1
|
Lee TW, Singleton DC, Harms JK, Lu M, McManaway SP, Lai A, Tercel M, Pruijn FB, Macann AMJ, Hunter FW, Wilson WR, Jamieson SMF. Clinical relevance and therapeutic predictive ability of hypoxia biomarkers in head and neck cancer tumour models. Mol Oncol 2024; 18:1885-1903. [PMID: 38426642 PMCID: PMC11306523 DOI: 10.1002/1878-0261.13620] [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: 10/02/2023] [Revised: 12/20/2023] [Accepted: 02/19/2024] [Indexed: 03/02/2024] Open
Abstract
Tumour hypoxia promotes poor patient outcomes, with particularly strong evidence for head and neck squamous cell carcinoma (HNSCC). To effectively target hypoxia, therapies require selection biomarkers and preclinical models that can accurately model tumour hypoxia. We established 20 patient-derived xenograft (PDX) and cell line-derived xenograft (CDX) models of HNSCC that we characterised for their fidelity to represent clinical HNSCC in gene expression, hypoxia status and proliferation and that were evaluated for their sensitivity to hypoxia-activated prodrugs (HAPs). PDX models showed greater fidelity in gene expression to clinical HNSCC than cell lines, as did CDX models relative to their paired cell lines. PDX models were significantly more hypoxic than CDX models, as assessed by hypoxia gene signatures and pimonidazole immunohistochemistry, and showed similar hypoxia gene expression to clinical HNSCC tumours. Hypoxia or proliferation status alone could not determine HAP sensitivity across our 20 HNSCC and two non-HNSCC tumour models by either tumour growth inhibition or killing of hypoxia cells in an ex vivo clonogenic assay. In summary, our tumour models provide clinically relevant HNSCC models that are suitable for evaluating hypoxia-targeting therapies; however, additional biomarkers to hypoxia are required to accurately predict drug sensitivity.
Collapse
Affiliation(s)
- Tet Woo Lee
- Auckland Cancer Society Research CentreUniversity of AucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of AucklandNew Zealand
| | - Dean C. Singleton
- Auckland Cancer Society Research CentreUniversity of AucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of AucklandNew Zealand
- Department of Molecular Medicine and PathologyUniversity of AucklandNew Zealand
| | - Julia K. Harms
- Auckland Cancer Society Research CentreUniversity of AucklandNew Zealand
| | - Man Lu
- Auckland Cancer Society Research CentreUniversity of AucklandNew Zealand
| | - Sarah P. McManaway
- Auckland Cancer Society Research CentreUniversity of AucklandNew Zealand
| | - Amy Lai
- Auckland Cancer Society Research CentreUniversity of AucklandNew Zealand
- Department of Pharmacology and Clinical PharmacologyUniversity of AucklandNew Zealand
| | - Moana Tercel
- Auckland Cancer Society Research CentreUniversity of AucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of AucklandNew Zealand
| | - Frederik B. Pruijn
- Auckland Cancer Society Research CentreUniversity of AucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of AucklandNew Zealand
| | | | - Francis W. Hunter
- Auckland Cancer Society Research CentreUniversity of AucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of AucklandNew Zealand
- Oncology Therapeutic AreaJanssen Research and DevelopmentSpring HousePAUSA
| | - William R. Wilson
- Auckland Cancer Society Research CentreUniversity of AucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of AucklandNew Zealand
| | - Stephen M. F. Jamieson
- Auckland Cancer Society Research CentreUniversity of AucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of AucklandNew Zealand
- Department of Pharmacology and Clinical PharmacologyUniversity of AucklandNew Zealand
| |
Collapse
|
2
|
Yasser N, Sroor FM, El-Shorbagy HM, Eissa SM, Hassaneen HM, Abdelhamid IA. Synthesis, anticancer evaluation of novel hybrid pyrazole-based chalcones, molecular docking, DNA fragmentation, and gene expression: in vitro studies. RSC Adv 2024; 14:21859-21873. [PMID: 38984258 PMCID: PMC11232109 DOI: 10.1039/d4ra03375b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024] Open
Abstract
A unique series of pyrazolyl-chalcone derivatives was synthesized via the method of Claisen-Schmidt condensation. The desired chalcone derivatives 7a-d and 9a-f were obtained in good yields by reacting the 4-acetyl-5-thiophene-pyrazole with the appropriate heteroaryl aldehyde derivatives. The novel chalcones have undergone complete elemental analysis, 1H-NMR, 13C-NMR, mass spectrometry, and IR characterization. The three human cancer cell lines MCF7 (human Caucasian breast adenocarcinoma), PC3 (prostatic cancer) and PACA2 (pancreatic carcinoma) as well as the normal cell line BJ1 (normal skin fibroblasts) were tested in vitro for the anti-cancer properties of the newly synthesized chalcone derivatives. When compared to the reference medicine doxorubicin (IC50 = 52.1 μM), compound 9e showed the most promise derivative (IC50 = 27.6 μM) against PACA2 cells, while compound 7d demonstrated anticancer efficacy (IC50 = 42.6 μM against MCF7 cells compared to the reference drug doxorubicin (IC50 = 48 μM). Using breast and pancreatic cell lines, the gene expression, DNA damage, and DNA fragmentation percentages for compounds 7d and 9e were evaluated. Moreover, the molecular docking study of compounds 7d and 9e was assessed. The binding affinities of compound 9e toward P53 mutant Y220C was -22 kcal per mole, while those of compound 7d towards Bcl2 and CDK4 were -27.81 and -26.9 kcal per mole, respectively, compared to the standard values (-15.82, -33.96 and -29.9 kcal per mole).
