1
|
Alzain AA, Elbadwi FA, Mukhtar RM, Shoaib TH, Abdelmoniem N, Miski SF, Ghazawi KF, Alsulaimany M, Mohamed SGA, Ainousah BE, Hussein HGA, Mohamed GA, Ibrahim SRM. Design of new Mcl-1 inhibitors for cancer using fragments hybridization, molecular docking, and molecular dynamics studies. J Biomol Struct Dyn 2025; 43:386-398. [PMID: 37962580 DOI: 10.1080/07391102.2023.2281637] [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: 08/09/2023] [Accepted: 11/04/2023] [Indexed: 11/15/2023]
Abstract
Apoptosis is a critical process that regulates cell survival and death and plays an essential role in cancer development. The Bcl-2 protein family, including myeloid leukemia 1 (Mcl-1), is a key regulator of the intrinsic apoptosis pathway, and its overexpression in many human cancers has prompted efforts to develop Mcl-1 inhibitors as potential anticancer agents. In this study, we aimed to design new Mcl-1 inhibitors using various computational techniques. First, we used the Mcl-1 receptor-ligand complex to build an e-pharmacophore hypothesis and screened a library of 567,000 fragments from the Enamine database. We obtained 410 fragments and used them to design 92,384 novel compounds, which we then docked into the Mcl-1 binding cavity using HTVS, SP, and XP docking modes of Glide. To assess their suitability as drug candidates, we conducted MM-GBSA calculations and ADME prediction, leading to the identification of 10 compounds with excellent binding affinity and favorable pharmacokinetic properties. To further investigate the interaction strength, we performed molecular dynamics simulations on the top three Mcl-1 receptor-ligand complexes to study their interaction stability. Overall, our findings suggest that these compounds have promising potential as anticancer agents, pending further experimental validation such as Mcl-1 apoptosis Assay. By combining experimental methods with various in silico approaches, these techniques prove to be invaluable for identifying novel drug candidates with distinct therapeutic applications using fragment-based drug design. This methodology has the potential to expedite the drug discovery process while also reducing its costs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Abdulrahim A Alzain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani, Sudan
| | - Fatima A Elbadwi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani, Sudan
| | - Rua M Mukhtar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani, Sudan
| | - Tagyedeen H Shoaib
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani, Sudan
| | - Nihal Abdelmoniem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani, Sudan
| | - Samar F Miski
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia
| | - Kholoud F Ghazawi
- Pharmacy Practice Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Marwa Alsulaimany
- Department of Pharmacognosy & Pharmaceutical Chemistry, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | | | - Bayan E Ainousah
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Hazem G A Hussein
- Preparatory Year Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Gamal A Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sabrin R M Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
2
|
Zhong C, Wang S, Xia L, Yang X, Fang L, Zhang X, Wang M, Zhao H, Wang G, Wu J, Guo R, Zhong M, Gohda E. The tubulin polymerization inhibitor gambogenic acid induces myelodysplastic syndrome cell apoptosis through upregulation of Fas expression mediated by the NF-κB signaling pathway. Cancer Biol Ther 2024; 25:2427374. [PMID: 39540618 PMCID: PMC11572293 DOI: 10.1080/15384047.2024.2427374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
The development of an effective treatment for myelodysplastic syndrome (MDS) is needed due to the insufficient efficacy of current therapies. Gambogenic acid (GNA) is a xanthone constituent of gamboge, a resin secreted by Garcinia hanburyi Hook. f. GNA exhibits antitumor and apoptosis-inducing activities against some cancer cells, but the mechanism is unknown. This study aimed to validate the anti-proliferative and apoptosis-inducing effects of GNA on MDS cells and to elucidate the mechanisms underlying those activities. Apoptosis, proliferation and cell cycle of MDS-L cells were assessed by the caspase 3/7 assay, cell counting and flow cytometry, respectively. The levels of apoptotic, tubulin, NF-κB pathways, and Fas proteins were determined by Western blotting. CRISPR/Cas9 knockout (KO) plasmids were used to generate KO cells of p65 and Fas. MDS cell growth in a xenograft model was evaluated by the AkaBLI system. GNA induced MDS cell apoptosis, accompanied by a reduction in the anti-apoptotic protein MCL-1 expression, and inhibited their growth in vitro and in vivo. GNA combined with the MCL-1 inhibitor MIK665 potently suppressed the proliferation of MDS cells. GNA interfered with tubulin polymerization, resulting in G2/M arrest. GNA induced NF-κB activation and upregulation of Fas, the latter of which was inhibited by p65 KO. GNA-induced apoptosis was attenuated in either p65 KO or Fas KO cells. These results demonstrate that GNA inhibited tubulin polymerization and induced apoptosis of MDS cells through upregulation of Fas expression mediated by the NF-κB signaling pathway, suggesting a chemotherapeutic strategy for MDS by microtubule dynamics disruption.
Collapse
Affiliation(s)
- Cheng Zhong
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Postdoctoral Research Workstation of Integrated Traditional Chinese and Western Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shijun Wang
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Postdoctoral Research Workstation of Integrated Traditional Chinese and Western Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Xia
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaoman Yang
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liguang Fang
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianyi Zhang
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengyue Wang
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haijun Zhao
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guanghui Wang
- Department of Pharmacology and Toxicology, School of Pharmacy, Jining Medical University, Rizhao, China
| | - Jinglong Wu
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, Japan
| | - Ruijian Guo
- Department of Pharmaceutics, College of pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming Zhong
- Department of Pharmacology and Toxicology, School of Pharmacy, Jining Medical University, Rizhao, China
| | - Eiichi Gohda
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
3
|
Khoder ZM, Mohamed MS, Awad SM, Gharib AF, Aly O, Khodair MAEF, Fatahala SS, El-Hameed RHA. Synthesis, Anti-Cancer Activity, Cell Cycle Arrest, Apoptosis Induction, and Docking Study of Fused Benzo[ h]chromeno[2,3- d]pyrimidine on Human Breast Cancer Cell Line MCF-7. Molecules 2024; 29:4697. [PMID: 39407625 PMCID: PMC11478142 DOI: 10.3390/molecules29194697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Breast cancer is the predominant form of cancer among women and ranks as the second most prevalent cancer globally, affecting both developed and less developed countries. Presently, accessible cancer treatment methods either employ recently created, secure, and efficient chemotherapeutic medications or directly target innovative pathways that cause apoptosis. One of the indirect strategies for treating this fatal illness has mostly depended on its essential role in cell cycle arrest and apoptosis induction, as well as the antagonistic interaction between the Bcl-2 and Mcl-1 proteins, in order to avert major health repercussions. We reported that newly synthesized fused chromenopyrimidines (3a and 4a) showed potential cell cycle arrest and dual Bcl-2 and Mcl-1 inhibitory characteristics. Bcl-2 and Mcl-1 were the targets of a molecular docking procedure. The previous docking results are in line with the biological data and suggest that 3a may have promising anti-cancer activity.
Collapse
Affiliation(s)
- Zainab M. Khoder
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Helwan, Cairo 11795, Egypt; (Z.M.K.); (M.S.M.); (S.M.A.); (R.H.A.E.-H.)
- Department of Chemistry, The State University of New York at Buffalo, New York, NY 14260, USA
| | - Mosaad S. Mohamed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Helwan, Cairo 11795, Egypt; (Z.M.K.); (M.S.M.); (S.M.A.); (R.H.A.E.-H.)
| | - Samir M. Awad
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Helwan, Cairo 11795, Egypt; (Z.M.K.); (M.S.M.); (S.M.A.); (R.H.A.E.-H.)
- Pharmacy Department, Al-zahrawi University College, Carbala 56001, Iraq
| | - Amal F. Gharib
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia;
| | - Omnia Aly
- Medical Biochemistry Department, National Research Centre, Doki, P.O. Box 12622 Cairo, Egypt;
| | - Marwa Abd El-Fattah Khodair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Helwan, Cairo 11795, Egypt; (Z.M.K.); (M.S.M.); (S.M.A.); (R.H.A.E.-H.)
| | - Samar S. Fatahala
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Helwan, Cairo 11795, Egypt; (Z.M.K.); (M.S.M.); (S.M.A.); (R.H.A.E.-H.)
| | - Rania H. Abd El-Hameed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Helwan, Cairo 11795, Egypt; (Z.M.K.); (M.S.M.); (S.M.A.); (R.H.A.E.-H.)
| |
Collapse
|
4
|
Iyer P, Jasdanwala SS, Bhatia K, Bhatt S. Mitochondria and Acute Leukemia: A Clinician's Perspective. Int J Mol Sci 2024; 25:9704. [PMID: 39273651 PMCID: PMC11395402 DOI: 10.3390/ijms25179704] [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: 07/19/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Acute leukemia is a group of aggressive hematological malignancies, with acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) being the most common types. The biology of acute leukemia involves complex genetic and epigenetic alterations that lead to uncontrolled cell proliferation and resistance to apoptosis. Mitochondrial dysfunction is a feature of acute leukemia that results in altered energy production, unregulated cell death pathways, and increased cancer cell survival. Apoptosis, particularly via the mitochondrial pathway, is crucial for cellular homeostasis and cancer prevention. In acute leukemia, disruption of apoptosis is pivotal in disease development and progression, with elevated levels of anti-apoptotic proteins conferring a survival advantage to leukemia cells and promoting resistance to conventional therapies. Targeting mitochondrial apoptosis using BH3 mimetics and anti-apoptotic protein inhibitors is a viable therapeutic strategy. Alterations in the mitochondrial membrane potential, metabolism, and dynamics also contribute to the pathogenesis of acute leukemia. Continued research is vital for developing novel therapies and enhancing survival outcomes in patients with acute leukemia while minimizing the long-term adverse effects of treatment. In this narrative review, we provide a birds-eye view of the available scientific literature on the importance of mitochondria in acute leukemia, and discuss the role of BH3 mimetics in targeting the mitochondrial internal apoptotic machinery.
Collapse
Affiliation(s)
- Prasad Iyer
- Children's Blood and Cancer Centre, KK Women's and Children's Hospital, Singapore 229899, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
| | | | - Karanpreet Bhatia
- Department of Hematology and Medical Oncology, School of Medicine, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Shruti Bhatt
- Department of Pharmacy, National University of Singapore, Singapore 119077, Singapore
| |
Collapse
|
5
|
Liu F, Ma M, Li L, Zhang Y, Shang Y, Yuan Q, Ju B, Wang Z. A Study of Sperm DNA Damage Mechanism Based on miRNA Sequencing. Am J Mens Health 2024; 18:15579883241286672. [PMID: 39462893 PMCID: PMC11528732 DOI: 10.1177/15579883241286672] [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: 05/27/2024] [Revised: 08/07/2024] [Accepted: 08/23/2024] [Indexed: 10/29/2024] Open
Abstract
To analyze the differential expression profiles of microRNAs (miRNAs) in spermatozoa of patients with sperm DNA damage and to investigate the role of miRNAs in sperm DNA damage. Male infertility patients with sperm DNA damage who attended the First Affiliated Hospital of Henan University of Chinese Medicine from October 2023 to December 2023 were selected and included in this study as a case group. Fertile healthy men who were seen at the health check-up center during the same period and diagnosed by examination were also included as a control group. Sperm miRNA expression was detected in patients with sperm DNA damage (case group, n = 5) and healthy medical check-ups (control group, n = 5) using high-throughput sequencing technology. The differentially expressed miRNAs between the two groups were bioinformatically analyzed to explore the main biological functions of the target genes. We found that 63 miRNAs were significantly changed in the spermatozoa of patients with sperm DNA damage,|log2 (foldchange)| ≥ 1, p < .05. Gene Ontology (GO) enrichment analysis indicated that these differential miRNAs might be involved in developmental process, anatomical structure development, cellular macromolecule metabolic process, multicellular organism development, system development, and so on. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that that they mainly affect the PI3K-AKT signaling pathway. The present study suggests that the altered expression of miR-1255a, miR-921, and miR-3156-5p may play an important role in the sperm DNA damage process, and the mechanism may involve the phosphatidylinositol-3'-kinase-AKT (PI3K-AKT) signaling pathway.
Collapse
Affiliation(s)
- Feng Liu
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Miaomiao Ma
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Luyu Li
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Yongtao Zhang
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Yihan Shang
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Quan Yuan
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Baojun Ju
- Department of Andrology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Zulong Wang
- Department of Andrology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
6
|
Han JM, Oh KY, Choi SJ, Lee WW, Jin BH, Kim JH, Yu HJ, Kim RJY, Yoon HJ, Lee JI, Hong SD, Cho SD. Antitumor activity of afatinib in EGFR T790M-negative human oral cancer therapeutically targets mTOR/Mcl-1 signaling axis. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00962-6. [PMID: 38888847 DOI: 10.1007/s13402-024-00962-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
PURPOSE This study investigates the role and effectiveness of the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) in oral cancer, focusing on the clinical relevance of EGFR and myeloid cell leukemia-1 (Mcl-1) in head and neck cancers (HNCs). It aims to explore the molecular mechanism of afatinib, a TKI, in treating human oral cancer. METHODS We conducted an in silico analysis using databases like The Cancer Genome Atlas, Gene Expression Omnibus, and Clinical Proteomic Tumor Analysis Consortium, along with immunohistochemistry staining, to study EGFR and Mcl-1 expression in HNCs. For investigating afatinib's anticancer properties, we performed various in vitro and in vivo analyses, including trypan blue exclusion assay, Western blotting, 4'-6-diamidino-2-phenylindole staining, flow cytometry, quantitative real-time PCR, Mitochondrial membrane potential assay, overexpression vector construction, transient transfection, and a tumor xenograft model. RESULTS Higher expression levels of EGFR and Mcl-1 were observed in HNC patient tissues compared to normal tissues, with their co-expression significantly linked to poor prognosis. There was a strong correlation between EGFR and Mcl-1 expressions in oral cancer patients. Afatinib treatment induced apoptosis and suppressed Mcl-1 in oral cancer cell lines without the EGFR T790M mutation. The mechanism of afatinib-induced apoptosis involved the EGFR/mTOR/Mcl-1 axis, as shown by the effects of mTOR activator MHY1485 and inhibitor rapamycin. Afatinib also increased Bim expression, mitochondrial membrane permeabilization, and cytochrome c release. It significantly lowered tumor volume without affecting body, liver, and kidney weights. CONCLUSION Afatinib, targeting the EGFR/mTOR/Mcl-1 axis, shows promise as a therapeutic strategy for oral cancer, especially in patients with high EGFR and Mcl-1 expressions.
