1
|
Sun J, Lou L, Zhu C, Chen P, Tang G, Gu M, Xia S, Dong X, Zhang ZM, Gao L, Yao SQ, Xiao Q. Rationally designed BCR-ABL kinase inhibitors for improved leukemia treatment via covalent and pro-/dual-drug targeting strategies. J Adv Res 2024:S2090-1232(24)00392-8. [PMID: 39255927 DOI: 10.1016/j.jare.2024.09.008] [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: 06/28/2024] [Revised: 08/08/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Chronic Myeloid Leukemia (CML) is a blood cancer that remains challenging to cure due to drug resistance and side effects from current BCR-ABL inhibitors. There is an urgent need for novel and more effective BCR-ABL targeting inhibitors and therapeutic strategies to combat this deadly disease. METHOD We disclose an "OH-implant" strategy to improve a noncovalent BCR-ABL inhibitor, PPY-A, by adding a hydroxyl group to its scaffold. By taking advantage of this OH "hot spot", we designed a panel of irreversible covalent kinase inhibitors and hypoxia-responsive pro-/dual-drugs, and their biological activities were studied in vitro, in cellulo and in vivo. RESULT The resulting compound B1 showed enhanced solubility and biological activity. B4 achieved sustained BCR-ABL inhibition by forming a stable covalent bond with ABL kinase. Hypoxia-responsive prodrug P1 and dual-drugs D1/D2/D3 demonstrated significant anti-tumor effects under hypoxic conditions. The in vivo studies using K562-xenografted mice showed that B1 displayed superior antitumor activity than PPY-A, while P1 and D3 offered better safety profiles alongside significant tumor control. CONCLUSION We have successfully developed a chemical biology approach to convert a known noncovalent BCR-ABL inhibitor into more potent and safer inhibitors through covalent and pro-/dual-drug targeting strategies. Our "OH-implant" approach and the resulting drug design strategies have general applicability and hold promise for improvement the performance of various other reported drugs/drug candidates, thereby providing advanced medicines for disease treatment.
Collapse
Affiliation(s)
- Jie Sun
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, and Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| | - Liang Lou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, and Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| | - Chengjun Zhu
- School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China.
| | - Peng Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, and Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| | - Guanghui Tang
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore.
| | - Mingxi Gu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, and Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| | - Shu Xia
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Xiao Dong
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Zhi-Min Zhang
- School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China.
| | - Liqian Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, and Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore.
| | - Qicai Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, and Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
2
|
Khan MS, Altwaijry N, Al-Bagmi MS, Alafaleq NO, Alokail MS, Shahwan M, Shamsi A. Structure-guided identification of potent inhibitors of ROS1 kinase for therapeutic development against non-small cell lung cancer. J Biomol Struct Dyn 2024; 42:3837-3847. [PMID: 37254309 DOI: 10.1080/07391102.2023.2217450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/12/2023] [Indexed: 06/01/2023]
Abstract
Proto-oncogene tyrosine-protein kinase ROS (ROS1) is a member of the sevenless receptor, which affects epithelial cell differentiation and is highly expressed in a variety of tumor cells. The elevated expression and dysfunction of ROS1 have been involved in various malignancies, such as non-small cell lung cancer (NSCLC), stomach cancer, ovarian, breast cancer, cholangiocarcinoma, colorectal cancer, adenosarcoma, oesophageal cancer, etc. ROS1 has been postulated as a potential drug target in anticancer therapeutics. In this study, we carried out a virtual screening of phytochemicals against ROS1 to identify its potential inhibitors. The virtual screening process was performed on the ROS1 structure, where two phytochemicals, Helioscopinolide C and Taiwanin C, were identified. These compounds resulted from filters like Lipinski rule of five, PAINS filter, binding affinities values, and all-atom molecular dynamics (MD) simulations followed by principal component analysis (PCA) and essential dynamics. The findings of this study highlight the role of ROS1 in multiple physiological candidates and its therapeutic targeting using phytochemicals. This study suggests Helioscopinolide C and Taiwanin C as potential compounds for therapeutic development targeting ROS1-associated non-small cell lung cancer for clinical applications. Further in vitro and in vivo experiments are required to validate these findings.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, KSA, Riyadh, Saudi Arabia
| | - Nojood Altwaijry
- Department of Biochemistry, College of Science, King Saud University, KSA, Riyadh, Saudi Arabia
| | - Moneera Saud Al-Bagmi
- Department of Biochemistry, College of Science, King Saud University, KSA, Riyadh, Saudi Arabia
| | - Nouf Omar Alafaleq
- Department of Biochemistry, College of Science, King Saud University, KSA, Riyadh, Saudi Arabia
| | - Majed S Alokail
- Department of Biochemistry, College of Science, King Saud University, KSA, Riyadh, Saudi Arabia
| | - Moyad Shahwan
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
3
|
Bordeaux ZA, Reddy SV, Choi J, Braun G, McKeel J, Lu W, Yossef SM, Ma EZ, West CE, Kwatra SG, Kwatra MM. Transcriptomic and proteomic analysis of tumor suppressive effects of GZ17-6.02 against mycosis fungoides. Sci Rep 2024; 14:1955. [PMID: 38263212 PMCID: PMC10805783 DOI: 10.1038/s41598-024-52544-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/19/2024] [Indexed: 01/25/2024] Open
Abstract
Mycosis fungoides (MF) is the most common form of cutaneous T-cell lymphoma (CTCL). Despite having a wide variety of therapeutic agents available for the treatment of MF, patients often suffer from a significant decrease in quality of life and rarely achieve long-term remission or complete cure, highlighting a need to develop novel therapeutic agents for this disease. The present study was undertaken to evaluate the efficacy of a novel anti-tumor agent, GZ17-6.02, which is composed of curcumin, harmine, and isovanillin, against MF in vitro and in murine models. Treatment of HH and MyLa cells with GZ17-6.02 inhibited the growth of both cell lines with IC50 ± standard errors for growth inhibition of 14.37 ± 1.19 µg/mL and 14.56 ± 1.35 µg/mL, respectively, and increased the percentage of cells in late apoptosis (p = .0304 for HH; p = .0301 for MyLa). Transcriptomic and proteomic analyses revealed that GZ17-6.02 suppressed several pathways, including tumor necrosis factor (TNF)-ɑ signaling via nuclear factor (NF)-kB, mammalian target of rapamycin complex (mTORC)1, and Pi3K/Akt/mTOR signaling. In a subcutaneous tumor model, GZ17-6.02 decreased tumor volume (p = .002) and weight (p = .009) compared to control conditions. Proteomic analysis of tumor samples showed that GZ17-6.02 suppressed the expression of several proteins that may promote CTCL growth, including mitogen-activated protein kinase (MAPK)1, MAPK3, Growth factor receptor bound protein (GRB)2, and Mediator of RAP80 interactions and targeting subunit of 40 kDa (MERIT)40.
Collapse
Affiliation(s)
- Zachary A Bordeaux
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Sriya V Reddy
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Justin Choi
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Gabriella Braun
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Jaimie McKeel
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Weiying Lu
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Selina M Yossef
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
| | - Emily Z Ma
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA
| | - Cameron E West
- Genzada Pharmaceuticals, Hutchinson, USA
- US Dermatology Partners, Wichita, USA
| | - Shawn G Kwatra
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 206 1550 Orleans Street, Baltimore, MD, 21231, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA.
| | - Madan M Kwatra
- Department of Anesthesiology, Duke University School of Medicine, Durham, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, USA
| |
Collapse
|
4
|
Wang H. Prediction of protein-ligand binding affinity via deep learning models. Brief Bioinform 2024; 25:bbae081. [PMID: 38446737 PMCID: PMC10939342 DOI: 10.1093/bib/bbae081] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/31/2024] [Indexed: 03/08/2024] Open
Abstract
Accurately predicting the binding affinity between proteins and ligands is crucial in drug screening and optimization, but it is still a challenge in computer-aided drug design. The recent success of AlphaFold2 in predicting protein structures has brought new hope for deep learning (DL) models to accurately predict protein-ligand binding affinity. However, the current DL models still face limitations due to the low-quality database, inaccurate input representation and inappropriate model architecture. In this work, we review the computational methods, specifically DL-based models, used to predict protein-ligand binding affinity. We start with a brief introduction to protein-ligand binding affinity and the traditional computational methods used to calculate them. We then introduce the basic principles of DL models for predicting protein-ligand binding affinity. Next, we review the commonly used databases, input representations and DL models in this field. Finally, we discuss the potential challenges and future work in accurately predicting protein-ligand binding affinity via DL models.
Collapse
Affiliation(s)
- Huiwen Wang
- School of Physics and Engineering, Henan University of Science and Technology, Luoyang 471023, China
| |
Collapse
|
5
|
Batool A, Rashid W, Fatima K, Khan SU. Mechanisms of Cancer Resistance to Various Therapies. DRUG RESISTANCE IN CANCER: MECHANISMS AND STRATEGIES 2024:31-75. [DOI: 10.1007/978-981-97-1666-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Lee JY, Lee JW, Park TG, Han SH, Yoo SY, Jung KM, Kim DM, Lee OJ, Kim D, Chi XZ, Kim EG, Lee YS, Bae SC. Runx3 Restoration Regresses K-Ras-Activated Mouse Lung Cancers and Inhibits Recurrence. Cells 2023; 12:2438. [PMID: 37887282 PMCID: PMC10605764 DOI: 10.3390/cells12202438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Oncogenic K-RAS mutations occur in approximately 25% of human lung cancers and are most frequently found in codon 12 (G12C, G12V, and G12D). Mutated K-RAS inhibitors have shown beneficial results in many patients; however, the inhibitors specifically target K-RASG12C and acquired resistance is a common occurrence. Therefore, new treatments targeting all kinds of oncogenic K-RAS mutations with a durable response are needed. RUNX3 acts as a pioneer factor of the restriction (R)-point, which is critical for the life and death of cells. RUNX3 is inactivated in most K-RAS-activated mouse and human lung cancers. Deletion of mouse lung Runx3 induces adenomas (ADs) and facilitates the development of K-Ras-activated adenocarcinomas (ADCs). In this study, conditional restoration of Runx3 in an established K-Ras-activated mouse lung cancer model regressed both ADs and ADCs and suppressed cancer recurrence, markedly increasing mouse survival. Runx3 restoration suppressed K-Ras-activated lung cancer mainly through Arf-p53 pathway-mediated apoptosis and partly through p53-independent inhibition of proliferation. This study provides in vivo evidence supporting RUNX3 as a therapeutic tool for the treatment of K-RAS-activated lung cancers with a durable response.
