1
|
Chaudhary B, Arya P, Sharma V, Kumar P, Singla D, Grewal AS. Targeting anti-apoptotic mechanisms in tumour cells: Strategies for enhancing Cancer therapy. Bioorg Chem 2025; 159:108388. [PMID: 40107036 DOI: 10.1016/j.bioorg.2025.108388] [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: 01/23/2025] [Revised: 03/05/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Anti-cancer drug's cytotoxicity is determined by their ability to induce predetermined cell demise, commonly called apoptosis. The cancer-causing cells are able to evade cell death, which has been affiliated with both malignancy as well as resistance to cancer treatments. In order to avoid cell death, cancerous tumour cells often produce an abundance of anti-apoptotic proteins, becoming "dependent" on them. Consequently, protein inhibitors of cell death may prove to be beneficial as pharmacological targets for the future creation of cancer therapies. This article examines the molecular routes of apoptosis, its clinical manifestations, anti-cancer therapy options that target the intrinsic mechanism of apoptosis, proteins that prevent cell death, and members of the B-lymphoma-2 subset. In addition, novel approaches to cell death are highlighted, including how curcumin mitigates chemotherapy-induced apoptosis in healthy tissues and the various ways melatonin modifies apoptosis to improve cancer treatment efficacy, particularly through the TNF superfamily. Cancer treatment-induced increases in anti-apoptotic proteins lead to drug resistance; yet, ligands that trigger cell death by inhibiting these proteins are expected to improve chemotherapy's efficacy. The potential of frequency-modulated dietary phytochemicals as a cancer therapeutic pathway, including autophagy and apoptosis, is also explored. This approach may be more efficient than inhibition alone in overcoming drug resistance. Consequently, this method has the potential to allow for lower medication concentrations, reducing cytotoxicity and unwanted side effects.
Collapse
Affiliation(s)
- Benu Chaudhary
- Shri Ram College of Pharmacy, Ramba, Karnal, Haryana, India
| | - Preeti Arya
- Shri Ram College of Pharmacy, Ramba, Karnal, Haryana, India
| | - Vikas Sharma
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar, Haryana, India
| | - Parveen Kumar
- NIMS Institute of Pharmacy, NIMS University, Jaipur, Rajasthan, India
| | - Deepak Singla
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar, Haryana, India
| | | |
Collapse
|
2
|
Wang L, Chen Y, Zhang M, Liu J, Li H, Liu M, Wu S, Zhang Y, Li W, Wang B. Chemical dissection of selective myeloid leukemia-1 inhibitors: How they were found and evolved. Eur J Med Chem 2025; 283:117168. [PMID: 39708769 DOI: 10.1016/j.ejmech.2024.117168] [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: 08/17/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
Myeloid cell leukemia-1 (MCL-1), a key anti-apoptotic protein within the BCL-2 family, is essential in regulating cell survival, particularly in cancer, where its overexpression is often linked to therapeutic resistance. This review begins with an overview of BCL-2-mediated apoptosis, highlighting the pivotal role of MCL-1 in cellular homeostasis. We then focus on the structure and function of MCL-1, elucidating how its unique structural features contribute to its function and interaction with pro-apoptotic proteins. The core of this review is a detailed structural analysis of selective MCL-1 inhibitors, tracing their development from initial discovery to stepwise optimization. We explore various classes of inhibitors, including those with distinct core structures, covalent inhibitors that reversibly/irreversibly bind to MCL-1, and innovative approaches such as metal-based inhibitors and proteolysis-targeting chimeras (PROTACs). The structural evolution of these inhibitors is discussed, with particular emphasis on the modifications that have enhanced their selectivity, potency, and pharmacokinetic profiles. Additionally, we summarize the synergistic potential of MCL-1 inhibitors when used in combination with other therapeutic agents, emphasizing their role in overcoming drug resistance. The review concludes with a discussion of current challenges in MCL-1 modulation and future perspectives, proposing alternative strategies for targeting this critical protein for cancer therapy.
Collapse
Affiliation(s)
- Luyao Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuxiang Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Maoqian Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jin Liu
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing 211162, China
| | - Haozhe Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Menghui Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shuyun Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yongmin Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, 75005 Paris, France; Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China; Fuyang Institute & School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311422, Zhejiang, China.
| | - Wei Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Bo Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
3
|
Ma L, Zhang J, Dai Z, Liao P, Guan J, Luo Z. Top 100 most-cited articles on apoptosis of non-small cell lung cancer over the past two decades: a bibliometrics analysis. Front Immunol 2025; 15:1512349. [PMID: 39872524 PMCID: PMC11770037 DOI: 10.3389/fimmu.2024.1512349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/09/2024] [Indexed: 01/30/2025] Open
Abstract
Background Recently there has been an increasing number of studies have explored apoptosis mechanisms in lung cancer (LC). However, no researchers have conducted a bibliometric analysis of the most cited articles in this field. Objective To examine the top 100 most influential and cited publications on apoptosis in non-small cell lung cancer (NSCLC) from 2004 to 2023, summarizing research trends and key focus areas. Methods This study utilized the Web of Science Core Database (WOSCC) to research NSCLC apoptosis from 2004 to 2023, using keyword selection and manual screening for article searches. Bibliometrix package of R software 4.3.1 was used to generate distribution statistics for the top ten institutions, journals and authors. Citespace6.2. R6 was used to create the visualization maps for keyword co-occurrence and clustering. VOSviewer1.6.19 was used to conduct cluster analysis of publishing countries (regions), with data exported to SCImago Graphica for geographic visualization and cooperation analysis. VOSviewer1.6.19 was used to produced co-citation maps of institutions, journals, authors, and references. Results From 2004 to 2023, 13316 articles were retrieved, and the top 100 most cited were chosen. These were authored by 934 individuals from 269 institutions across 18 countries and appeared in 45 journals. Citations ranged from 150 to 1,389, with a median of 209.5. The most influential articles appeared in 2005 and 2007 (n=13). The leading countries (regions), institutions, journals and authors were identified as the United States (n=60), Harvard University (n=64), CANCER RESEARCH (n=15), SUN M and YANG JS (n=6). The top five keywords were "expression", "activation", "apoptosis", "pathway" and "gefitinib". This study indicates that enhancing apoptosis through circular RNA regulation and targeting the Nrf2 signaling pathway could become a key research focus in recent years. Conclusion Apoptosis has been the subject of extensive research over many years, particularly in relation to its role in the pathogenesis, diagnosis, and treatment of NSCLC. This study aims to identify highly influential articles and forecast emerging research trends, thereby offering insights into novel therapeutic targets and strategies to overcome drug resistance. The findings are intended to serve as a valuable reference for scholars engaged in this field of study.
