFR mutant-driven NSCLC.
Collapse
Affiliation(s)
- Cheng Wang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xin Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xiaoxue Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Baorui Tian
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Sihe Zhang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Tianqi Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yakun Ma
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yan Fan
- Eye Institute, Nankai University, 94 Weijin Road, Tianjin 300071, China; School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| |
Collapse
|
4
|
Jeon J, Jang SY, Kwak EJ, Lee SH, Byun JY, Kim YY, Ahn YG, Singh P, Moon K, Kim IS. Design and synthesis of 4th generation EGFR inhibitors against human triple (Del19/T790M/C797S) mutation. Eur J Med Chem 2023; 261:115840. [PMID: 37783102 DOI: 10.1016/j.ejmech.2023.115840] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/04/2023]
Abstract
Epidermal growth factor receptor (EGFR)-targeted therapy is used to treat EGFR mutation-induced non-small cell lung cancer (NSCLC). However, its efficacy does not last beyond a certain period due to the development of primary and secondary resistance. First and second-generation inhibitors (e.g., gefitinib, erlotinib, and afatinib) induce EGFR T790M mutations, while third-generation inhibitors (e.g., osimertinib) induce C797S as a major target resistance mutation. Therefore, the C797S mutation is being actively researched. In this study, we investigated the structure-activity relationship of several synthesized compounds as fourth-generation inhibitors against the C797S mutation. We identified a compound 13k that displayed nanomolar potency and high selectivity. Moreover, we used a triple mutant xenograft mouse model to evaluate the in vivo efficacy of 13k in inhibiting EGFR C797S, which demonstrated exceptional profiles and satisfactory EGFR C797S inhibition efficacy. Based on its excellent in vitro and in vivo profiles, compound 13k can be considered a promising candidate for treating EGFR C797S mutations.
Collapse
Affiliation(s)
- Jiyoung Jeon
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong, 18469, Republic of Korea
| | - Sun Young Jang
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong, 18469, Republic of Korea
| | - Eun Joo Kwak
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong, 18469, Republic of Korea
| | - Sun Hoe Lee
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong, 18469, Republic of Korea
| | - Joo-Yun Byun
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong, 18469, Republic of Korea
| | - Yu-Yon Kim
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong, 18469, Republic of Korea
| | - Young Gil Ahn
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong, 18469, Republic of Korea
| | - Pargat Singh
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyeongwon Moon
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
5
|
Zhang N, Li Y. Receptor tyrosine kinases: biological functions and anticancer targeted therapy. MedComm (Beijing) 2023; 4:e446. [PMID: 38077251 PMCID: PMC10701465 DOI: 10.1002/mco2.446] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 10/16/2024] Open
Abstract
Receptor tyrosine kinases (RTKs) are a class of protein kinases that play crucial roles in various cellular processes, including cell migration, morphological differentiation, cell growth, and angiogenesis. In humans, 58 RTKs have been identified and categorized into 20 distinct families based on the composition of their extracellular regions. RTKs are primarily activated by specific ligands that bind to their extracellular region. They not only regulate tumor transformation, proliferation, metastasis, drug resistance, and angiogenesis, but also initiate and maintain the self-renewal and cloning ability of cancer stem cells. Accurate diagnosis and grading of tumors with dysregulated RTKs are essential in clinical practice. There is a growing body of evidence supporting the benefits of RTKs-targeted therapies for cancer patients, and researchers are actively exploring new targets and developing targeted agents. However, further optimization of RTK inhibitors is necessary to effectively target the diverse RTK alterations observed in human cancers. This review provides insights into the classification, structure, activation mechanisms, and expression of RTKs in tumors. It also highlights the research advances in RTKs targeted anticancer therapy and emphasizes their significance in optimizing cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Nan Zhang
- Chongqing University Cancer Hospital, School of MedicineChongqing UniversityChongqingChina
| | - Yongsheng Li
- Chongqing University Cancer Hospital, School of MedicineChongqing UniversityChongqingChina
- Department of Medical OncologyChongqing University Cancer HospitalChongqingChina
| |
Collapse
|
6
|
Li J, Gu A, Nong XM, Zhai S, Yue ZY, Li MY, Liu Y. Six-Membered Aromatic Nitrogen Heterocyclic Anti-Tumor Agents: Synthesis and Applications. CHEM REC 2023; 23:e202300293. [PMID: 38010365 DOI: 10.1002/tcr.202300293] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/19/2023] [Indexed: 11/29/2023]
Abstract
Cancer stands as a serious malady, posing substantial risks to human well-being and survival. This underscores the paramount necessity to explore and investigate novel antitumor medications. Nitrogen-containing compounds, especially those derived from natural sources, form a highly significant category of antitumor agents. Among these, antitumor agents with six-membered aromatic nitrogen heterocycles have consistently attracted the attention of chemists and pharmacologists. Accordingly, we present a comprehensive summary of synthetic strategies and clinical implications of these compounds in this review. This entails an in-depth analysis of synthesis pathways for pyridine, quinoline, pyrimidine, and quinazoline. Additionally, we explore the historical progression, targets, mechanisms of action, and clinical effectiveness of small molecule inhibitors possessing these structural features.