Collapse
Affiliation(s)
- Ja-Yeol Lee
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.-Y.L.); (J.-W.L.); (T.-G.P.); (S.-H.H.); (S.-Y.Y.); (K.-M.J.); (D.-M.K.); (X.-Z.C.); (E.-G.K.)
| | - Jung-Won Lee
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.-Y.L.); (J.-W.L.); (T.-G.P.); (S.-H.H.); (S.-Y.Y.); (K.-M.J.); (D.-M.K.); (X.-Z.C.); (E.-G.K.)
| | - Tae-Geun Park
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.-Y.L.); (J.-W.L.); (T.-G.P.); (S.-H.H.); (S.-Y.Y.); (K.-M.J.); (D.-M.K.); (X.-Z.C.); (E.-G.K.)
| | - Sang-Hyun Han
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.-Y.L.); (J.-W.L.); (T.-G.P.); (S.-H.H.); (S.-Y.Y.); (K.-M.J.); (D.-M.K.); (X.-Z.C.); (E.-G.K.)
| | - Seo-Yeong Yoo
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.-Y.L.); (J.-W.L.); (T.-G.P.); (S.-H.H.); (S.-Y.Y.); (K.-M.J.); (D.-M.K.); (X.-Z.C.); (E.-G.K.)
| | - Kyoung-Mi Jung
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.-Y.L.); (J.-W.L.); (T.-G.P.); (S.-H.H.); (S.-Y.Y.); (K.-M.J.); (D.-M.K.); (X.-Z.C.); (E.-G.K.)
| | - Da-Mi Kim
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.-Y.L.); (J.-W.L.); (T.-G.P.); (S.-H.H.); (S.-Y.Y.); (K.-M.J.); (D.-M.K.); (X.-Z.C.); (E.-G.K.)
| | - Ok-Jun Lee
- Department of Pathology, School of Medicine, Chungbuk National University and Hospital, Cheongju 28644, Republic of Korea;
| | - Dohun Kim
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Chungbuk National University and Hospital, Cheongju 28644, Republic of Korea;
| | - Xin-Zi Chi
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.-Y.L.); (J.-W.L.); (T.-G.P.); (S.-H.H.); (S.-Y.Y.); (K.-M.J.); (D.-M.K.); (X.-Z.C.); (E.-G.K.)
| | - Eung-Gook Kim
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.-Y.L.); (J.-W.L.); (T.-G.P.); (S.-H.H.); (S.-Y.Y.); (K.-M.J.); (D.-M.K.); (X.-Z.C.); (E.-G.K.)
| | - You-Soub Lee
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.-Y.L.); (J.-W.L.); (T.-G.P.); (S.-H.H.); (S.-Y.Y.); (K.-M.J.); (D.-M.K.); (X.-Z.C.); (E.-G.K.)
| | - Suk-Chul Bae
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.-Y.L.); (J.-W.L.); (T.-G.P.); (S.-H.H.); (S.-Y.Y.); (K.-M.J.); (D.-M.K.); (X.-Z.C.); (E.-G.K.)
| |
Collapse
|
7
|
Piteša N, Kurtović M, Bartoniček N, Gkotsi DS, Čonkaš J, Petrić T, Musani V, Ozretić P, Riobo-Del Galdo NA, Sabol M. Signaling Switching from Hedgehog-GLI to MAPK Signaling Potentially Serves as a Compensatory Mechanism in Melanoma Cell Lines Resistant to GANT-61. Biomedicines 2023; 11:1353. [PMID: 37239024 PMCID: PMC10216463 DOI: 10.3390/biomedicines11051353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Melanoma represents the deadliest skin cancer due to its cell plasticity which results in high metastatic potential and chemoresistance. Melanomas frequently develop resistance to targeted therapy; therefore, new combination therapy strategies are required. Non-canonical signaling interactions between HH-GLI and RAS/RAF/ERK signaling were identified as one of the drivers of melanoma pathogenesis. Therefore, we decided to investigate the importance of these non-canonical interactions in chemoresistance, and examine the potential for HH-GLI and RAS/RAF/ERK combined therapy. METHODS We established two melanoma cell lines resistant to the GLI inhibitor, GANT-61, and characterized their response to other HH-GLI and RAS/RAF/ERK inhibitors. RESULTS We successfully established two melanoma cell lines resistant to GANT-61. Both cell lines showed HH-GLI signaling downregulation and increased invasive cell properties like migration potential, colony forming capacity, and EMT. However, they differed in MAPK signaling activity, cell cycle regulation, and primary cilia formation, suggesting different potential mechanisms responsible for resistance occurrence. CONCLUSIONS Our study provides the first ever insights into cell lines resistant to GANT-61 and shows potential mechanisms connected to HH-GLI and MAPK signaling which may represent new hot spots for noncanonical signaling interactions.
Collapse
Affiliation(s)
- Nikolina Piteša
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| | - Matea Kurtović
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| | - Nenad Bartoniček
- The Garvan Institute of Medical Research, Genome Informatics, Genomics & Epigenetics Division, 384 Victoria St., Darlinghurst, NSW 2010, Australia;
- The Kinghorn Centre for Clinical Genomics, 370 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Danai S. Gkotsi
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK; (D.S.G.); (N.A.R.-D.G.)
- Astbury Centre for Molecular Structural Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Josipa Čonkaš
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| | - Tina Petrić
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| | - Vesna Musani
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| | - Petar Ozretić
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| | - Natalia A. Riobo-Del Galdo
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK; (D.S.G.); (N.A.R.-D.G.)
- Astbury Centre for Molecular Structural Biology, University of Leeds, Leeds LS2 9JT, UK
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK
- Leeds Cancer Research Centre, University of Leeds, Leeds LS2 9JT, UK
| | - Maja Sabol
- Ruđer Bošković Institute, Division of Molecular Medicine, 10 000 Zagreb, Croatia; (N.P.); (M.K.); (J.Č.); (T.P.); (V.M.); (P.O.)
| |
Collapse
|
8
|
Maji S, Barman S, Panda G. An Efficient Approach Towards the Synthesis of Nintedanib. ChemistrySelect 2023. [DOI: 10.1002/slct.202300968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Affiliation(s)
- Saroj Maji
- Medicinal and Process Chemistry Division CSIR-Central Drug Research Institute Sector 10 Jankipuram Extension Sitapur Road Lucknow 226031 UP India. Academy of Scientific and Innovative Research (AcSIR) Ghaziabad Uttar Pradesh 201002 India
| | - Souvik Barman
- Medicinal and Process Chemistry Division CSIR-Central Drug Research Institute Sector 10 Jankipuram Extension Sitapur Road Lucknow 226031 UP India. Academy of Scientific and Innovative Research (AcSIR) Ghaziabad Uttar Pradesh 201002 India
| | - Gautam Panda
- Medicinal and Process Chemistry Division CSIR-Central Drug Research Institute Sector 10 Jankipuram Extension Sitapur Road Lucknow 226031 UP India. Academy of Scientific and Innovative Research (AcSIR) Ghaziabad Uttar Pradesh 201002 India
| |
Collapse
|
9
|
Capogiri M, De Micheli AJ, Lassaletta A, Muñoz DP, Coppé JP, Mueller S, Guerreiro Stucklin AS. Response and resistance to BRAF V600E inhibition in gliomas: Roadblocks ahead? Front Oncol 2023; 12:1074726. [PMID: 36698391 PMCID: PMC9868954 DOI: 10.3389/fonc.2022.1074726] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/12/2022] [Indexed: 01/11/2023] Open
Abstract
BRAFV600E represents the most common BRAF mutation in all human cancers. Among central nervous system (CNS) tumors, BRAFV600E is mostly found in pediatric low-grade gliomas (pLGG, ~20%) and, less frequently, in pediatric high-grade gliomas (pHGG, 5-15%) and adult glioblastomas (GBM, ~5%). The integration of BRAF inhibitors (BRAFi) in the treatment of patients with gliomas brought a paradigm shift to clinical care. However, not all patients benefit from treatment due to intrinsic or acquired resistance to BRAF inhibition. Defining predictors of response, as well as developing strategies to prevent resistance to BRAFi and overcome post-BRAFi tumor progression/rebound growth are some of the main challenges at present in the field. In this review, we outline current achievements and limitations of BRAF inhibition in gliomas, with a special focus on potential mechanisms of resistance. We discuss future directions of targeted therapy for BRAFV600E mutated gliomas, highlighting how insights into resistance to BRAFi could be leveraged to improve outcomes.
Collapse
Affiliation(s)
- Monica Capogiri
- Department of Oncology and Children’s Research Center, University Children’s Hospital of Zurich, Zurich, Switzerland
| | - Andrea J. De Micheli
- Department of Oncology and Children’s Research Center, University Children’s Hospital of Zurich, Zurich, Switzerland
| | - Alvaro Lassaletta
- Department of Pediatric Hematology and Oncology, Hospital Universitario Niño Jesús, Madrid, Spain
| | - Denise P. Muñoz
- Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States
| | - Jean-Philippe Coppé
- Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States
| | - Sabine Mueller
- Department of Oncology and Children’s Research Center, University Children’s Hospital of Zurich, Zurich, Switzerland,Department of Neurology, Neurosurgery and Pediatrics, University of California, San Francisco, United States
| | - Ana S. Guerreiro Stucklin
- Department of Oncology and Children’s Research Center, University Children’s Hospital of Zurich, Zurich, Switzerland,*Correspondence: Ana S. Guerreiro Stucklin,
| |
Collapse
|
10
|
Pratap Reddy Gajulapalli V, Lee J, Sohn I. Ligand-Based Pharmacophore Modelling in Search of Novel Anaplastic Lymphoma Kinase Inhibitors. RESULTS IN CHEMISTRY 2022. [DOI: 10.1016/j.rechem.2022.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
11
|
Hardy S, Choo YM, Hamann M, Cray J. Manzamine-A Alters In Vitro Calvarial Osteoblast Function. Mar Drugs 2022; 20:647. [PMID: 36286470 PMCID: PMC9604769 DOI: 10.3390/md20100647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
Manzamine-A is a marine-derived alkaloid which has anti-viral and anti-proliferative properties and is currently being investigated for its efficacy in the treatment of certain viruses (malaria, herpes, HIV-1) and cancers (breast, cervical, colorectal). Manzamine-A has been found to exert effects via modulation of SIX1 gene expression, a gene critical to craniofacial development via the WNT, NOTCH, and PI3K/AKT pathways. To date little work has focused on Manzamine-A and how its use may affect bone. We hypothesize that Manzamine-A, through SIX1, alters bone cell activity. Here, we assessed the effects of Manzamine-A on cells that are responsible for the generation of bone, pre-osteoblasts and osteoblasts. PCR, qrtPCR, MTS cell viability, Caspase 3/7, and functional assays were used to test the effects of Manzamine-A on these cells. Our data suggests Six1 is highly expressed in osteoblasts and their progenitors. Further, osteoblast progenitors and osteoblasts exhibit great sensitivity to Manzamine-A treatment exhibited by a significant decrease in cell viability, increase in cellular apoptosis, and decrease in alkaline phosphatase activity. In silico binding experiment showed that manzamine A potential as an inhibitor of cell proliferation and survival proteins, i.e., Iκb, JAK2, AKT, PKC, FAK, and Bcl-2. Overall, our data suggests Manzamine-A may have great effects on bone health overall and may disrupt skeletal development, homeostasis, and repair.
Collapse
Affiliation(s)
- Samantha Hardy
- Department of Biomedical Education and Anatomy, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Yeun-Mun Choo
- Chemistry Department, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Mark Hamann
- Departments of Drug Discovery and Biomedical Sciences and Public Health, Colleges of Pharmacy and Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - James Cray
- Department of Biomedical Education and Anatomy, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Biosciences, The Ohio State College of Dentistry, Columbus, OH 43210, USA
| |
Collapse
|
12
|
Wang Y, Chen R, Hu Y, Jiao S, Zou Z. Synthesis, Molecular Docking Study, and Anticancer Activity of 7-Azaindole-1,2,3-triazol Bearing N-Benzamide Derivatives. RUSS J GEN CHEM+ 2022. [DOI: 10.1134/s1070363222100255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
13
|
Li X, Li M, Huang M, Lin Q, Fang Q, Liu J, Chen X, Liu L, Zhan X, Shan H, Lu D, Li Q, Li Z, Zhu X. The multi-molecular mechanisms of tumor-targeted drug resistance in precision medicine. Biomed Pharmacother 2022; 150:113064. [PMID: 35658234 DOI: 10.1016/j.biopha.2022.113064] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 11/02/2022] Open
Abstract
Clinically, cancer drug therapy is still dominated by chemotherapy drugs. Although the emergence of targeted drugs has greatly improved the survival rate of patients with advanced cancer, drug resistance has always been a difficult problem in clinical cancer treatment. At the current level of medicine, most drugs cannot escape the fate of drug resistance. With the emergence and development of gene detection, liquid biopsy ctDNA technology, and single-cell sequencing technology, the molecular mechanism of tumor drug resistance has gradually emerged. Drugs can also be updated in response to drug resistance mechanisms and bring higher survival benefits. The use of new drugs often leads to new mechanisms of resistance. In this review, the multi-molecular mechanisms of drug resistance are introduced, and the overcoming of drug resistance is discussed from the perspective of the tumor microenvironment.