Collapse
Affiliation(s)
- Norhan Yasser
- Department of Zoology, Faculty of Science, Cairo University 12613 Giza Egypt
- Faculty of Biotechnology, October University for Modern Science and Arts 6th October Giza Egypt
| | - Farid M Sroor
- Organometallic and Organometalloid Chemistry Department, National Research Centre 12622 Cairo Egypt
| | - Haidan M El-Shorbagy
- Department of Zoology, Faculty of Science, Cairo University 12613 Giza Egypt
- Faculty of Biotechnology, October University for Modern Science and Arts 6th October Giza Egypt
| | - Shaymaa M Eissa
- Department of Zoology, Faculty of Science, Cairo University 12613 Giza Egypt
| | - Hamdi M Hassaneen
- Department of Chemistry, Faculty of Science, Cairo University Giza Egypt
| | | |
Collapse
|
3
|
Mushtaq A, Asif R, Humayun WA, Naseer MM. Novel isatin-triazole based thiosemicarbazones as potential anticancer agents: synthesis, DFT and molecular docking studies. RSC Adv 2024; 14:14051-14067. [PMID: 38686286 PMCID: PMC11057040 DOI: 10.1039/d4ra01937g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024] Open
Abstract
Thiosemicarbazones of isatin have been found to exhibit versatile bioactivities. In this study, two distinct types of isatin-triazole hybrids 3a and 3b were accessed via copper-catalyzed azide-alkyne cycloaddition reaction (CuAAC), together with their mono and bis-thiosemicarbazone derivatives 4a-h and 5a-h. In addition to the characterization by physical, spectral and analytical data, a DFT study was carried out to obtain the optimized geometries of all thiosemicarbazones. The global reactivity values showed that among the synthesized derivatives, 4c, 4g and 5c having nitro substituents are the most soft compounds, with compound 5c having the highest electronegativity and electrophilicity index values among the synthesized series, thus possessing strong binding ability with biomolecules. Molecular docking studies were performed to explore the inhibitory ability of the selected compounds against the active sites of the anticancer protein of phosphoinositide 3-kinase (PI3K). Among the synthesized derivatives, 4-nitro substituted bisthiosemicarbazone 5c showed the highest binding energy of -10.3 kcal mol-1. These findings demonstrated that compound 5c could be used as a favored anticancer scaffold via the mechanism of inhibition against the PI3K signaling pathways.
Collapse
Affiliation(s)
- Alia Mushtaq
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan
| | - Rabbia Asif
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan
| | - Waqar Ahmed Humayun
- Department of Medical Oncology & Radiotherapy, King Edward Medical University Lahore 54000 Pakistan
| | | |
Collapse
|
4
|
Natangelo S, Trapani D, Koukoutzeli C, Boscolo Bielo L, Marvaso G, Jereczek-Fossa BA, Curigliano G. Radiation therapy, tissue radiosensitization, and potential synergism in the era of novel antibody-drug conjugates. Crit Rev Oncol Hematol 2024; 195:104270. [PMID: 38272150 DOI: 10.1016/j.critrevonc.2024.104270] [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: 09/14/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Antibody-drug conjugates (ADCs) represent a therapeutic class of agents designed to selectively deliver cytotoxic payloads to cancer cells. With the increasingly positioning of ADCs in the clinical practice, combinations with other treatment modalities, including radiation therapy (RT), will open new opportunities but also challenges. This review evaluates ADC-RT interactions, examining therapeutic synergies and potential caveats. ADC payloads can be radiosensitizing, enhancing cytotoxicity when used in combination with RT. Antigens targeted by ADCs can have various tissue expressions, resulting in possible off-target toxicities by tissue radiosensitization. Notably, the HER-2-directed ADC trastuzumab emtansine has appeared to increase the risk of radionecrosis when used concomitantly with brain RT, as glial cells can express HER2, too. Other possible organ-specific effects are discussed, such as pulmonary and cardiac toxicities. The lack of robust clinical data on the ADC-RT combination raises concerns regarding specific side effects and the ultimate trade-off of toxicity and safety of some combined approaches. Clinical studies are needed to assess ADC-RT combination safety and efficacy.