Collapse
Affiliation(s)
- Jiatong Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Ao Gu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Xiao-Mei Nong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Shuyang Zhai
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Zhu-Ying Yue
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Meng-Yao Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Yingbin Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| |
Collapse
|
7
|
Li P, Li B, Yang N, Xu T, Zheng Z. The next generation of EGFR inhibitors: a patenting perspective of PROTACs based EGFR degraders. Expert Opin Ther Pat 2023; 33:477-492. [PMID: 37873645 DOI: 10.1080/13543776.2023.2262176] [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: 05/22/2023] [Accepted: 09/19/2023] [Indexed: 10/25/2023]
Abstract
INTRODUCTION Abnormal expression of epidermal growth factor receptor (EGFR) contributes to tumor development, especially in non-small cell lung cancer (NSCLC). Although multiple inhibitors have been developed to target diverse EGFR mutations and several have been approved, the inevitable drug resistance and side effect remain a challenge, which motivates novel strategies. Proteolysis-targeting chimeras (PROTACs) have been gaining momentum for their potential as novel therapeutics for human diseases by triggering protein degradation. To date, various potent and specific EGFR PROTACs have been discovered and some of them have entered clinical trials. AREAS COVERED This review provides an overview of EGFR degraders in patents from 2016 to 2022. It provides an update of the discovery strategies, chemical structures, and molecular profiling of all available EGFR PROTACs. SciFinder, PubMed, Web of Science, EPO, and CNIPA databases were used for searching the literature and patents for EGFR PROTACs. EXPERT OPINION By employing the PROTAC technology, highly potent and selective EGFR degraders based on four generation EGFR inhibitors have been developed, which offer a new strategy to target EGFR mutations and overcome the drug resistance. Despite the satisfactory result in vitro and in vivo studies, their therapeutic value awaits more rigorous preclinical testing and clinical investigation.
Collapse
Affiliation(s)
- Pengyun Li
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Bingkun Li
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ning Yang
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Tingting Xu
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhibing Zheng
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
8
|
Puhl AC, Godoy AS, Noske GD, Nakamura AM, Gawriljuk VO, Fernandes RS, Oliva G, Ekins S. Discovery of PL pro and M pro Inhibitors for SARS-CoV-2. ACS OMEGA 2023; 8:22603-22612. [PMID: 37387790 PMCID: PMC10275482 DOI: 10.1021/acsomega.3c01110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023]
Abstract
There are very few small-molecule antivirals for SARS-CoV-2 that are either currently approved (or emergency authorized) in the US or globally, including remdesivir, molnupiravir, and paxlovid. The increasing number of SARS-CoV-2 variants that have appeared since the outbreak began over three years ago raises the need for continual development of updated vaccines and orally available antivirals in order to fully protect or treat the population. The viral main protease (Mpro) and the papain-like protease (PLpro) are key for viral replication; therefore, they represent valuable targets for antiviral therapy. We herein describe an in vitro screen performed using the 2560 compounds from the Microsource Spectrum library against Mpro and PLpro in an attempt to identify additional small-molecule hits that could be repurposed for SARS-CoV-2. We subsequently identified 2 hits for Mpro and 8 hits for PLpro. One of these hits was the quaternary ammonium compound cetylpyridinium chloride with dual activity (IC50 = 2.72 ± 0.09 μM for PLpro and IC50 = 7.25 ± 0.15 μM for Mpro). A second inhibitor of PLpro was the selective estrogen receptor modulator raloxifene (IC50 = 3.28 ± 0.29 μM for PLpro and IC50 = 42.8 ± 6.7 μM for Mpro). We additionally tested several kinase inhibitors and identified olmutinib (IC50 = 0.54 ± 0.04 μM), bosutinib (IC50 = 4.23 ± 0.28 μM), crizotinib (IC50 = 3.81 ± 0.04 μM), and dacominitinib (IC50 = IC50 3.33 ± 0.06 μM) as PLpro inhibitors for the first time. In some cases, these molecules have also been tested by others for antiviral activity for this virus, or we have used Calu-3 cells infected with SARS-CoV-2. The results suggest that approved drugs can be identified with promising activity against these proteases, and in several cases we or others have validated their antiviral activity. The additional identification of known kinase inhibitors as molecules targeting PLpro may provide new repurposing opportunities or starting points for chemical optimization.
Collapse
Affiliation(s)
- Ana C. Puhl
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Andre S. Godoy
- Sao
Carlos Institute of Physics, University
of Sao Paulo, Av. Joao
Dagnone, 1100—Jardim Santa Angelina, Sao Carlos 13563-120, Brazil
| | - Gabriela D. Noske
- Sao
Carlos Institute of Physics, University
of Sao Paulo, Av. Joao
Dagnone, 1100—Jardim Santa Angelina, Sao Carlos 13563-120, Brazil
| | - Aline M. Nakamura
- Sao
Carlos Institute of Physics, University
of Sao Paulo, Av. Joao
Dagnone, 1100—Jardim Santa Angelina, Sao Carlos 13563-120, Brazil
| | - Victor O. Gawriljuk
- Sao
Carlos Institute of Physics, University
of Sao Paulo, Av. Joao
Dagnone, 1100—Jardim Santa Angelina, Sao Carlos 13563-120, Brazil
| | - Rafaela S. Fernandes
- Sao
Carlos Institute of Physics, University
of Sao Paulo, Av. Joao
Dagnone, 1100—Jardim Santa Angelina, Sao Carlos 13563-120, Brazil
| | - Glaucius Oliva
- Sao
Carlos Institute of Physics, University
of Sao Paulo, Av. Joao
Dagnone, 1100—Jardim Santa Angelina, Sao Carlos 13563-120, Brazil
| | - Sean Ekins
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
9
|
Wang GQ, Ren XX, Wei JJ, Wang AJ, Zhao T, Feng JJ, Yun Cheang T. Ultrasensitive PEC cytosensor for breast cancer cells detection and inhibitor screening based on plum-branched CdS/Bi 2S 3 heterostructures. Bioelectrochemistry 2023; 152:108442. [PMID: 37060704 DOI: 10.1016/j.bioelechem.2023.108442] [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: 02/17/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/17/2023]
Abstract
Breast cancer is the most common malignant tumor in women, which seriously threatens the life and health of patients. Therefore, facile and sensitive detection of human breast cancer cells is crucial for cancer diagnosis. In this work, plum-branched CdS/Bi2S3 heterostructures (CdS/Bi2S3 HSs) were synthesized under hydrothermal condition, whose photoelectrochemical (PEC) property and biocompatibility were scrutinously investigated. In parallel, a signal amplification strategy was designed based on immune recognition between epidermal growth factor receptor (EGFR) overexpressed on membrane of breast cancer cells MDA-MB-231 and its aptamer. Integration of the above together, a highly sensitive PEC cytosensor was developed for analysis of target MDA-MB-231 cells, exhibiting a wider linear range of 1 × 102 ∼ 3 × 105 cells mL-1 with a limit of detection (LOD) down to 6 cells mL-1 (S/N = 3). Further, the biosensor was explored for anticancer drug (e.g., dacomitinib) screening by monitoring the variations in the PEC signals of the expressed EGFR upon drug stimulation. The obtained CdS/Bi2S3 HSs are identified as promising and feasible photoactive material for determination of cancer cells and drug screening in clinic and related research.