Collapse
Affiliation(s)
- Leshi Ma
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Zhang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zi Dai
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pei Liao
- Department of Oncology, Chongqing Hospital, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Chongqing, China
| | - Jieshan Guan
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Oncology, Shenshan Hospital, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Shanwei, China
| | - Zhijie Luo
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
4
|
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
|
5
|
Boroujeni AF, Ates-Alagoz Z. Pioneering the Battle Against Breast Cancer: The Promise of New Bcl-2 Family. Anticancer Agents Med Chem 2025; 25:164-178. [PMID: 39313901 DOI: 10.2174/0118715206320224240910054728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024]
Abstract
Currently, breast cancer is the most common cancer type, accounting for 1 in every 4 cancer cases. Leading both in mortality and incidence, breast cancer causes 1 in 4 cancer deaths. To decrease the burden of breast cancer, novel therapeutic agents which target the key hallmarks of cancer, are being explored. The Bcl-2 family of proteins has a crucial role in governing cell death, making them an attractive target for cancer therapy. As cancer chemotherapies lead to oncogenic stress, cancer cells upregulate the Bcl-2 family to overcome apoptosis, leading to failure of treatment. To fix this issue, Bcl-2 family inhibitors, which can cause cell death, have been introduced as novel therapeutic agents. Members of this group have shown promising results in in-vitro studies, and some are currently in clinical trials. In this review, we will investigate Bcl-2 family inhibitors, which are already in trials as monotherapy or combination therapy for breast cancer, and we will also highlight the result of in vitro studies of novel Bcl-2 family inhibitors on breast cancer cells. The findings of these studies have yielded encouraging outcomes regarding the identification of novel Bcl-2 family inhibitors. These compounds hold significant potential as efficacious agents for employment in both monotherapy and combination therapy settings.
Collapse
Affiliation(s)
- Ali Farhang Boroujeni
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Zeynep Ates-Alagoz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
6
|
Suo S, Sun S, Nguyen LXT, Qian J, Li F, Zhao D, Yu W, Lou Y, Zhu H, Tong H, Yang M, Huang X, Zhao S, Qiao J, Liang C, Wang H, Zhang Y, Zhang X, Hoang DH, Chen F, Kang H, Valerio M, Sun J, Ghoda L, Li L, Marcucci G, Zhang B, Jin J. Homoharringtonine synergizes with venetoclax in early T cell progenitor acute lymphoblastic leukemia: Bench and bed. MED 2024; 5:1510-1524.e4. [PMID: 39151422 DOI: 10.1016/j.medj.2024.07.018] [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: 08/14/2023] [Revised: 04/26/2024] [Accepted: 07/22/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Early T cell precursor acute lymphoblastic leukemia (ETP-ALL) is a distinct subtype of T-ALL with a poor prognosis. To find a cure, we examined the synergistic effect of homoharringtonine (HHT) in combination with the BCL-2 inhibitor venetoclax (VEN) in ETP-ALL. METHODS Using in vitro cellular assays and ETP-ALL xenograft models, we first investigated the synergistic activity of HHT and VEN in ETP-ALL. Next, to explore the underlying mechanism, we employed single-cell RNA sequencing of primary ETP-ALL cells treated with HHT or VEN alone or in combination and validated the results with western blot assays. Based on the promising preclinical results and given that both drugs have been approved for clinical use, we then assessed this combination in clinical practice. FINDINGS Our results showed that HHT synergizes strongly with VEN both in vitro and in vivo in ETP-ALL. Mechanistic studies demonstrated that the HHT/VEN combination concurrently downregulated key anti-apoptotic proteins, i.e., MCL1, leading to enhanced apoptosis. Importantly, the clinical results were very promising. Six patients with ETP-ALL with either refractory/relapsed (R/R) or newly diagnosed disease were treated with an HHT/VEN-based regimen. All patients achieved complete remission (CR) after only one cycle of treatment. CONCLUSIONS Our findings demonstrate that a combination of HHT/VEN is effective on ETP-ALL and represents the "backbone" of a promising and safe regimen for newly diagnosed and R/R patients with ETP-ALL. FUNDING This work was funded by the National Cancer Institute, Gehr Family Foundation, George Hoag Family Foundation, National Natural Science Foundation of China, and Key Research and Development Program of Zhejiang Province of China.
Collapse
Affiliation(s)
- Shanshan Suo
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China; Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA; Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China
| | - Shu Sun
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China; Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China
| | - Le Xuan Truong Nguyen
- Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA
| | - Jiejing Qian
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China; Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China
| | - Fenglin Li
- Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Dandan Zhao
- Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA
| | - Wenjuan Yu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China; Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China
| | - Yinjun Lou
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China; Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China
| | - Honghu Zhu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China; Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China
| | - Hongyan Tong
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China; Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China
| | - Min Yang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China; Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China
| | - Xin Huang
- Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Shuqi Zhao
- Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Junjing Qiao
- Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA
| | - Chen Liang
- Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA
| | - Huafeng Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China; Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China
| | - Yi Zhang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China; Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China
| | - Xiang Zhang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China; Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China
| | - Dinh Hoa Hoang
- Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA
| | - Fang Chen
- Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA
| | - Hyunjun Kang
- Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA
| | - Melissa Valerio
- Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA
| | - Jie Sun
- Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China
| | - Lucy Ghoda
- Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA
| | - Ling Li
- Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA
| | - Guido Marcucci
- Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA.
| | - Bin Zhang
- Hematology Malignancies Research Institute, Gehr Family Center for Leukemia Research, City of Hope Medical Center, Duarte, CA, USA.