Collapse
Affiliation(s)
- Xinming Li
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China; Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Mingdong Li
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Meiying Huang
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Qianyi Lin
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Qiuping Fang
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Jianjiang Liu
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Xiaohui Chen
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Lin Liu
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Xuliang Zhan
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Huisi Shan
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Deshuai Lu
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Qinlan Li
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Zesong Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors,Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China.
| | - Xiao Zhu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China; Cancer Research Center, Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
14
|
de Sá ÉRA, Souza JL, Costa RKM, Barros RO, de Lima CEB, Lima FDCA, Ramos RM. Computational investigation of the alkaloids of Pilocarpus microphyllus species as phytopharmaceuticals for the inhibition of sterol 14α-demethylase protease of Trypanosoma cruzi. J Biomol Struct Dyn 2022; 41:2555-2573. [PMID: 35132947 DOI: 10.1080/07391102.2022.2035819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Trypanosoma cruzi is a protozoan transmitted by the insect Triatoma infestans, popularly known as kissing bug. This protozoan causes the Chagas disease, a Neglected Tropical Disease. This study aimed to investigate, through DFT method and B3LYP hybrid functional, the physicochemical, pharmacokinetic, and pharmacodynamic properties of the alkaloids present in the leaves of the species Pilocarpus microphyllus (jaborandi) as a potential inhibitory activity on the protease sterol 14α-demethylase of T. cruzi associated with the techniques of molecular docking, molecular dynamics, MM-PBSA and ADMET predictions. The molecules of isopilosine, epiisopiloturine, epiisopilosine, and pilosine showed up the lowest binding energies by molecular docking, good human intestinal absorption, low penetration in the blood-brain barrier, antiprotozoal and anticarcinogenic activities in ADMET studies. It has been observed a better binding affinity of the sterol 14α-demethylase protease with isopilosine in molecular dynamics and MM-PBSA studies, which indicates it as a potential drug candidate for Chagas disease.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ézio R. A. de Sá
- Federal Institute of Education, Science and Technology of Piauí, IFPI, Picos, Piauí, Brazil
- Graduate Program in Chemistry, Federal University of Piauí, PPGQ/UFPI, Teresina, Piauí, Brazil
- Research Laboratory of the Computational Quantum Chemistry and Drug Planning Group, Chemistry Department, State University of Piauí, GQQC&PF/UESPI, Teresina, Piauí, Brazil
- Research Laboratory in Information Systems, Information Department, Environment, Health and Food Production, Federal Institute of Education, Science and Technology of Piauí, LaPeSI/IFPI, Teresina, Piauí, Brazil
| | - Janilson L. Souza
- Federal Institute of Education, Science and Technology of Maranhão, IFMA, Bacabal, Maranhão, Brazil
| | - Rayla K. M. Costa
- Research Laboratory of the Computational Quantum Chemistry and Drug Planning Group, Chemistry Department, State University of Piauí, GQQC&PF/UESPI, Teresina, Piauí, Brazil
| | - Rômulo O. Barros
- Research Laboratory in Information Systems, Information Department, Environment, Health and Food Production, Federal Institute of Education, Science and Technology of Piauí, LaPeSI/IFPI, Teresina, Piauí, Brazil
| | - Carlos E. B. de Lima
- University Hospital, Federal University of Piauí, Discipline of Cardiology, Department of General Practice - Cardiology and Health Sciences Center, DCG/CCS/UFPI, Teresina, Piauí, Brazil
| | - Francisco das C. A. Lima
- Research Laboratory of the Computational Quantum Chemistry and Drug Planning Group, Chemistry Department, State University of Piauí, GQQC&PF/UESPI, Teresina, Piauí, Brazil
| | - Ricardo M. Ramos
- Research Laboratory in Information Systems, Information Department, Environment, Health and Food Production, Federal Institute of Education, Science and Technology of Piauí, LaPeSI/IFPI, Teresina, Piauí, Brazil
| |
Collapse
|
15
|
Ravanmehr V, Blau H, Cappelletti L, Fontana T, Carmody L, Coleman B, George J, Reese J, Joachimiak M, Bocci G, Hansen P, Bult C, Rueter J, Casiraghi E, Valentini G, Mungall C, Oprea TI, Robinson PN. Supervised learning with word embeddings derived from PubMed captures latent knowledge about protein kinases and cancer. NAR Genom Bioinform 2021; 3:lqab113. [PMID: 34888523 PMCID: PMC8652379 DOI: 10.1093/nargab/lqab113] [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: 07/10/2021] [Revised: 10/14/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
Inhibiting protein kinases (PKs) that cause cancers has been an important topic in cancer therapy for years. So far, almost 8% of >530 PKs have been targeted by FDA-approved medications, and around 150 protein kinase inhibitors (PKIs) have been tested in clinical trials. We present an approach based on natural language processing and machine learning to investigate the relations between PKs and cancers, predicting PKs whose inhibition would be efficacious to treat a certain cancer. Our approach represents PKs and cancers as semantically meaningful 100-dimensional vectors based on word and concept neighborhoods in PubMed abstracts. We use information about phase I-IV trials in ClinicalTrials.gov to construct a training set for random forest classification. Our results with historical data show that associations between PKs and specific cancers can be predicted years in advance with good accuracy. Our tool can be used to predict the relevance of inhibiting PKs for specific cancers and to support the design of well-focused clinical trials to discover novel PKIs for cancer therapy.
Collapse
Affiliation(s)
- Vida Ravanmehr
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Hannah Blau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Luca Cappelletti
- AnacletoLab, Dipartimento di Informatica, Università degli Studi di Milano, Italy
| | - Tommaso Fontana
- AnacletoLab, Dipartimento di Informatica, Università degli Studi di Milano, Italy
| | - Leigh Carmody
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Ben Coleman
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
- University of Connecticut Health Center, Department of Genetics and Genome Sciences, Farmington, CT 06030, USA
| | - Joshy George
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Justin Reese
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
| | - Marcin Joachimiak
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
| | - Giovanni Bocci
- Department of Internal Medicine and UNM Comprehensive Cancer Center, UNM School of, Medicine, Albuquerque, NM 87102, USA
| | - Peter Hansen
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Carol Bult
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Jens Rueter
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Elena Casiraghi
- AnacletoLab, Dipartimento di Informatica, Università degli Studi di Milano, Italy
| | - Giorgio Valentini
- AnacletoLab, Dipartimento di Informatica, Università degli Studi di Milano, Italy
| | - Christopher Mungall
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94710, USA
| | - Tudor I Oprea
- Department of Internal Medicine and UNM Comprehensive Cancer Center, UNM School of, Medicine, Albuquerque, NM 87102, USA
| | - Peter N Robinson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, CT 06032, USA
| |
Collapse
|
16
|
Balogun TA, Ige OM, Alausa AO, Onyeani CO, Tiamiyu ZA, Omoboyowa DA, Saibu OA, Abdullateef OT. Receptor tyrosine kinases as a therapeutic target by natural compounds in cancer treatment. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00346-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Receptor tyrosine kinases (RTKs) are single-pass transmembrane proteins that play significant roles in regulating cellular processes, including cell division and growth. Overexpression and mutations of RTKs have been found in clinical manifestations of different forms of cancer. Therefore, RTKs have received considerable interest as a therapeutic biomarker in the treatment of cancer cells.
Main body of the abstract
Comprehensive data on RTKs, pharmacological and biological properties of natural compounds were systematically searched up to 2021 using relevant keywords from various databases, such as Google Scholar, PubMed, Web of Science, and Scopus. The scientific search by various standard electronic resources and databases unveils the effectiveness of medicinal plants in the treatment of various cancers. In vitro and in vivo studies suggested that bioactive compounds such as flavonoids, phenols, alkaloids, and many others can be used pharmacologically as RTKs inhibitors (RTKI) either by competing with ATP at the ATP binding site of the tyrosine kinase domain or competing for the receptor extracellular domain. Additionally, studies conducted on animal models indicated that inhibition of RTKs catalytic activity by natural compounds is one of the most effective ways to block the activation of RTKs signaling cascades, thereby hampering the proliferation of cancer cells. Furthermore, various pharmacological experiments, transcriptomic, and proteomic data also reported that cancer cells treated with different plants extracts or isolated phytochemicals exhibited better anticancer properties with minimal side effects than synthetic drugs. Clinically, natural compounds have demonstrated significant anti-proliferative effect via induction of cell apoptosis in cancer cell lines.
Short conclusion
An in-depth knowledge of the mechanism of inhibition and structural characterization of RTKs is important to the design of novel and selective RTKIs. This review focuses on the molecular mechanisms and structures of natural compounds RTKI targeting vascular endothelial growth factor, epidermal growth factor receptor, insulin receptor, and platelet-derived growth factor while also giving future directions to ameliorate the scientific burden of cancer.
Graphic abstract
Collapse
|
17
|
Liu X, Fields R, Schweppe DK, Paulo JA. Strategies for mass spectrometry-based phosphoproteomics using isobaric tagging. Expert Rev Proteomics 2021; 18:795-807. [PMID: 34652972 DOI: 10.1080/14789450.2021.1994390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Protein phosphorylation is a primary mechanism of signal transduction in cellular systems. Isobaric tagging can be used to investigate alterations in phosphorylation events in sample multiplexing experiments where quantification extends across all conditions. As such, innovations in tandem mass tag methods can facilitate the expansion of the depth and breadth of phosphoproteomic analyses. AREAS COVERED This review discusses the current state of tandem mass tag-centric phosphoproteomics and highlights advances in reagent chemistry, instrumentation, data acquisition, and data analysis. We stress that approaches for phosphoproteomic investigations require high-specificity enrichment, sensitive detection, and accurate phosphorylation site localization. EXPERT OPINION Tandem mass tag-centric phosphoproteomics will continue to be an important conduit for our understanding of signal transduction in living organisms. We anticipate that progress in phosphopeptide enrichment methodologies, enhancements in instrumentation and data acquisition technologies, and further refinements in analytical strategies will be key to the discovery of biologically relevant findings from phosphoproteomics studies.
Collapse
Affiliation(s)
- Xinyue Liu
- Department of Cell Biology, Harvard Medical School, Boston, USA
| | - Rose Fields
- Department of Genome Sciences, University of Washington, Seattle, USA
| | - Devin K Schweppe
- Department of Genome Sciences, University of Washington, Seattle, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, USA
| |
Collapse
|
18
|
Marek L, Váňa J, Svoboda J, Hanusek J. Synthesis of the Kinase Inhibitors Nintedanib, Hesperadin, and Their Analogues Using the Eschenmoser Coupling Reaction. J Org Chem 2021; 86:10621-10629. [PMID: 34269051 DOI: 10.1021/acs.joc.1c01269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A novel synthetic approach involving an Eschenmoser coupling reaction of substituted 3-bromooxindoles (H, 6-Cl, 6-COOMe, 5-NO2) with two substituted thiobenzanilides in dimethylformamide or acetonitrile was used for the synthesis of eight kinase inhibitors including Nintedanib and Hesperadin in yields exceeding 76%. Starting compounds for the synthesis are also easily available in good yields. 3-Bromooxindoles were prepared either from corresponding isatins using a three-step synthesis in an average overall yield of 65% or by direct bromination of oxindoles (yield of 65-86%). Starting N-(4-piperidin-1-ylmethyl-phenyl)-thiobenzamide was prepared by thionation of the corresponding benzanilide in an 86% yield and N-methyl-N-(4-thiobenzoylaminophenyl)-2-(4-methylpiperazin-1-yl)acetamide was prepared by thioacylation of the corresponding aniline with methyl dithiobenzoate in an 86% yield.