Collapse
Affiliation(s)
- Gui-Qing Wang
- Department of Breast Care Centre, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China; College of Geography and Environmental Sciences, College of Life Science, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua 321004, China
| | - Xin-Xin Ren
- Department of Breast Care Centre, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China; College of Geography and Environmental Sciences, College of Life Science, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua 321004, China
| | - Jing-Jing Wei
- College of Geography and Environmental Sciences, College of Life Science, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua 321004, China
| | - Ai-Jun Wang
- College of Geography and Environmental Sciences, College of Life Science, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua 321004, China
| | - Tiejun Zhao
- College of Geography and Environmental Sciences, College of Life Science, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua 321004, China; School of Medicine, Hangzhou City University, Hangzhou 310015, China.
| | - Jiu-Ju Feng
- College of Geography and Environmental Sciences, College of Life Science, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua 321004, China.
| | - Tuck Yun Cheang
- Department of Breast Care Centre, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China.
| |
Collapse
|
10
|
Fan Y, Li W, Nie W, Yao H, Ren Y, Wang M, Nie H, Gu C, Liu J, An B. Novel Dual-Target Kinase Inhibitors of EGFR and ALK Were Designed, Synthesized, and Induced Cell Apoptosis in Non-Small Cell Lung Cancer. Molecules 2023; 28:molecules28052006. [PMID: 36903251 PMCID: PMC10004195 DOI: 10.3390/molecules28052006] [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: 01/19/2023] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
ALK-positive NSCLC coexisting with EGFR mutations is a frequently occurring clinical phenomenon. Targeting ALK and EGFR simultaneously may be an effective way to treat these cancer patients. In this study, we designed and synthesized ten new dual-target EGFR/ALK inhibitors. Among them, the optimal compound 9j exhibited good activity with IC50 values of 0.07829 ± 0.03 μM and 0.08183 ± 0.02 μM against H1975 (EGFR T790M/L858R) and H2228 (EML4-ALK) cells, respectively. Immunofluorescence assays indicated that the compound could simultaneously inhibit the expression of phosphorylated EGFR and ALK proteins. A kinase assay demonstrated that compound 9j could inhibit both EGFR and ALK kinases; thus, exerting an antitumor effect. Additionally, compound 9j induced apoptosis in a dose-dependent manner and inhibited the invasion and migration of tumor cells. All of these results indicate that 9j is worthy of further study.
Collapse
Affiliation(s)
- Yangyang Fan
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Wei Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wenyan Nie
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Han Yao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuanyuan Ren
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Mengxuan Wang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Haoran Nie
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Chenxi Gu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiadai Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Baijiao An
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Yantai 264003, China
- Correspondence:
| |
Collapse
|
11
|
Bruggemann L, Falls Z, Mangione W, Schwartz SA, Battaglia S, Aalinkeel R, Mahajan SD, Samudrala R. Multiscale Analysis and Validation of Effective Drug Combinations Targeting Driver KRAS Mutations in Non-Small Cell Lung Cancer. Int J Mol Sci 2023; 24:ijms24020997. [PMID: 36674513 PMCID: PMC9867122 DOI: 10.3390/ijms24020997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 01/06/2023] Open
Abstract
Pharmacogenomics is a rapidly growing field with the goal of providing personalized care to every patient. Previously, we developed the Computational Analysis of Novel Drug Opportunities (CANDO) platform for multiscale therapeutic discovery to screen optimal compounds for any indication/disease by performing analytics on their interactions using large protein libraries. We implemented a comprehensive precision medicine drug discovery pipeline within the CANDO platform to determine which drugs are most likely to be effective against mutant phenotypes of non-small cell lung cancer (NSCLC) based on the supposition that drugs with similar interaction profiles (or signatures) will have similar behavior and therefore show synergistic effects. CANDO predicted that osimertinib, an EGFR inhibitor, is most likely to synergize with four KRAS inhibitors.Validation studies with cellular toxicity assays confirmed that osimertinib in combination with ARS-1620, a KRAS G12C inhibitor, and BAY-293, a pan-KRAS inhibitor, showed a synergistic effect on decreasing cellular proliferation by acting on mutant KRAS. Gene expression studies revealed that MAPK expression is strongly correlated with decreased cellular proliferation following treatment with KRAS inhibitor BAY-293, but not treatment with ARS-1620 or osimertinib. These results indicate that our precision medicine pipeline may be used to identify compounds capable of synergizing with inhibitors of KRAS G12C, and to assess their likelihood of becoming drugs by understanding their behavior at the proteomic/interactomic scales.