Collapse
Affiliation(s)
- Jung-Min Han
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Kyu-Young Oh
- Department of Oral Pathology, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Su-Jung Choi
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Won-Woo Lee
- Laboratory Animal Center, CHA University, CHA Biocomplex, Sampyeong-Dong, Seongnam, 13488, Republic of Korea
| | - Bo-Hwan Jin
- Laboratory Animal Center, CHA University, CHA Biocomplex, Sampyeong-Dong, Seongnam, 13488, Republic of Korea
| | - Ji-Hoon Kim
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hyun-Ju Yu
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Ryan Jin Young Kim
- Department of Dental Science, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hye-Jung Yoon
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Jae-Il Lee
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seong-Doo Hong
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea.
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea.
| |
Collapse
|
7
|
Hekking KFW, Maroto S, van Kekem K, Haasjes FS, Slootweg JC, Oude Alink PGB, Dirks R, Sardana M, Bolster MG, Kuijpers B, Smith D, Doodeman R, Scheepstra M, Zech B, Mulvihill M, Renzetti LM, Babiss L, Centrella PA, Clark MA, Cuozzo JW, Guié MA, Sigel E, Habeshian S, Hupp CD, Liu J, Thomson HA, Zhang Y, Keefe AD, Müller G, Gremmen S. Development of Potent Mcl-1 Inhibitors: Structural Investigations on Macrocycles Originating from a DNA-Encoded Chemical Library Screen. J Med Chem 2024; 67:3039-3065. [PMID: 38306405 DOI: 10.1021/acs.jmedchem.3c02206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2024]
Abstract
Evasion of apoptosis is critical for the development and growth of tumors. The pro-survival protein myeloid cell leukemia 1 (Mcl-1) is an antiapoptotic member of the Bcl-2 family, associated with tumor aggressiveness, poor survival, and drug resistance. Development of Mcl-1 inhibitors implies blocking of protein-protein interactions, generally requiring a lengthy optimization process of large, complex molecules. Herein, we describe the use of DNA-encoded chemical library synthesis and screening to directly generate complex, yet conformationally privileged macrocyclic hits that serve as Mcl-1 inhibitors. By applying a conceptual combination of conformational analysis and structure-based design in combination with a robust synthetic platform allowing rapid analoging, we optimized in vitro potency of a lead series into the low nanomolar regime. Additionally, we demonstrate fine-tuning of the physicochemical properties of the macrocyclic compounds, resulting in the identification of lead candidates 57/59 with a balanced profile, which are suitable for future development toward therapeutic use.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ron Dirks
- Symeres, 6546BB Nijmegen, The Netherlands
| | | | | | | | | | | | | | - Birgit Zech
- X-Rx, Inc., New York, New York 10016, United States
| | | | | | - Lee Babiss
- X-Rx, Inc., New York, New York 10016, United States
| | | | | | - John W Cuozzo
- X-Chem, Inc., Waltham, Massachusetts 02453, United States
| | | | - Eric Sigel
- X-Chem, Inc., Waltham, Massachusetts 02453, United States
| | | | | | - Julie Liu
- X-Chem, Inc., Waltham, Massachusetts 02453, United States
| | | | - Ying Zhang
- X-Chem, Inc., Waltham, Massachusetts 02453, United States
| | | | | | | |
Collapse
|
8
|
Shin GC, Lee HM, Kim N, Seo SU, Kim KP, Kim KH. PRKCSH contributes to TNFSF resistance by extending IGF1R half-life and activation in lung cancer. Exp Mol Med 2024; 56:192-209. [PMID: 38200153 PMCID: PMC10834952 DOI: 10.1038/s12276-023-01147-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 09/25/2023] [Accepted: 10/23/2023] [Indexed: 01/12/2024] Open
Abstract
Tumor necrosis factor superfamily (TNFSF) resistance contributes to the development and progression of tumors and resistance to various cancer therapies. Tumor-intrinsic alterations involved in the adaptation to the TNFSF response remain largely unknown. Here, we demonstrate that protein kinase C substrate 80K-H (PRKCSH) abundance in lung cancers boosts oncogenic IGF1R activation, leading to TNFSF resistance. PRKCSH abundance is correlated with IGF1R upregulation in lung cancer tissues. Specifically, PRKCSH interacts with IGF1R and extends its half-life. The PRKCSH-IGF1R axis in tumor cells impairs caspase-8 activation, increases Mcl-1 expression, and inhibits caspase-9, leading to an imbalance between cell death and survival. PRKCSH deficiency augmented the antitumor effects of natural killer (NK) cells, representative TNFSF effector cells, in a tumor xenograft IL-2Rg-deficient NOD/SCID (NIG) mouse model. Our data suggest that PRKCSH plays a critical role in TNFSF resistance and may be a potential target to improve the efficacy of NK cell-based cancer therapy.
Collapse
Affiliation(s)
- Gu-Choul Shin
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| | - Hyeong Min Lee
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, 446-701, Republic of Korea
- Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, 02453, Republic of Korea
| | - Nayeon Kim
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, 446-701, Republic of Korea
- Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, 02453, Republic of Korea
| | - Kyun-Hwan Kim
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
9
|
Beltrán-Visiedo M, Jiménez-Alduán N, Díez R, Cuenca M, Benedi A, Serrano-Del Valle A, Azaceta G, Palomera L, Peperzak V, Anel A, Naval J, Marzo I. Dinaciclib synergizes with BH3 mimetics targeting BCL-2 and BCL-X L in multiple myeloma cell lines partially dependent on MCL-1 and in plasma cells from patients. Mol Oncol 2023; 17:2507-2525. [PMID: 37704591 DOI: 10.1002/1878-0261.13522] [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: 05/08/2023] [Revised: 08/01/2023] [Accepted: 09/12/2023] [Indexed: 09/15/2023] Open
Abstract
A better understanding of multiple myeloma (MM) biology has led to the development of novel therapies. However, MM is still an incurable disease and new pharmacological strategies are needed. Dinaciclib, a multiple cyclin-dependent kinase (CDK) inhibitor, which inhibits CDK1, 2, 5 and 9, displays significant antimyeloma activity as found in phase II clinical trials. In this study, we have explored the mechanism of dinaciclib-induced death and evaluated its enhancement by different BH3 mimetics in MM cell lines as well as in plasma cells from MM patients. Our results indicate a synergistic effect of dinaciclib-based combinations with B-cell lymphoma 2 or B-cell lymphoma extra-large inhibitors, especially in MM cell lines with partial dependence on myeloid cell leukemia sequence 1 (MCL-1). Simultaneous treatment with dinaciclib and BH3 mimetics ABT-199 or A-1155463 additionally showed a synergistic effect in plasma cells from MM patients, ex vivo. Altered MM cytogenetics did not affect dinaciclib response ex vivo, alone or in combined treatment, suggesting that these combinations could be a suitable therapeutic option for patients bearing cytogenetic alterations and poor prognosis. This work also opens the possibility to explore cyclin-dependent kinase 9 inhibition as a targeted therapy in MM patients overexpressing or with high dependence on MCL-1.
Collapse
Affiliation(s)
| | | | - Rosana Díez
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Spain
- Hematology Service, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Marta Cuenca
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Andrea Benedi
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Spain
| | | | - Gemma Azaceta
- Hematology Service, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
- HCU-Lozano Blesa-Hematology Research Group, IIS Aragón, Instituto Aragonés de Ciencias de la Salud, Zaragoza, Spain
| | - Luis Palomera
- Hematology Service, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
- HCU-Lozano Blesa-Hematology Research Group, IIS Aragón, Instituto Aragonés de Ciencias de la Salud, Zaragoza, Spain
| | - Victor Peperzak
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Alberto Anel
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Spain
| | - Javier Naval
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Spain
| | - Isabel Marzo
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Spain
| |
Collapse
|
10
|
Tantawy SI, Timofeeva N, Sarkar A, Gandhi V. Targeting MCL-1 protein to treat cancer: opportunities and challenges. Front Oncol 2023; 13:1226289. [PMID: 37601693 PMCID: PMC10436212 DOI: 10.3389/fonc.2023.1226289] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/03/2023] [Indexed: 08/22/2023] Open
Abstract
Evading apoptosis has been linked to tumor development and chemoresistance. One mechanism for this evasion is the overexpression of prosurvival B-cell lymphoma-2 (BCL-2) family proteins, which gives cancer cells a survival advantage. Mcl-1, a member of the BCL-2 family, is among the most frequently amplified genes in cancer. Targeting myeloid cell leukemia-1 (MCL-1) protein is a successful strategy to induce apoptosis and overcome tumor resistance to chemotherapy and targeted therapy. Various strategies to inhibit the antiapoptotic activity of MCL-1 protein, including transcription, translation, and the degradation of MCL-1 protein, have been tested. Neutralizing MCL-1's function by targeting its interactions with other proteins via BCL-2 interacting mediator (BIM)S2A has been shown to be an equally effective approach. Encouraged by the design of venetoclax and its efficacy in chronic lymphocytic leukemia, scientists have developed other BCL-2 homology (BH3) mimetics-particularly MCL-1 inhibitors (MCL-1i)-that are currently in clinical trials for various cancers. While extensive reviews of MCL-1i are available, critical analyses focusing on the challenges of MCL-1i and their optimization are lacking. In this review, we discuss the current knowledge regarding clinically relevant MCL-1i and focus on predictive biomarkers of response, mechanisms of resistance, major issues associated with use of MCL-1i, and the future use of and maximization of the benefits from these agents.
Collapse
Affiliation(s)
- Shady I. Tantawy
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Natalia Timofeeva
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Aloke Sarkar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
11
|
Demin S, Peschiulli A, Velter AI, Vos A, De Boeck B, Miller B, Rombouts FJR, Reuillon T, Lento W, Blanco MD, Jouffroy M, Steyvers H, Bekkers M, Altrocchi C, Pietrak B, Koo SJ, Szewczuk L, Attar R, Philippar U. Macrocyclic Carbon-Linked Pyrazoles As Novel Inhibitors of MCL-1. ACS Med Chem Lett 2023; 14:955-961. [PMID: 37465311 PMCID: PMC10351060 DOI: 10.1021/acsmedchemlett.3c00141] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023] Open
Abstract
Myeloid cell leukemia-1 (MCL-1) is a member of the antiapoptotic BCL-2 proteins family and a key regulator of mitochondrial homeostasis. Overexpression of MCL-1 is found in many cancer cells and contributes to tumor progression, which makes it an attractive therapeutic target. Pursuing our previous study of macrocyclic indoles for the inhibition of MCL-1, we report herein the impact of both pyrazole and indole isomerism on the potency and overall properties of this family of compounds. We demonstrated that the incorporation of a fluorine atom on the naphthalene moiety was a necessary step to improve cellular potency and that, combined with the introduction of various side chains on the pyrazole, it enhanced solubility significantly. This exploration culminated in the discovery of compounds (Ra)-10 and (Ra)-15, possessing remarkable cellular potency and properties.
Collapse
Affiliation(s)
- Samuël Demin
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Aldo Peschiulli
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Adriana I. Velter
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Ann Vos
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Benoît De Boeck
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Bradley Miller
- Janssen
Research & Development LLC, 1400 McKean Road (Welsh Road), Spring House, Pennsylvania 19477, United States
| | - Frederik J. R. Rombouts
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Tristan Reuillon
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| | - William Lento
- Janssen
Research & Development LLC, 1400 McKean Road (Welsh Road), Spring House, Pennsylvania 19477, United States
| | - Maria Dominguez Blanco
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Matthieu Jouffroy
- Chemical
Process R&D, Discovery Process Research, Janssen Pharmaceutica N.V., Beerse B-2340, Belgium
| | - Helena Steyvers
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Mariette Bekkers
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Cristina Altrocchi
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Beth Pietrak
- Janssen
Research & Development LLC, 1400 McKean Road (Welsh Road), Spring House, Pennsylvania 19477, United States
| | - Seong Joo Koo
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| | - Lawrence Szewczuk
- Janssen
Research & Development LLC, 1400 McKean Road (Welsh Road), Spring House, Pennsylvania 19477, United States
| | - Ricardo Attar
- Janssen
Research & Development LLC, 1400 McKean Road (Welsh Road), Spring House, Pennsylvania 19477, United States
| | - Ulrike Philippar
- Janssen
Research & Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, Beerse B-2340, Belgium
| |
Collapse
|
12
|
Jayappa KD, Tran B, Gordon VL, Morris C, Saha S, Farrington CC, O’Connor CM, Zawacki KP, Isaac KM, Kester M, Bender TP, Williams ME, Portell CA, Weber MJ, Narla G. PP2A modulation overcomes multidrug resistance in chronic lymphocytic leukemia via mPTP-dependent apoptosis. J Clin Invest 2023; 133:e155938. [PMID: 37166997 PMCID: PMC10313372 DOI: 10.1172/jci155938] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 05/09/2023] [Indexed: 05/12/2023] Open
Abstract
Targeted therapies such as venetoclax (VEN) (Bcl-2 inhibitor) have revolutionized the treatment of chronic lymphocytic leukemia (CLL). We previously reported that persister CLL cells in treated patients overexpress multiple antiapoptotic proteins and display resistance to proapoptotic agents. Here, we demonstrated that multidrug-resistant CLL cells in vivo exhibited apoptosis restriction at a pre-mitochondrial level due to insufficient activation of the Bax and Bak (Bax/Bak) proteins. Co-immunoprecipitation analyses with selective BH domain antagonists revealed that the pleiotropic proapoptotic protein (Bim) was prevented from activating Bax/Bak by "switching" interactions to other upregulated antiapoptotic proteins (Mcl-1, Bcl-xL, Bcl-2). Hence, treatments that bypass Bax/Bak restriction are required to deplete these resistant cells in patients. Protein phosphatase 2A (PP2A) contributes to oncogenesis and treatment resistance. We observed that small-molecule activator of PP2A (SMAP) induced cytotoxicity in multiple cancer cell lines and CLL samples, including multidrug-resistant leukemia and lymphoma cells. The SMAP (DT-061) activated apoptosis in multidrug-resistant CLL cells through induction of mitochondrial permeability transition pores, independent of Bax/Bak. DT-061 inhibited the growth of wild-type and Bax/Bak double-knockout, multidrug-resistant CLL cells in a xenograft mouse model. Collectively, we discovered multidrug-resistant CLL cells in patients and validated a pharmacologically tractable pathway to deplete this reservoir.