Collapse
Affiliation(s)
- Lukáš Marek
- Institute of Organic Chemistry and Technology, Faculty of Chemical Technology, University of Pardubice, Studentská 573, CZ-532 10 Pardubice, The Czech Republic
| | - Jiří Váňa
- Institute of Organic Chemistry and Technology, Faculty of Chemical Technology, University of Pardubice, Studentská 573, CZ-532 10 Pardubice, The Czech Republic
| | - Jan Svoboda
- Institute of Organic Chemistry and Technology, Faculty of Chemical Technology, University of Pardubice, Studentská 573, CZ-532 10 Pardubice, The Czech Republic
| | - Jiří Hanusek
- Institute of Organic Chemistry and Technology, Faculty of Chemical Technology, University of Pardubice, Studentská 573, CZ-532 10 Pardubice, The Czech Republic
| |
Collapse
|
19
|
Pacini L, Jenks AD, Lima NC, Huang PH. Targeting the Fibroblast Growth Factor Receptor (FGFR) Family in Lung Cancer. Cells 2021; 10:1154. [PMID: 34068816 PMCID: PMC8151052 DOI: 10.3390/cells10051154] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the most common cause of cancer-related deaths globally. Genetic alterations, such as amplifications, mutations and translocations in the fibroblast growth factor receptor (FGFR) family have been found in non-small cell lung cancer (NSCLC) where they have a role in cancer initiation and progression. FGFR aberrations have also been identified as key compensatory bypass mechanisms of resistance to targeted therapy against mutant epidermal growth factor receptor (EGFR) and mutant Kirsten rat sarcoma 2 viral oncogene homolog (KRAS) in lung cancer. Targeting FGFR is, therefore, of clinical relevance for this cancer type, and several selective and nonselective FGFR inhibitors have been developed in recent years. Despite promising preclinical data, clinical trials have largely shown low efficacy of these agents in lung cancer patients with FGFR alterations. Preclinical studies have highlighted the emergence of multiple intrinsic and acquired resistance mechanisms to FGFR tyrosine kinase inhibitors, which include on-target FGFR gatekeeper mutations and activation of bypass signalling pathways and alternative receptor tyrosine kinases. Here, we review the landscape of FGFR aberrations in lung cancer and the array of targeted therapies under clinical evaluation. We also discuss the current understanding of the mechanisms of resistance to FGFR-targeting compounds and therapeutic strategies to circumvent resistance. Finally, we highlight our perspectives on the development of new biomarkers for stratification and prediction of FGFR inhibitor response to enable personalisation of treatment in patients with lung cancer.
Collapse
Affiliation(s)
| | | | | | - Paul H. Huang
- Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK; (L.P.); (A.D.J.); (N.C.L.)
| |
Collapse
|
20
|
Geskovski N, Matevska-Geshkovska N, Dimchevska Sazdovska S, Glavas Dodov M, Mladenovska K, Goracinova K. The impact of molecular tumor profiling on the design strategies for targeting myeloid leukemia and EGFR/CD44-positive solid tumors. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:375-401. [PMID: 33981532 PMCID: PMC8093552 DOI: 10.3762/bjnano.12.31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 04/12/2021] [Indexed: 05/21/2023]
Abstract
Nanomedicine has emerged as a novel cancer treatment and diagnostic modality, whose design constantly evolves towards increasing the safety and efficacy of the chemotherapeutic and diagnostic protocols. Molecular diagnostics, which create a great amount of data related to the unique molecular signatures of each tumor subtype, have emerged as an important tool for detailed profiling of tumors. They provide an opportunity to develop targeting agents for early detection and diagnosis, and to select the most effective combinatorial treatment options. Alongside, the design of the nanoscale carriers needs to cope with novel trends of molecular screening. Also, multiple targeting ligands needed for robust and specific interactions with the targeted cell populations have to be introduced, which should result in substantial improvements in safety and efficacy of the cancer treatment. This article will focus on novel design strategies for nanoscale drug delivery systems, based on the unique molecular signatures of myeloid leukemia and EGFR/CD44-positive solid tumors, and the impact of novel discoveries in molecular tumor profiles on future chemotherapeutic protocols.
Collapse
Affiliation(s)
- Nikola Geskovski
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Nadica Matevska-Geshkovska
- Center for Pharmaceutical Biomolecular Analyses, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Simona Dimchevska Sazdovska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
- Department of Nanobiotechnology, Institute of Biotechnology, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Marija Glavas Dodov
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Kristina Mladenovska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Katerina Goracinova
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
- College of Pharmacy, Qatar University, PO Box 2713, Doha, Qatar
| |
Collapse
|
21
|
Lee YS, Lee JY, Song SH, Kim DM, Lee JW, Chi XZ, Ito Y, Bae SC. K-Ras-Activated Cells Can Develop into Lung Tumors When Runx3-Mediated Tumor Suppressor Pathways Are Abrogated. Mol Cells 2020; 43:889-897. [PMID: 33115981 PMCID: PMC7604022 DOI: 10.14348/molcells.2020.0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 11/27/2022] Open
Abstract
K-RAS is frequently mutated in human lung adenocarcinomas (ADCs), and the p53 pathway plays a central role in cellular defense against oncogenic K-RAS mutation. However, in mouse lung cancer models, oncogenic K-RAS mutation alone can induce ADCs without p53 mutation, and loss of p53 does not have a significant impact on early K-RAS-induced lung tumorigenesis. These results raise the question of how K-RAS-activated cells evade oncogene surveillance mechanisms and develop into lung ADCs. RUNX3 plays a key role at the restriction (R)-point, which governs multiple tumor suppressor pathways including the p14ARF-p53 pathway. In this study, we found that K-RAS activation in a very limited number of cells, alone or in combination with p53 inactivation, failed to induce any pathologic lesions for up to 1 year. By contrast, when Runx3 was inactivated and K-RAS was activated by the same targeting method, lung ADCs and other tumors were rapidly induced. In a urethane-induced mouse lung tumor model that recapitulates the features of K-RAS-driven human lung tumors, Runx3 was inactivated in both adenomas (ADs) and ADCs, whereas K-RAS was activated only in ADCs. Together, these results demonstrate that the R-point-associated oncogene surveillance mechanism is abrogated by Runx3 inactivation in AD cells and these cells cannot defend against K-RAS activation, resulting in the transition from AD to ADC. Therefore, K-RAS-activated lung epithelial cells do not evade oncogene surveillance mechanisms; instead, they are selected if they occur in AD cells in which Runx3 has been inactivated.
Collapse
Affiliation(s)
- You-Soub Lee
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| | - Ja-Yeol Lee
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| | - Soo-Hyun Song
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| | - Da-Mi Kim
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| | - Jung-Won Lee
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| | - Xin-Zi Chi
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| | - Yoshiaki Ito
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599
| | - Suk-Chul Bae
- Department of Biochemistry, School of Medicine, Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
22
|
Chung CT, Yeh KC, Lee CH, Chen YY, Ho PJ, Chang KY, Chen CH, Lai YK, Chen CT. Molecular profiling of afatinib-resistant non-small cell lung cancer cells in vivo derived from mice. Pharmacol Res 2020; 161:105183. [PMID: 32896579 DOI: 10.1016/j.phrs.2020.105183] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 01/28/2023]
Abstract
Non-small-cell lung cancer (NSCLC) is a leading cause of cancer-related death worldwide. NSCLC patients with overexpressed or mutated epidermal growth factor receptor (EGFR) related to disease progression are treated with EGFR-tyrosine kinase inhibitors (EGFR-TKIs). Acquired drug resistance after TKI treatments has been a major focus for development of NSCLC therapies. This study aimed to establish afatinib-resistant cell lines from which afatinib resistance-associated genes are identified and the underlying mechanisms of multiple-TKI resistance in NSCLC can be further investigated. Nude mice bearing subcutaneous NSCLC HCC827 tumors were administered with afatinib at different dose intensities (5-100 mg/kg). We established three HCC827 sublines resistant to afatinib (IC50 > 1 μM) with cross-resistance to gefitinib (IC50 > 5 μM). cDNA microarray revealed several of these sublines shared 27 up- and 13 down-regulated genes. The mRNA expression of selective novel genes - such as transmembrane 4 L six family member 19 (TM4SF19), suppressor of cytokine signaling 2 (SOCS2), and quinolinate phosphoribosyltransferase (QPRT) - are responsive to afatinib treatments only at high concentrations. Furthermore, c-MET amplification and activations of a subset of tyrosine kinase receptors were observed in all three resistant cells. PHA665752, a c-MET inhibitor, remarkably increased the sensitivity of these resistant cells to afatinib (IC50 = 12-123 nM). We established afatinib-resistant lung cancer cell lines and here report genes associated with afatinib resistance in human NSCLC. These cell lines and the identified genes serve as useful investigational tools, prognostic biomarkers of TKI therapies, and promising molecule targets for development of human NSCLC therapeutics.
Collapse
Affiliation(s)
- Cheng-Ta Chung
- Graduate Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan; Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Kai-Chia Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chia-Huei Lee
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yun-Yu Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Pai-Jiun Ho
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Kai-Yen Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chieh-Hsin Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yiu-Kay Lai
- Graduate Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan.
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan.
| |
Collapse
|
23
|
The Multidrug Resistance-Reversing Activity of a Novel Antimicrobial Peptide. Cancers (Basel) 2020; 12:cancers12071963. [PMID: 32707710 PMCID: PMC7409168 DOI: 10.3390/cancers12071963] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/07/2020] [Accepted: 07/17/2020] [Indexed: 11/16/2022] Open
Abstract
The overexpression of ATP-binding cassette (ABC) transporters is a common cause of multidrug resistance (MDR) in cancers. The intracellular drug concentration of cancer cells can be decreased relative to their normal cell counterparts due to increased expression of ABC transporters acting as efflux pumps of anticancer drugs. Over the past decades, antimicrobial peptides have been investigated as a new generation of anticancer drugs and some of them were reported to have interactions with ABC transporters. In this article, we investigated several novel antimicrobial peptides to see if they could sensitize ABCB1-overexpressing cells to the anticancer drugs paclitaxel and doxorubicin, which are transported by ABCB1. It was found that peptide XH-14C increased the intracellular accumulation of ABCB1 substrate paclitaxel, which demonstrated that XH-14C could reverse ABCB1-mediated MDR. Furthermore, XH-14C could stimulate the ATPase activity of ABCB1 and the molecular dynamic simulation revealed a stable binding pose of XH-14C-ABCB1 complex. There was no change on the expression level or the location of ABCB1 transporter with the treatment of XH-14C. Our results suggest that XH-14C in combination with conventional anticancer agents could be used as a novel strategy for cancer treatment.
Collapse
|
24
|
Williams DE, Andersen RJ. Biologically active marine natural products and their molecular targets discovered using a chemical genetics approach. Nat Prod Rep 2020; 37:617-633. [PMID: 31750842 PMCID: PMC7874888 DOI: 10.1039/c9np00054b] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Covering: 2000 to 2019The discovery of new natural products that have some combination of unprecedented chemical structures, biological activities of therapeutic interest for urgent medical needs, and new molecular targets provides the fuel that sustains the vitality of natural products chemistry research. Unfortunately, finding these important new compounds is neither routine or trivial and a major challenge is finding effective discovery paradigms. This review presents examples that illustrate the effectiveness of a chemical genetics approach to marine natural product (MNP) discovery that intertwines compound discovery, molecular target identification, and phenotypic response/biological activity. The examples include MNPs that have complex unprecedented structures, new or understudied molecular targets, and potent biological activities of therapeutic interest. A variety of methods to identify molecular targets are also featured.