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China; Zhejiang Provincial Key Lab of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China; Zhejiang University Cancer Center, Hangzhou, Zhejiang, P.R. China.
| |
Collapse
|
7
|
Wang P, Cui J, Cheng G, Zhang D. Theoretical study on the selective binding of BH3-only protein BAD to anti-apoptotic protein BCL- xL instead of MCL-1. Phys Chem Chem Phys 2024; 26:25480-25487. [PMID: 39324232 DOI: 10.1039/d4cp02936d] [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: 09/27/2024]
Abstract
In this study, molecular dynamics simulations were used to systematically explore the reason why BH3-only protein BAD binds to anti-apoptotic protein BCL-xL but not to MCL-1 to give more theoretical hints for the design of BAD mimetic inhibitors for the dual-targeting of BCL-xL and MCL-1. Starting with the difference in residue-based binding energy contributions, a series of analyses were conducted to identify the hotspot residues in MCL-1 that significantly affect the interaction with BAD. Among them, the insertion of the T residue in the loop between α4 and α5 domains of MCL-1 is considered to be the main cause of BAD selective binding. The inserted T residue reduces the stability of the loop and weakens the hydrogen bond interactions that originally bound E19 of BAD in BCL-xL/BAD, and the freed E19 severely interferes with the salt bridge between D16 and Arg53 by electrostatic repulsion. This salt-bridge is believed to be critical for maintaining the binding between BCL-xL and BAD. By clarifying the reasons for differential binding, we can more specifically optimize the BAD sequence to target both BCL-xL and MCL-1.
Collapse
Affiliation(s)
- Panpan Wang
- School of Physics and Engineering, Henan University of Science and Technology, Luoyang 471023, P. R. China.
| | - Jinglan Cui
- School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637551, Singapore.
| | - Guojie Cheng
- School of Physics and Engineering, Henan University of Science and Technology, Luoyang 471023, P. R. China.
| | - Dawei Zhang
- School of Physics and Engineering, Henan University of Science and Technology, Luoyang 471023, P. R. China.
| |
Collapse
|
8
|
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
|
9
|
Singla S, Sreedharanunni S, Singh A, Singh C, Bose P, Kumar A, Balakrishnan A, Jain A, Khadwal A, Lad D, Prakash G, Sharma P, Mallik N, Sachdeva MUS, Das R, Malhotra P. Flow cytometric expression of Bcl-2, Mcl-1, and their ratios correlates with primary and secondary cytogenetic changes and their combinations in multiple myeloma. Ann Hematol 2024:10.1007/s00277-024-06004-3. [PMID: 39264433 DOI: 10.1007/s00277-024-06004-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
Response to BH3 mimetics in multiple myeloma (MM) correlates with CCND1-rearrangement or expression of anti-apoptotic molecules, particularly Bcl-2 and Mcl-1. Our study investigates the relationship between cytogenetic abnormalities (CGAs) and intracellular Bcl-2 and Mcl-1 expression in myeloma plasma cells (MPCs) using flow cytometry (FCM). We measured median fluorescence intensity (MFI) of Bcl-2 and Mcl-1 in 163 bone marrow samples (143 MM, 20 controls) across various cell types. Both Bcl-2MFI and Mcl-1MFI were significantly higher in MPCs compared to other cells, with Bcl-2 MFI exceeding Mcl-1 MFI in MPCs. Bcl-2 expression peaked in CCND1-rearranged cases, while Mcl-1 expression was highest in cases with 1q21 gain/amplification. Notably, 65-74% of cases with other CGAs exhibited moderate to strong Bcl-2 or Mcl-1 expression, indicating potential utility of BH3 mimetics in this group, while 25% showed dim to absent expression of one or both markers, suggesting potential futility in these patients. Our study highlights FCM's potential for rapid Bcl-2 and Mcl-1 quantification, surpassing traditional methods. We propose that direct measurement of Bcl-2 and Mcl-1 expression in PCs by FCM, combined with cytogenetic characterization, could improve therapeutic decision-making regarding the use of BH3 mimetics in MM, potentially enhancing outcomes and overcoming resistance.
Collapse
Affiliation(s)
- Shelly Singla
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sreejesh Sreedharanunni
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Archana Singh
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Charanpreet Singh
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Parveen Bose
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arun Kumar
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anand Balakrishnan
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arihant Jain
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Khadwal
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepesh Lad
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gaurav Prakash
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Praveen Sharma
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Nabhajit Mallik
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Man Updesh Singh Sachdeva
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Reena Das
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Malhotra
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
10
|
Palominos C, Fuentes-Retamal S, Salazar JP, Guzmán-Rivera D, Correa P, Mellado M, Araya-Maturana R, Urra FA. Mitochondrial bioenergetics as a cell fate rheostat for responsive to Bcl-2 drugs: New cues for cancer chemotherapy. Cancer Lett 2024; 594:216965. [PMID: 38788967 DOI: 10.1016/j.canlet.2024.216965] [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: 03/05/2024] [Revised: 05/03/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024]
Abstract
Pro-survival BCL-2 proteins prevent the initiation of intrinsic apoptosis (mitochondria-dependent pathway) by inhibiting the pro-apoptotic proteins BAX and BAK, while BH3-only proteins promote apoptosis by blocking pro-survival BCL-2 proteins. Disruptions in this delicate balance contribute to cancer cell survival and chemoresistance. Recent advances in cancer therapeutics involve a new generation of drugs known as BH3-mimetics, which are small molecules designed to mimic the action of BH3-only proteins. Promising effects have been observed in patients with hematological and solid tumors undergoing treatment with these agents. However, the rapid emergence of mitochondria-dependent resistance to BH3-mimetics has been reported. This resistance involves increased mitochondrial respiration, altered mitophagy, and mitochondria with higher and tighter cristae. Conversely, mutations in isocitrate dehydrogenase 1 and 2, catalyzing R-2-hydroxyglutarate production, promote sensitivity to venetoclax. This evidence underscores the urgency for comprehensive studies on bioenergetics-based adaptive responses in both BH3 mimetics-sensitive and -resistant cancer cells. Ongoing clinical trials are evaluating BH3-mimetics in combination with standard chemotherapeutics. In this article, we discuss the role of mitochondrial bioenergetics in response to BH3-mimetics and explore potential therapeutic opportunities through metabolism-targeting strategies.