Collapse
Affiliation(s)
- Stefano Natangelo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Chrysanthi Koukoutzeli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
| | - Luca Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Giulia Marvaso
- Division of Radiation Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy; Division of Radiation Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy.
| |
Collapse
|
5
|
Fraire-Soto I, Araujo-Huitrado JG, Granados-López AJ, Segura-Quezada LA, Ortiz-Alvarado R, Herrera MD, Gutiérrez-Hernández R, Reyes-Hernández CA, López-Hernández Y, Tapia-Juárez M, Negrete-Díaz JV, Chacón-García L, Solorio-Alvarado CR, López JA. Differential Effect of 4 H-Benzo[ d] [1, 3]oxazines on the Proliferation of Breast Cancer Cell Lines. Curr Med Chem 2024; 31:6306-6318. [PMID: 38676529 DOI: 10.2174/0109298673292365240422104456] [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: 12/21/2023] [Revised: 02/20/2024] [Accepted: 03/11/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND A family of 4H-benzo[d][1,3]oxazines were obtained from a group of N-(2-alkynyl)aryl benzamides precursors via gold(I) catalysed chemoselective 6-exo-dig C-O cyclization. METHOD The precursors and oxazines obtained were studied in breast cancer cell lines MCF-7, CAMA-1, HCC1954 and SKBR-3 with differential biological activity showing various degrees of inhibition with a notable effect for those that had an aryl substituted at C-2 of the molecules. 4H-benzo[d][1,3]oxazines showed an IC50 rating from 0.30 to 157.4 µM in MCF-7, 0.16 to 139 in CAMA-1, 0.09 to 93.08 in SKBR-3, and 0.51 to 157.2 in HCC1954 cells. RESULTS We observed that etoposide is similar to benzoxazines while taxol effect is more potent. Four cell lines responded to benzoxazines while SKBR-3 cell line responded to precursors and benzoxazines. Compounds 16, 24, 25 and 26 have the potent effect in cell proliferation inhibition in the 4 cell lines tested and correlated with oxidant activity suggesting a possible mechanism by ROS generation. CONCLUSION These compounds represent possible drug candidates for the treatment of breast cancer. However, further trials are needed to elucidate its full effect on cellular and molecular features of cancer.
Collapse
Affiliation(s)
- Ixamail Fraire-Soto
- Laboratorio de MicroRNAs y Cáncer, Universidad Autónoma de Zacatecas, Av. Preparatoria S/N, Agronómica, Campus II, Zacatecas, Zac., 98066, México
| | - Jorge Gustavo Araujo-Huitrado
- Laboratorio de MicroRNAs y Cáncer, Universidad Autónoma de Zacatecas, Av. Preparatoria S/N, Agronómica, Campus II, Zacatecas, Zac., 98066, México
| | - Angelica Judith Granados-López
- Laboratorio de MicroRNAs y Cáncer, Universidad Autónoma de Zacatecas, Av. Preparatoria S/N, Agronómica, Campus II, Zacatecas, Zac., 98066, México
| | - Luis A Segura-Quezada
- Departamento de Química, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Campus Guanajuato, Noria Alta S/N, Guanajuato, 36050, México
| | - Rafael Ortiz-Alvarado
- Departamento de Química, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Campus Guanajuato, Noria Alta S/N, Guanajuato, 36050, México
| | - Mayra Denise Herrera
- Laboratorio de MicroRNAs y Cáncer, Universidad Autónoma de Zacatecas, Av. Preparatoria S/N, Agronómica, Campus II, Zacatecas, Zac., 98066, México
- Campo Experimental Zacatecas (CEZAC-INIFAP), Carretera Zacatecas-Fresnillo Km 24.5, Calera de VR, Zacatecas, 98500, Mexico
| | - Rosalinda Gutiérrez-Hernández
- Laboratorio de MicroRNAs y Cáncer, Universidad Autónoma de Zacatecas, Av. Preparatoria S/N, Agronómica, Campus II, Zacatecas, Zac., 98066, México
| | - Claudia Araceli Reyes-Hernández
- Laboratorio de MicroRNAs y Cáncer, Universidad Autónoma de Zacatecas, Av. Preparatoria S/N, Agronómica, Campus II, Zacatecas, Zac., 98066, México
| | - Yamilé López-Hernández
- Laboratorio de Metabolómica y Proteómica Universidad Autónoma de Zacatecas, Av. Preparatoria S/N, Agronómica, Campus II, Zacatecas, Zac., 98066, México
| | - Melissa Tapia-Juárez
- Laboratorio de Diseño Molecular, Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, Morelia, Mich., 58033, México
| | - José Vicente Negrete-Díaz
- Laboratory of Brain Plasticity and Integrative Neuroscience, Program of Clinical Psychology, University of Guanajuato, Guanajuato, 38060, México
| | - Luis Chacón-García
- Laboratorio de Diseño Molecular, Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Ciudad Universitaria, Morelia, Mich., 58033, México
| | - César R Solorio-Alvarado
- Departamento de Química, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Campus Guanajuato, Noria Alta S/N, Guanajuato, 36050, México
| | - Jesús Adrián López
- Laboratorio de MicroRNAs y Cáncer, Universidad Autónoma de Zacatecas, Av. Preparatoria S/N, Agronómica, Campus II, Zacatecas, Zac., 98066, México
| |
Collapse
|
6
|
Weng C, Yang H, Loh BS, Wong MW, Ang WH. Targeting Pathogenic Formate-Dependent Bacteria with a Bioinspired Metallo-Nitroreductase Complex. J Am Chem Soc 2023; 145:6453-6461. [PMID: 36881731 DOI: 10.1021/jacs.3c00237] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Nitroreductases (NTRs) constitute an important class of oxidoreductase enzymes that have evolved to metabolize nitro-containing compounds. Their unique characteristics have spurred an array of potential uses in medicinal chemistry, chemical biology, and bioengineering toward harnessing nitro caging groups and constructing NTR variants for niche applications. Inspired by how they carry out enzymatic reduction via a cascade of hydride transfer reactions, we sought to develop a synthetic small-molecule NTR system based on transfer hydrogenation mediated by transition metal complexes harnessing native cofactors. We report the first water-stable Ru-arene complex capable of selectively and fully reducing nitroaromatics into anilines in a biocompatible buffered aqueous environment using formate as the hydride source. We further demonstrated its application to activate nitro-caged sulfanilamide prodrug in formate-abundant bacteria, specifically pathogenic methicillin-resistant Staphylococcus aureus. This proof of concept paves the way for a new targeted antibacterial chemotherapeutic approach leveraging on redox-active metal complexes for prodrug activation via bioinspired nitroreduction.