Collapse
Affiliation(s)
- David E Williams
- Departments of Chemistry and Earth, Ocean & Atmospheric Science, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada.
| | | |
Collapse
|
25
|
Shah AP, Patel CN. Virtual Screening of Novel Hybrid Non-Steroidal Anti-Inflammatory Drugs (NSAIDs): Exploring Multiple Targeted Cancer Therapy by an In Silico Approach. CURRENT CANCER THERAPY REVIEWS 2020. [DOI: 10.2174/1573394715666190618114748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Dual-targeting/Multi-targeting of oncoproteins by a single drug molecule
represents an efficient, logical and alternative approach to drug combinations. In silico methods
are useful tool for the search and design of selective multi-target agents.
Objective:
The objective of the present study was to design new hybrid compounds by linking the
main structural unit of the NSAIDs with the benzothiazole and thiadiazole ring and to discover
new hybrid NSAIDs as multi targeted anticancer agents through in silico approach.
Method:
Structure-based virtual screening was performed by applying ADMET filtration and
Glide docking using Virtual screening Workflow. The docking studies were performed on three
different types of receptors TNF-α, COX-II and protein kinase. Bioactivity prediction of screened
compounds were done using Molinspiration online software tool.
Results:
Out of 54 designed compounds eighteen were screened on the basis of binding affinity on various receptors and ADMET filtration. Bioactivity prediction reveals that screened compounds may act through kinase inhibition or enzyme inhibition. Compounds 2sa, 5sa, 6sa and 7sa shows higher binding affinity with all three receptors.
Conclusion:
The study concluded that compound 2sa, 5sa, 6sa, and 7sa could be further explored
for multiple targeted cancer therapy.
Collapse
Affiliation(s)
| | - Chhagan N. Patel
- Department of Pharmaceutical Chemistry, Shree Sarvajanik Pharmacy College, Mehsana, India
| |
Collapse
|
26
|
Wang H, Guan Z, Qiu J, Jia Y, Zeng C, Zhao Y. Novel method to identify group-specific non-catalytic pockets of human kinome for drug design. RSC Adv 2020; 10:2004-2015. [PMID: 35494619 PMCID: PMC9047066 DOI: 10.1039/c9ra07471f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/27/2019] [Indexed: 01/11/2023] Open
Abstract
Kinase proteins have been intensively investigated as drug targets for decades because of their crucial involvement in many biological pathways. We developed hybrid approach to identify non-catalytic pockets and will benefit the kinome drug design.
Collapse
Affiliation(s)
- Huiwen Wang
- Department of Physics
- Institute of Biophysics
- Central China Normal University
- Wuhan 430079
- China
| | - Zeyu Guan
- Department of Physics
- Institute of Biophysics
- Central China Normal University
- Wuhan 430079
- China
| | - Jiadi Qiu
- Department of Physics
- Institute of Biophysics
- Central China Normal University
- Wuhan 430079
- China
| | - Ya Jia
- Department of Physics
- Institute of Biophysics
- Central China Normal University
- Wuhan 430079
- China
| | - Chen Zeng
- Department of Physics
- Institute of Biophysics
- Central China Normal University
- Wuhan 430079
- China
| | - Yunjie Zhao
- Department of Physics
- Institute of Biophysics
- Central China Normal University
- Wuhan 430079
- China
| |
Collapse
|
27
|
Wang H, Qiu J, Liu H, Xu Y, Jia Y, Zhao Y. HKPocket: human kinase pocket database for drug design. BMC Bioinformatics 2019; 20:617. [PMID: 31783725 PMCID: PMC6884818 DOI: 10.1186/s12859-019-3254-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 11/15/2019] [Indexed: 01/06/2023] Open
Abstract
Background The kinase pocket structural information is important for drug discovery targeting cancer or other diseases. Although some kinase sequence, structure or drug databases have been developed, the databases cannot be directly used in the kinase drug study. Therefore, a comprehensive database of human kinase protein pockets is urgently needed to be developed. Results Here, we have developed HKPocket, a comprehensive Human Kinase Pocket database. This database provides sequence, structure, hydrophilic-hydrophobic, critical interactions, and druggability information including 1717 pockets from 255 kinases. We further divided these pockets into 91 pocket clusters using structural and position features in each kinase group. The pocket structural information would be useful for preliminary drug screening. Then, the potential drugs can be further selected and optimized by analyzing the sequence conservation, critical interactions, and hydrophobicity of identified drug pockets. HKPocket also provides online visualization and pse files of all identified pockets. Conclusion The HKPocket database would be helpful for drug screening and optimization. Besides, drugs targeting the non-catalytic pockets would cause fewer side effects. HKPocket is available at http://zhaoserver.com.cn/HKPocket/HKPocket.html.
Collapse
Affiliation(s)
- Huiwen Wang
- Department of Physics, Central China Normal University, Wuhan, 430079, China
| | - Jiadi Qiu
- Department of Physics, Central China Normal University, Wuhan, 430079, China
| | - Haoquan Liu
- Department of Physics, Central China Normal University, Wuhan, 430079, China
| | - Ying Xu
- Department of Physics, Central China Normal University, Wuhan, 430079, China
| | - Ya Jia
- Department of Physics, Central China Normal University, Wuhan, 430079, China
| | - Yunjie Zhao
- Department of Physics, Central China Normal University, Wuhan, 430079, China.
| |
Collapse
|
28
|
Golonko A, Lewandowska H, Świsłocka R, Jasińska U, Priebe W, Lewandowski W. Curcumin as tyrosine kinase inhibitor in cancer treatment. Eur J Med Chem 2019; 181:111512. [DOI: 10.1016/j.ejmech.2019.07.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022]
|
29
|
Hussain S, Singh A, Nazir SU, Tulsyan S, Khan A, Kumar R, Bashir N, Tanwar P, Mehrotra R. Cancer drug resistance: A fleet to conquer. J Cell Biochem 2019; 120:14213-14225. [PMID: 31037763 DOI: 10.1002/jcb.28782] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/08/2019] [Accepted: 01/14/2019] [Indexed: 12/18/2022]
Abstract
Cancer is a disease that claims millions of lives each year across the world. Despite advancement in technologies and therapeutics for treating the disease, these modes are often found to turn ineffective during the course of treatment. The resistance against drugs in cancer patients stems from multiple factors, which constitute genetic heterogeneity like gene mutations, tumor microenvironment, exosomes, miRNAs, high rate of drug efflux from cells, and so on. This review attempts to collate all such known and reported factors that influence cancer drug resistance and may help researchers with information that might be useful in developing better therapeutics in near future to enable better management of several cancers across the world.
Collapse
Affiliation(s)
- Showket Hussain
- Division of Cellular and Molecular Diagnostics, National Institute of Cancer Prevention and Research, Noida, India
| | - Ankita Singh
- Division of Cellular and Molecular Diagnostics, National Institute of Cancer Prevention and Research, Noida, India
| | - Sheeraz Un Nazir
- Division of Cellular and Molecular Diagnostics, National Institute of Cancer Prevention and Research, Noida, India
| | - Sonam Tulsyan
- Division of Preventive Oncology, National Institute of Cancer Prevention and Research, Noida, India
| | - Asiya Khan
- Department of Lab Oncology, AIIMS, New Delhi, India
| | - Ramesh Kumar
- Department of Biochemistry, Bundelkhand University, Jhansi, India
| | - Nasreena Bashir
- College of Applied Medicine, King Khalid University, Abha, Saudi Arabia
| | | | - Ravi Mehrotra
- Division of Preventive Oncology, National Institute of Cancer Prevention and Research, Noida, India
| |
Collapse
|
30
|
Jenardhanan P, Panneerselvam M, Mathur PP. Targeting Kinase Interaction Networks: A New Paradigm in PPI Based Design of Kinase Inhibitors. Curr Top Med Chem 2019; 19:467-485. [PMID: 31184298 DOI: 10.2174/1568026619666190304155711] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/20/2019] [Accepted: 02/06/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Kinases are key modulators in regulating diverse range of cellular activities and are an essential part of the protein-protein interactome. Understanding the interaction of kinases with different substrates and other proteins is vital to decode the cell signaling machinery as well as causative mechanism for disease onset and progression. OBJECTIVE The objective of this review is to present all studies on the structure and function of few important kinases and highlight the protein-protein interaction (PPI) mechanism of kinases and the kinase specific interactome databases and how such studies could be utilized to develop anticancer drugs. METHODS The article is a review of the detailed description of the various domains in kinases that are involved in protein-protein interactions and specific inhibitors developed targeting these PPI domains. RESULTS The review has surfaced in depth the interacting domains in key kinases and their features and the roles of PPI in the human kinome and the various signaling cascades that are involved in certain types of cancer. CONCLUSION The insight availed into the mechanism of existing peptide inhibitors and peptidomimetics against kinases will pave way for the design and generation of domain specific peptide inhibitors with better productivity and efficiency and the various software and servers available can be of great use for the identification and analysis of protein-protein interactions.
Collapse
Affiliation(s)
| | - Manivel Panneerselvam
- Department of Biotechnology, BJM School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Premendu P Mathur
- Department of Biochemistry & Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| |
Collapse
|
31
|
Jin J, Ye X, Boateng D, Dai K, Ye F, Du P, Yu H. Identification and characterization of potent and selective inhibitors targeting protein tyrosine phosphatase 1B (PTP1B). Bioorg Med Chem Lett 2019; 29:2358-2363. [PMID: 31221555 DOI: 10.1016/j.bmcl.2019.06.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 06/03/2019] [Accepted: 06/06/2019] [Indexed: 11/20/2022]
Abstract
Protein tyrosine phosphatase 1B (PTP1B) plays an important role in the negative regulation of insulin and leptin signaling. The development of small molecular inhibitors targeting PTP1B has been validated as a potential therapeutic strategy for Type 2 diabetes (T2D). In this work, we have identified a series of compounds containing dihydropyridine thione and particular chiral structure as novel PTP1B inhibitors. Among those, compound 4b showed moderate activity with IC50 value of 3.33 μM and meanwhile with good selectivity (>30-fold) against TCPTP. The further MOA study of PTP1B demonstrated that compounds 4b is a substrate-competitive inhibitor. The binding mode analysis suggested that compound 4b simultaneously occupies the active site and the second phosphotyrosine (pTyr) binding site of PTP1B. Furthermore, the cell viability assay of compound 4b showed tolerable cytotoxicity in L02 cells, thus 4b may be prospectively used to further in vivo study.
Collapse
Affiliation(s)
- Jia Jin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou 310018, China.
| | - Xiaoqing Ye
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materta Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Derrick Boateng
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Kaili Dai
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Fei Ye
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Pengfei Du
- Department of Endocrinology, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, China.
| | - Han Yu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China.
| |
Collapse
|
32
|
Coppé JP, Mori M, Pan B, Yau C, Wolf DM, Ruiz-Saenz A, Brunen D, Prahallad A, Cornelissen-Steijger P, Kemper K, Posch C, Wang C, Dreyer CA, Krijgsman O, Lee PRE, Chen Z, Peeper DS, Moasser MM, Bernards R, van 't Veer LJ. Mapping phospho-catalytic dependencies of therapy-resistant tumours reveals actionable vulnerabilities. Nat Cell Biol 2019; 21:778-790. [PMID: 31160710 DOI: 10.1038/s41556-019-0328-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 04/09/2019] [Indexed: 12/21/2022]
Abstract
Phosphorylation networks intimately regulate mechanisms of response to therapies. Mapping the phospho-catalytic profile of kinases in cells or tissues remains a challenge. Here, we introduce a practical high-throughput system to measure the enzymatic activity of kinases using biological peptide targets as phospho-sensors to reveal kinase dependencies in tumour biopsies and cell lines. A 228-peptide screen was developed to detect the activity of >60 kinases, including ABLs, AKTs, CDKs and MAPKs. Focusing on BRAFV600E tumours, we found mechanisms of intrinsic resistance to BRAFV600E-targeted therapy in colorectal cancer, including targetable parallel activation of PDPK1 and PRKCA. Furthermore, mapping the phospho-catalytic signatures of melanoma specimens identifies RPS6KB1 and PIM1 as emerging druggable vulnerabilities predictive of poor outcome in BRAFV600E patients. The results show that therapeutic resistance can be caused by the concerted upregulation of interdependent pathways. Our kinase activity-mapping system is a versatile strategy that innovates the exploration of actionable kinases for precision medicine.