Collapse
Affiliation(s)
- Charlotte Palominos
- Metabolic Plasticity and Bioenergetics Laboratory, Clinical and Molecular Pharmacology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, 8380453, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, 8380453, Chile; Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca, 3480094, Chile
| | - Sebastián Fuentes-Retamal
- Metabolic Plasticity and Bioenergetics Laboratory, Clinical and Molecular Pharmacology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, 8380453, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, 8380453, Chile; Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca, 3480094, Chile; Universidad Andrés Bello. Escuela de Química y Farmacia, Facultad de Medicina, 8320000, Santiago, Chile
| | - Juan Pablo Salazar
- Metabolic Plasticity and Bioenergetics Laboratory, Clinical and Molecular Pharmacology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, 8380453, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, 8380453, Chile; Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca, 3480094, Chile
| | - Daniela Guzmán-Rivera
- Metabolic Plasticity and Bioenergetics Laboratory, Clinical and Molecular Pharmacology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, 8380453, Chile; Universidad Andrés Bello. Escuela de Química y Farmacia, Facultad de Medicina, 8320000, Santiago, Chile
| | - Pablo Correa
- Metabolic Plasticity and Bioenergetics Laboratory, Clinical and Molecular Pharmacology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, 8380453, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, 8380453, Chile; Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca, 3480094, Chile
| | - Mathias Mellado
- Metabolic Plasticity and Bioenergetics Laboratory, Clinical and Molecular Pharmacology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, 8380453, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, 8380453, Chile
| | - Ramiro Araya-Maturana
- Network for Snake Venom Research and Drug Discovery, Santiago, 8380453, Chile; Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca, 3480094, Chile; Instituto de Química de Recursos Naturales, Universidad de Talca, Talca, 3460000, Chile
| | - Félix A Urra
- Metabolic Plasticity and Bioenergetics Laboratory, Clinical and Molecular Pharmacology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, 8380453, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, 8380453, Chile; Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Talca, 3480094, Chile; Interuniversity Center for Healthy Aging (CIES), Consortium of Universities of the State of Chile (CUECH), Santiago, 8320216, Chile.
| |
Collapse
|
11
|
Nocquet L, Roul J, Lefebvre CC, Duarte L, Campone M, Juin PP, Souazé F. Low BCL-xL expression in triple-negative breast cancer cells favors chemotherapy efficacy, and this effect is limited by cancer-associated fibroblasts. Sci Rep 2024; 14:14177. [PMID: 38898061 PMCID: PMC11187150 DOI: 10.1038/s41598-024-64696-z] [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: 03/01/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024] Open
Abstract
Triple negative breast cancers (TNBC) present a poor prognosis primarily due to their resistance to chemotherapy. This resistance is known to be associated with elevated expression of certain anti-apoptotic members within the proteins of the BCL-2 family (namely BCL-xL, MCL-1 and BCL-2). These regulate cell death by inhibiting pro-apoptotic protein activation through binding and sequestration and they can be selectively antagonized by BH3 mimetics. Yet the individual influences of BCL-xL, MCL-1, and BCL-2 on the sensitivity of TNBC cells to chemotherapy, and their regulation by cancer-associated fibroblasts (CAFs), major components of the tumor stroma and key contributors to therapy resistance remain to be delineated. Using gene editing or BH3 mimetics to inhibit anti-apoptotic BCL-2 family proteins in TNBC line MDA-MB-231, we show that BCL-xL and MCL-1 promote cancer cell survival through compensatory mechanisms. This cell line shows limited sensitivity to chemotherapy, in line with the clinical resistance observed in TNBC patients. We elucidate that BCL-xL plays a pivotal role in therapy response, as its depletion or pharmacological inhibition heightened chemotherapy effectiveness. Moreover, BCL-xL expression is associated with chemotherapy resistance in patient-derived tumoroids where its pharmacological inhibition enhances ex vivo response to chemotherapy. In a co-culture model of cancer cells and CAFs, we observe that even in a context where BCL-xL reduced expression renders cancer cells more susceptible to chemotherapy, those in contact with CAFs display reduced sensitivity to chemotherapy. Thus CAFs exert a profound pro-survival effect in breast cancer cells, even in a setting highly favoring cell death through combined chemotherapy and absence of the main actor of chemoresistance, BCL-xL.
Collapse
Affiliation(s)
- Lisa Nocquet
- INSERM, CNRS, CRCI2NA, Université de Nantes, 44000, Nantes, France
- Equipe Labellisée LIGUE Contre le Cancer, Paris, France
- SIRIC ILIAD, Nantes, Angers, France
| | - Julie Roul
- INSERM, CNRS, CRCI2NA, Université de Nantes, 44000, Nantes, France
- Equipe Labellisée LIGUE Contre le Cancer, Paris, France
- SIRIC ILIAD, Nantes, Angers, France
- ICO René Gauducheau, Saint Herblain, France
| | - Chloé C Lefebvre
- INSERM, CNRS, CRCI2NA, Université de Nantes, 44000, Nantes, France
- Equipe Labellisée LIGUE Contre le Cancer, Paris, France
- SIRIC ILIAD, Nantes, Angers, France
| | - Laurine Duarte
- INSERM, CNRS, CRCI2NA, Université de Nantes, 44000, Nantes, France
- Equipe Labellisée LIGUE Contre le Cancer, Paris, France
- SIRIC ILIAD, Nantes, Angers, France
| | - Mario Campone
- INSERM, CNRS, CRCI2NA, Université de Nantes, 44000, Nantes, France
- SIRIC ILIAD, Nantes, Angers, France
- ICO René Gauducheau, Saint Herblain, France
| | - Philippe P Juin
- INSERM, CNRS, CRCI2NA, Université de Nantes, 44000, Nantes, France.
- Equipe Labellisée LIGUE Contre le Cancer, Paris, France.
- SIRIC ILIAD, Nantes, Angers, France.