Collapse
MESH Headings
- Humans
- Animals
- Mice
- bcl-2-Associated X Protein/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Protein Phosphatase 2/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
- Proto-Oncogene Proteins c-bcl-2
- Apoptosis/physiology
- Apoptosis Regulatory Proteins/metabolism
- Drug Resistance, Multiple
Collapse
Affiliation(s)
- Kallesh D. Jayappa
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, Charlottesville, Virginia, USA
- Department of Pharmacology, Charlottesville, Virginia, USA
| | - Brian Tran
- Division of Genetic Medicine, Department of Internal Medicine, the University of Michigan, Ann Arbor, Michigan, USA
| | - Vicki L. Gordon
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, Charlottesville, Virginia, USA
| | - Christopher Morris
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, Charlottesville, Virginia, USA
| | - Shekhar Saha
- Department of Biochemistry and Molecular Genetics, Charlottesville, Virginia, USA
| | - Caroline C. Farrington
- Division of Genetic Medicine, Department of Internal Medicine, the University of Michigan, Ann Arbor, Michigan, USA
| | - Caitlin M. O’Connor
- Division of Genetic Medicine, Department of Internal Medicine, the University of Michigan, Ann Arbor, Michigan, USA
| | - Kaitlin P. Zawacki
- Division of Genetic Medicine, Department of Internal Medicine, the University of Michigan, Ann Arbor, Michigan, USA
| | - Krista M. Isaac
- Division of Hematology/Oncology, UVA School of Medicine, Charlottesville, Virginia, USA
| | - Mark Kester
- Department of Pharmacology, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, UVA, Charlottesville, Virginia, USA
| | - Timothy P. Bender
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, Charlottesville, Virginia, USA
| | - Michael E. Williams
- Division of Hematology/Oncology, UVA School of Medicine, Charlottesville, Virginia, USA
- Cancer Center, UVA, Charlottesville, Virginia, USA
| | - Craig A. Portell
- Division of Hematology/Oncology, UVA School of Medicine, Charlottesville, Virginia, USA
- Cancer Center, UVA, Charlottesville, Virginia, USA
| | - Michael J. Weber
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, Charlottesville, Virginia, USA
- Cancer Center, UVA, Charlottesville, Virginia, USA
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, the University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
13
|
Wang J, He Y, Yang C, Luo Q, Wang B. Myeloid cell leukemia-1 as a candidate prognostic biomarker in cancers: a systematic review and meta-analysis. Expert Rev Anticancer Ther 2023; 23:1017-1027. [PMID: 37467344 DOI: 10.1080/14737140.2023.2238900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION Studies have shown that myeloma cell leukemia-1 (MCL-1) is associated with the prognosis of patients with cancer. To further validate the prognostic value of MCL-1 in cancer, a meta-analysis was conducted. METHODS Six databases were searched using Boolean logic search formulas. Data were extracted from the included literature, and pooled odds ratio, hazard ratio, and 95% confidence interval were calculated to determine the relationship between MCL-1 levels and clinicopathological characteristics and prognosis of patients with cancer. When heterogeneity was found to be significant, a random effects model was used, otherwise, a fixed effects model was used. RESULTS Twelve articles were included in this meta-analysis, totaling 2208 patients with cancer across 14 studies. A high MCL-1 expression level was associated with patients with high T stage, M stage, and TNM stage in some cancers. Additionally, high MCL-1 expression was likely to be observed in patients with poorly differentiated digestive system tumors and patients with lung adenocarcinoma. Notably, a higher expression of MCL-1 was found to be associated with shorter overall survival in patients with hematological tumors, digestive system tumors, and lung cancer. CONCLUSION MCL-1 may be a prognostic biomarker in patients with some types of cancer.
Collapse
Affiliation(s)
- Jianrong Wang
- Department of Respiratory and Critical Care Medicine, Ya'an People's Hospital, Ya'an, Sichuan, China
| | - Yu He
- Department of Respiratory and Critical Care Medicine, Ya'an People's Hospital, Ya'an, Sichuan, China
| | - Chao Yang
- Department of Respiratory and Critical Care Medicine, Ya'an People's Hospital, Ya'an, Sichuan, China
| | - Qiurui Luo
- Department of Respiratory and Critical Care Medicine, Ya'an People's Hospital, Ya'an, Sichuan, China
| | - Bingchi Wang
- Department of Respiratory and Critical Care Medicine, Ya'an People's Hospital, Ya'an, Sichuan, China
| |
Collapse
|
14
|
Li J, Li F, Zhao Y, Jin D. Integrating network pharmacology and experimental validation to explore the effect and mechanism of AD-1 in the treatment of colorectal cancer. Front Pharmacol 2023; 14:1159712. [PMID: 37284306 PMCID: PMC10239872 DOI: 10.3389/fphar.2023.1159712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/11/2023] [Indexed: 06/08/2023] Open
Abstract
20 (R)-25-methoxyl-dammarane-3β, 12β, 20-triol (AD-1), a novel ginsenoside isolated from stem and leaf of Panax Notoginseng, has anticancer activity against a variety of malignant tumors. However, the pharmacological mechanism of AD-1 on colorectal cancer (CRC) remains unclear. The purpose of this study was to verify the potential mechanism of action of AD-1 against CRC through network pharmacology and experiments. A total of 39 potential targets were obtained based on the intersection of AD-1 and CRC targets, and key genes were analyzed and identified from the PPI network using Cytoscape software. 39 targets were significantly enriched in 156 GO terms and 138 KEGG pathways, among which PI3K-Akt signaling pathway was identified as one of the most enriched pathways. Based on experimental results, AD-1 can inhibit the proliferation and migration of SW620 and HT-29 cells, and induce their apoptosis. Subsequently, the HPA and UALCAN databases showed that PI3K and Akt were highly expressed in CRC. AD-1 also decreased the expressions of PI3K and Akt. In summary, these results suggest that AD-1 can play an anti-tumor role by inducing cell apoptosis and regulating PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Jiawei Li
- Immunology Biology Key Laboratory, Yanbian University, Yanji, China
| | - Fangfang Li
- Immunology Biology Key Laboratory, Yanbian University, Yanji, China
| | - Yuqing Zhao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
| | - Dan Jin
- Immunology Biology Key Laboratory, Yanbian University, Yanji, China
| |
Collapse
|
15
|
Zantut-Wittmann DE, Barreto IS, Laus AC, Moreno DA, Moma CA, Maia FFR, Assumpção LVMD, Reis RM. PD-L1 and MCL-1 markers and the relationship with prognostic characteristics of differentiated thyroid carcinoma. Mol Cell Endocrinol 2023; 570:111931. [PMID: 37072108 DOI: 10.1016/j.mce.2023.111931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/13/2023] [Accepted: 04/15/2023] [Indexed: 04/20/2023]
Abstract
OBJECTIVES MCL-1 and PD-L1 proteins are related to carcinogenesis mechanisms in differentiated thyroid carcinoma(DTC). Tumor antigens stimulate the expression of PD-1 in immune cells, which binds to PD-L1 of tumor cells, inducing immune escape from the tumor. MCL-1, an anti-apoptotic member of the BCL-2 family, is necessary for the survival of T and B lymphocytes and has a high oncogenic potential. We aim to evaluate the clinical utility and relevance of MCL-1 and PD-L1 in the long-term prognosis of DTC. METHODS 120 DTC patients after total thyroidectomy and radioiodine therapy followed for a minimum of 2 years were included. Demographic features, tumor histopathology, persistence/recurrence risk, factors associated with outcome, initial response to therapy, persistence or disease-free at the follow-up were related to MCL-1 and PD-L1 immunohistochemical expression and BRAFV600E mutation. RESULTS 100(83.3%) were women, 46.64 ± 16.73 years old at diagnosis; 37(30.8%) patients were at high, 45(37.5%) of intermediate and 38(31.7%) of low disease recurrence/persistence risk. At the end of follow-up of 124.86 ± 65.36 months, 48(42.5%) had persistent disease. 103(85.8%) patients had papillary thyroid carcinoma (PTC), 17(14.2%) follicular thyroid carcinoma (FTC). In PTC, moderate/strong PD-L1 and MCL-1 expressions were associated to BRAFV600E (p=0.0467; p=0.0044). PD-L1 was also associated with tall cell subtype (p=0.0274). In FTC, weak PD-L1 expression was associated to the largest nodule diameter (p=0.0100). Strong/moderate PD-L1 expression was associated to T2 and the weak expression with T3 in TNM classification (p=0.0490). Moderate MCL-1 expression was associated to smoking (p=0.0350). CONCLUSIONS PDL-1, marker of progression of tumor cells and MCL-1, anti-apoptotic marker, were associated with PTC carrying BRAFV600E mutation, while PDL-1 was associated with more aggressive PTC subtype. MCL-1 and PD-L1 could be useful in composing a panel to assess the prognosis of PTC patients. On the other hand, both markers seemed to have lower relevance to FTC patients.
Collapse
Affiliation(s)
- Denise Engelbrecht Zantut-Wittmann
- Endocrinology Division, Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil.
| | - Icleia Siqueira Barreto
- Department of Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Ana Carolina Laus
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, Brazil
| | | | - Camila Aparecida Moma
- Endocrinology Division, Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Frederico Fernandes Ribeiro Maia
- Endocrinology Division, Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Lígia Vera Montali da Assumpção
- Endocrinology Division, Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, Brazil; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
16
|
Sarkar A, Paul A, Banerjee T, Maji A, Saha S, Bishayee A, Maity TK. Therapeutic advancements in targeting BCL-2 family proteins by epigenetic regulators, natural, and synthetic agents in cancer. Eur J Pharmacol 2023; 944:175588. [PMID: 36791843 DOI: 10.1016/j.ejphar.2023.175588] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/21/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023]
Abstract
Cancer is amongst the deadliest and most disruptive disorders, having a much higher death rate than other diseases worldwide. Human cancer rates continue to rise, thereby posing the most significant concerns for medical health professionals. In the last two decades, researchers have gone past several milestones in tackling cancer while gaining insight into the role of apoptosis in cancer or targeting various biomarker tools for prognosis and diagnosis. Apoptosis which is still a topic full of complexities, can be controlled considerably by B-cell lymphoma 2 (BCL-2) and its family members. Therefore, targeting proteins of this family to prevent tumorigenesis, is essential to focus on the pharmacological features of the anti-apoptotic and pro-apoptotic members, which will help to develop and manage this disorder. This review deals with the advancements of various epigenetic regulators to target BCL-2 family proteins, including the mechanism of several microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). Similarly, a rise in natural and synthetic molecules' research over the last two decades has allowed us to acquire insights into understanding and managing the transcriptional alterations that have led to apoptosis and treating various neoplastic diseases. Furthermore, several inhibitors targeting anti-apoptotic proteins and inducers or activators targeting pro-apoptotic proteins in preclinical and clinical stages have been summarized. Overall, agonistic and antagonistic mechanisms of BCL-2 family proteins conciliated by epigenetic regulators, natural and synthetic agents have proven to be an excellent choice in developing cancer therapeutics.
Collapse
Affiliation(s)
- Arnab Sarkar
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata, 700032, India.
| | - Abhik Paul
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata, 700032, India.
| | - Tanmoy Banerjee
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata, 700032, India.
| | - Avik Maji
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata, 700032, India.
| | - Sanjukta Saha
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata, 700032, India.
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| | - Tapan Kumar Maity
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata, 700032, India.
| |
Collapse
|
17
|
Peng Z, Gillissen B, Richter A, Sinnberg T, Schlaak MS, Eberle J. Enhanced Apoptosis and Loss of Cell Viability in Melanoma Cells by Combined Inhibition of ERK and Mcl-1 Is Related to Loss of Mitochondrial Membrane Potential, Caspase Activation and Upregulation of Proapoptotic Bcl-2 Proteins. Int J Mol Sci 2023; 24:ijms24054961. [PMID: 36902392 PMCID: PMC10002974 DOI: 10.3390/ijms24054961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Targeting of MAP kinase pathways by BRAF inhibitors has evolved as a key therapy for BRAF-mutated melanoma. However, it cannot be applied for BRAF-WT melanoma, and also, in BRAF-mutated melanoma, tumor relapse often follows after an initial phase of tumor regression. Inhibition of MAP kinase pathways downstream at ERK1/2, or inhibitors of antiapoptotic Bcl-2 proteins, such as Mcl-1, may serve as alternative strategies. As shown here, the BRAF inhibitor vemurafenib and the ERK inhibitor SCH772984 showed only limited efficacy in melanoma cell lines, when applied alone. However, in combination with the Mcl-1 inhibitor S63845, the effects of vemurafenib were strongly enhanced in BRAF-mutated cell lines, and the effects of SCH772984 were enhanced in both BRAF-mutated and BRAF-WT cells. This resulted in up to 90% loss of cell viability and cell proliferation, as well as in induction of apoptosis in up to 60% of cells. The combination of SCH772984/S63845 resulted in caspase activation, processing of poly (ADP-ribose) polymerase (PARP), phosphorylation of histone H2AX, loss of mitochondrial membrane potential, and cytochrome c release. Proving the critical role of caspases, a pan-caspase inhibitor suppressed apoptosis induction, as well as loss of cell viability. As concerning Bcl-2 family proteins, SCH772984 enhanced expression of the proapoptotic Bim and Puma, as well as decreased phosphorylation of Bad. The combination finally resulted in downregulation of antiapoptotic Bcl-2 and enhanced expression of the proapoptotic Noxa. In conclusion, combined inhibition of ERK and Mcl-1 revealed an impressive efficacy both in BRAF-mutated and WT melanoma cells, and may thus represent a new strategy for overcoming drug resistance.