Collapse
Affiliation(s)
- Cheng Weng
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Hui Yang
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Boon Shing Loh
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Ming Wah Wong
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
- NUS Graduate School─Integrative Sciences and Engineering Programme, National University of Singapore, 21 Lower Kent Ridge Road, Singapore 119077, Singapore
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
- NUS Graduate School─Integrative Sciences and Engineering Programme, National University of Singapore, 21 Lower Kent Ridge Road, Singapore 119077, Singapore
| |
Collapse
|
7
|
Pramanik S, Saha P, Ghosh P, Mukhopadhyay C. Steric-Hindrance-Induced Diastereoselective Radical Nitration of 3-Alkylidene-2-oxindoles Followed by Tosylhydrazine-Mediated Sulfonation. J Org Chem 2023; 88:3386-3402. [PMID: 36847251 DOI: 10.1021/acs.joc.2c01523] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Metal-free radical nitration of the β C-H bond of 3-alkylidene-2-oxindoles with tert-butyl nitrite (TBN) has been explored. Interestingly, (E)-3-(2-(aryl)-2-oxoethylidene)oxindole and (E)-3-ylidene oxindole give different diastereomers on nitration. The mechanistic investigation revealed that the diastereoselectivity was controlled by the size of the functional group. Another transformation of 3-(nitroalkylidene) oxindole into 3-(tosylalkylidene) oxindole was performed through metal and oxidant-free tosylhydrazine-mediated sulfonation. Both methods have the advantages of readily available starting materials and operational simplicity.
Collapse
Affiliation(s)
- Sayan Pramanik
- Department of Chemistry, University of Calcutta, 92 APC Road, Kolkata 700009, India
| | - Pinaki Saha
- Department of Chemistry, R. K. Mission Residential College, Narendrapur, Kolkata 700103, India
| | - Prasanta Ghosh
- Department of Chemistry, R. K. Mission Residential College, Narendrapur, Kolkata 700103, India
| | - Chhanda Mukhopadhyay
- Department of Chemistry, University of Calcutta, 92 APC Road, Kolkata 700009, India
| |
Collapse
|
8
|
Nitroaromatic Hypoxia-Activated Prodrugs for Cancer Therapy. Pharmaceuticals (Basel) 2022; 15:ph15020187. [PMID: 35215299 PMCID: PMC8878295 DOI: 10.3390/ph15020187] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/24/2022] [Accepted: 02/01/2022] [Indexed: 02/04/2023] Open
Abstract
The presence of “hypoxic” tissue (with O2 levels of <0.1 mmHg) in solid tumours, resulting in quiescent tumour cells distant from blood vessels, but capable of being reactivated by reoxygenation following conventional therapy (radiation or drugs), have long been known as a limitation to successful cancer chemotherapy. This has resulted in a sustained effort to develop nitroaromatic “hypoxia-activated prodrugs” designed to undergo enzyme-based nitro group reduction selectively in these hypoxic regions, to generate active drugs. Such nitro-based prodrugs can be classified into two major groups; those activated either by electron redistribution or by fragmentation following nitro group reduction, relying on the extraordinary difference in electron demand between an aromatic nitro group and its reduction products. The vast majority of hypoxia-activated fall into the latter category and are discussed here classed by the nature of their nitroaromatic trigger units.