- ICO René Gauducheau, Saint Herblain, France.
| | - Frédérique Souazé
- INSERM, CNRS, CRCI2NA, Université de Nantes, 44000, Nantes, France.
- Equipe Labellisée LIGUE Contre le Cancer, Paris, France.
- SIRIC ILIAD, Nantes, Angers, France.
| |
Collapse
|
12
|
Alboreggia G, Udompholkul P, Baggio C, Muzzarelli K, Assar Z, Pellecchia M. Histidine-Covalent Stapled Alpha-Helical Peptides Targeting hMcl-1. J Med Chem 2024; 67:8172-8185. [PMID: 38695666 PMCID: PMC11129181 DOI: 10.1021/acs.jmedchem.4c00277] [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: 01/30/2024] [Revised: 03/28/2024] [Accepted: 04/24/2024] [Indexed: 05/24/2024]
Abstract
Several novel and effective cysteine targeting (Cys) covalent drugs are in clinical use. However, the target area containing a druggable Cys residue is limited. Therefore, methods for creating covalent drugs that target different residues are being looked for; examples of such ligands include those that target the residues lysine (Lys) and tyrosine (Tyr). Though the histidine (His) side chain is more frequently found in protein binding locations and has higher desirable nucleophilicity, surprisingly limited research has been done to specifically target this residue, and there are not many examples of His-targeting ligands that have been rationally designed. In the current work, we created novel stapled peptides that are intended to target hMcl-1 His 252 covalently. We describe the in vitro (biochemical, NMR, and X-ray) and cellular design and characterization of such agents. Our findings further suggest that the use of electrophiles to specifically target His residues is warranted.
Collapse
Affiliation(s)
- Giulia Alboreggia
- Division
of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, California 92521, United States
| | - Parima Udompholkul
- Division
of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, California 92521, United States
| | - Carlo Baggio
- Division
of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, California 92521, United States
| | - Kendall Muzzarelli
- Cayman
Chemical Co., 1180 E. Ellsworth road, Ann Arbor, Michigan 48108, United States
| | - Zahra Assar
- Cayman
Chemical Co., 1180 E. Ellsworth road, Ann Arbor, Michigan 48108, United States
| | - Maurizio Pellecchia
- Division
of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, California 92521, United States
| |
Collapse
|
13
|
Kaplan Ö, Gök MK, Pekmez M, Erden Tayhan S, Özgümüş S, Gökçe İ, Arda N. Development of recombinant protein-based nanoparticle systems for inducing tumor cell apoptosis: In vitro evaluation of their cytotoxic and apoptotic effects on cancer cells. J Drug Deliv Sci Technol 2024; 95:105565. [DOI: 10.1016/j.jddst.2024.105565] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Deng H, Han Y, Liu L, Zhang H, Liu D, Wen J, Huang M, Zhao L. Targeting Myeloid Leukemia-1 in Cancer Therapy: Advances and Directions. J Med Chem 2024; 67:5963-5998. [PMID: 38597264 DOI: 10.1021/acs.jmedchem.3c01998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
As a tripartite cell death switch, B-cell lymphoma protein 2 (Bcl-2) family members precisely regulate the endogenous apoptosis pathway in response to various cell signal stresses through protein-protein interactions. Myeloid leukemia-1 (Mcl-1), a key anti-apoptotic Bcl-2 family member, is positioned downstream in the endogenous apoptotic pathway and plays a central role in regulating mitochondrial function. Mcl-1 is highly expressed in a variety of hematological malignancies and solid tumors, contributing to tumorigenesis, poor prognosis, and chemoresistance, making it an attractive target for cancer treatment. This Perspective aims to discuss the mechanism by which Mcl-1 regulates apoptosis and non-apoptotic functions in cancer cells and to outline the discovery and optimization process of potent Mcl-1 modulators. In addition, we summarize the structural characteristics of potent inhibitors that bind to Mcl-1 through multiple co-crystal structures and analyze the cardiotoxicity caused by current Mcl-1 inhibitors, providing prospects for rational targeting of Mcl-1.
Collapse
Affiliation(s)
- Hongguang Deng
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Han
- 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
| | - Hong Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, 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
| | - Jiachen Wen
- 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
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
15
|
Wright T, Turnis ME, Grace CR, Li X, Brakefield LA, Wang YD, Xu H, Kaminska E, Climer LK, Mukiza TO, Chang CL, Moldoveanu T, Opferman JT. Anti-apoptotic MCL-1 promotes long-chain fatty acid oxidation through interaction with ACSL1. Mol Cell 2024; 84:1338-1353.e8. [PMID: 38503284 PMCID: PMC11017322 DOI: 10.1016/j.molcel.2024.02.035] [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: 08/11/2023] [Revised: 12/19/2023] [Accepted: 02/27/2024] [Indexed: 03/21/2024]
Abstract
MCL-1 is essential for promoting the survival of many normal cell lineages and confers survival and chemoresistance in cancer. Beyond apoptosis regulation, MCL-1 has been linked to modulating mitochondrial metabolism, but the mechanism(s) by which it does so are unclear. Here, we show in tissues and cells that MCL-1 supports essential steps in long-chain (but not short-chain) fatty acid β-oxidation (FAO) through its binding to specific long-chain acyl-coenzyme A (CoA) synthetases of the ACSL family. ACSL1 binds to the BH3-binding hydrophobic groove of MCL-1 through a non-conventional BH3-domain. Perturbation of this interaction, via genetic loss of Mcl1, mutagenesis, or use of selective BH3-mimetic MCL-1 inhibitors, represses long-chain FAO in cells and in mouse livers and hearts. Our findings reveal how anti-apoptotic MCL-1 facilitates mitochondrial metabolism and indicate that disruption of this function may be associated with unanticipated cardiac toxicities of MCL-1 inhibitors in clinical trials.