Collapse
Affiliation(s)
- Liana Bruggemann
- Department of Biomedical Informatics, University at Buffalo, Buffalo, NY 14260, USA
| | - Zackary Falls
- Department of Biomedical Informatics, University at Buffalo, Buffalo, NY 14260, USA
| | - William Mangione
- Department of Biomedical Informatics, University at Buffalo, Buffalo, NY 14260, USA
| | | | | | | | - Supriya D. Mahajan
- Department of Medicine, University at Buffalo, Buffalo, NY 14260, USA
- Correspondence: (S.D.M.); (R.S.)
| | - Ram Samudrala
- Department of Biomedical Informatics, University at Buffalo, Buffalo, NY 14260, USA
- Correspondence: (S.D.M.); (R.S.)
| |
Collapse
|
12
|
Van De Stadt E, Yaqub M, Jahangir AA, Hendrikse H, Bahce I. Radiolabeled EGFR TKI as predictive imaging biomarkers in NSCLC patients – an overview. Front Oncol 2022; 12:900450. [PMID: 36313723 PMCID: PMC9597357 DOI: 10.3389/fonc.2022.900450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/28/2022] [Indexed: 12/03/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) has one of the highest cancer-related mortality rates worldwide. In a subgroup of NSCLC, tumor growth is driven by epidermal growth factor receptors (EGFR) that harbor an activating mutation. These patients are best treated with EGFR tyrosine kinase inhibitors (EGFR TKI). Identifying the EGFR mutational status on a tumor biopsy or a liquid biopsy using tumor DNA sequencing techniques is the current approach to predict tumor response on EGFR TKI therapy. However, due to difficulty in reaching tumor sites, and varying inter- and intralesional tumor heterogeneity, biopsies are not always possible or representative of all tumor lesions, highlighting the need for alternative biomarkers that predict tumor response. Positron emission tomography (PET) studies using EGFR TKI-based tracers have shown that EGFR mutational status could be identified, and that tracer uptake could potentially be used as a biomarker for tumor response. However, despite their likely predictive and monitoring value, the EGFR TKI-PET biomarkers are not yet qualified to be used in the routine clinical practice. In this review, we will discuss the currently investigated EGFR-directed PET biomarkers, elaborate on the typical biomarker development process, and describe how the advances, challenges, and opportunities of EGFR PET biomarkers relate to this process on their way to qualification for routine clinical practice.
Collapse
Affiliation(s)
- Eveline Van De Stadt
- Department of Pulmonology, Amsterdam University Medical Centers (UMC), VU University Medical Center, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC), Amsterdam, Netherlands
- *Correspondence: Eveline Van De Stadt,
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), VU University Medical Center, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC), Amsterdam, Netherlands
| | - A. A. Jahangir
- Department of Pulmonology, Amsterdam University Medical Centers (UMC), VU University Medical Center, Amsterdam, Netherlands
| | - Harry Hendrikse
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers (UMC), VU University Medical Center, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC), Amsterdam, Netherlands
| | - Idris Bahce
- Department of Pulmonology, Amsterdam University Medical Centers (UMC), VU University Medical Center, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam University Medical Centers (UMC), Amsterdam, Netherlands
| |
Collapse
|
13
|
Min HY, Lee HY. Molecular targeted therapy for anticancer treatment. Exp Mol Med 2022; 54:1670-1694. [PMID: 36224343 PMCID: PMC9636149 DOI: 10.1038/s12276-022-00864-3] [Citation(s) in RCA: 177] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 02/07/2023] Open
Abstract
Since the initial clinical approval in the late 1990s and remarkable anticancer effects for certain types of cancer, molecular targeted therapy utilizing small molecule agents or therapeutic monoclonal antibodies acting as signal transduction inhibitors has served as a fundamental backbone in precision medicine for cancer treatment. These approaches are now used clinically as first-line therapy for various types of human cancers. Compared to conventional chemotherapy, targeted therapeutic agents have efficient anticancer effects with fewer side effects. However, the emergence of drug resistance is a major drawback of molecular targeted therapy, and several strategies have been attempted to improve therapeutic efficacy by overcoming such resistance. Herein, we summarize current knowledge regarding several targeted therapeutic agents, including classification, a brief biology of target kinases, mechanisms of action, examples of clinically used targeted therapy, and perspectives for future development.