Collapse
Affiliation(s)
- Jean-Philippe Coppé
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| | - Miki Mori
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.,Department of Breast Surgical Oncology, Showa University, Tokyo, Japan
| | - Bo Pan
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.,Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Christina Yau
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Denise M Wolf
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Ana Ruiz-Saenz
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Diede Brunen
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anirudh Prahallad
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Kristel Kemper
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Christian Posch
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.,Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany.,School of Medicine, Sigmund Freud University, Vienna, Austria
| | - Changjun Wang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.,Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Courtney A Dreyer
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Oscar Krijgsman
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Pei Rong Evelyn Lee
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Zhongzhong Chen
- The State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Daniel S Peeper
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mark M Moasser
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - René Bernards
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Laura J van 't Veer
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
33
|
Roberti M, Schipani F, Bagnolini G, Milano D, Giacomini E, Falchi F, Balboni A, Manerba M, Farabegoli F, De Franco F, Robertson J, Minucci S, Pallavicini I, Di Stefano G, Girotto S, Pellicciari R, Cavalli A. Rad51/BRCA2 disruptors inhibit homologous recombination and synergize with olaparib in pancreatic cancer cells. Eur J Med Chem 2019; 165:80-92. [DOI: 10.1016/j.ejmech.2019.01.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/03/2019] [Accepted: 01/04/2019] [Indexed: 10/27/2022]
|
34
|
|
35
|
Kaushal T, Srivastava G, Sharma A, Singh Negi A. An insight into medicinal chemistry of anticancer quinoxalines. Bioorg Med Chem 2018; 27:16-35. [PMID: 30502116 DOI: 10.1016/j.bmc.2018.11.021] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/06/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
Abstract
Quinoxalines are benzopyrazines containing benzene and pyrazine rings fused together. In the recent past, quinoxalines have attracted Medicinal Chemists considerably for their syntheses and chemistry due to their distinct pharmacological activities. Diverse synthetic protocols have been developed via multicomponent reactions, single pot synthesis and combinatorial approach using efficient catalysts, reagents, and nano-composites etc. Further, the versatility of the quinoxaline core and its reasonable chemical simplicity devise it extremely promising source of bioactive compounds. Therefore, a wide variety of bioactive quinoxalines has been realised as antitumour, antifungal, anti-inflammatory, antimicrobial, and antiviral agents. Already, a few of them are clinical drugs while many more are under various phases of clinical trials. Present review focuses on chemistry and pharmacology (both efficacy and safety) of quinoxalines and also provides some insight in to their structure-activity relationship.
Collapse
Affiliation(s)
- Tanu Kaushal
- CSIR-Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP), P.O. CIMAP, Kukrail Picnic Spot Road, Lucknow 226 015, UP, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India
| | - Gaurava Srivastava
- CSIR-Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP), P.O. CIMAP, Kukrail Picnic Spot Road, Lucknow 226 015, UP, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India
| | - Ashok Sharma
- CSIR-Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP), P.O. CIMAP, Kukrail Picnic Spot Road, Lucknow 226 015, UP, India
| | - Arvind Singh Negi
- CSIR-Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP), P.O. CIMAP, Kukrail Picnic Spot Road, Lucknow 226 015, UP, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India.
| |
Collapse
|
36
|
Deng S, Zhang G, Kuai J, Fan P, Wang X, Zhou P, Yang D, Zheng X, Liu X, Wu Q, Huang Y. Lentinan inhibits tumor angiogenesis via interferon γ and in a T cell independent manner. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:260. [PMID: 30373628 PMCID: PMC6206909 DOI: 10.1186/s13046-018-0932-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 10/17/2018] [Indexed: 12/15/2022]
Abstract
Background Antiangiogenic agents are commonly used in lung and colon cancer treatments, however, rapid development of drug resistance limits their efficacy. Methods Lentinan (LNT) is a biologically active compound extracted from Lentinus edodes. The effects of LNT on tumor angiogenesis were evaluated by immunohistochemistry in murine LAP0297 lung and CT26 colorectal tumor models. The impacts of LNT on immune cells and gene expression in tumor tissues were determined by flow cytometry, qPCR, and ELISA. Nude mice and IFNγ blockade were used to investigate the mechanism of LNT affecting on tumor angiogenesis. The data sets were compared using two-tailed student’s t tests or ANOVA. Results We found that LNT inhibited tumor angiogenesis and the growth of lung and colon cancers. LNT treatments elevated the expression of angiostatic factors such as IFNγ and also increased tumor infiltration of IFNγ-expressing T cells and myeloid cells. Interestingly, IFNγ blockade, but not T cell deficiency, reversed the effects of LNT treatments on tumor blood vessels. Moreover, long-lasting LNT administration persistently suppressed tumor angiogenesis and inhibited tumor growth. Conclusions LNT inhibits tumor angiogenesis by increasing IFNγ production and in a T cell-independent manner. Our findings suggest that LNT could be developed as a new antiangiogenic agent for long-term treatment of lung and colon cancers. Electronic supplementary material The online version of this article (10.1186/s13046-018-0932-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shengming Deng
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, 199 Ren-Ai Road, Suzhou, 215123, Jiangsu, China
| | - Guoxi Zhang
- Nanjing Luye Pharmaceutical Co., Ltd, Nanjing, 210061, Jiangsu, China
| | - Jiajie Kuai
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, 199 Ren-Ai Road, Suzhou, 215123, Jiangsu, China
| | - Peng Fan
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, 199 Ren-Ai Road, Suzhou, 215123, Jiangsu, China
| | - Xuexiang Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, 199 Ren-Ai Road, Suzhou, 215123, Jiangsu, China
| | - Pei Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, 199 Ren-Ai Road, Suzhou, 215123, Jiangsu, China
| | - Dan Yang
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Xichen Zheng
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, 199 Ren-Ai Road, Suzhou, 215123, Jiangsu, China
| | - Xiaomei Liu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, 199 Ren-Ai Road, Suzhou, 215123, Jiangsu, China
| | - Qunli Wu
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| | - Yuhui Huang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, 199 Ren-Ai Road, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
37
|
Zhi LQ, Yang YX, Yao SX, Qing Z, Ma JB. Identification of Novel Target for Osteosarcoma by Network Analysis. Med Sci Monit 2018; 24:5914-5924. [PMID: 30144309 PMCID: PMC6120164 DOI: 10.12659/msm.909973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Osteosarcoma (OS) is a highly complicated bone cancer involving imbalance of signaling transduction networks in cells. Development of new anti-osteosarcoma drugs is very challenging, mainly due to lack of known key targets. Material/Method In this study, we attempted to reveal more promising targets for drug design by “Target-Pathway” network analysis, providing the new therapeutic strategy of osteosarcoma. The potential targets used for the treatment of OS were selected from 4 different sources: DrugBank, TCRD database, dbDEMC database, and recent scientific literature papers. Cytoscape was used for the establishment of the “Target-Pathway” network. Results The obtained results suggest that tankyrase 2 (TNKS2) might be a very good potential protein target for the treatment of osteosarcoma. An in vitro MTT assay proved that it is an available option against OS by targeting the TNKS2 protein. Subsequently, cell cycle and apoptosis assay by flow cytometry showed the TNKS2 inhibitor can obviously induce cell cycle arrest, apoptosis, and mitotic cell death. Conclusions Tankyrase 2 (TNKS2), a member of the multifunctional poly(ADP-ribose) polymerases (PARPs), could be a very useful protein target for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Li-Qiang Zhi
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Yi-Xin Yang
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (mainland)
| | - Shu-Xin Yao
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Zhong Qing
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Jian-Bing Ma
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
38
|
Vishwakarma V, New J, Kumar D, Snyder V, Arnold L, Nissen E, Hu Q, Cheng N, Miller D, Thomas AR, Shnayder Y, Kakarala K, Tsue TT, Girod DA, Thomas SM. Potent Antitumor Effects of a Combination of Three Nutraceutical Compounds. Sci Rep 2018; 8:12163. [PMID: 30111862 PMCID: PMC6093880 DOI: 10.1038/s41598-018-29683-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 07/11/2018] [Indexed: 01/05/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is associated with low survival, and the current aggressive therapies result in high morbidity. Nutraceuticals are dietary compounds with few side effects. However, limited antitumor efficacy has restricted their application for cancer therapy. Here, we examine combining nutraceuticals, establishing a combination therapy that is more potent than any singular component, and delineate the mechanism of action. Three formulations were tested: GZ17-S (combined plant extracts from Arum palaestinum, Peganum harmala and Curcuma longa); GZ17-05.00 (16 synthetic components of GZ17-S); and GZ17-6.02 (3 synthetic components of GZ17S; curcumin, harmine and isovanillin). We tested the formulations on HNSCC proliferation, migration, invasion, angiogenesis, macrophage viability and infiltration into the tumor and tumor apoptosis. GZ17-6.02, the most effective formulation, significantly reduced in vitro assessments of HNSCC progression. When combined with cisplatin, GZ17-6.02 enhanced anti-proliferative effects. Molecular signaling cascades inhibited by GZ17-6.02 include EGFR, ERK1/2, and AKT, and molecular docking analyses demonstrate GZ17-6.02 components bind at distinct binding sites. GZ17-6.02 significantly inhibited growth of HNSCC cell line, patient-derived xenografts, and murine syngeneic tumors in vivo (P < 0.001). We demonstrate GZ17-6.02 as a highly effective plant extract combination and pave the way for future clinical application in HNSCC.
Collapse
Affiliation(s)
- Vikalp Vishwakarma
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Jacob New
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Dhruv Kumar
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.,Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Uttar Pradesh, Noida, India
| | - Vusala Snyder
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Levi Arnold
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Emily Nissen
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Qingting Hu
- Department of Pathology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Nikki Cheng
- Department of Pathology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - David Miller
- Department of Mechanical Engineering Technology, Pittsburg State University, Pittsburg, KS, 66762, USA
| | - Ahia Rael Thomas
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Yelizaveta Shnayder
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Kiran Kakarala
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Terance Ted Tsue
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Douglas A Girod
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Sufi Mary Thomas
- Department of Otolaryngology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA. .,Department of Anatomy and Cell Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA. .,Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| |
Collapse
|
39
|
Zhang Z, Guo X, To KK, Chen Z, Fang X, Luo M, Ma C, Xu J, Yan S, Fu L. Olmutinib (HM61713) reversed multidrug resistance by inhibiting the activity of ATP-binding cassette subfamily G member 2 in vitro and in vivo. Acta Pharm Sin B 2018; 8:563-574. [PMID: 30109181 PMCID: PMC6089862 DOI: 10.1016/j.apsb.2018.06.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/30/2018] [Accepted: 06/04/2018] [Indexed: 01/29/2023] Open
Abstract
Overexpressing of ATP-binding cassette (ABC) transporters is the essential cause of multidrug resistance (MDR), which is a significant hurdle to the success of chemotherapy in many cancers. Therefore, inhibiting the activity of ABC transporters may be a logical approach to circumvent MDR. Olmutinib is an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI), which has been approved in South Korea for advanced EGFR T790M-positive non-small cell lung cancer (NSCLC). Here, we found that olmutinib significantly increased the sensitivity of chemotherapy drug in ABCG2-overexpressing cells. Furthermore, olmutinib could also increase the retention of doxorubicin (DOX) and rhodamine 123 (Rho 123) in ABC transporter subfamily G member 2 (ABCG2)-overexpressing cells. In addition, olmutinib was found to stimulate ATPase activity and inhibit photolabeling of ABCG2 with [125I]-iodoarylazidoprazosin (IAAP). However, olmutinib neither altered ABCG2 expression at protein and mRNA levels nor blocked EGFR, Her-2 downstream signaling of AKT and ERK. Importantly, olmutinib enhanced the efficacy of topotecan on the inhibition of S1-MI-80 cell xenograft growth. All the results suggest that olmutinib reverses ABCG2-mediated MDR by binding to ATP bind site of ABCG2 and increasing intracellular chemotherapeutic drug accumulation. Our findings encouraged to further clinical investigation on combination therapy of olmutinib with conventional chemotherapeutic drugs in ABCG2-overexpressing cancer patients.