Collapse
Affiliation(s)
- Tristen Wright
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Meghan E Turnis
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christy R Grace
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xiao Li
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Lauren A Brakefield
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Integrated Program in Biomedical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Haiyan Xu
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ewa Kaminska
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Leslie K Climer
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Tresor O Mukiza
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Chi-Lun Chang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Tudor Moldoveanu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Joseph T Opferman
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
16
|
Nguyen TT, Torrini C, Shang E, Shu C, Mun JY, Gao Q, Humala N, Akman HO, Zhang G, Westhoff MA, Karpel-Massler G, Bruce JN, Canoll P, Siegelin MD. OGDH and Bcl-xL loss causes synthetic lethality in glioblastoma. JCI Insight 2024; 9:e172565. [PMID: 38483541 PMCID: PMC11141877 DOI: 10.1172/jci.insight.172565] [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: 05/24/2023] [Accepted: 03/13/2024] [Indexed: 04/23/2024] Open
Abstract
Glioblastoma (GBM) remains an incurable disease, requiring more effective therapies. Through interrogation of publicly available CRISPR and RNAi library screens, we identified the α-ketoglutarate dehydrogenase (OGDH) gene, which encodes an enzyme that is part of the tricarboxylic acid (TCA) cycle, as essential for GBM growth. Moreover, by combining transcriptome and metabolite screening analyses, we discovered that loss of function of OGDH by the clinically validated drug compound CPI-613 was synthetically lethal with Bcl-xL inhibition (genetically and through the clinically validated BH3 mimetic, ABT263) in patient-derived xenografts as well neurosphere GBM cultures. CPI-613-mediated energy deprivation drove an integrated stress response with an upregulation of the BH3-only domain protein, Noxa, in an ATF4-dependent manner, as demonstrated by genetic loss-of-function experiments. Consistently, silencing of Noxa attenuated cell death induced by CPI-613 in model systems of GBM. In patient-derived xenograft models of GBM in mice, the combination treatment of ABT263 and CPI-613 suppressed tumor growth and extended animal survival more potently than each compound on its own. Therefore, combined inhibition of Bcl-xL along with disruption of the TCA cycle might be a treatment strategy for GBM.
Collapse
Affiliation(s)
- Trang Tt Nguyen
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - Consuelo Torrini
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - Enyuan Shang
- Department of Biological Sciences, Bronx Community College, City University of New York, New York, USA
| | - Chang Shu
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - Jeong-Yeon Mun
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - Qiuqiang Gao
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | | | - Hasan O Akman
- Department of Neurology, Columbia University Medical Center, New York, New York, USA
| | - Guoan Zhang
- Proteomics and Metabolomics Core Facility, Weill Cornell Medicine, New York, New York, USA
| | | | | | | | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
17
|
Gong Q, Wang H, Zhou M, Zhou L, Wang R, Li Y. B-cell lymphoma-2 family proteins in the crosshairs: Small molecule inhibitors and activators for cancer therapy. Med Res Rev 2024; 44:707-737. [PMID: 37983840 DOI: 10.1002/med.21999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023]
Abstract
The B-cell lymphoma-2 (BCL-2) family of proteins plays a crucial role in the regulation of apoptosis, offering a dual mechanism for its control. Numerous studies have established a strong association between gene disorders of these proteins and the proliferation of diverse cancer cell types. Consequently, the identification and development of drugs targeting BCL-2 family proteins have emerged as a prominent area in antitumor therapy. Over the last two decades, several small-molecules have been designed to modulate the protein-protein interactions between anti- and proapoptotic BCL-2 proteins, effectively suppressing tumor growth and metastasis in vivo. The primary focus of research has been on developing BCL-2 homology 3 (BH3) mimetics to target antiapoptotic BCL-2 proteins, thereby competitively releasing proapoptotic BCL-2 proteins and restoring the blocked intrinsic apoptotic program. Additionally, for proapoptotic BCL-2 proteins, exogenous small molecules have been explored to activate cell apoptosis by directly interacting with executioner proteins such as BCL-2-associated X protein (BAX) or BCL-2 homologous antagonist/killer protein (BAK). In this comprehensive review, we summarize the inhibitors and activators (sensitizers) of BCL-2 family proteins developed over the past decades, highlighting their discovery, optimization, preclinical and clinical status, and providing an overall landscape of drug development targeting these proteins for therapeutic purposes.
Collapse
Affiliation(s)
- Qineng Gong
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Haojie Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Mi Zhou
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Lu Zhou
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Renxiao Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Yan Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
18
|
Hoogenboezem EN, Patel SS, Lo JH, Cavnar AB, Babb LM, Francini N, Gbur EF, Patil P, Colazo JM, Michell DL, Sanchez VM, McCune JT, Ma J, DeJulius CR, Lee LH, Rosch JC, Allen RM, Stokes LD, Hill JL, Vickers KC, Cook RS, Duvall CL. Structural optimization of siRNA conjugates for albumin binding achieves effective MCL1-directed cancer therapy. Nat Commun 2024; 15:1581. [PMID: 38383524 PMCID: PMC10881965 DOI: 10.1038/s41467-024-45609-0] [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: 03/14/2023] [Accepted: 01/29/2024] [Indexed: 02/23/2024] Open
Abstract
The high potential of siRNAs to silence oncogenic drivers remains largely untapped due to the challenges of tumor cell delivery. Here, divalent lipid-conjugated siRNAs are optimized for in situ binding to albumin to improve pharmacokinetics and tumor delivery. Systematic variation of the siRNA conjugate structure reveals that the location of the linker branching site dictates tendency toward albumin association versus self-assembly, while the lipid hydrophobicity and reversibility of albumin binding also contribute to siRNA intracellular delivery. The lead structure increases tumor siRNA accumulation 12-fold in orthotopic triple negative breast cancer (TNBC) tumors over the parent siRNA. This structure achieves approximately 80% silencing of the anti-apoptotic oncogene MCL1 and yields better survival outcomes in three TNBC models than an MCL-1 small molecule inhibitor. These studies provide new structure-function insights on siRNA-lipid conjugate structures that are intravenously injected, associate in situ with serum albumin, and improve pharmacokinetics and tumor treatment efficacy.