Collapse
Affiliation(s)
- Hye-Young Min
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ho-Young Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Zhou T, Xiong Q, Hong C, Wang Q, Wang W, Xu C, Cai J. A novel EGFR exon 21 indel mutation in lung adenocarcinoma and response to dacomitinib: A case report. Medicine (Baltimore) 2022; 101:e30269. [PMID: 36042660 PMCID: PMC9410616 DOI: 10.1097/md.0000000000030269] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
RATIONALE Uncommon epidermal growth factor receptor (EGFR) mutations are increasingly being identified in non-small cell lung cancer. Insertion and deletion mutations have been detected in exons 18, 19, and 20, but not in exon 21. In patients with uncommon mutations, the second-generation EGFR tyrosine kinase inhibitor afatinib has shown good efficacy, whereas that of dacomitinib, another second-generation EGFR-tyrosine kinase inhibitor, remains unknown. Here, we reported a patient with a novel EGFR21 exon insertion-deletion (indel) mutation and demonstrated the efficacy of dacomitinib. PATIENT CONCERNS A 59-year-old nonsmoking Chinese male was admitted to the hospital with lung cancer after a chest computed tomography for coughing and sputum. The patient's condition progressed after multiple treatments including surgery, chemotherapy, and radiotherapy. DIAGNOSIS The patient had clinical manifestations of cough and sputum and was pathologically confirmed to have T2bN1M0 (stage IIB) lung adenocarcinoma according to the seventh edition of tumor-node-metastasis staging. The patient underwent a second operation after detection of recurrence, and postoperative pathology confirmed adenocarcinoma of the lung. The patient progressed again after surgery, and the tumor-node-metastasis stage was changed to T4N0M1a (stage IVA) before treatment with dacomitinib. INTERVENTIONS After detection of the EGFR exon 21 indel mutation, the patient began treatment with dacomitinib (45 mg once a day) on March 12, 2021. OUTCOMES After 1 month of targeted therapy, the patient showed a partial response to dacomitinib. As of March 19, 2022, his condition remained stable and he continued to receive dacomitinib. Progression-free survival reached 12.4 months. The patient experienced mild adverse reactions of pruritus during the use of dacomitinib, but recovered after drug treatment. LESSON We reported a novel EGFR exon 21 indel mutation in a lung adenocarcinoma patient. Dacomitinib showed efficacy in the treatment of a patient with this mutation, suggesting that its efficacy in patients with uncommon mutations should be explored further. The next-generation sequencing is recommended as a guiding tool for the treatment of advanced non-small cell lung cancer.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qiang Xiong
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Molecular Center, Nanchang, China
| | - Chen Hong
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qian Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Wenxian Wang
- Department of Medical Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Chunwei Xu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jing Cai
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Molecular Center, Nanchang, China
- *Correspondence: Jing Cai, Department of Oncology, Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi Province 330006, China (e-mail: )
| |
Collapse
|
15
|
Abstract
EGFR is a member of the ERBB family. It plays a significant role in cellular processes such as growth, survival and differentiation via the activation of various signaling pathways. EGFR deregulation is implicated in various human malignancies, and therefore EGFR has emerged as an attractive anticancer target. EGFR inhibition using strategies such as tyrosine kinase inhibitors and monoclonal antibodies hinders cellular proliferation and promotes apoptosis in cancer cells in vitro and in vivo. EGFR inhibition by tyrosine kinase inhibitors has been shown to be a better treatment option than chemotherapy for advanced-stage EGFR-driven non-small-cell lung cancer, yet de novo and acquired resistance limits the clinical benefit of these therapeutic molecules. This review discusses the cellular signaling pathways activated by EGFR. Further, current therapeutic strategies to target aberrant EGFR signaling in cancer and mechanisms of resistance to them are highlighted.
Collapse
|
16
|
Guo Y, Gao B, Gao P, Fang L, Gou S. Novel anilinopyrimidine derivatives as potential EGFR T790M/C797S Inhibitors: Design, Synthesis, biological activity study. Bioorg Med Chem 2022; 70:116907. [PMID: 35810715 DOI: 10.1016/j.bmc.2022.116907] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/25/2022]
Abstract
EGFRT790M/C797S is an important target for the development of new generation of EGFR kinase inhibitors without drug resistance. In this work, a series of anilinopyrimidine derivatives that targeting EGFRT790M/C797S were designed, synthesized, and evaluated in vitro for the inhibitory effect on triple mutations kinases and cell lines. Based on the pharmacology data, the anilinopyrimidine derivatives showed high inhibitory activity on triple mutations kinases (EGFRdel 18/T790M/C797S and EGFRL858R/T790M/C797S) as well as the cell line Ba/F3 with highly expression of EGFRdel 18/T790M/C797S. In addition, the anilinopyrimidine derivatives had a more than 50-fold selectivity towards EGFRdel 18/T790M/C797S as compared with EGFRWT. In vivo antitumor activity test also indicated that 8j had good pharmacokinetic parameters, low toxicity and better inhibitory activity. Overall, the anilinopyrimidine derivatives could be regarded as promising candidates for the further development of novel EGFRT790M/C797S inhibitors for clinical application.
Collapse
Affiliation(s)
- Yanliang Guo
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China; Jiangsu Hansoh Pharmaceutical Group CO., LTD., Lianyungang 222000, China
| | - Biao Gao
- Jiangsu Hansoh Pharmaceutical Group CO., LTD., Lianyungang 222000, China
| | - Peng Gao
- Jiangsu Hansoh Pharmaceutical Group CO., LTD., Lianyungang 222000, China
| | - Lei Fang
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| | - Shaohua Gou
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| |
Collapse
|
17
|
Li HS, Yang GJ, Cai Y, Li JL, Xu HY, Zhang T, Zhou LQ, Wang YY, Wang JL, Hu XS, Yan X, Wang Y. Dacomitinib for Advanced Non-small Cell Lung Cancer Patients Harboring Major Uncommon EGFR Alterations: A Dual-Center, Single-Arm, Ambispective Cohort Study in China. Front Pharmacol 2022; 13:919652. [PMID: 35770100 PMCID: PMC9234690 DOI: 10.3389/fphar.2022.919652] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/18/2022] [Indexed: 11/20/2022] Open
Abstract
Objective: Dacomitinib has been approved for non-small-cell lung cancer (NSCLC) patients harboring classical epidermal growth factor receptor (EGFR) mutations; however, clinical evidence of its activity on major uncommon EGFR mutations is currently limited. Materials and methods: This was a dual-center, single-arm, ambispective cohort study in China. Patients with histologically confirmed metastatic or recurrent NSCLC harboring major uncommon EGFR mutations were eligible for the study. The objective response rate and disease control rate were determined by RECIST 1.1 every 1–2 months. Adverse events were assessed by CTCAE 5.0. Results: In total, 32 NSCLC patients were enrolled between July 2020 and January 2022, and 18 (56.3%) patients received dacomitinib as first-line therapy. Median age was 64 years, and 20 (62.5%) were female. The mutations identified were G719X (n = 24; 75%), followed by L861X (n = 10; 31.3%), and S768I (n = 8; 25%). In the first-line setting, 72.2% of patients (13/18) had a confirmed partial response and 100% (18/18) had disease control, and the median progression-free survival (PFS) and overall survival (OS) were unreached. In the whole cohort, 56.3% of patients (18/32) had a confirmed partial response and 90.6% (29/32) had disease control, and the median PFS was 10.3 months (95% confidence interval, 6.1–14.5) and the median OS was 36.5 months. Except for one case not available for brain re-evaluation, control of the intracranial metastases was observed in 13 patients (13/14, 92.9%). No grade 4–5 adverse events (AEs) occurred, but all patients had grade 1–2 AEs, and 12.5% (4/32) patients required a dosage reduction due to intolerable AEs. Conclusions: Dacomitinib demonstrated favorable activity with manageable toxicity in patients with NSCLC harboring major uncommon EGFR mutations.