Collapse
|
9
|
Seen SB, Gong Y, Ashton M. The application of the Fischer indole synthesis in medicinal chemistry. ADVANCES IN HETEROCYCLIC CHEMISTRY 2022. [DOI: 10.1016/bs.aihch.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
10
|
Zheng YN, Zheng H, Li T, Wei WT. Recent Advances in Copper-Catalyzed C-N Bond Formation Involving N-Centered Radicals. CHEMSUSCHEM 2021; 14:5340-5358. [PMID: 34750973 DOI: 10.1002/cssc.202102243] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/09/2021] [Indexed: 06/13/2023]
Abstract
C-N bonds are pervasive throughout organic-based materials, natural products, pharmaceutical compounds, and agricultural chemicals. Considering the widespread importance of C-N bonds, the development of greener and more convenient ways to form C-N bonds, especially in late-stage synthesis, has become one of the hottest research goals in synthetic chemistry. Copper-catalyzed radical reactions involving N-centered radicals have emerged as a sustainable and promising approach to build C-N bonds. As a chemically popular and diverse radical species, N-centered radicals have been used for all kinds of reactions for C-N bond formation by taking advantage of their inherently incredible reactive flexibility. Copper is also the most abundant and economic catalyst with the most relevant activity for facilitating the synthesis of valuable compounds. Therefore, the aim of the present Review was to illustrate recent and significant advances in C-N bond formation methods and to understand the unique advantages of copper catalysis in the generation of N-centered radicals since 2016. To provide an ease of understanding for the readers, this Review was organized based on the types of nitrogen sources (amines, amides, sulfonamides, oximes, hydrazones, azides, and tert-butyl nitrite).
Collapse
Affiliation(s)
- Yan-Nan Zheng
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Hongxing Zheng
- Institution of Functional Organic Molecules and Materials, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong, 252059, P. R. China
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang, Henan, 473061, P. R. China
| | - Ting Li
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang, Henan, 473061, P. R. China
| | - Wen-Ting Wei
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| |
Collapse
|
11
|
Therapeutic targeting of the hypoxic tumour microenvironment. Nat Rev Clin Oncol 2021; 18:751-772. [PMID: 34326502 DOI: 10.1038/s41571-021-00539-4] [Citation(s) in RCA: 213] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
Hypoxia is prevalent in human tumours and contributes to microenvironments that shape cancer evolution and adversely affect therapeutic outcomes. Historically, two different tumour microenvironment (TME) research communities have been discernible. One has focused on physicochemical gradients of oxygen, pH and nutrients in the tumour interstitium, motivated in part by the barrier that hypoxia poses to effective radiotherapy. The other has focused on cellular interactions involving tumour and non-tumour cells within the TME. Over the past decade, strong links have been established between these two themes, providing new insights into fundamental aspects of tumour biology and presenting new strategies for addressing the effects of hypoxia and other microenvironmental features that arise from the inefficient microvascular system in solid tumours. This Review provides a perspective on advances at the interface between these two aspects of the TME, with a focus on translational therapeutic opportunities relating to the elimination and/or exploitation of tumour hypoxia.
Collapse
|
12
|
Spatially-resolved pharmacokinetic/pharmacodynamic modelling of bystander effects of a nitrochloromethylbenzindoline hypoxia-activated prodrug. Cancer Chemother Pharmacol 2021; 88:673-687. [PMID: 34245333 DOI: 10.1007/s00280-021-04320-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/23/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Hypoxia-activated prodrugs (HAPs) have the potential for eliminating chemo- and radiation-resistant hypoxic tumour cells, but their activity is often compromised by limited penetration into hypoxic zones. Nitrochloromethylbenzindoline (nitroCBI) HAPs are reduced in hypoxic cells to highly cytotoxic DNA minor groove alkylating aminoCBI metabolites. In this study, we investigate whether a lead nitroCBI, SN30548, generates a significant bystander effect through the diffusion of its aminoCBI metabolite and whether this compensates for any diffusion limitations of the prodrug in tumour tissue. METHODS Metabolism and uptake of the nitroCBI in oxic and anoxic cells, and diffusion through multicellular layer cultures, was characterised by LC-MS/MS. To quantify bystander effects, clonogenic cell killing of HCT116 cells was assessed in multicellular spheroid co-cultures comprising cells transfected with cytochrome P450 oxidoreductase (POR) or E. coli nitroreductase NfsA. Spatially-resolved pharmacokinetic/pharmacodynamic (PK/PD) models, parameterised by the above measurements, were developed for spheroids and tumours using agent-based and Green's function modelling, respectively. RESULTS NitroCBI was reduced to aminoCBI by POR under anoxia and by NfsA under oxia, and was the only significant cytotoxic metabolite in both cases. In spheroid co-cultures comprising 30% NfsA-expressing cells, non-metabolising cells were as sensitive as the NfsA cells, demonstrating a marked bystander effect. Agent-based PK/PD models provided good prediction of cytotoxicity in spheroids, while use of the same parameters in a Green's function model for a tumour microregion demonstrated that local diffusion of aminoCBI overcomes the penetration limitation of the prodrug. CONCLUSIONS The nitroCBI HAP SN30548 generates a highly efficient bystander effect through local diffusion of its active metabolite in tumour tissue.