Collapse
Affiliation(s)
- Ella N Hoogenboezem
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Shrusti S Patel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Justin H Lo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ashley B Cavnar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lauren M Babb
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Nora Francini
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Eva F Gbur
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Prarthana Patil
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Juan M Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Danielle L Michell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Violeta M Sanchez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua T McCune
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jinqi Ma
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Carlisle R DeJulius
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Linus H Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jonah C Rosch
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ryan M Allen
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Larry D Stokes
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jordan L Hill
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kasey C Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Rebecca S Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
19
|
Li X, Xu SJ, Jin B, Lu HS, Zhao SK, Ding XF, Xu LL, Li HJ, Liu SC, Chen J, Chen G. Heparanase inhibitor OGT 2115 induces prostate cancer cell apoptosis via the downregulation of MCL‑1. Oncol Lett 2024; 27:83. [PMID: 38249815 PMCID: PMC10797316 DOI: 10.3892/ol.2024.14217] [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: 07/28/2023] [Accepted: 10/31/2023] [Indexed: 01/23/2024] Open
Abstract
Heparanase (HPSE), an endo-β-D-glucuronidase, cleaves heparan sulfate and serves an important role in the tumor microenvironment and thus in tumorigenesis. HPSE is known to promote tumor cell evasion of apoptosis. However, the underlying mechanism of this requires further study. In the present study, the results demonstrated that myeloid cell leukemia-1 (MCL-1), an antiapoptotic protein, and HPSE were upregulated in prostate cancer tissues compared with adjacent normal tissues. In addition, the HPSE inhibitor, OGT 2115, inhibited PC-3 and DU-145 prostate cancer cell viability in a dose-dependent manner, with IC50 values of 20.2 and 97.2 µM, respectively. Furthermore, annexin V/PI double-staining assays demonstrated that OGT 2115 induced apoptosis in prostate cancer cells. OGT 2115 treatment markedly decreased MCL-1 protein expression levels, whereas RNA interference-mediated downregulation of MCL-1 and OGT 2115 drug treatment synergistically induced apoptosis in PC-3 and DU-145 cells. In vivo, OGT 2115 40 mg/kg (ig) significantly inhibited PC-3 cell xenograft growth in nude mice and increased the positive TUNEL staining rate of xenograft tissues. It was therefore hypothesized that MCL-1 was an important signaling molecule in OGT 2115-induced apoptosis. The results of the present study also demonstrated that the proteasome inhibitor, MG-132, markedly inhibited the downregulation of MCL-1 protein expression levels induced by OGT 2115. However, the protein synthesis inhibitor, cycloheximide, did not affect the role of OGT 2115 in regulating MCL-1. In summary, the results of the present study demonstrated that the proapoptotic activity of OGT 2115 was achieved by downregulating MCL-1.
Collapse
Affiliation(s)
- Xin Li
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Shuai-Jun Xu
- Graduate School of Medicine, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Bin Jin
- Graduate School of Medicine, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Hong-Sheng Lu
- Department of Pathology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang 318000, P.R. China
| | - Shan-Kun Zhao
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Xiao-Fei Ding
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Ling-Long Xu
- Department of Hematology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang 318000, P.R. China
| | - Hai-Jun Li
- Department of Neurology, Taizhou Second People's Hospital, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Shuang-Chun Liu
- Laboratory Department, Municipal Hospital Affiliated to Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Jie Chen
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Guang Chen
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
- Department of Hematology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang 318000, P.R. China
| |
Collapse
|
20
|
Bigos KJA, Quiles CG, Lunj S, Smith DJ, Krause M, Troost EGC, West CM, Hoskin P, Choudhury A. Tumour response to hypoxia: understanding the hypoxic tumour microenvironment to improve treatment outcome in solid tumours. Front Oncol 2024; 14:1331355. [PMID: 38352889 PMCID: PMC10861654 DOI: 10.3389/fonc.2024.1331355] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Hypoxia is a common feature of solid tumours affecting their biology and response to therapy. One of the main transcription factors activated by hypoxia is hypoxia-inducible factor (HIF), which regulates the expression of genes involved in various aspects of tumourigenesis including proliferative capacity, angiogenesis, immune evasion, metabolic reprogramming, extracellular matrix (ECM) remodelling, and cell migration. This can negatively impact patient outcomes by inducing therapeutic resistance. The importance of hypoxia is clearly demonstrated by continued research into finding clinically relevant hypoxia biomarkers, and hypoxia-targeting therapies. One of the problems is the lack of clinically applicable methods of hypoxia detection, and lack of standardisation. Additionally, a lot of the methods of detecting hypoxia do not take into consideration the complexity of the hypoxic tumour microenvironment (TME). Therefore, this needs further elucidation as approximately 50% of solid tumours are hypoxic. The ECM is important component of the hypoxic TME, and is developed by both cancer associated fibroblasts (CAFs) and tumour cells. However, it is important to distinguish the different roles to develop both biomarkers and novel compounds. Fibronectin (FN), collagen (COL) and hyaluronic acid (HA) are important components of the ECM that create ECM fibres. These fibres are crosslinked by specific enzymes including lysyl oxidase (LOX) which regulates the stiffness of tumours and induces fibrosis. This is partially regulated by HIFs. The review highlights the importance of understanding the role of matrix stiffness in different solid tumours as current data shows contradictory results on the impact on therapeutic resistance. The review also indicates that further research is needed into identifying different CAF subtypes and their exact roles; with some showing pro-tumorigenic capacity and others having anti-tumorigenic roles. This has made it difficult to fully elucidate the role of CAFs within the TME. However, it is clear that this is an important area of research that requires unravelling as current strategies to target CAFs have resulted in worsened prognosis. The role of immune cells within the tumour microenvironment is also discussed as hypoxia has been associated with modulating immune cells to create an anti-tumorigenic environment. Which has led to the development of immunotherapies including PD-L1. These hypoxia-induced changes can confer resistance to conventional therapies, such as chemotherapy, radiotherapy, and immunotherapy. This review summarizes the current knowledge on the impact of hypoxia on the TME and its implications for therapy resistance. It also discusses the potential of hypoxia biomarkers as prognostic and predictive indictors of treatment response, as well as the challenges and opportunities of targeting hypoxia in clinical trials.