Collapse
Affiliation(s)
- Zhe Peng
- Skin Cancer Centre Charité, Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Clinical Medicine, University of South China, Hengyang 421001, China
| | - Bernhard Gillissen
- Department of Hematology, Oncology, and Tumor Immunology, Charité—Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Antje Richter
- Department of Hematology, Oncology, and Tumor Immunology, Charité—Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Tobias Sinnberg
- Skin Cancer Centre Charité, Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Division of Dermatooncology, Department of Dermatology, University Tübingen, 72076 Tübingen, Germany
| | - Max S. Schlaak
- Skin Cancer Centre Charité, Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jürgen Eberle
- Skin Cancer Centre Charité, Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Correspondence:
| |
Collapse
|
18
|
Tantawy SI, Sarkar A, Hubner S, Tan Z, Wierda WG, Eldeib A, Zhang S, Kornblau S, Gandhi V. Mechanisms of MCL-1 Protein Stability Induced by MCL-1 Antagonists in B-Cell Malignancies. Clin Cancer Res 2023; 29:446-457. [PMID: 36346691 PMCID: PMC9852224 DOI: 10.1158/1078-0432.ccr-22-2088] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/19/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE Several MCL-1 inhibitors (MCL-1i), including AMG-176 and AZD5991, have shown promise in preclinical studies and are being tested for the treatment of hematologic malignancies. A unique feature of these agents is induction and stability of Mcl-1 protein; however, the precise mechanism is unknown. We aim to study the mechanism of MCL-1i-induced Mcl-1 protein stability. EXPERIMENTAL DESIGN Using several B-cell leukemia and lymphoma cell lines and primary chronic lymphocytic leukemia (CLL) lymphocytes, we evaluated molecular events associated with Mcl-1 protein stability including protein half-life, reverse-phase protein array, protein-protein interaction, phosphorylation, ubiquitination, and de-ubiquitination, followed by molecular simulation and modeling. RESULTS Using both in vivo and in vitro analysis, we demonstrate that MCL-1i-induced Mcl-1 protein stability is predominantly associated with defective Mcl-1 ubiquitination and concurrent apoptosis induction in both cell lines and primary CLL subjects. These MCL1i also induced ERK-mediated Mcl-1Thr163 phosphorylation, which partially contributed to Mcl-1 stability. Disruption of Mcl-1:Noxa interaction followed by Noxa degradation, enhanced Mcl-1 de-ubiquitination by USP9x, and Mule destabilization are the major effects of these inhibitors. However, unlike other BH3 proteins, Mule:Mcl-1 interaction was unaffected by MCL-1i. WP1130, a global deubiquitinase (DUB) inhibitor, abrogated Mcl-1 induction reaffirming a critical role of DUBs in the observed Mcl-1 protein stability. Further, in vitro ubiquitination studies of Mcl-1 showed distinct difference among these inhibitors. CONCLUSIONS We conclude that MCL-1i blocked Mcl-1 ubiquitination via enhanced de-ubiquitination and dissociation of Mcl-1 from Noxa, Bak and Bax, and Mule de-stabilization. These are critical events associated with increased Mcl-1 protein stability with AMG-176 and AZD5991.
Collapse
Affiliation(s)
- Shady I. Tantawy
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas,Department of Internal Medicine, College of Medicine, Suez Canal University, Ismailia, Egypt
| | - Aloke Sarkar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stefan Hubner
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhi Tan
- Center for Drug Discovery, Department of Pathology and Immunology, Department of Pharmacology and Chemical Biology, Baylor College of Medicine
| | - William G. Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abdelraouf Eldeib
- Center for Drug Discovery, Department of Pathology and Immunology, Department of Pharmacology and Chemical Biology, Baylor College of Medicine
| | - Shuxing Zhang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven Kornblau
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
19
|
Tannoury M, Garnier D, Susin SA, Bauvois B. Current Status of Novel Agents for the Treatment of B Cell Malignancies: What's Coming Next? Cancers (Basel) 2022; 14:6026. [PMID: 36551511 PMCID: PMC9775488 DOI: 10.3390/cancers14246026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/29/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Resistance to death is one of the hallmarks of human B cell malignancies and often contributes to the lack of a lasting response to today's commonly used treatments. Drug discovery approaches designed to activate the death machinery have generated a large number of inhibitors of anti-apoptotic proteins from the B-cell lymphoma/leukemia 2 family and the B-cell receptor (BCR) signaling pathway. Orally administered small-molecule inhibitors of Bcl-2 protein and BCR partners (e.g., Bruton's tyrosine kinase and phosphatidylinositol-3 kinase) have already been included (as monotherapies or combination therapies) in the standard of care for selected B cell malignancies. Agonistic monoclonal antibodies and their derivatives (antibody-drug conjugates, antibody-radioisotope conjugates, bispecific T cell engagers, and chimeric antigen receptor-modified T cells) targeting tumor-associated antigens (TAAs, such as CD19, CD20, CD22, and CD38) are indicated for treatment (as monotherapies or combination therapies) of patients with B cell tumors. However, given that some patients are either refractory to current therapies or relapse after treatment, novel therapeutic strategies are needed. Here, we review current strategies for managing B cell malignancies, with a focus on the ongoing clinical development of more effective, selective drugs targeting these molecules, as well as other TAAs and signaling proteins. The observed impact of metabolic reprogramming on B cell pathophysiology highlights the promise of targeting metabolic checkpoints in the treatment of these disorders.
Collapse
Affiliation(s)
| | | | | | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France
| |
Collapse
|
20
|
Pallerla SR, Hoan NX, Rachakonda S, Meyer CG, Van Tong H, Toan NL, Linh LTK, Giang DP, Kremsner PG, Bang MH, Song LH, Velavan TP. Custom gene expression panel for evaluation of potential molecular markers in hepatocellular carcinoma. BMC Med Genomics 2022; 15:235. [PMID: 36345011 PMCID: PMC9641913 DOI: 10.1186/s12920-022-01386-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 10/14/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related mortality worldwide. It is a highly heterogeneous disease with poor prognosis and limited treatment options, which highlights the need for reliable biomarkers. This study aims to explore molecular markers that allow stratification of HCC and may lead to better prognosis and treatment prediction. MATERIALS AND METHODS We studied 20 candidate genes (HCC hub genes, potential drug target genes, predominant somatic mutant genes) retrieved from literature and public databases with potential to be used as the molecular markers. We analysed expression of the genes by RT-qPCR in 30 HCC tumour and adjacent non-tumour paired samples from Vietnamese patients. Fold changes in expression were then determined using the 2-∆∆CT method, and unsupervised hierarchical clustering was generated using Cluster v3.0 software. RESULTS Clustering of expression data revealed two subtypes of tumours (proliferative and normal-like) and four clusters for genes. The expression profiles of the genes TOP2A, CDK1, BIRC5, GPC3, IGF2, and AFP were strongly correlated. Proliferative tumours were characterized by high expression of the c-MET, ARID1A, CTNNB1, RAF1, LGR5, and GLUL1 genes. TOP2A, CDK1, and BIRC5 HCC hub genes were highly expressed (> twofold) in 90% (27/30), 83% (25/30), and 83% (24/30) in the tissue samples, respectively. Among the drug target genes, high expression was observed in the GPC3, IGF2 and c-MET genes in 77% (23/30), 63% (19/30), and 37% (11/30), respectively. The somatic mutant Wnt/ß-catenin genes (CTNNB1, GLUL and LGR5) and TERT were highly expressed in 40% and 33% of HCCs, respectively. Among the HCC marker genes, a higher percentage of tumours showed GPC3 expression compared to AFP expression [73% (23/30) vs. 43% (13/30)]. CONCLUSION The custom panel and molecular markers from this study may be useful for diagnosis, prognosis, biomarker-guided clinical trial design, and prediction of treatment outcomes.
Collapse
Affiliation(s)
- Srinivas Reddy Pallerla
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Universität Tübingen, Wilhelmstr 27, 72074, Tübingen, Germany.
| | - Nghiem Xuan Hoan
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam.
- Department of Molecular Biology, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam.
| | - Sivaramakrishna Rachakonda
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Universität Tübingen, Wilhelmstr 27, 72074, Tübingen, Germany
| | - Christian G Meyer
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Universität Tübingen, Wilhelmstr 27, 72074, Tübingen, Germany
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
| | | | | | - Le Thi Kieu Linh
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Universität Tübingen, Wilhelmstr 27, 72074, Tübingen, Germany
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
| | - Dao Phuong Giang
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
- Department of Molecular Biology, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam
| | - Peter G Kremsner
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Universität Tübingen, Wilhelmstr 27, 72074, Tübingen, Germany
- Centre de Recherches Medicales de Lambarene, Lambaréné, Gabon
| | - Mai Hong Bang
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
- Faculty of Gastroenterology, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam
| | - Le Huu Song
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
- Department of Molecular Biology, 108 Institute of Clinical Medical and Pharmaceutical Sciences, Hanoi, Vietnam
| | - Thirumalaisamy P Velavan
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Universität Tübingen, Wilhelmstr 27, 72074, Tübingen, Germany
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
| |
Collapse
|
21
|
Pal P, Zhang P, Poddar SK, Zheng G. Patent landscape of inhibitors and PROTACs of the anti-apoptotic BCL-2 family proteins. Expert Opin Ther Pat 2022; 32:1003-1026. [PMID: 35993382 PMCID: PMC9942934 DOI: 10.1080/13543776.2022.2116311] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 08/19/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The anti-apoptotic BCL-2 family proteins, such as BCL-2, BCL-XL, and MCL-1, are excellent cancer therapeutic targets. The FDA approval of BCL-2 selective inhibitor venetoclax in 2016 validated the strategy of targeting these proteins with BH3 mimetic small molecule inhibitors. AREAS COVERED This review provides an overview of the patent literature between 2016 and 2021 covering inhibitors and PROTACs of the anti-apoptotic BCL-2 proteins. EXPERT OPINION Since the FDA approval of venetoclax, tremendous efforts have been made to develop its analogues with improved drug properties. These activities will likely result in new drugs in coming years. Significant progress on MCL-1 inhibitors has also been made, with multiple compounds entering clinical trials. However, MCL-1 inhibition could cause on-target toxicity to normal tissues especially the heart. Similar issue exists with BCL-XL inhibitors, which cause on-target platelet toxicity. To overcome this issue, several strategies have been applied, including prodrug, dendrimer-based drug delivery, antibody-drug conjugate (ADC), and proteolysis targeting chimera (PROTAC); and amazingly, each of these approaches has resulted in a drug candidate entering clinical trials. We envision technologies like ADC and PROTAC could also be utilized to increase the therapeutic index of MCL-1 inhibitors.
Collapse
Affiliation(s)
- Pratik Pal
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Peiyi Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Saikat K Poddar
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| |
Collapse
|
22
|
Chan AM, Goodis CC, Pommier EG, Fletcher S. Recent applications of covalent chemistries in protein-protein interaction inhibitors. RSC Med Chem 2022; 13:921-928. [PMID: 36092144 PMCID: PMC9384789 DOI: 10.1039/d2md00112h] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/30/2022] [Indexed: 10/17/2023] Open
Abstract
Protein-protein interactions (PPIs) are large, often featureless domains whose modulations by small-molecules are challenging. Whilst there are some notable successes, such as the BCL-2 inhibitor venetoclax, the requirement for larger ligands to achieve the desired level of potency and selectivity may result in poor "drug-like" properties. Covalent chemistry is presently enjoying a renaissance. In particular, targeted covalent inhibition (TCI), in which a weakly electrophilic "warhead" is installed onto a protein ligand scaffold, is a powerful strategy to develop potent inhibitors of PPIs that are smaller/more drug-like yet have enhanced affinities by virtue of the reinforcing effect on the existing non-covalent interactions by the resulting protein-ligand covalent bond. Furthermore, the covalent bond delivers sustained inhibition, which may translate into significantly reduced therapeutic dosing. Herein, we discuss recent applications of a spectrum of TCIs, as well as covalent screening strategies, in the discovery of more effective inhibitors of PPIs using the HDM2 and BCL-2 protein families as case studies.
Collapse
Affiliation(s)
- Alexandria M Chan
- University of Maryland School of Pharmacy, Department of Pharmaceutical Sciences 20 N. Pine St Baltimore MD 21201 USA
| | - Christopher C Goodis
- University of Maryland School of Pharmacy PharmD Program, 20 N. Pine St Baltimore MD 21201 USA
| | - Elie G Pommier
- University of Maryland School of Pharmacy PharmD Program, 20 N. Pine St Baltimore MD 21201 USA
| | - Steven Fletcher
- University of Maryland School of Pharmacy, Department of Pharmaceutical Sciences 20 N. Pine St Baltimore MD 21201 USA
- University of Maryland Greenebaum Cancer Center 20 S. Greene St Baltimore MD 21201 USA
| |
Collapse
|
23
|
Peng F, Liao M, Qin R, Zhu S, Peng C, Fu L, Chen Y, Han B. Regulated cell death (RCD) in cancer: key pathways and targeted therapies. Signal Transduct Target Ther 2022; 7:286. [PMID: 35963853 PMCID: PMC9376115 DOI: 10.1038/s41392-022-01110-y] [Citation(s) in RCA: 320] [Impact Index Per Article: 106.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023] Open
Abstract
Regulated cell death (RCD), also well-known as programmed cell death (PCD), refers to the form of cell death that can be regulated by a variety of biomacromolecules, which is distinctive from accidental cell death (ACD). Accumulating evidence has revealed that RCD subroutines are the key features of tumorigenesis, which may ultimately lead to the establishment of different potential therapeutic strategies. Hitherto, targeting the subroutines of RCD with pharmacological small-molecule compounds has been emerging as a promising therapeutic avenue, which has rapidly progressed in many types of human cancers. Thus, in this review, we focus on summarizing not only the key apoptotic and autophagy-dependent cell death signaling pathways, but the crucial pathways of other RCD subroutines, including necroptosis, pyroptosis, ferroptosis, parthanatos, entosis, NETosis and lysosome-dependent cell death (LCD) in cancer. Moreover, we further discuss the current situation of several small-molecule compounds targeting the different RCD subroutines to improve cancer treatment, such as single-target, dual or multiple-target small-molecule compounds, drug combinations, and some new emerging therapeutic strategies that would together shed new light on future directions to attack cancer cell vulnerabilities with small-molecule drugs targeting RCD for therapeutic purposes.