Collapse
Key Words
- ABC, adenosine triphosphate (ATP)-binding cassette
- ABCG2
- ABCG2, ABC transporter subfamily G member 2
- ATPase
- Chemotherapy
- DDP, cisplatin
- DMEM, Dulbecco׳s modified Eagle׳s medium
- DMSO, dimethyl sulfoxide
- DOX, doxorubicin
- FTC, fumitremorgin C
- IAAP, iodoarylazidoprazosin
- MDR, multidrug resistance
- MTT, 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazoliumbromide
- MX, methotrexate
- Multidrug resistance
- Olmutinib
- PCR, polymerase chain reaction
- Rho 123, rhodamine 123
- TKI, tyrosine kinase inhibitor
- Tyrosine kinase inhibitor
- VRP, verapamil
Collapse
|
40
|
Helgadottir H, Rocha Trocoli Drakensjö I, Girnita A. Personalized Medicine in Malignant Melanoma: Towards Patient Tailored Treatment. Front Oncol 2018; 8:202. [PMID: 29946532 PMCID: PMC6006716 DOI: 10.3389/fonc.2018.00202] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/21/2018] [Indexed: 12/24/2022] Open
Abstract
Despite enormous international efforts, skin melanoma is still a major clinical challenge. Melanoma takes a top place among the most common cancer types and it has one of the most rapidly increasing incidences in many countries around the world. Until recent years, there have been limited options for effective systemic treatment of disseminated melanoma. However, lately, we have experienced a rapid advancement in the understanding of the biology and molecular background of the disease. This has led to new molecular classifications and the development of more effective targeted therapies adapted to distinct melanoma subtypes. Not only are these treatments more effective but they can be rationally prescribed to the patients standing to benefit. As such, melanoma management has now become one of the most developed for personalized medicine. The aim of the present paper is to summarize the current knowledge on melanoma molecular classification, predictive markers, combination therapies, as well as emerging new treatments.
Collapse
Affiliation(s)
- Hildur Helgadottir
- Skin Tumor Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden.,Cancer Centrum Karolinska, Karolinska Institutet Stockholm, Stockholm, Sweden
| | - Iara Rocha Trocoli Drakensjö
- Skin Tumor Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden.,Cancer Centrum Karolinska, Karolinska Institutet Stockholm, Stockholm, Sweden
| | - Ada Girnita
- Skin Tumor Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden.,Cancer Centrum Karolinska, Karolinska Institutet Stockholm, Stockholm, Sweden
| |
Collapse
|
41
|
Abstract
Resistance to chemotherapeutic drugs exemplifies the greatest hindrance to effective treatment of cancer patients. The molecular mechanisms responsible have been investigated for over 50 years and have revealed the lack of a single cause, but instead, multiple mechanisms including induced expression of membrane transporters that pump drugs out of cells (multidrug resistance (MDR) phenotype), changes in the glutathione system, and altered metabolism. Treatment of cancer patients/cancer cells with chemotherapeutic agents and/or molecularly targeted drugs is accompanied by acquisition of resistance to the treatment administered. Chemotherapeutic agent resistance was initially assumed to be due to induction of mutations leading to a resistant phenotype. While this has occurred for molecularly targeted drugs, it is clear that drugs selectively targeting tyrosine kinases (TKs) cause the acquisition of mutational changes and resistance to inhibition. The first TK to be targeted, Bcr-Abl, led to the generation of several drugs including imatinib, dasatinib, and sunitinib that provided a rich understanding of this phenomenon. It became clear that mutations alone were not the only cause of resistance. Additional mechanisms were involved, including alternative splicing, alternative/compensatory signaling pathways, and epigenetic changes. This review will focus on resistance to tyrosine kinase inhibitors (TKIs), receptor TK (RTK)-directed antibodies, and antibodies that inactivate specific RTK ligands. New approaches and concepts aimed at avoiding the generation of drug resistance will be examined. Many RTKs, including the IGF-1R, are dependence receptors that induce ligand-independent apoptosis. How this signaling paradigm has implications on therapeutic strategies will also be considered.
Collapse
|
42
|
Zheng P, Li J, Kros JM. Breakthroughs in modern cancer therapy and elusive cardiotoxicity: Critical research-practice gaps, challenges, and insights. Med Res Rev 2018; 38:325-376. [PMID: 28862319 PMCID: PMC5763363 DOI: 10.1002/med.21463] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 07/14/2017] [Accepted: 07/15/2017] [Indexed: 12/16/2022]
Abstract
To date, five cancer treatment modalities have been defined. The three traditional modalities of cancer treatment are surgery, radiotherapy, and conventional chemotherapy, and the two modern modalities include molecularly targeted therapy (the fourth modality) and immunotherapy (the fifth modality). The cardiotoxicity associated with conventional chemotherapy and radiotherapy is well known. Similar adverse cardiac events are resurging with the fourth modality. Aside from the conventional and newer targeted agents, even the most newly developed, immune-based therapeutic modalities of anticancer treatment (the fifth modality), e.g., immune checkpoint inhibitors and chimeric antigen receptor (CAR) T-cell therapy, have unfortunately led to potentially lethal cardiotoxicity in patients. Cardiac complications represent unresolved and potentially life-threatening conditions in cancer survivors, while effective clinical management remains quite challenging. As a consequence, morbidity and mortality related to cardiac complications now threaten to offset some favorable benefits of modern cancer treatments in cancer-related survival, regardless of the oncologic prognosis. This review focuses on identifying critical research-practice gaps, addressing real-world challenges and pinpointing real-time insights in general terms under the context of clinical cardiotoxicity induced by the fourth and fifth modalities of cancer treatment. The information ranges from basic science to clinical management in the field of cardio-oncology and crosses the interface between oncology and onco-pharmacology. The complexity of the ongoing clinical problem is addressed at different levels. A better understanding of these research-practice gaps may advance research initiatives on the development of mechanism-based diagnoses and treatments for the effective clinical management of cardiotoxicity.
Collapse
Affiliation(s)
- Ping‐Pin Zheng
- Cardio‐Oncology Research GroupErasmus Medical CenterRotterdamthe Netherlands
- Department of PathologyErasmus Medical CenterRotterdamthe Netherlands
| | - Jin Li
- Department of OncologyShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Johan M Kros
- Department of PathologyErasmus Medical CenterRotterdamthe Netherlands
| |
Collapse
|
43
|
Cantisani MC, Parascandolo A, Perälä M, Allocca C, Fey V, Sahlberg N, Merolla F, Basolo F, Laukkanen MO, Kallioniemi OP, Santoro M, Castellone MD. A loss-of-function genetic screening identifies novel mediators of thyroid cancer cell viability. Oncotarget 2017; 7:28510-22. [PMID: 27058903 PMCID: PMC5053742 DOI: 10.18632/oncotarget.8577] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 03/02/2016] [Indexed: 12/31/2022] Open
Abstract
RET, BRAF and other protein kinases have been identified as major molecular players in thyroid cancer. To identify novel kinases required for the viability of thyroid carcinoma cells, we performed a RNA interference screening in the RET/PTC1(CCDC6-RET)-positive papillary thyroid cancer cell line TPC1 using a library of synthetic small interfering RNAs (siRNAs) targeting the human kinome and related proteins. We identified 14 hits whose silencing was able to significantly reduce the viability and the proliferation of TPC1 cells; most of them were active also in BRAF-mutant BCPAP (papillary thyroid cancer) and 8505C (anaplastic thyroid cancer) and in RAS-mutant CAL62 (anaplastic thyroid cancer) cells. These included members of EPH receptor tyrosine kinase family as well as SRC and MAPK (mitogen activated protein kinases) families. Importantly, silencing of the identified hits did not affect significantly the viability of Nthy-ori 3-1 (hereafter referred to as NTHY) cells derived from normal thyroid tissue, suggesting cancer cell specificity. The identified proteins are worth exploring as potential novel druggable thyroid cancer targets.
Collapse
Affiliation(s)
| | - Alessia Parascandolo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universita' Federico II, Naples, Italy
| | - Merja Perälä
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.,Center for Biotechnology, University of Turku, Turku, Finland
| | - Chiara Allocca
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universita' Federico II, Naples, Italy
| | - Vidal Fey
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.,Center for Biotechnology, University of Turku, Turku, Finland
| | - Niko Sahlberg
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.,Center for Biotechnology, University of Turku, Turku, Finland
| | - Francesco Merolla
- Dipartimento di Scienze Biomediche Avanzate, Università Federico II, Naples, Italy
| | - Fulvio Basolo
- Division of Pathology, Department of Surgery, University of Pisa, Pisa, Italy
| | | | - Olli Pekka Kallioniemi
- FIMM-Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Massimo Santoro
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universita' Federico II, Naples, Italy.,Istituto di Endocrinologia ed Oncologia Sperimentale "G. Salvatore" (IEOS), C.N.R., Naples, Italy
| | | |
Collapse
|
44
|
Wang WL, Chen XY, Gao Y, Gao LX, Sheng L, Zhu J, Xu L, Ding ZZ, Zhang C, Li JY, Li J, Zhou YB. Benzo[ c ][1,2,5]thiadiazole derivatives: A new class of potent Src homology-2 domain containing protein tyrosine phosphatase-2 (SHP2) inhibitors. Bioorg Med Chem Lett 2017; 27:5154-5157. [DOI: 10.1016/j.bmcl.2017.10.059] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 11/16/2022]
|
45
|
Khattab M, Wang F, Clayton AHA. A pH-induced conformational switch in a tyrosine kinase inhibitor identified by electronic spectroscopy and quantum chemical calculations. Sci Rep 2017; 7:16271. [PMID: 29176733 PMCID: PMC5701190 DOI: 10.1038/s41598-017-16583-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 11/14/2017] [Indexed: 01/18/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are a major class of drug utilised in the clinic. During transit to their cognate kinases, TKIs will encounter different pH environments that could have a major influence on TKI structure. To address this, we report UV-Vis spectroscopic and computational studies of the TKI, AG1478, as a function of pH. The electronic absorption spectrum of AG1478 shifted by 10 nm (from 342 nm to 332 nm) from acid to neutral pH and split into two peaks (at 334 nm and 345 nm) in highly alkaline conditions. From these transitions, the pKa value was calculated as 5.58 ± 0.01. To compute structures and spectra, time-dependent density functional theory (TD-DFT) calculations were performed along with conductor-like polarizable continuum model (CPCM) to account for implicit solvent effect. On the basis of the theoretical spectra, we could assign the AG1478 experimental spectrum at acidic pH to a mixture of two twisted conformers (71% AG1478 protonated at quinazolyl nitrogen N(1) and 29% AG1478 protonated at quinazolyl nitrogen N(3)) and at neutral pH to the neutral planar conformer. The AG1478 absorption spectrum (pH 13.3) was fitted to a mixture of neutral (70%) and NH-deprotonated species (30%). These studies reveal a pH-induced conformational transition in a TKI.