Collapse
|
13
|
Annatelli M, Trapasso G, Salaris C, Salata C, Castellano S, Aricò F. Mustard Carbonate Analogues as Sustainable Reagents for the Aminoalkylation of Phenols. European J Org Chem 2021. [DOI: 10.1002/ejoc.202100328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Mattia Annatelli
- Department of Environmental Sciences Informatics and Statistics Ca' Foscari University Campus Scientifico, Via Torino 155 30172 Venezia Mestre Italy
| | - Giacomo Trapasso
- Department of Environmental Sciences Informatics and Statistics Ca' Foscari University Campus Scientifico, Via Torino 155 30172 Venezia Mestre Italy
| | - Claudio Salaris
- Department of Molecular Medicine Padua University via Gabelli 63 35121 Padova Italy
| | - Cristiano Salata
- Department of Molecular Medicine Padua University via Gabelli 63 35121 Padova Italy
| | - Sabrina Castellano
- Department of Pharmacy University of Salerno Via Giovanni Paolo II, 132 84084 Fisciano, Salerno Italy
| | - Fabio Aricò
- Department of Environmental Sciences Informatics and Statistics Ca' Foscari University Campus Scientifico, Via Torino 155 30172 Venezia Mestre Italy
| |
Collapse
|
14
|
Jukes Z, Morais GR, Loadman PM, Pors K. How can the potential of the duocarmycins be unlocked for cancer therapy? Drug Discov Today 2020; 26:577-584. [PMID: 33232841 DOI: 10.1016/j.drudis.2020.11.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/03/2020] [Accepted: 11/16/2020] [Indexed: 12/31/2022]
Abstract
The duocarmycins belong to a class of agent that has fascinated scientists for over four decades. Their exquisite potency, unique mechanism of action, and efficacy in multidrug-resistant tumour models makes them attractive to medicinal chemists and drug hunters. However, despite great advances in fine-tuning biological activity through structure-activity relationship studies (SARS), no duocarmycin-based therapeutic has reached clinical approval. In this review, we provide an overview of the most promising strategies currently used and include both tumour-targeted prodrug approaches and antibody-directed technologies.
Collapse
Affiliation(s)
- Zoë Jukes
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Goreti Ribeiro Morais
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Paul M Loadman
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Klaus Pors
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, UK.
| |
Collapse
|
15
|
Zhou M, Xie Y, Xu S, Xin J, Wang J, Han T, Ting R, Zhang J, An F. Hypoxia-activated nanomedicines for effective cancer therapy. Eur J Med Chem 2020; 195:112274. [PMID: 32259703 DOI: 10.1016/j.ejmech.2020.112274] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/12/2020] [Accepted: 03/24/2020] [Indexed: 12/27/2022]
Abstract
Hypoxia, a common characteristic in solid tumors, is found in phenotypically aggressive cancers that display resistance to typical cancer interventions. Due to its important role in tumor progression, tumor hypoxia has been considered as a primary target for cancer diagnosis and treatment. An advantage of hypoxia-activated nanomedicines is that they are inactive in normoxic cells. In hypoxic tumor tissues and cells, these nanomedicines undergo reduction by activated enzymes (usually through 1 or 2 electron oxidoreductases) to produce cytotoxic substances. In this review, we will focus on approaches to design nanomedicines that take advantage of tumor hypoxia. These approaches include: i) inhibitors of hypoxia-associated signaling pathways; ii) prodrugs activated by hypoxia; iii) nanocarriers responsive to hypoxia, and iv) bacteria mediated hypoxia targeting therapy. These strategies have guided and will continue to guide nanoparticle design in the near future. These strategies have the potential to overcome tumor heterogeneity to improve the efficiency of radiotherapy, chemotherapy and diagnosis.
Collapse
Affiliation(s)
- Mengjiao Zhou
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Yuqi Xie
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Shujun Xu
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Jingqi Xin
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, PR China
| | - Jin Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, PR China
| | - Tao Han
- College of Chemistry and Life Science, Institute of Functional Molecules, Chengdu Normal University, Chengdu, 611130, PR China
| | - Richard Ting
- Department of Radiology, Weill Cornell Medicine, 413E, 69th St, New York, NY, 10065, USA
| | - Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, PR China.
| | - Feifei An
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, PR China.