Collapse
Affiliation(s)
- Fu Peng
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Minru Liao
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shiou Zhu
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Leilei Fu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Yi Chen
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
24
|
Wang X, Liu X, Huang J, Liu C, Li H, Wang C, Hong Q, Lei Y, Xia J, Yu Z, Dong R, Xu J, Tu Z, Duan C, Li S, Lu T, Tang W, Chen Y. Discovery of 2H-benzo[b][1,4]oxazin-3(4H)-one derivatives as potent and selective CDK9 inhibitors that enable transient target engagement for the treatment of hematologic malignancies. Eur J Med Chem 2022; 238:114461. [PMID: 35605362 DOI: 10.1016/j.ejmech.2022.114461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022]
Abstract
Cyclin-dependent kinase 9 (CDK9) is a transcriptional regulator and a potential therapeutic target in hematologic malignancies. Selective and transient CDK9 inhibition reduces Mcl-1 expression and induces apoptosis in Mcl-1-dependent tumor cells for survival. Here, we describe our efforts to discover a novel series of 2H-benzo[b][1,4]oxazin-3(4H)-one as CDK9 inhibitors. Compound 32k was identified as a selective CDK9 inhibitor with short pharmacokinetic and physicochemical properties suitable for intravenous administration. Short-term treatment with 32k resulted in a rapid dose-dependent decrease in cellular p-Ser2-RNAPII, Mcl-1 and c-Myc, leading to apoptosis in the MV4-11 cell line. Correspondingly, significant in vivo antitumor efficacy was observed in xenograft models derived from multiple hematological tumors with intermittent 32k dosing. These results provide evidence that selective transient CDK9 inhibitors could be used for the treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Xinren Wang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Xiaoyue Liu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Jianhang Huang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Chenhe Liu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Hongmei Li
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Cong Wang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Qianqian Hong
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Yan Lei
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Jiawei Xia
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Ziheng Yu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Ruinan Dong
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Junyu Xu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Zhenlin Tu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - ChunQi Duan
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Shuwen Li
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China
| | - Tao Lu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Weifang Tang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Yadong Chen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, PR China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China.
| |
Collapse
|
25
|
Uthale A, Anantram A, Sulkshane P, Degani M, Teni T. Identification of bicyclic compounds that act as dual inhibitors of Bcl-2 and Mcl-1. Mol Divers 2022:10.1007/s11030-022-10494-6. [PMID: 35909144 DOI: 10.1007/s11030-022-10494-6] [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: 04/30/2022] [Accepted: 07/02/2022] [Indexed: 10/16/2022]
Abstract
Elevated expression of anti-apoptotic proteins, such as Bcl-2 and Mcl-1 contributes to poor prognosis and resistance to current treatment modalities in multiple cancers. Here, we report the design, synthesis and characterization of benzimidazole chalcone and flavonoid scaffold-derived bicyclic compounds targeting both Bcl-2 and Mcl-1 by optimizing the structural differences in the binding sites of both these proteins. Initial docking screen of Bcl-2 and Mcl-1 with pro-apoptotic protein Bim revealed possible hits with optimal binding energies. All the optimized bicyclic compounds were screened for their in vitro cytotoxic activity against two oral cancer cell lines (AW8507 and AW13516) which express high levels of Bcl-2 and Mcl-1. Compound 4d from the benzimidazole chalcone series and compound 6d from the flavonoid series exhibited significant cytotoxic activity (IC50 7.12 μM and 17.18 μM, respectively) against AW13516 cell line. Time Resolved-Fluorescence Resonance Energy Transfer (TR-FRET) analysis further demonstrated that compound 4d and compound 6d could effectively inhibit the Bcl-2 and Mcl-1 proteins by displacing their BH3 binding partners. Both compounds exhibited potent activation of canonical pathway of apoptosis evident from appearance of cleaved Caspase-3 and PARP. Further, treatment of oral cancer cells with the inhibitors induced dissociation of the BH3 only protein Bim from Mcl-1 and Bak from Bcl-2 but failed to release Bax from Bcl-xL thereby confirming the nature of compounds as BH3-mimetics selectively targeting Bcl-2 and Mcl-1. Our study thus identifies bicyclic compounds as promising candidates for anti-apoptotic Bcl-2/Mcl-1 dual inhibitors with a potential for further development.
Collapse
Affiliation(s)
- Abhay Uthale
- Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410 210, India.,Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Aarti Anantram
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, 400 019, India
| | - Prasad Sulkshane
- Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410 210, India.,Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Mariam Degani
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, 400 019, India.
| | - Tanuja Teni
- Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410 210, India. .,Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India.
| |
Collapse
|
26
|
Deng H, Huang M, Liu H, Zhang H, Liu L, Gao B, Li X, Li J, Niu Q, Zhang Z, Luan S, Zhang J, Jing Y, Liu D, Zhao L. Development of a series of novel Mcl-1 inhibitors bearing an indole carboxylic acid moiety. Bioorg Chem 2022; 127:106018. [PMID: 35901526 DOI: 10.1016/j.bioorg.2022.106018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/03/2022] [Accepted: 07/07/2022] [Indexed: 11/25/2022]
Abstract
The B cell lymphoma protein 2 (Bcl-2) family proteins regulate cell apoptosis by participating in the endogenous apoptosis pathway. As an important anti-apoptotic protein, Myeloid cell leukemia 1 (Mcl-1) is overexpressed in a variety of tumor cells, and targeting this protein has been a promising strategy for cancer therapy. Herein, based on the 1H-indole-5-carboxylic acid structure previously discovered, we have developed a series of novel compounds with increased affinities and selectivity toward Mcl-1 through structure-based drug design. Among those compounds, 26 exerted relatively better affinity and selectivity for Mcl-1 with moderate inhibition in HL-60 cells. Mechanism studies showed that compound 26 could induce cancer cells apoptosis in an Mcl-1-dependent manner. It also exhibited good microsomal and plasma stability with acceptable pharmacokinetics profiles. Furthermore, treatment with target compound in a 4T1 xenograft mouse model significantly suppressed the tumor growth. Overall, the small molecule described herein represents a promising Mcl-1 inhibitor for further study.
Collapse
Affiliation(s)
- Hongguang Deng
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Min Huang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hui Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Liaoning Key Laboratory of Targeting Drugs for Hematological Malignancies, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hong Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Liang Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bensheng Gao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xianlu Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qun Niu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhenwei Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shenglin Luan
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingyi Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Liaoning Key Laboratory of Targeting Drugs for Hematological Malignancies, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yongkui Jing
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Liaoning Key Laboratory of Targeting Drugs for Hematological Malignancies, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
27
|
Mukherjee N, Dart CR, Amato CM, Honig-Frand A, Lambert JR, Lambert KA, Robinson WA, Tobin RP, McCarter MD, Couts KL, Fujita M, Norris DA, Shellman YG. Expression Differences in BCL2 Family Members between Uveal and Cutaneous Melanomas Account for Varying Sensitivity to BH3 Mimetics. J Invest Dermatol 2022; 142:1912-1922.e7. [PMID: 34942200 PMCID: PMC9635014 DOI: 10.1016/j.jid.2021.11.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/04/2021] [Accepted: 11/19/2021] [Indexed: 10/19/2022]
Abstract
Uveal melanoma (UM) is a subtype of melanoma. Although they share a melanocytic origin with cutaneous melanoma (CM), patients with UM have few treatment options. BCL2 homologous 3 mimetics are small-molecule drugs that mimic proapoptotic BCL2 family members. We compared BCL2 family member expression between UM and CM using immunoblot and The Cancer Genome Atlas transcriptomic analysis. UM has a unique signature of low BFL1 and high PUMA proteins compared with CM and 30 other cancer types, making them an attractive candidate for BCL2 homologous 3 protein mimetics. We tested the efficacy of a BCL2 inhibitor and MCL1 inhibitor (MCL1i) in UM, with viability assays, live-cell imaging, sphere assays, and mouse xenograft models. UM had a higher sensitivity to MCL1i than CM. Overexpression of BFL1 or knockdown of PUMA made the UM more resistant to MCL1i. In contrast, MAPK/extracellular signal‒regulated kinase inhibitor treatment in CM made them more sensitive to MCL1i. However, MCL1i-alone treatment was not very effective to reduce the UM initiating cells; to overcome this, we employed a combination of MCL1i with BCL2 inhibitor that synergistically inhibited UM initiating cell's capacity to expand. Overall, we identify a distinct expression profile of BCL2 family members for UM that makes them susceptible to BCL2 homologous 3 mimetics.
Collapse
Affiliation(s)
- Nabanita Mukherjee
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Chiara R Dart
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Carol M Amato
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Adam Honig-Frand
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - James R Lambert
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Karoline A Lambert
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - William A Robinson
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard P Tobin
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Martin D McCarter
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kasey L Couts
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mayumi Fujita
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Dermatology Section, U.S. Department of Veterans Affairs Medical Center, Denver, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David A Norris
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Dermatology Section, U.S. Department of Veterans Affairs Medical Center, Denver, Colorado, USA
| | - Yiqun G Shellman
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
28
|
Byadi S, Sadik K, Hachim ME, Daoudi M, Podlipnik Č, Aboulmouhajir A. Discovery of a New Mcl‐1 Protein Inhibitor through the QSAR Approach and Molecular Docking Study. ADVANCED THEORY AND SIMULATIONS 2022. [DOI: 10.1002/adts.202100590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Said Byadi
- Organic Synthesis Extraction and Valorization Laboratory Team of Extraction Spectroscopy and Valorization Sciences Faculty of Ain Chock Hassan II University Casablanca 20100 Morocco
| | - Karima Sadik
- Team of Molecular Modelling and Spectroscopy Sciences Faculty Chouaib Doukkali University El Jadida 24000 Morocco
| | - Mouhi Eddine Hachim
- Team of Molecular Modelling and Spectroscopy Sciences Faculty Chouaib Doukkali University El Jadida 24000 Morocco
| | - Mohamed Daoudi
- Laboratory of Organic and Bio‐Organic Chemistry and the Environment Sciences Faculty Chouaib Doukkali University El Jadida 24000 Morocco
| | - Črtomir Podlipnik
- Faculty of Chemistry and Chemical Technology University of Ljubljana Ljubljana 1000 Slovenia
| | - Aziz Aboulmouhajir
- Organic Synthesis Extraction and Valorization Laboratory Team of Extraction Spectroscopy and Valorization Sciences Faculty of Ain Chock Hassan II University Casablanca 20100 Morocco
- Team of Molecular Modelling and Spectroscopy Sciences Faculty Chouaib Doukkali University El Jadida 24000 Morocco
| |
Collapse
|
29
|
Sulkshane P, Teni T. Myeloid cell leukemia-1: a formidable barrier to anticancer therapeutics and the quest of targeting it. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:278-296. [PMID: 36045907 PMCID: PMC9400788 DOI: 10.37349/etat.2022.00083] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/16/2022] [Indexed: 11/22/2022] Open
Abstract
The antiapoptotic B cell lymphoma-2 (Bcl-2) family members are apical regulators of the intrinsic pathway of apoptosis that orchestrate mitochondrial outer membrane permeabilization (MOMP) through interactions with their proapoptotic counterparts. Overexpression of antiapoptotic Bcl-2 family proteins has been linked to therapy resistance and poor prognosis in diverse cancers. Among the antiapoptotic Bcl-2 family members, predominant overexpression of the prosurvival myeloid cell leukemia-1 (Mcl-1) has been reported in a myriad of hematological malignancies and solid tumors, contributing to therapy resistance and poor outcomes, thus making it a potential druggable target. The unique structure of Mcl-1 and its complex regulatory mechanism makes it an adaptive prosurvival switch that ensures tumor cell survival despite therapeutic intervention. This review focusses on diverse mechanisms adopted by tumor cells to maintain sustained elevated levels of Mcl-1 and how high Mcl-1 levels contribute to resistance in conventional as well as targeted therapies. Moreover, recent developments in the Mcl-1-targeted therapeutics and the underlying challenges and considerations in designing novel Mcl-1 inhibitors are also discussed.
Collapse
Affiliation(s)
- Prasad Sulkshane
- Glickman Laboratory, Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Tanuja Teni
- Teni Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Mumbai 400094, India
| |
Collapse
|
30
|
He W, Li X, Morsch M, Ismail M, Liu Y, Rehman FU, Zhang D, Wang Y, Zheng M, Chung R, Zou Y, Shi B. Brain-Targeted Codelivery of Bcl-2/Bcl-xl and Mcl-1 Inhibitors by Biomimetic Nanoparticles for Orthotopic Glioblastoma Therapy. ACS NANO 2022; 16:6293-6308. [PMID: 35353498 DOI: 10.1021/acsnano.2c00320] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Glioblastoma (GBM) is among the most treatment-resistant solid tumors and often recurrs after resection. One of the mechanisms through which GBM escapes various treatment modalities is the overexpression of anti-apoptotic Bcl-2 family proteins (e.g., Bcl-2, Bcl-xl, and Mcl-1) in tumor cells. Small-molecule inhibitors such as ABT-263 (ABT), which can promote mitochondrial-mediated cell apoptosis by selectively inhibiting the function of Bcl-2 and Bcl-xl, have been proven to be promising anticancer agents in clinical trials. However, the therapeutic prospects of ABT for GBM treatment are hampered by its limited blood-brain barrier (BBB) penetration, dose-dependent thrombocytopenia, and the drug resistance driven by Mcl-1, which is overexpressed in GBM cells and further upregulated upon treatment with ABT. Herein, we reported that the Mcl-1-specific inhibitor A-1210477 (A12) can act synergistically with ABT to induce potent cell apoptosis in U87 MG cells, drug-resistant U251 cells, and patient-derived GBM cancer stem cells. We further designed a biomimetic nanomedicine, based on the apolipoprotein E (ApoE) peptide-decorated red blood cell membrane and pH-sensitive dextran nanoparticles, for the brain-targeted delivery of ABT and A12. The synergistic anti-GBM effect was retained after encapsulation in the nanomedicine. Additionally, the obtained nanomedicine possessed good biocompatibility, exhibited efficient BBB penetration, and could effectively suppress tumor growth and prolong the survival time of mice bearing orthotopic GBM xenografts without inducing detectable adverse effects.