Collapse
Affiliation(s)
- Muhammad Khattab
- Centre for Micro-Photonics, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria, 3122, Australia
| | - Feng Wang
- Molecular Model Discovery Laboratory, Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria, 3122, Australia.
- School of Chemistry (Bio21 Institute), University of Melbourne, Parkville, Victoria, 3052, Australia.
- School of Physics, University of Melbourne, Parkville, Victoria, 3052, Australia.
| | - Andrew H A Clayton
- Centre for Micro-Photonics, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria, 3122, Australia.
| |
Collapse
|
46
|
Ma X, Lv X, Zhang J. Exploiting polypharmacology for improving therapeutic outcome of kinase inhibitors (KIs): An update of recent medicinal chemistry efforts. Eur J Med Chem 2017; 143:449-463. [PMID: 29202407 DOI: 10.1016/j.ejmech.2017.11.049] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/12/2017] [Accepted: 11/18/2017] [Indexed: 12/23/2022]
Abstract
Polypharmacology has been increasingly advocated for the therapeutic intervention in complex pathological conditions, exemplified by cancer. Although kinase inhibitors (KIs) have revolutionized the treatment for certain types of malignancies, some major medical needs remain unmet due to the relentless advance of drug resistance and insufficient efficacy of mono-target KIs. Hence, "multiple targets, multi-dimensional activities" represents an emerging paradigm for innovative anti-cancer drug discovery. Over recent years, considerable leaps have been made in pursuit of kinase-centric polypharmacological anti-cancer therapeutics, providing avenues to tackling the limitation of mono-target KIs. In the review, we summarize the clinically important mechanisms inducing KI resistance and depict a landscape of recent medicinal chemistry efforts on exploring kinase-centric polypharmacological anti-cancer agents that targeting multiple cancer-related processes. In parallel, some inevitable challenges are emphasized for the sake of more accurate and efficient drug discovery in the field.
Collapse
Affiliation(s)
- Xiaodong Ma
- Department of Medicinal Chemistry, School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Department of Medicinal Chemistry, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Xiaoqing Lv
- College of Medicine, Jiaxing University, Jiaxing 314001, China.
| | - Jiankang Zhang
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou 310023, China.
| |
Collapse
|
47
|
Fan Y, Arechederra M, Richelme S, Daian F, Novello C, Calderaro J, Di Tommaso L, Morcrette G, Rebouissou S, Donadon M, Morenghi E, Zucman-Rossi J, Roncalli M, Dono R, Maina F. A phosphokinome-based screen uncovers new drug synergies for cancer driven by liver-specific gain of nononcogenic receptor tyrosine kinases. Hepatology 2017; 66:1644-1661. [PMID: 28586114 DOI: 10.1002/hep.29304] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 05/24/2017] [Accepted: 06/02/2017] [Indexed: 12/14/2022]
Abstract
UNLABELLED Genetic mutations leading to oncogenic variants of receptor tyrosine kinases (RTKs) are frequent events during tumorigenesis; however, the cellular vulnerability to nononcogenic RTK fluctuations has not been characterized. Here, we demonstrated genetically that in the liver subtle increases in wild-type Met RTK levels are sufficient for spontaneous tumors in mice (Alb-R26Met ), conceptually illustrating how the shift from physiological to pathological conditions results from slight perturbations in signaling dosage. By analyzing 96 different genes in a panel of tumor samples, we demonstrated that liver tumorigenesis modeled by Alb-R26Met mice corresponds to a subset of hepatocellular carcinoma (HCC) patients, thus establishing the clinical relevance of this HCC mouse model. We elucidated the regulatory networks underlying tumorigenesis by combining a phosphokinome screen with bioinformatics analysis. We then used the signaling diversity results obtained from Alb-R26Met HCC versus control livers to design an "educated guess" drug screen, which led to the identification of new, deleterious synthetic lethal interactions. In particular, we report synergistic effects of mitogen-activated protein kinase kinase, ribosomal S6 kinase, and cyclin-dependent kinase 1/2 in combination with Bcl-XL inhibition on a panel of liver cancer cells. Focusing on mitogen-activated protein kinase kinase and Bcl-XL targeting, we mechanistically demonstrated concomitant down-regulation of phosphorylated extracellular signal-regulated kinase and myeloid cell leukemia 1 levels. Of note, a phosphorylated extracellular signal-regulated kinase+/BCL-XL+ /myeloid cell leukemia 1+ signature, deregulated in Alb-R26Met tumors, characterizes a subgroup of HCC patients with poor prognosis. CONCLUSION Our genetic studies highlight the heightened vulnerability of liver cells to subtle changes in nononcogenic RTK levels, allowing them to acquire a molecular profile that facilitates the full tumorigenic program; furthermore, our outcomes uncover new synthetic lethal interactions as potential therapies for a cluster of HCC patients. (Hepatology 2017;66:1644-1661).
Collapse
Affiliation(s)
- Yannan Fan
- Aix Marseille Univ., CNRS, Institute of Developmental Biology of Marseille, Parc Scientifique de Luminy, Marseille, France
| | - Maria Arechederra
- Aix Marseille Univ., CNRS, Institute of Developmental Biology of Marseille, Parc Scientifique de Luminy, Marseille, France
| | - Sylvie Richelme
- Aix Marseille Univ., CNRS, Institute of Developmental Biology of Marseille, Parc Scientifique de Luminy, Marseille, France
| | - Fabrice Daian
- Aix Marseille Univ., CNRS, Institute of Developmental Biology of Marseille, Parc Scientifique de Luminy, Marseille, France
| | - Chiara Novello
- Pathology Unit, Humanitas Clinical and Research Center, and Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
| | - Julien Calderaro
- Département de Pathologie, APHP, Groupe Hospitalier Henri Mondor.,INSERM U955, Team 18, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Luca Di Tommaso
- Pathology Unit, Humanitas Clinical and Research Center, and Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
| | - Guillaume Morcrette
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR674, Génomique Fonctionnelle des Tumeurs Solides, Institut Universitaire d'Hematologie, Paris, France
| | - Sandra Rebouissou
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR674, Génomique Fonctionnelle des Tumeurs Solides, Institut Universitaire d'Hematologie, Paris, France
| | - Matteo Donadon
- Hepatobiliary and General Surgery, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Emanuela Morenghi
- Biostatistics Unit, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Jessica Zucman-Rossi
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR674, Génomique Fonctionnelle des Tumeurs Solides, Institut Universitaire d'Hematologie, Paris, France
| | - Massimo Roncalli
- Pathology Unit, Humanitas Clinical and Research Center, and Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
| | - Rosanna Dono
- Aix Marseille Univ., CNRS, Institute of Developmental Biology of Marseille, Parc Scientifique de Luminy, Marseille, France
| | - Flavio Maina
- Aix Marseille Univ., CNRS, Institute of Developmental Biology of Marseille, Parc Scientifique de Luminy, Marseille, France
| |
Collapse
|
48
|
Wu X, Wang L, Qiu Y, Zhang B, Hu Z, Jin R. Cooperation of IRAK1/4 inhibitor and ABT-737 in nanoparticles for synergistic therapy of T cell acute lymphoblastic leukemia. Int J Nanomedicine 2017; 12:8025-8034. [PMID: 29184402 PMCID: PMC5673049 DOI: 10.2147/ijn.s146875] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is caused by clonal expansion of variant T cell progenitors and is considered as a high risk leukemia. Contemporary single chemotherapy has a limited effect due to dynamic and versatile properties of T-ALL. Here IRAK1/4 inhibitor and ABT-737 were co-encapsulated into polyethylene glycol modified poly (lactic-co-glycolic acid) nanoparticles (IRAK/ABT-NP) to enhance synergistic therapy of T-ALL. The formulation was optimized to achieve high drug loading using Box-Behnken design and response surface methodology. The optimal parameter comprised 2.98% polymer in acetonitrile, a ratio of oil phase to water phase of 1:8.33, and 2.12% emulsifier concentration. High drug loading and uniform spherical shape was achieved. In vitro release study showed sustained release of IRAK1/4 inhibitor for 72 hours as well as sustained release of ABT-737 for more than 120 hours. Uptake efficiency of IRAK/ABT-NP and induced apoptotic T-ALL fraction by IRAK/ABT-NP were much higher than the IRAK1/4 and ABT-737 combined solution. IC50 of IRAK/ABT-NP was two-fold lower than free drug combination in Jurkat cells. Additionally, we conducted in vivo experiments in which IRAK/ABT-NP exhibited greater cytotoxicity toward T-ALL cells, the capacity to significantly restore white blood cell number in peripheral blood, and improved survival time of T-ALL mouse model compared to the IRAK1/4 and ABT-737 combined solution.
Collapse
Affiliation(s)
- Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
| | - Lin Wang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
| | - Yining Qiu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
| | - Bingyu Zhang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
| | - Zhenhua Hu
- Department of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
| |
Collapse
|
49
|
68 Ga labeled Erlotinib: A novel PET probe for imaging EGFR over-expressing tumors. Bioorg Med Chem Lett 2017; 27:4552-4557. [DOI: 10.1016/j.bmcl.2017.08.065] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/07/2017] [Accepted: 08/25/2017] [Indexed: 01/13/2023]
|
50
|
Adam E, Kim HN, Gang EJ, Schnair C, Lee S, Lee S, Khazal S, Kosoyan O, Konopleva M, Parekh C, Bhojwani D, Wayne AS, Abdel-Azim H, Heisterkamp N, Kim YM. The PI3Kδ Inhibitor Idelalisib Inhibits Homing in an in Vitro and in Vivo Model of B ALL. Cancers (Basel) 2017; 9:cancers9090121. [PMID: 28891959 PMCID: PMC5615336 DOI: 10.3390/cancers9090121] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/06/2017] [Accepted: 09/06/2017] [Indexed: 11/16/2022] Open
Abstract
The quest continues for targeted therapies to reduce the morbidity of chemotherapy and to improve the response of resistant leukemia. Adhesion of acute lymphoblastic leukemia (ALL) cells to bone marrow stromal cells triggers intracellular signals that promote cell-adhesion-mediated drug resistance (CAM-DR). Idelalisib, an U.S. Food and Drug Administration (FDA)-approved PI3Kδ-specific inhibitor has been shown to be effective in CLL in down-regulating p-Akt and prolonging survival in combination with Rituximab; herein we explore the possibility of its use in B ALL and probe the mechanism of action. Primary B ALL in contact with OP9 stromal cells showed increased p-Aktser473. Idelalisib decreased p-Akt in patient samples of ALL with diverse genetic lesions. Addition of idelalisib to vincristine inhibited proliferation when compared to vincristine monotherapy in a subset of samples tested. Idelalisib inhibited ALL migration to SDF-1α in vitro and blocked homing of ALL cells to the bone marrow in vivo. This report tests PI3Kδ inhibitors in a more diverse group of ALL than has been previously reported and is the first published report of idelalisib inhibiting homing of ALL cells to bone marrow. Our data support further pre-clinical evaluation of idelalisib for the therapy of B ALL.
Collapse
Affiliation(s)
- Etai Adam
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| | - Hye Na Kim
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| | - Eun Ji Gang
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| | - Caitlin Schnair
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| | - Solomon Lee
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| | - Solah Lee
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| | - Sajad Khazal
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Osanna Kosoyan
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| | - Marina Konopleva
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Chintan Parekh
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| | - Deepa Bhojwani
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| | - Alan S Wayne
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| | - Hisham Abdel-Azim
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| | - Nora Heisterkamp
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| | - Yong-Mi Kim
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA.
| |
Collapse
|