Collapse
Affiliation(s)
- Hong-Shuai Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guang-Jian Yang
- Department of Respiratory Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, China
| | - Yi Cai
- Independent Researcher, Ellicott City, MD, United States
| | - Jun-Ling Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hai-Yan Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Qiang Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-Ying Wang
- Department of Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jin-Liang Wang
- Department of Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xing-Sheng Hu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiang Yan
- Department of Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Xiang Yan, ; Yan Wang,
| | - Yan Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Xiang Yan, ; Yan Wang,
| |
Collapse
|
18
|
Wang C, Wang X, Huang Z, Wang T, Nie Y, Yang S, Xiang R, Fan Y. Discovery and structural optimization of potent epidermal growth factor receptor (EGFR) inhibitors against L858R/T790M/C797S resistance mutation for lung cancer treatment. Eur J Med Chem 2022; 237:114381. [DOI: 10.1016/j.ejmech.2022.114381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/18/2022] [Accepted: 04/09/2022] [Indexed: 12/01/2022]
|
19
|
Borsari C, Keles E, McPhail JA, Schaefer A, Sriramaratnam R, Goch W, Schaefer T, De Pascale M, Bal W, Gstaiger M, Burke JE, Wymann MP. Covalent Proximity Scanning of a Distal Cysteine to Target PI3Kα. J Am Chem Soc 2022; 144:6326-6342. [PMID: 35353516 PMCID: PMC9011356 DOI: 10.1021/jacs.1c13568] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
Covalent protein
kinase inhibitors exploit currently noncatalytic
cysteines in the adenosine 5′-triphosphate (ATP)-binding site
via electrophiles directly appended to a reversible-inhibitor scaffold.
Here, we delineate a path to target solvent-exposed cysteines at a
distance >10 Å from an ATP-site-directed core module and produce
potent covalent phosphoinositide 3-kinase α (PI3Kα) inhibitors.
First, reactive warheads are used to reach out to Cys862 on PI3Kα,
and second, enones are replaced with druglike warheads while linkers
are optimized. The systematic investigation of intrinsic warhead reactivity
(kchem), rate of covalent bond formation
and proximity (kinact and reaction space
volume Vr), and integration of structure
data, kinetic and structural modeling, led to the guided identification
of high-quality, covalent chemical probes. A novel stochastic approach
provided direct access to the calculation of overall reaction rates
as a function of kchem, kinact, Ki, and Vr, which was validated with compounds with varied linker
lengths. X-ray crystallography, protein mass spectrometry (MS), and
NanoBRET assays confirmed covalent bond formation of the acrylamide
warhead and Cys862. In rat liver microsomes, compounds 19 and 22 outperformed the rapidly metabolized CNX-1351,
the only known PI3Kα irreversible inhibitor. Washout experiments
in cancer cell lines with mutated, constitutively activated PI3Kα
showed a long-lasting inhibition of PI3Kα. In SKOV3 cells, compounds 19 and 22 revealed PI3Kβ-dependent signaling,
which was sensitive to TGX221. Compounds 19 and 22 thus qualify as specific chemical probes to explore PI3Kα-selective
signaling branches. The proposed approach is generally suited to develop
covalent tools targeting distal, unexplored Cys residues in biologically
active enzymes.
Collapse
Affiliation(s)
- Chiara Borsari
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Erhan Keles
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Jacob A McPhail
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
| | - Alexander Schaefer
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland
| | - Rohitha Sriramaratnam
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Wojciech Goch
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Thorsten Schaefer
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Martina De Pascale
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Wojciech Bal
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland
| | - John E Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
| | - Matthias P Wymann
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| |
Collapse
|
20
|
Du RL, Sun N, Fung YH, Zheng YY, Chen YW, Chan PH, Wong WL, Wong KY. Discovery of FtsZ inhibitors by virtual screening as antibacterial agents and study of the inhibition mechanism. RSC Med Chem 2022; 13:79-89. [PMID: 35224498 PMCID: PMC8792978 DOI: 10.1039/d1md00249j] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/07/2021] [Indexed: 09/29/2023] Open
Abstract
Inhibition of bacterial cell division is a novel mechanistic action in the development of new antimicrobial agents. The FtsZ protein is an important antimicrobial drug target because of its essential role in bacterial cell division. In the present study, potential inhibitors of FtsZ were identified by virtual screening followed by in vivo and in vitro bioassays. One of the candidates, Dacomitinib (S2727), shows for the first time its potent inhibitory activity against the MRSA strains. The binding mode of Dacomitinib in FtsZ was analyzed by docking, and Asp199 and Thr265 are thought to be essential residues involved in the interactions.