Collapse
Affiliation(s)
| | | | - Marco Morsch
- Center for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | | | | | | | | | | | | | - Roger Chung
- Center for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Yan Zou
- Center for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Bingyang Shi
- Center for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
31
|
Sellin M, Mack R, Rhodes MC, Zhang L, Berg S, Joshi K, Liu S, Wei W, S. J. PB, Larsen P, Taylor RE, Zhang J. Molecular mechanisms by which splice modulator GEX1A inhibits leukaemia development and progression. Br J Cancer 2022; 127:223-236. [PMID: 35422078 PMCID: PMC9296642 DOI: 10.1038/s41416-022-01796-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/18/2022] [Accepted: 03/17/2022] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION Splice modulators have been assessed clinically in treating haematologic malignancies exhibiting splice factor mutations and acute myeloid leukaemia. However, the mechanisms by which such modulators repress leukaemia remain to be elucidated. OBJECTIVES The primary goal of this assessment was to assess the molecular mechanism by which the natural splice modulator GEX1A kills leukaemic cells in vitro and within in vivo mouse models. METHODS Using human leukaemic cell lines, we assessed the overall sensitivity these cells have to GEX1A via EC50 analysis. We subsequently analysed its effects using in vivo xenograft mouse models and examined whether cell sensitivities were correlated to genetic characteristics or protein expression levels. We also utilised RT-PCR and RNAseq analyses to determine splice change and RNA expression level differences between sensitive and resistant leukaemic cell lines. RESULTS We found that, in vitro, GEX1A induced an MCL-1 isoform shift to pro-apoptotic MCL-1S in all leukaemic cell types, though sensitivity to GEX1A-induced apoptosis was negatively associated with BCL-xL expression. In BCL-2-expressing leukaemic cells, GEX1A induced BCL-2-dependent apoptosis by converting pro-survival BCL-2 into a cell killer. Thus, GEX1A + selective BCL-xL inhibition induced synergism in killing leukaemic cells, while GEX1A + BCL-2 inhibition showed antagonism in BCL-2-expressing leukaemic cells. In addition, GEX1A sensitised FLT3-ITD+ leukaemic cells to apoptosis by inducing aberrant splicing and repressing the expression of FLT3-ITD. Consistently, in in vivo xenografts, GEX1A killed the bulk of leukaemic cells via apoptosis when combined with BCL-xL inhibition. Furthermore, GEX1A repressed leukaemia development by targeting leukaemia stem cells through inhibiting FASTK mitochondrial isoform expression across sensitive and non-sensitive leukaemia types. CONCLUSION Our study suggests that GEX1A is a potent anti-leukaemic agent in combination with BCL-xL inhibitors, which targets leukaemic blasts and leukaemia stem cells through distinct mechanisms.
Collapse
|
32
|
MEK and MCL-1 sequential inhibition synergize to enhance rhabdomyosarcoma treatment. Cell Death Dis 2022; 8:172. [PMID: 35393436 PMCID: PMC8989976 DOI: 10.1038/s41420-022-00959-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 02/18/2022] [Accepted: 03/16/2022] [Indexed: 11/09/2022]
Abstract
Targeted agents have emerged as promising molecules for cancer treatment, but most of them fail to achieve complete tumor regression or attain durable remissions due to tumor adaptations. We used dynamic BH3 profiling to identify targeted agents effectiveness and anti-apoptotic adaptations upon targeted treatment in rhabdomyosarcoma. We focused on studying the use of BH3 mimetics to specifically inhibit pro-survival BCL-2 family proteins, overwhelm resistance to therapy and prevent relapse. We observed that the MEK1/2 inhibitor trametinib rapidly depleted the pro-apoptotic protein NOXA, thus increasing MCL-1 availability. Indeed, we found that the MCL-1 inhibitor S63845 synergistically enhanced trametinib cytotoxicity in rhabdomyosarcoma cells in vitro and in vivo. In conclusion, our findings indicate that the combination of a BH3 mimetic targeting MCL-1 with trametinib improves efficiency on rhabdomyosarcoma by blocking tumor adaptation to treatment.
Collapse
|
33
|
Bourafai-Aziez A, Benabderrahmane M, Paysant H, Weiswald LB, Poulain L, Carlier L, Ravault D, Jouanne M, Coadou G, Oulyadi H, Voisin-Chiret AS, Sopková-de Oliveira Santos J, Sebban M. Drug Repurposing: Deferasirox Inhibits the Anti-Apoptotic Activity of Mcl-1. Drug Des Devel Ther 2021; 15:5035-5059. [PMID: 34949914 PMCID: PMC8688747 DOI: 10.2147/dddt.s323077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/27/2021] [Indexed: 11/30/2022] Open
Abstract
Introduction With the aim of repositioning commercially available drugs for the inhibition of the anti-apoptotic myeloid cell leukemia protein, Mcl-1, implied in various cancers, five molecules, highlighted from a published theoretical screening, were selected to experimentally validate their affinity toward Mcl-1. Results A detailed NMR study revealed that only two of the five tested drugs, Torsemide and Deferasirox, interacted with Mcl-1. NMR data analysis allowed the complete characterization of the binding mode of both drugs to Mcl-1, including the estimation of their affinity for Mcl-1. Biological assays evidenced that the biological activity of Torsemide was lower as compared to the Deferasirox, which was able to efficiently and selectively inhibit the anti-apoptotic activity of Mcl-1. Finally, docking and molecular dynamics led to a 3D model for the Deferasirox:Mcl-1 complex and revealed the positioning of the drug in the Mcl-1 P2/P3 pockets as well as almost all synthetic Mcl-1 inhibitors. Interestingly, contrary to known synthetic Mcl-1 inhibitors which interact through Arg263, Deferasirox, establishes a salt bridge with Lys234. Conclusion Deferasirox could be a potential candidate for drug repositioning as Mcl-1 inhibitor.
Collapse
Affiliation(s)
- Asma Bourafai-Aziez
- Normandie Université, UNIROUEN, INSA de Rouen, CNRS Laboratoire COBRA (UMR 6014 & FR 3038), Rouen, 76000, France
| | | | - Hippolyte Paysant
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE «Interdisciplinary Research Unit for Cancer Prevention and Treatment», Biology and Innovative Therapeutics for Ovarian Cancers Group (BioTICLA), Centre de Lutte Contre le Cancer F. Baclesse, Caen, 14076, France.,UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, Caen, 14076, France
| | - Louis-Bastien Weiswald
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE «Interdisciplinary Research Unit for Cancer Prevention and Treatment», Biology and Innovative Therapeutics for Ovarian Cancers Group (BioTICLA), Centre de Lutte Contre le Cancer F. Baclesse, Caen, 14076, France.,UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, Caen, 14076, France
| | - Laurent Poulain
- Normandie Université, UNICAEN, Inserm U1086 ANTICIPE «Interdisciplinary Research Unit for Cancer Prevention and Treatment», Biology and Innovative Therapeutics for Ovarian Cancers Group (BioTICLA), Centre de Lutte Contre le Cancer F. Baclesse, Caen, 14076, France.,UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, Caen, 14076, France
| | - Ludovic Carlier
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Paris, France
| | - Delphine Ravault
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Paris, France
| | | | - Gaël Coadou
- Normandie Université, UNIROUEN, INSA de Rouen, CNRS Laboratoire COBRA (UMR 6014 & FR 3038), Rouen, 76000, France
| | - Hassan Oulyadi
- Normandie Université, UNIROUEN, INSA de Rouen, CNRS Laboratoire COBRA (UMR 6014 & FR 3038), Rouen, 76000, France
| | | | | | - Muriel Sebban
- Normandie Université, UNIROUEN, INSA de Rouen, CNRS Laboratoire COBRA (UMR 6014 & FR 3038), Rouen, 76000, France
| |
Collapse
|
34
|
Nie W, Wang B, Mi X, Chen J, Yu T, Miao J, Lin Y, Yang T, Ran M, Hong Z, Liu X, Liang X, Qian Z, Gao X. Co-Delivery of Paclitaxel and shMCL-1 by Folic Acid-Modified Nonviral Vector to Overcome Cancer Chemotherapy Resistance. SMALL METHODS 2021; 5:e2001132. [PMID: 34928100 DOI: 10.1002/smtd.202001132] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/05/2021] [Indexed: 02/05/2023]
Abstract
Acquired chemoresistance presents a major clinical impediment, which is an urgent problem to be solved. Interestingly, myeloma cell leukemia-1 (MCL-1) and folate receptor expression levels are higher in chemotherapy-resistant patients than in pretreatment patients. In this study, a multifunctional folic acid (FA)-targeting core-shell structure is presented for simultaneous delivery of shMCL-1 and paclitaxel (PTX). The transfection efficiency of shMCL-1 with the FA-targeting delivery system is higher than with a nontargeting delivery system in Skov3 and A2780T cells. The FA-targeting system significantly inhibits cell growth, blocks cell cycles, and promotes apoptosis of cancer cells in vitro. The mechanisms involved in inhibiting growth are related to Bcl-2/Bax and cdc2/Cyclin B1 pathways. An analysis of RNA sequencing suggests that shMCL-1 reverses chemoresistance through regulating genes such as regulator of chromosome condensation 2 (RCC2). The synergetic effect of shMCL-1 and PTX effectively inhibits tumor growth in both PTX-resistant and normal cancer models by inducing tumor apoptosis, inhibiting proliferation, and limiting tumor angiogenesis. The study results indicate that a FA-targeting delivery system combining shMCL-1 with PTX can simultaneously target tumor sites and restore the sensitivity of chemotherapy-resistant cancer to PTX. These findings have important implications for patients with normal or PTX-resistant cancer.
Collapse
Affiliation(s)
- Wen Nie
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Xue Mi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Jing Chen
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Ting Yu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Junming Miao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, P. R. China
| | - Yunzhu Lin
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Tingting Yang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Mengni Ran
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Zehuo Hong
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiaoxiao Liu
- Department of Radiation Oncology, Cancer Center, Affiliated Hospital of Xuzhou Medical University, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, P. R. China
| | - Xiao Liang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| |
Collapse
|
35
|
Continuous Flow Synthesis of Anticancer Drugs. Molecules 2021; 26:molecules26226992. [PMID: 34834084 PMCID: PMC8625794 DOI: 10.3390/molecules26226992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/27/2022] Open
Abstract
Continuous flow chemistry is by now an established and valued synthesis technology regularly exploited in academic and industrial laboratories to bring about the improved preparation of a variety of molecular structures. Benefits such as better heat and mass transfer, improved process control and safety, a small equipment footprint, as well as the ability to integrate in-line analysis and purification tools into telescoped sequences are often cited when comparing flow to analogous batch processes. In this short review, the latest developments regarding the exploitation of continuous flow protocols towards the synthesis of anticancer drugs are evaluated. Our efforts focus predominately on the period of 2016-2021 and highlight key case studies where either the final active pharmaceutical ingredient (API) or its building blocks were produced continuously. It is hoped that this manuscript will serve as a useful synopsis showcasing the impact of continuous flow chemistry towards the generation of important anticancer drugs.
Collapse
|
36
|
Jeon BE, Kwon CS, Lee JE, Moon K, Cha J, Park I, Koh S, Yoon M, Kim SW, Kim JN. Anticancer Activity of Continentalic Acid in B-Cell Lymphoma. Molecules 2021; 26:molecules26226845. [PMID: 34833935 PMCID: PMC8625780 DOI: 10.3390/molecules26226845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/24/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
Aralia continentalis has been used in Korea as a folk remedy for arthralgia, rheumatism, and inflammation. However, its anti-lymphoma effect remains uncharacterized. Here, we demonstrate that A. continentalis extract and its three diterpenes efficiently kill B-lymphoma cells. Our in vitro and in vivo results suggest that the cytotoxic activities of continentalic acid, a major diterpene from A. continentalis extract, are specific towards cancer cells while leaving normal murine cells and tissues unharmed. Mechanistically, continentalic acid represses the expression of pro-survival Bcl-2 family members, such as Mcl-1 and Bcl-xL. It dissociates the mitochondrial membrane potential, leading to the stimulation of effector caspase 3/7 activities and, ultimately, cell death. Intriguingly, this agent therapeutically synergizes with roflumilast, a pan-PDE4 inhibitor that has been successfully repurposed for the treatment of aggressive B-cell malignancies in recent clinical tests. Our findings unveiled that A. continentalis extract and three of the plant’s diterpenes exhibit anti-cancer activities. We also demonstrate the synergistic inhibitory effect of continentalic acid on the survival of B-lymphoma cells when combined with roflumilast. Taken in conjunction, continentalic acid may hold significant potential for the treatment of B-cell lymphoma.
Collapse
Affiliation(s)
- Byeol-Eun Jeon
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (B.-E.J.); (C.-S.K.); (J.-E.L.); (K.M.); (J.C.)
| | - Chan-Seong Kwon
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (B.-E.J.); (C.-S.K.); (J.-E.L.); (K.M.); (J.C.)
| | - Ji-Eun Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (B.-E.J.); (C.-S.K.); (J.-E.L.); (K.M.); (J.C.)
| | - Keumok Moon
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (B.-E.J.); (C.-S.K.); (J.-E.L.); (K.M.); (J.C.)
| | - Jaeho Cha
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (B.-E.J.); (C.-S.K.); (J.-E.L.); (K.M.); (J.C.)