| |
Collapse
|
16
|
Li D, Wang Y, Jia Z, Ou Z, Dong Y, Lv C, Fu G, Liang D. Cu-Mediated Synthesis of Indolines and Dihydroisoquinolinones through Arylperfluoroalkylation of Unactivated Alkenes. European J Org Chem 2019. [DOI: 10.1002/ejoc.201900680] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Dandan Li
- School of Chemistry and Chemical Engineering; Xuchang University; No. 88, Bayi Road, Xuchang, Henan 461000 P. R. China
| | - Yan Wang
- Department of Chemistry; Kunming University; 650214 Kunming P. R. China
| | - Zhenzhen Jia
- School of Chemistry and Chemical Engineering; Xuchang University; No. 88, Bayi Road, Xuchang, Henan 461000 P. R. China
| | - Zhaocheng Ou
- School of Chemistry and Chemical Engineering; Xuchang University; No. 88, Bayi Road, Xuchang, Henan 461000 P. R. China
| | - Yongrui Dong
- Department of Chemistry; Kunming University; 650214 Kunming P. R. China
| | - Cunjie Lv
- School of Chemistry and Chemical Engineering; Xuchang University; No. 88, Bayi Road, Xuchang, Henan 461000 P. R. China
| | - Guangbin Fu
- School of Chemistry and Chemical Engineering; Xuchang University; No. 88, Bayi Road, Xuchang, Henan 461000 P. R. China
| | - Deqiang Liang
- Department of Chemistry; Kunming University; 650214 Kunming P. R. China
| |
Collapse
|
17
|
Shen QK, Deng H, Wang SB, Tian YS, Quan ZS. Synthesis, and evaluation of in vitro and in vivo anticancer activity of 14-substituted oridonin analogs: A novel and potent cell cycle arrest and apoptosis inducer through the p53-MDM2 pathway. Eur J Med Chem 2019; 173:15-31. [DOI: 10.1016/j.ejmech.2019.04.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 04/01/2019] [Accepted: 04/01/2019] [Indexed: 02/07/2023]
|
18
|
Wang Y, Liu BY, Yang G, Chai Z. Synthesis of 2-Aminophosphates via S N2-Type Ring Openings of Aziridines with Organophosphoric Acids. Org Lett 2019; 21:4475-4479. [PMID: 31184161 DOI: 10.1021/acs.orglett.9b01302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The synthesis of 2-aminophosphates is achieved by a SN2-type ring opening reaction of various N-protected or free aziridines with phosphoric acids in a regiospecific and/or enantiospecific way. A one-pot, two-step procedure is also developed enabling direct access to 2-aminophosphates from olefins without isolation of the aziridine intermediates.
Collapse
Affiliation(s)
- Yang Wang
- MOE Key Laboratory of Functionalized Molecular Solids, Anhui Key Laboratory of Molecule-Based Materials (State Key Laboratory Cultivation Base), College of Chemistry and Materials Science , Anhui Normal University , Wuhu , Anhui 241002 , China
| | - Bing-Yi Liu
- MOE Key Laboratory of Functionalized Molecular Solids, Anhui Key Laboratory of Molecule-Based Materials (State Key Laboratory Cultivation Base), College of Chemistry and Materials Science , Anhui Normal University , Wuhu , Anhui 241002 , China
| | - Gaosheng Yang
- MOE Key Laboratory of Functionalized Molecular Solids, Anhui Key Laboratory of Molecule-Based Materials (State Key Laboratory Cultivation Base), College of Chemistry and Materials Science , Anhui Normal University , Wuhu , Anhui 241002 , China
| | - Zhuo Chai
- MOE Key Laboratory of Functionalized Molecular Solids, Anhui Key Laboratory of Molecule-Based Materials (State Key Laboratory Cultivation Base), College of Chemistry and Materials Science , Anhui Normal University , Wuhu , Anhui 241002 , China
| |
Collapse
|
19
|
Lee HH, Dickson BD, Stevenson RJ, Yang S, Tercel M. Optimised synthesis of a nitroCBI hypoxia-activated prodrug with substantial anticancer activity. Tetrahedron 2019. [DOI: 10.1016/j.tet.2019.04.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
Sharma A, Arambula JF, Koo S, Kumar R, Singh H, Sessler JL, Kim JS. Hypoxia-targeted drug delivery. Chem Soc Rev 2019; 48:771-813. [PMID: 30575832 PMCID: PMC6361706 DOI: 10.1039/c8cs00304a] [Citation(s) in RCA: 319] [Impact Index Per Article: 53.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hypoxia is a state of low oxygen tension found in numerous solid tumours. It is typically associated with abnormal vasculature, which results in a reduced supply of oxygen and nutrients, as well as impaired delivery of drugs. The hypoxic nature of tumours often leads to the development of localized heterogeneous environments characterized by variable oxygen concentrations, relatively low pH, and increased levels of reactive oxygen species (ROS). The hypoxic heterogeneity promotes tumour invasiveness, metastasis, angiogenesis, and an increase in multidrug-resistant proteins. These factors decrease the therapeutic efficacy of anticancer drugs and can provide a barrier to advancing drug leads beyond the early stages of preclinical development. This review highlights various hypoxia-targeted and activated design strategies for the formulation of drugs or prodrugs and their mechanism of action for tumour diagnosis and treatment.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Chemistry, Korea University, Seoul, 02841, Korea.
| | | | | | | | | | | | | |
Collapse
|
21
|
Ai Y, Liu P, Liang R, Liu Y, Li F. The N-alkylation of sulfonamides with alcohols in water catalyzed by a water-soluble metal–ligand bifunctional iridium complex [Cp*Ir(biimH2)(H2O)][OTf]2. NEW J CHEM 2019. [DOI: 10.1039/c9nj02542a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A water-soluble dinuclear Cp*Ir complex bearing 4,4′,6,6′-tetrahydroxy-2,2′-bipyrimidine as a bridging ligand was found to be a highly effective catalyst for the N-alkylation of ketones with alcohols in water.