Collapse
Affiliation(s)
- Ruo-Lan Du
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Ning Sun
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Yik-Hong Fung
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Yuan-Yuan Zheng
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Yu-Wei Chen
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Pak-Ho Chan
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| | - Kwok-Yin Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University Hunghom Kowloon Hong Kong P.R. China
| |
Collapse
|
21
|
Overhoff B, Falls Z, Mangione W, Samudrala R. A Deep-Learning Proteomic-Scale Approach for Drug Design. Pharmaceuticals (Basel) 2021; 14:1277. [PMID: 34959678 PMCID: PMC8709297 DOI: 10.3390/ph14121277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 12/26/2022] Open
Abstract
Computational approaches have accelerated novel therapeutic discovery in recent decades. The Computational Analysis of Novel Drug Opportunities (CANDO) platform for shotgun multitarget therapeutic discovery, repurposing, and design aims to improve their efficacy and safety by employing a holistic approach that computes interaction signatures between every drug/compound and a large library of non-redundant protein structures corresponding to the human proteome fold space. These signatures are compared and analyzed to determine if a given drug/compound is efficacious and safe for a given indication/disease. In this study, we used a deep learning-based autoencoder to first reduce the dimensionality of CANDO-computed drug-proteome interaction signatures. We then employed a reduced conditional variational autoencoder to generate novel drug-like compounds when given a target encoded "objective" signature. Using this approach, we designed compounds to recreate the interaction signatures for twenty approved and experimental drugs and showed that 16/20 designed compounds were predicted to be significantly (p-value ≤ 0.05) more behaviorally similar relative to all corresponding controls, and 20/20 were predicted to be more behaviorally similar relative to a random control. We further observed that redesigns of objectives developed via rational drug design performed significantly better than those derived from natural sources (p-value ≤ 0.05), suggesting that the model learned an abstraction of rational drug design. We also show that the designed compounds are structurally diverse and synthetically feasible when compared to their respective objective drugs despite consistently high predicted behavioral similarity. Finally, we generated new designs that enhanced thirteen drugs/compounds associated with non-small cell lung cancer and anti-aging properties using their predicted proteomic interaction signatures. his study represents a significant step forward in automating holistic therapeutic design with machine learning, enabling the rapid generation of novel, effective, and safe drug leads for any indication.
Collapse
Affiliation(s)
| | | | | | - Ram Samudrala
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; (B.O.); (Z.F.); (W.M.)
| |
Collapse
|
22
|
Ferlenghi F, Scalvini L, Vacondio F, Castelli R, Bozza N, Marseglia G, Rivara S, Lodola A, La Monica S, Minari R, Petronini PG, Alfieri R, Tiseo M, Mor M. A sulfonyl fluoride derivative inhibits EGFR L858R/T790M/C797S by covalent modification of the catalytic lysine. Eur J Med Chem 2021; 225:113786. [PMID: 34464874 DOI: 10.1016/j.ejmech.2021.113786] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/13/2021] [Accepted: 08/15/2021] [Indexed: 10/20/2022]
Abstract
The emergence of the C797S mutation in EGFR is a frequent mechanism of resistance to osimertinib in the treatment of non-small cell lung cancer (NSCLC). In the present work, we report the design, synthesis and biochemical characterization of UPR1444 (compound 11), a new sulfonyl fluoride derivative which potently and irreversibly inhibits EGFRL858R/T790M/C797S through the formation of a sulfonamide bond with the catalytic residue Lys745. Enzymatic assays show that compound 11 displayed an inhibitory activity on EGFRWT comparable to that of osimertinib, and it resulted more selective than the sulfonyl fluoride probe XO44, recently reported to inhibit a significant part of the kinome. Neither compound 11 nor XO44 inhibited EGFRdel19/T790M/C797S triple mutant. When tested in Ba/F3 cells expressing EGFRL858R/T790M/C797S, compound 11 resulted significantly more potent than osimertinib at inhibiting both EGFR autophosphorylation and proliferation, even if the inhibition of EGFR autophosphorylation by compound 11 in Ba/F3 cells was not long lasting.
Collapse
Affiliation(s)
| | - Laura Scalvini
- Department of Food and Drug, University of Parma, Parma, Italy
| | | | | | - Nicole Bozza
- Department of Food and Drug, University of Parma, Parma, Italy
| | | | - Silvia Rivara
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Alessio Lodola
- Department of Food and Drug, University of Parma, Parma, Italy.
| | - Silvia La Monica
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Roberta Minari
- Medical Oncology, University Hospital of Parma, Parma, Italy
| | | | - Roberta Alfieri
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology, University Hospital of Parma, Parma, Italy
| | - Marco Mor
- Department of Food and Drug, University of Parma, Parma, Italy
| |
Collapse
|
23
|
Papini F, Sundaresan J, Leonetti A, Tiseo M, Rolfo C, Peters GJ, Giovannetti E. Hype or hope - Can combination therapies with third-generation EGFR-TKIs help overcome acquired resistance and improve outcomes in EGFR-mutant advanced/metastatic NSCLC? Crit Rev Oncol Hematol 2021; 166:103454. [PMID: 34455092 DOI: 10.1016/j.critrevonc.2021.103454] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/07/2021] [Accepted: 08/10/2021] [Indexed: 02/08/2023] Open
Abstract
Three generations of epidermal growth factor receptor - tyrosine kinase inhibitors (EGFR-TKIs) have been developed for treating advanced/metastatic non-small cell lung cancer (NSCLC) patients harboring EGFR-activating mutations, while a fourth generation is undergoing preclinical assessment. Although initially effective, acquired resistance to EGFR-TKIs usually arises within a year due to the emergence of clones harboring multiple resistance mechanisms. Therefore, the combination of EGFR-TKIs with other therapeutic agents has emerged as a potential strategy to overcome resistance and improve clinical outcomes. However, results obtained so far are ambiguous and ideal therapies for patients who experience disease progression during treatment with EGFR-TKIs remain elusive. This review provides an updated landscape of EGFR-TKIs, along with a description of the mechanisms causing resistance to these drugs. Moreover, it discusses the current knowledge, limitations, and future perspective regarding the use of EGFR-TKIs in combination with other anticancer agents, supporting the need for bench-to-bedside approaches in selected populations.