- Department of Microbiology, Pusan National University, Busan 46241, Korea
| | - Inmyoung Park
- Department of Asian Food and Culinary Arts, Youngsan University, Busan 48015, Korea;
| | - Sara Koh
- Department of Biological Sciences, Southern Methodist University, Dallas, TX 75206, USA;
| | - Myunghee Yoon
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, Biomedical Research Institute, Pusan National University, Busan 46241, Korea;
| | - Sang-Woo Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (B.-E.J.); (C.-S.K.); (J.-E.L.); (K.M.); (J.C.)
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea
- Correspondence: (S.-W.K.); (J.N.K.); Tel.: +82-51-510-2260 (S.-W.K.); +82-51-510-2269 (J.N.K.)
| | - Jeong Nam Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (B.-E.J.); (C.-S.K.); (J.-E.L.); (K.M.); (J.C.)
- Department of Microbiology, Pusan National University, Busan 46241, Korea
- Correspondence: (S.-W.K.); (J.N.K.); Tel.: +82-51-510-2260 (S.-W.K.); +82-51-510-2269 (J.N.K.)
| |
Collapse
|
37
|
Lan H, Sun Y. Tumor Suppressor FBXW7 and Its Regulation of DNA Damage Response and Repair. Front Cell Dev Biol 2021; 9:751574. [PMID: 34760892 PMCID: PMC8573206 DOI: 10.3389/fcell.2021.751574] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/05/2021] [Indexed: 12/21/2022] Open
Abstract
The proper DNA damage response (DDR) and repair are the central molecular mechanisms for the maintenance of cellular homeostasis and genomic integrity. The abnormality in this process is frequently observed in human cancers, and is an important contributing factor to cancer development. FBXW7 is an F-box protein serving as the substrate recognition component of SCF (SKP1-CUL1-F-box protein) E3 ubiquitin ligase. By selectively targeting many oncoproteins for proteasome-mediated degradation, FBXW7 acts as a typical tumor suppressor. Recent studies have demonstrated that FBXW7 also plays critical roles in the process of DDR and repair. In this review, we first briefly introduce the processes of protein ubiquitylation by SCFFBXW7 and DDR/repair, then provide an overview of the molecular characteristics of FBXW7. We next discuss how FBXW7 regulates the process of DDR and repair, and its translational implication. Finally, we propose few future perspectives to further elucidate the role of FBXW7 in regulation of a variety of biological processes and tumorigenesis, and to design a number of approaches for FBXW7 reactivation in a subset of human cancers for potential anticancer therapy.
Collapse
Affiliation(s)
- Huiyin Lan
- Department of Thoracic Radiation Oncology, Zhejiang Cancer Hospital, Cancer Hospital of University of Chinese Academy of Sciences, Hangzhou, China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, China
| | - Yi Sun
- Cancer Institute of the Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
38
|
Vasconcelos JCD, Siqueira IBD, Maia FFR, Parisi MCR, Zantut-Wittmann DE. Influence of thyroid hormone in the expression of the marker pro-apoptosis BID, in spite of the predominance of anti-apoptosis activation in intratiroidal lymphocytic infiltration in Hashimoto's thyroiditis. Mol Cell Endocrinol 2021; 537:111421. [PMID: 34389447 DOI: 10.1016/j.mce.2021.111421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 11/25/2022]
Abstract
UNLABELLED Cell destruction in Hashimoto's thyroiditis (HT) involves autoantibodies and cytotoxic T lymphocytes. Thyrocytes maintenance occurs by pro-apoptotic, anti-apoptotic and cell proliferation balance. OBJECTIVES To characterize factors related to the mechanisms of apoptosis and cell proliferation in thyroid cells and intrathyroid lymphocytic infiltrate in HT. METHODS We assessed lymphocytic infiltrate and thyroid cells from HT and normal thyroid by immunohistochemical analysis of cell proliferation (Ki-67), antiproliferation (p27Kip1), pro-apoptosis (Fas, Fas-ligand, BID) and anti-apoptosis (MCL-1, BCL2) markers. RESULTS Lymphocytic infiltrate presented BCL2 and MCL-1 higher expression, Ki-67 and p27kip1 balance. Thyrocytes exhibited Fas and FasL balance, higher BID expression; MCL-1, BCL-2, Ki-67 similar to the normal thyroid. T4 and higher lymphocytes BID expression were associated. CONCLUSIONS In lymphocytic infiltrate predominated anti-apoptosis in relation to pro-apoptosis except for BID. Thyrocytes presented pro-apoptosis and anti-apoptosis balance and cell proliferation similar to normal thyroid. T4-associated BID expression in HT lymphocytes suggests the influence of thyroid hormone as a signal to up-regulate the BID pro-apoptotic protein and thus increase lymphocytic apoptosis rates.
Collapse
Affiliation(s)
- Jessica Castro de Vasconcelos
- Endocrinology Division, Department of Internal Medicine, School of Medical Sciences, University of Campinas, São Paulo, Brazil.
| | | | | | - Maria Cândida Ribeiro Parisi
- Endocrinology Division, Department of Internal Medicine, School of Medical Sciences, University of Campinas, São Paulo, Brazil.
| | | |
Collapse
|
39
|
Yakymiv Y, Augeri S, Bracci C, Marchisio S, Aydin S, D'Ardia S, Massaia M, Ferrero E, Ortolan E, Funaro A. CD157 signaling promotes survival of acute myeloid leukemia cells and modulates sensitivity to cytarabine through regulation of anti-apoptotic Mcl-1. Sci Rep 2021; 11:21230. [PMID: 34707185 PMCID: PMC8551154 DOI: 10.1038/s41598-021-00733-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/18/2021] [Indexed: 12/17/2022] Open
Abstract
CD157/BST-1 (a member of the ADP-ribosyl cyclase family) is expressed at variable levels in 97% of patients with acute myeloid leukemia (AML), and is currently under investigation as a target for antibody-based immunotherapy. We used peripheral blood and bone marrow samples from patients with AML to analyse the impact of CD157-directed antibodies in AML survival and in response to cytarabine (AraC) ex vivo. The study was extended to the U937, THP1 and OCI-AML3 AML cell lines of which we engineered CD157-low versions by shRNA knockdown. CD157-targeting antibodies enhanced survival, decreased apoptosis and reduced AraC toxicity in AML blasts and cell lines. CD157 signaling activated the PI3K/AKT/mTOR and MAPK/ERK pathways and increased expression of Mcl-1 and Bcl-XL anti-apoptotic proteins, while decreasing expression of Bax pro-apoptotic protein, thus preventing Caspase-3 activation. The primary CD157-mediated anti-apoptotic mechanism was Bak sequestration by Mcl-1. Indeed, the Mcl-1-specific inhibitor S63845 restored apoptosis by disrupting the interaction of Mcl-1 with Bim and Bak and significantly increased AraC toxicity in CD157-high but not in CD157-low AML cells. This study provides a new role for CD157 in AML cell survival, and indicates a potential role of CD157 as a predictive marker of response to therapies exploiting Mcl-1 pharmacological inhibition.
Collapse
Affiliation(s)
- Yuliya Yakymiv
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Via Santena 19, 10126, Torino, Italy
| | - Stefania Augeri
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Via Santena 19, 10126, Torino, Italy
| | - Cristiano Bracci
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Via Santena 19, 10126, Torino, Italy
| | - Sara Marchisio
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Via Santena 19, 10126, Torino, Italy
| | - Semra Aydin
- Department of Oncology, Hematology, Immuno-Oncology and Rheumatology, University of Bonn, Bonn, Germany
| | - Stefano D'Ardia
- Division of Hematology, Department of Oncology, Presidio Molinette, AOU Città della Salute e della Scienza, Torino, Italy
| | | | - Enza Ferrero
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Via Santena 19, 10126, Torino, Italy
| | - Erika Ortolan
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Via Santena 19, 10126, Torino, Italy.
| | - Ada Funaro
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, Via Santena 19, 10126, Torino, Italy.
| |
Collapse
|
40
|
Aslam M, Kanthlal SK, Panonummal R. Peptides: A Supercilious Candidate for Activating Intrinsic Apoptosis by Targeting Mitochondrial Membrane Permeability for Cancer Therapy. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10297-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
41
|
Hetherington K, Dutt S, Ibarra AA, Cawood EE, Hobor F, Woolfson DN, Edwards TA, Nelson A, Sessions RB, Wilson AJ. Towards optimizing peptide-based inhibitors of protein-protein interactions: predictive saturation variation scanning (PreSaVS). RSC Chem Biol 2021; 2:1474-1478. [PMID: 34704051 PMCID: PMC8495968 DOI: 10.1039/d1cb00137j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/30/2021] [Indexed: 12/21/2022] Open
Abstract
A simple-to-implement and experimentally validated computational workflow for sequence modification of peptide inhibitors of protein–protein interactions (PPIs) is described. An experimentally validated approach for in silico modification of peptide based protein–protein interaction inhibitors is described.![]()
Collapse
Affiliation(s)
- Kristina Hetherington
- Astbury Centre for Structural Molecular Biology, University of Leeds Woodhouse Lane Leeds LS2 9JT UK .,School of Chemistry, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
| | - Som Dutt
- Astbury Centre for Structural Molecular Biology, University of Leeds Woodhouse Lane Leeds LS2 9JT UK .,School of Chemistry, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
| | - Amaurys A Ibarra
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TD UK
| | - Emma E Cawood
- Astbury Centre for Structural Molecular Biology, University of Leeds Woodhouse Lane Leeds LS2 9JT UK .,School of Chemistry, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
| | - Fruzsina Hobor
- Astbury Centre for Structural Molecular Biology, University of Leeds Woodhouse Lane Leeds LS2 9JT UK .,School of Molecular and Cellular Biology, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
| | - Derek N Woolfson
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TD UK .,School of Chemistry, University of Bristol, Cantock's Close Bristol BS8 1TS UK.,BrisSynBio, University of Bristol, Life Sciences Building Tyndall Avenue Bristol BS8 1TQ UK
| | - Thomas A Edwards
- Astbury Centre for Structural Molecular Biology, University of Leeds Woodhouse Lane Leeds LS2 9JT UK .,School of Molecular and Cellular Biology, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
| | - Adam Nelson
- Astbury Centre for Structural Molecular Biology, University of Leeds Woodhouse Lane Leeds LS2 9JT UK .,School of Chemistry, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
| | - Richard B Sessions
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk Bristol BS8 1TD UK .,BrisSynBio, University of Bristol, Life Sciences Building Tyndall Avenue Bristol BS8 1TQ UK
| | - Andrew J Wilson
- Astbury Centre for Structural Molecular Biology, University of Leeds Woodhouse Lane Leeds LS2 9JT UK .,School of Chemistry, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
| |
Collapse
|
42
|
In vivo inducible reverse genetics in patients' tumors to identify individual therapeutic targets. Nat Commun 2021; 12:5655. [PMID: 34580292 PMCID: PMC8476619 DOI: 10.1038/s41467-021-25963-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/09/2021] [Indexed: 01/18/2023] Open
Abstract
High-throughput sequencing describes multiple alterations in individual tumors, but their functional relevance is often unclear. Clinic-close, individualized molecular model systems are required for functional validation and to identify therapeutic targets of high significance for each patient. Here, we establish a Cre-ERT2-loxP (causes recombination, estrogen receptor mutant T2, locus of X-over P1) based inducible RNAi- (ribonucleic acid interference) mediated gene silencing system in patient-derived xenograft (PDX) models of acute leukemias in vivo. Mimicking anti-cancer therapy in patients, gene inhibition is initiated in mice harboring orthotopic tumors. In fluorochrome guided, competitive in vivo trials, silencing of the apoptosis regulator MCL1 (myeloid cell leukemia sequence 1) correlates to pharmacological MCL1 inhibition in patients´ tumors, demonstrating the ability of the method to detect therapeutic vulnerabilities. The technique identifies a major tumor-maintaining potency of the MLL-AF4 (mixed lineage leukemia, ALL1-fused gene from chromosome 4) fusion, restricted to samples carrying the translocation. DUX4 (double homeobox 4) plays an essential role in patients’ leukemias carrying the recently described DUX4-IGH (immunoglobulin heavy chain) translocation, while the downstream mediator DDIT4L (DNA-damage-inducible transcript 4 like) is identified as therapeutic vulnerability. By individualizing functional genomics in established tumors in vivo, our technique decisively complements the value chain of precision oncology. Being broadly applicable to tumors of all kinds, it will considerably reinforce personalizing anti-cancer treatment in the future. Preclinical molecular models are useful that mimic a patient´s response to targeted therapy. Here, the authors establish an in vivo inducible RNAi-mediated gene silencing system in patient-derived xenograft models of acute leukemia to identify individual vulnerabilities and therapeutic targets.
Collapse
|
43
|
Extrinsic interactions in the microenvironment in vivo activate an antiapoptotic multidrug-resistant phenotype in CLL. Blood Adv 2021; 5:3497-3510. [PMID: 34432864 DOI: 10.1182/bloodadvances.2020003944] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/06/2021] [Indexed: 12/17/2022] Open
Abstract
The Bcl-2 inhibitor venetoclax has yielded exceptional clinical responses in chronic lymphocytic leukemia (CLL). However, de novo resistance can result in failure to achieve negative minimal residual disease and predicts poor treatment outcomes. Consequently, additional proapoptotic drugs, such as inhibitors of Mcl-1 and Bcl-xL, are in development. By profiling antiapoptotic proteins using flow cytometry, we find that leukemic B cells that recently emigrated from the lymph node (CD69+/CXCR4Low) in vivo are enriched for cell clusters simultaneously overexpressing multiple antiapoptotic proteins (Mcl-1High/Bcl-xLHigh/Bcl-2High) in both treated and treatment-naive CLL patients. These cells exhibited antiapoptotic resistance to multiple BH-domain antagonists, including inhibitors of Bcl-2, Mcl-1, and Bcl-xL, when tested as single agents in a flow cytometry-based functional assay. Antiapoptotic multidrug resistance declines ex vivo, consistent with resistance being generated in vivo by extrinsic microenvironmental interactions. Surviving "persister" cells in patients undergoing venetoclax treatment are enriched for CLL cells displaying the functional and molecular properties of microenvironmentally induced multidrug resistance. Overcoming this resistance required simultaneous inhibition of multiple antiapoptotic proteins, with potential for unwanted toxicities. Using a drug screen performed using patient peripheral blood mononuclear cells cultured in an ex vivo microenvironment model, we identify novel venetoclax drug combinations that induce selective cytotoxicity in multidrug-resistant CLL cells. Thus, we demonstrate that antiapoptotic multidrug-resistant CLL cells exist in patients de novo and show that these cells persist during proapoptotic treatment, such as venetoclax. We validate clinically actionable approaches to selectively deplete this reservoir in patients.