Collapse
Affiliation(s)
- Yao Ai
- School of Chemical Engineering
- Nanjing University of Science & Technology
- Nanjing 210094
- P. R. China
| | - Pengcheng Liu
- School of Chemical Engineering
- Nanjing University of Science & Technology
- Nanjing 210094
- P. R. China
| | - Ran Liang
- School of Chemical Engineering
- Nanjing University of Science & Technology
- Nanjing 210094
- P. R. China
| | - Yan Liu
- School of Chemical Engineering
- Nanjing University of Science & Technology
- Nanjing 210094
- P. R. China
| | - Feng Li
- School of Chemical Engineering
- Nanjing University of Science & Technology
- Nanjing 210094
- P. R. China
- State Key Laboratory of Fine Chemicals
| |
Collapse
|
22
|
Abstract
The nitro group is considered to be a versatile and unique functional group in medicinal chemistry. Despite a long history of use in therapeutics, the nitro group has toxicity issues and is often categorized as a structural alert or a toxicophore, and evidence related to drugs containing nitro groups is rather contradictory. In general, drugs containing nitro groups have been extensively associated with mutagenicity and genotoxicity. In this context, efforts toward the structure-mutagenicity or structure-genotoxicity relationships have been undertaken. The current Perspective covers various aspects of agents that contain nitro groups, their bioreductive activation mechanisms, their toxicities, and approaches to combat their toxicity issues. In addition, recent advances in the field of anticancer, antitubercular and antiparasitic agents containing nitro groups, along with a patent survey on hypoxia-activated prodrugs containing nitro groups, are also covered.
Collapse
Affiliation(s)
- Kunal Nepali
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| | - Hsueh-Yun Lee
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| |
Collapse
|
23
|
Hong CR, Dickson BD, Jaiswal JK, Pruijn FB, Hunter FW, Hay MP, Hicks KO, Wilson WR. Cellular pharmacology of evofosfamide (TH-302): A critical re-evaluation of its bystander effects. Biochem Pharmacol 2018; 156:265-280. [PMID: 30134191 DOI: 10.1016/j.bcp.2018.08.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022]
Abstract
Evofosfamide (TH-302) is a clinical-stage hypoxia-activated prodrug with proven efficacy against hypoxic cells in preclinical tumour models. TH-302 is designed to release the DNA crosslinking agent bromo-isophosphoramide mustard (Br-IPM) when reduced in hypoxic tissue. Br-IPM is considered to diffuse locally from hypoxic regions, eliciting additional tumour cell killing, but the latter 'bystander effect' has not been demonstrated directly. Previous studies with multicellular co-cultures that included cells expressing the E. coli nitroreductase NfsA as a model TH-302 reductase have provided clear evidence of a bystander effect (which we confirm in the present study). However, NfsA is an oxygen-insensitive two-electron reductase that is not expected to generate the nitro radical intermediate that has been demonstrated to fragment to release Br-IPM. Here, we use mass spectrometry methods to characterise TH-302 metabolites generated by one-electron reduction (steady-state radiolysis by ionising radiation and cellular metabolism under hypoxia, including HCT116 cells that overexpress P450 oxidoreductase, POR) or by NfsA expressed in HCT116 cells under oxic conditions, and investigate the stability and cytotoxicity of these products. Br-IPM is shown to have very low cytotoxic potency when added to extracellular culture medium and to be rapidly converted to other hydrophilic products including dichloro-isophosphoramide mustard (IPM). Only traces of Br-IPM or IPM were detected in the extracellular medium when generated by cellular metabolism of TH-302. We identify, in NfsA-expressing cells, the hydroxylamine metabolite of TH-302, and downstream products resulting from rearrangement or hydration of the imidazole ring, and demonstrate that formation of these candidate bystander effect mediators is suppressed by hypoxia. This characterisation of the cellular pharmacology of TH-302 implies that bystander effects from hypoxic activation of TH-302 are unlikely to contribute to its anticancer activity.
Collapse
Affiliation(s)
- Cho Rong Hong
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Benjamin D Dickson
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Jagdish K Jaiswal
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Frederik B Pruijn
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Francis W Hunter
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Michael P Hay
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Kevin O Hicks
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - William R Wilson
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
24
|
Liang D, Ge D, Lv Y, Huang W, Wang B, Li W. Silver-Catalyzed Radical Arylphosphorylation of Unactivated Alkenes: Synthesis of 3-Phosphonoalkyl Indolines. J Org Chem 2018; 83:4681-4691. [DOI: 10.1021/acs.joc.8b00450] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Deqiang Liang
- Department of Chemistry, Kunming University, Kunming 650214, China
- Yunnan Engineering Technology Research Center for Plastic Films, Kunming 650214, China
| | - Dandan Ge
- Department of Chemistry, Kunming University, Kunming 650214, China
| | - Yanping Lv
- Department of Chemistry, Kunming University, Kunming 650214, China
| | - Wenzhong Huang
- Yunnan Engineering Technology Research Center for Plastic Films, Kunming 650214, China
| | - Baoling Wang
- Yunnan Engineering Technology Research Center for Plastic Films, Kunming 650214, China
| | - Weili Li
- Yunnan Engineering Technology Research Center for Plastic Films, Kunming 650214, China
| |
Collapse
|