Collapse
Affiliation(s)
- Filippo Papini
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, the Netherlands; Fondazione Pisana per la Scienza, Pisa, Italy
| | - Janani Sundaresan
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Alessandro Leonetti
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Christian Rolfo
- The Center of Thoracic Oncology at the Tisch Cancer Institute, Mount Sinai, NYC, United States
| | - Godefridus J Peters
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Biochemistry, Medical University of Gdansk, Poland
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, VU University, Cancer Center Amsterdam, Amsterdam, the Netherlands; Fondazione Pisana per la Scienza, Pisa, Italy.
| |
Collapse
|
24
|
Van Luan P, Tien ND, Hai NM, Tien ND, Duyen TT. Real-world analysis of the effect of gefitinib as a first-line therapy in patients with advanced non-small cell lung cancer with EGFR mutations. Ther Adv Med Oncol 2021; 13:1758835921992977. [PMID: 33680095 PMCID: PMC7900790 DOI: 10.1177/1758835921992977] [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: 09/12/2020] [Accepted: 01/15/2021] [Indexed: 11/25/2022] Open
Abstract
Background: There have been few studies on the efficacy of tyrosine kinase inhibitors in
lung carcinomas. The purpose of this study was to evaluate the effect of
gefitinib as a first-line therapy in patients with advanced non-small cell
lung cancer (NSCLC) who were positive for epidermal growth factor receptor
(EGFR) mutations. Methods: This prospective analysis included 120 patients with advanced NSCLC with
EGFR mutations who were administered gefitinib as the
first-line therapy. Patient follow-up and evaluation were performed every
3 months or when there were symptoms of progressive disease. The main
criteria for the analysis of response were progression-free survival (PFS)
and overall response rate (ORR). The secondary criteria were overall
survival (OS) and disease control rate (DCR). In addition, the relationship
of OS with sex, smoking history, and performance status (PS), as well as
gefitinib toxicity were analyzed. Results: The ORR and DCR were 59.2% and 95.8%, respectively. The median PFS was
14.5 months and the median OS was 33 months. The longer OS was statistically
significant in women and non-smokers, and the patients had a good PS.
Adverse events occurred in 59.2% patients, but most of them were grade 1 and
2 events. Conclusion: This study conducted in Vietnam suggests the effectiveness of gefitinib as a
first-line treatment option in patients with advanced NSCLC and positive
EGFR mutations regardless of whether the patients have
a good PS or not. In particular, targeted therapy with gefitinib improved
the OS in women and non-smokers.
Collapse
Affiliation(s)
- Pham Van Luan
- Department of Respiratory Medicine, 108 Military Central Hospital, Tran Hung Dao Street, Hai Ba Trung, Hanoi, Vietnam
| | - Nguyen Dinh Tien
- Department of Respiratory Medicine, 108 Military Central Hospital, Hai Ba Trung, Hanoi, Vietnam
| | - Nguyen Minh Hai
- Department of Respiratory Medicine, 108 Military Central Hospital, Hai Ba Trung, Hanoi, Vietnam
| | - Nguyen Dao Tien
- Department of Respiratory Medicine, 108 Military Central Hospital, Hai Ba Trung, Hanoi, Vietnam
| | - Thi Thi Duyen
- Department of Respiratory Medicine, 108 Military Central Hospital, Hai Ba Trung, Hanoi, Vietnam
| |
Collapse
|
25
|
Kabir ML, Backler F, Clayton AHA, Wang F. Deducing the Conformational Properties of a Tyrosine Kinase Inhibitor in Solution by Optical Spectroscopy and Computational Chemistry. Front Chem 2020; 8:596. [PMID: 32850633 PMCID: PMC7399232 DOI: 10.3389/fchem.2020.00596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/08/2020] [Indexed: 01/03/2023] Open
Abstract
Dacomitinib (PF-00299804) was recently approved by the Food and Drug Administration (FDA) as a tyrosine kinase inhibitor (TKI). Unfortunately, side effects and disease resistance eventually result from its use. Off-target effects in some kinase inhibitors have arisen from drug conformational plasticity; however, the conformational states of Dacomitinib in solution are presently unknown. To fill this gap, we have used computational chemistry to explore optimized molecular geometry, properties, and ultraviolet-visible (UV-Vis) absorption spectra of Dacomitinib in dimethyl sulfoxide (DMSO) solution. Potential energy scans led to the discovery of two planar and two twisted conformers of Dacomitinib. The simulated UV-Vis spectral signatures of the planar conformers reproduced the two experimental spectral bands at 275 and 343 nm in solution. It was further discovered that Dacomitinib forms conformers through its three flexible linkers of two C-NH-C bridges, which control the orientations of the 3-chloro-4-fluoroaniline ring (Ring C) and the quinazoline ring (Rings A and B) and the 4-piperidin-1-yl-buten-2-nal side chain, and one C-O-C local bridge which controls the methoxy group locally. When in isolation, these flexible linkers form close hexagon and pentagon loops through strong intramolecular hydrogen bonding so that the "planar" conformers Daco-P1 and Daco-P2 are more stable in isolation. Such flexibility of the ligand and its ability to dock and bind with protein also depend on their interaction with the environment, in addition to their energy and spectra in isolation. However, an accurate quantum mechanical study on drug/ligand conformers in isolation provides necessary reference information for the ability to form a complex with proteins.
Collapse
Affiliation(s)
- Md. Lutful Kabir
- Department of Physics and Astronomy, Optical Sciences Centre, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Frederick Backler
- Department of Chemistry and Biotechnology, Centre for Translatonal Atomaterials, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Andrew H. A. Clayton
- Department of Physics and Astronomy, Optical Sciences Centre, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Feng Wang
- Department of Chemistry and Biotechnology, Centre for Translatonal Atomaterials, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, VIC, Australia
| |
Collapse
|