Collapse
|
44
|
Lamie PF, Philoppes JN. Design, synthesis, stereochemical determination, molecular docking study, in silico pre-ADMET prediction and anti-proliferative activities of indole-pyrimidine derivatives as Mcl-1 inhibitors. Bioorg Chem 2021; 116:105335. [PMID: 34509795 DOI: 10.1016/j.bioorg.2021.105335] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/13/2021] [Accepted: 09/03/2021] [Indexed: 10/20/2022]
Abstract
In this study, fourteen novel indole-pyrimidine hybrids were designed and synthesized. Their chemical structures were confirmed using different spectroscopic techniques (1H NMR, 13C NMR, IR and mass). Their (E) stereochemical configuration was determined theoretically (MM2 property) and experimentally using 2D NMR technique (NOESY experiment). The prepared compounds were subjected to preliminary biological studies as Mcl-1 inhibitors. Most of the compounds exhibited good abilities for targeting Mcl-1 protein, especially, 7d, 7e, 7i and 7k (Ki = 11.19-15.21 nM). These derivatives were further evaluated against Bcl-XL and Bcl-2 proteins. Some compounds were found to have dual Mcl-1/Bcl-XL such as 7i, or Bcl-XL/Bcl-2 inhibitory activity as 7d. The most potent derivatives as Mcl-1 inhibitors were chosen as representative examples for determination of in-vitro anti-proliferative activity against PC-3, K-562 and MDA-MB-231 cell lines. They possessed excellent to good anti-proliferative activities. All of the synthesized compounds were docked into Mcl-1 active site. Drug-likeness properties and in silico pre-ADMET characters were also predicted.
Collapse
Affiliation(s)
- Phoebe F Lamie
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - John N Philoppes
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
45
|
Li F, Aljahdali IAM, Zhang R, Nastiuk KL, Krolewski JJ, Ling X. Kidney cancer biomarkers and targets for therapeutics: survivin (BIRC5), XIAP, MCL-1, HIF1α, HIF2α, NRF2, MDM2, MDM4, p53, KRAS and AKT in renal cell carcinoma. J Exp Clin Cancer Res 2021; 40:254. [PMID: 34384473 PMCID: PMC8359575 DOI: 10.1186/s13046-021-02026-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence of renal cell carcinoma (RCC) is increasing worldwide with an approximate 20% mortality rate. The challenge in RCC is the therapy-resistance. Cancer resistance to treatment employs multiple mechanisms due to cancer heterogeneity with multiple genetic and epigenetic alterations. These changes include aberrant overexpression of (1) anticancer cell death proteins (e.g., survivin/BIRC5), (2) DNA repair regulators (e.g., ERCC6) and (3) efflux pump proteins (e.g., ABCG2/BCRP); mutations and/or deregulation of key (4) oncogenes (e.g., MDM2, KRAS) and/or (5) tumor suppressor genes (e.g., TP5/p53); and (6) deregulation of redox-sensitive regulators (e.g., HIF, NRF2). Foci of tumor cells that have these genetic alterations and/or deregulation possess survival advantages and are selected for survival during treatment. We will review the significance of survivin (BIRC5), XIAP, MCL-1, HIF1α, HIF2α, NRF2, MDM2, MDM4, TP5/p53, KRAS and AKT in treatment resistance as the potential therapeutic biomarkers and/or targets in RCC in parallel with our analized RCC-relevant TCGA genetic results from each of these gene/protein molecules. We then present our data to show the anticancer drug FL118 modulation of these protein targets and RCC cell/tumor growth. Finally, we include additional data to show a promising FL118 analogue (FL496) for treating the specialized type 2 papillary RCC.
Collapse
Affiliation(s)
- Fengzhi Li
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263 USA
- Genitourinary Disease Site Research Group, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263 USA
- Kidney Cancer Research Interest Group, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263 USA
- Developmental Therapeutics (DT) Program, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263 USA
| | - Ieman A. M. Aljahdali
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263 USA
- Department of Cellular & Molecular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263 USA
| | - Renyuan Zhang
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263 USA
| | - Kent L. Nastiuk
- Genitourinary Disease Site Research Group, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263 USA
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263 USA
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263 USA
| | - John J. Krolewski
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263 USA
| | - Xiang Ling
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263 USA
- Canget BioTekpharma LLC, Buffalo, New York 14203 USA
| |
Collapse
|
46
|
Sakuma Y, Hirai S, Sumi T, Tada M, Kojima T, Niki T, Yamaguchi M. MCL1 inhibition enhances the efficacy of docetaxel against airway-derived squamous cell carcinoma cells. Exp Cell Res 2021; 406:112763. [PMID: 34358524 DOI: 10.1016/j.yexcr.2021.112763] [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: 04/14/2021] [Revised: 07/19/2021] [Accepted: 08/01/2021] [Indexed: 10/20/2022]
Abstract
MCL1 is an anti-apoptotic BCL2 family member that is often overexpressed in various malignant tumors. However, few reports have described the role of MCL1 in squamous cell carcinoma (SqCC) derived from airways including the lung. In this study, we examined whether MCL1 could be a novel druggable target for airway-derived SqCC, for which effective molecular targeted drugs are unavailable. We searched the Kaplan-Meier Plotter database and found that high MCL1 mRNA expression was significantly associated with shorter survival in patients with lower airway (lung) or upper airway (head and neck) derived SqCC. We also explored the Expression Atlas database and learned that authentic lung SqCC cell lines expressing both TP63 and KRT5 mRNA were extremely sparse among the publicly available "lung SqCC cell lines", with an exception being HARA cells. HARA cells were highly dependent on MCL1 for survival, and MCL1-depleted cells were not able to grow, and even declined in number, upon docetaxel (DTX) exposure in vitro and in vivo. Similar in vitro experimental findings, including those in a 3D culture model, were also obtained using Detroit 562 pharyngeal SqCC cells. These findings suggested that combined treatment with MCL1 silencing plus DTX appears highly effective against airway-derived SqCC.
Collapse
Affiliation(s)
- Yuji Sakuma
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, South 1, West 17, Chuo-ku, Sapporo, 060-8556, Japan.
| | - Sachie Hirai
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, South 1, West 17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Toshiyuki Sumi
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, South 1, West 17, Chuo-ku, Sapporo, 060-8556, Japan; Department of Respiratory Medicine and Allergology Sapporo Medical University School of Medicine, South 1, West 17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Makoto Tada
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, South 1, West 17, Chuo-ku, Sapporo, 060-8556, Japan; Department of Thoracic Surgery, Sapporo Medical University School of Medicine, South 1, West 17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, South 1, West 17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Toshiro Niki
- Division of Integrative Pathology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Miki Yamaguchi
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, South 1, West 17, Chuo-ku, Sapporo, 060-8556, Japan
| |
Collapse
|
47
|
Mittal P, Singh S, Sinha R, Shrivastava A, Singh A, Singh IK. Myeloid cell leukemia 1 (MCL-1): Structural characteristics and application in cancer therapy. Int J Biol Macromol 2021; 187:999-1018. [PMID: 34339789 DOI: 10.1016/j.ijbiomac.2021.07.166] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/20/2022]
Abstract
Apoptosis, a major hallmark of cancer cells, regulates cellular fate and homeostasis. BCL-2 (B-cell CLL/Lymphoma 2) protein family is popularly known to mediate the intrinsic mode of apoptosis, of which MCL-1 is a crucial member. Myeloid cell leukemia 1 (MCL-1) is an anti-apoptotic oncoprotein and one of the most investigated members of the BCL-2 family. It is commonly known to be genetically altered, aberrantly overexpressed, and primarily associated with drug resistance in various human cancers. Recent advancements in the development of selective MCL-1 inhibitors and evaluating their effectiveness in cancer treatment establish its popularity as a molecular target. The overall aim is the selective induction of apoptosis in cancer cells by using a single or combination of BCL-2 family inhibitors. Delineating the precise molecular mechanisms associated with MCL-1-mediated cancer progression will certainly improve the efficacy of clinical interventions aimed at MCL-1 and hence patient survival. This review is structured to highlight the structural characteristics of MCL-1, its specific interactions with NOXA, MCL-1-regulatory microRNAs, and at the same time focus on the emerging therapeutic strategies targeting our protein of interest (MCL-1), alone or in combination with other treatments.
Collapse
Affiliation(s)
- Pooja Mittal
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India
| | - Sujata Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India
| | - Rajesh Sinha
- Department of Dermatology, University of Alabama, Birmingham 35205, United States of America
| | - Anju Shrivastava
- Department of Zoology, University of Delhi, New Delhi, 110007, India
| | - Archana Singh
- Department of Botany, Hans Raj College, University of Delhi, New Delhi 110007, India.
| | - Indrakant Kumar Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India.
| |
Collapse
|
48
|
A Novel Regimen for Treating Melanoma: MCL1 Inhibitors and Azacitidine. Pharmaceuticals (Basel) 2021; 14:ph14080749. [PMID: 34451846 PMCID: PMC8399604 DOI: 10.3390/ph14080749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 12/22/2022] Open
Abstract
Although treatment options for melanoma patients have expanded in recent years with the approval of immunotherapy and targeted therapy, there is still an unmet need for new treatment options for patients that are ineligible for, or resistant to these therapies. BH3 mimetics, drugs that mimic the activity of pro-apoptotic BCL2 family proteins, have recently achieved remarkable success in the clinical setting. The combination of BH3 mimetic ABT-199 (venetoclax) plus azacitidine has shown substantial benefit in treating acute myelogenous leukemia. We evaluated the efficacy of various combinations of BH3 mimetic + azacitidine in fourteen human melanoma cell lines from cutaneous, mucosal, acral and uveal subtypes. Using a combination of cell viability assay, BCL2 family knockdown cell lines, live cell imaging, and sphere formation assay, we found that combining inhibition of MCL1, an anti-apoptotic BCL2 protein, with azacitidine had substantial pro-apoptotic effects in multiple melanoma cell lines. Specifically, this combination reduced cell viability, proliferation, sphere formation, and induced apoptosis. In addition, this combination is highly effective at reducing cell viability in rare mucosal and uveal subtypes. Overall, our data suggest this combination as a promising therapeutic option for some patients with melanoma and should be further explored in clinical trials.
Collapse
|
49
|
Alcon C, Zañudo JGT, Albert R, Wagle N, Scaltriti M, Letai A, Samitier J, Montero J. ER+ Breast Cancer Strongly Depends on MCL-1 and BCL-xL Anti-Apoptotic Proteins. Cells 2021; 10:1659. [PMID: 34359829 PMCID: PMC8304651 DOI: 10.3390/cells10071659] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/18/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is the most frequent type of cancer and the major cause of mortality in women. The rapid development of various therapeutic options has led to the improvement of treatment outcomes; nevertheless, one-third of estrogen receptor (ER)-positive patients relapse due to cancer cell acquired resistance. Here, we use dynamic BH3 profiling (DBP), a functional predictive assay that measures net changes in apoptotic priming, to find new effective treatments for ER+ breast cancer. We observed anti-apoptotic adaptations upon treatment that pointed to metronomic therapeutic combinations to enhance cytotoxicity and avoid resistance. Indeed, we found that the anti-apoptotic proteins BCL-xL and MCL-1 are crucial for ER+ breast cancer cells resistance to therapy, as they exert a dual inhibition of the pro-apoptotic protein BIM and compensate for each other. In addition, we identified the AKT inhibitor ipatasertib and two BH3 mimetics targeting these anti-apoptotic proteins, S63845 and A-1331852, as new potential therapies for this type of cancer. Therefore, we postulate the sequential inhibition of both proteins using BH3 mimetics as a new treatment option for refractory and relapsed ER+ breast cancer tumors.
Collapse
Affiliation(s)
- Clara Alcon
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; (C.A.); (J.S.)
| | | | - Reka Albert
- Department of Biology, The Pennsylvania State University, University Park, PA 16802-6300, USA;
| | - Nikhil Wagle
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; (J.G.T.Z.); (N.W.)
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA;
| | - Maurizio Scaltriti
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA;
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; (C.A.); (J.S.)
- Department of Electronics and Biomedical Engineering, University of Barcelona (UB), 08028 Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Joan Montero
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; (C.A.); (J.S.)
| |
Collapse
|
50
|
Basu A. The interplay between apoptosis and cellular senescence: Bcl-2 family proteins as targets for cancer therapy. Pharmacol Ther 2021; 230:107943. [PMID: 34182005 DOI: 10.1016/j.pharmthera.2021.107943] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/30/2021] [Indexed: 02/07/2023]
Abstract
Cell death by apoptosis and permanent cell cycle arrest by senescence serve as barriers to the development of cancer. Chemotherapeutic agents not only induce apoptosis, they can also induce senescence known as therapy-induced senescence (TIS). There are, however, controversies whether TIS improves or worsens therapeutic outcome. Unlike apoptosis, which permanently removes cancer cells, senescent cells are metabolically active, and can contribute to tumor progression and relapse. If senescent cells are not cleared by the immune system or if cancer cells escape senescence, they may acquire resistance to apoptotic stimuli and become highly aggressive. Thus, there have been significant efforts in developing senolytics, drugs that target these pro-survival molecules to eliminate senescent cells. The anti-apoptotic Bcl-2 family proteins not only protect against cell death by apoptosis, but they also allow senescent cells to survive. While combining senolytics with chemotherapeutic drugs is an attractive approach, there are also limitations. Moreover, members of the Bcl-2 family have distinct effects on apoptosis and senescence. The purpose of this review article is to discuss recent literatures on how members of the Bcl-2 family orchestrate the interplay between apoptosis and senescence, and the challenges and progress in targeting these Bcl-2 family proteins for cancer therapy.
Collapse
Affiliation(s)
- Alakananda Basu
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| |
Collapse
|