Collapse
Affiliation(s)
- Kamilla JA. Bigos
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Conrado G. Quiles
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Sapna Lunj
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Danielle J. Smith
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Mechthild Krause
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- School of Medicine, Technische Universitat Dresden, Dresden, Germany
| | - Esther GC. Troost
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- School of Medicine, Technische Universitat Dresden, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Institute of Radiooncology – OncoRay, Helmholtz-Zentrum Dresden-Rossendorf, Rossendorf, Germany
| | - Catharine M. West
- Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, United Kingdom
| | - Peter Hoskin
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Mount Vernon Cancer Centre, Northwood, United Kingdom
| | - Ananya Choudhury
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Christie Hospital NHS Foundation Trust, Manchester, Germany
| |
Collapse
|
21
|
Kumar G, Pandey DM, Ghosh M, Dall'Acqua S, Gupta R, Tiwari NP, Siddique UM, Vishwakrama L, Guleri SK, Lal UR, Dubey S. Karanjin, A Promising Bioactive Compound Possessing Anti-cancer Activity against Experimental Model of Non-small Cell Lung Cancer Cells. Anticancer Agents Med Chem 2024; 24:317-333. [PMID: 37936467 DOI: 10.2174/0118715206255557231024095245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/07/2023] [Accepted: 09/25/2023] [Indexed: 11/09/2023]
Abstract
AIMS The aim of this study is to isolate the Millettia pinnata (Karanj) leaf extract for pure compound with anticancer properties and to study the molecular target of the isolates in non-small cell lung cancer cell lines. BACKGROUND In our earlier research Millettia pinnata leaf extract has demonstrated potential anticancer activities. Thus, in pursuit of the bioactive compounds, the most potential active extract from our previous study was purified. Furthermore, the anticancer properties of the isolated compound karanjin was studied and aimed for apoptosis and restraining growth. METHODS A novel method was developed through column chromatography for isolation and purification of the compound karanjin from leaf chloroform extract. The purified component was then characterised using FTIR, mass spectrometry, and NMR. An MTT-based cytotoxicity assay was used to analyse cell cytotoxicity, whereas fluorescence staining was used for apoptosis and reactive oxygen species inhibition quantification. Furthermore, the real-time PCR assay was used to determine the molecular mechanism of action in cells causing cytotoxicity induced by karanjin dosing. RESULTS The anticancer activity of karanjin in A549 cell line exhibited prominent activity revealing IC50 value of 4.85 μM. Conferring the predicted molecular pathway study, karanjin restrains the proliferation of cancer cells through apoptosis, which is controlled by extrinsic pathway proteins FAS/FADD/Caspases 8/3/9. Downregulation of KRAS and dependent gene expression also stopped cell proliferation. CONCLUSION Karanjin has been identified as a compound with potential effect in non-small cell lung cancer cells. Molecular mechanism for apoptosis and inhibition of reactive oxygen species induced through H2O2 were observed, concluding karanjin have medicinal and antioxidant properties.
Collapse
Affiliation(s)
- Gourav Kumar
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
- Birsa Munda Government Medical College, Shahdol, India
- Department of Pharmaceutical and Pharmacological Science, University of Padova, Italy
| | - Dev Mani Pandey
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Manik Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Stefano Dall'Acqua
- Department of Pharmaceutical and Pharmacological Science, University of Padova, Italy
| | - Rashmi Gupta
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
- Department of Pharmaceutical and Pharmacological Science, University of Padova, Italy
| | - Nishi Prakash Tiwari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Usman Mohd Siddique
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | | | | | - Uma Ranjan Lal
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
- Department of Natural Products, NIPER Ahmedabad, India
| | - Supriya Dubey
- Department of Chemistry, Kanya Gurukul Campus, Gurukul Kangri (Deemed to be University), Haridwar, India
| |
Collapse
|
22
|
Desai SR, Chakraborty S, Shastri A. Mechanisms of resistance to hypomethylating agents and BCL-2 inhibitors. Best Pract Res Clin Haematol 2023; 36:101521. [PMID: 38092478 DOI: 10.1016/j.beha.2023.101521] [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] [Indexed: 12/18/2023]
Abstract
Myeloid malignancies such as myelodysplastic syndrome (MDS) & acute myeloid leukemia (AML) are clonal diseases that emerge and progress due to the expansion of disease-initiating aberrant hematopoietic stem cells, that are not eliminated by conventional cytotoxic therapies. Hypomethylating agents(HMA), azacytidine and decitabine are the first line agents for treatment of MDS and a combination with BCL-2 inhibitor, venetoclax, is approved for AML induction in patients above 75 years and is also actively being investigated for use in high risk MDS. Resistance to these drugs has become a significant clinical challenge in treatment of myeloid malignancies. In this review, we discuss molecular mechanisms underlying the development of resistance to HMA and venetoclax. Insights into these mechanisms can help identify potential biomarkers for resistance prediction, aid in the development of combination therapies and strategies to prevent resistance and advance the field of cancer therapeutics.
Collapse
Affiliation(s)
- Sudhamsh Reddy Desai
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Samarpana Chakraborty
- Department of Medicine (Oncology), Department of Molecular & Developmental Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Aditi Shastri
- Department of Medicine (Oncology), Department of Molecular & Developmental Biology, Albert Einstein College of Medicine & Division of Hemato-Oncology, Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
23
|
Huang L, Yang H, Chen K, Yuan J, Li J, Dai G, Gu M, Shi Y. The suppressive efficacy of THZ1 depends on KRAS mutation subtype and is associated with super-enhancer activity and the PI3K/AKT/mTOR signalling in pancreatic ductal adenocarcinoma: A hypothesis-generating study. Clin Transl Med 2023; 13:e1500. [PMID: 38037549 PMCID: PMC10689978 DOI: 10.1002/ctm2.1500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 10/24/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Inhibition of CDK7, a potent transcription regulator, may bring new hope for treating pancreatic ductal adenocarcinoma (PDAC), which is featured by large genetic heterogeneity and abundant KRAS mutations. This investigation aimed at exploring the discrepant efficacies of THZ1, a small-molecule covalent CDK7 inhibitor, on PDACs with different KRAS mutations and the underlying mechanisms. METHODS Associations of CDK7 expression with survival by KRAS mutations were first assessed. Effects of THZ1 on PDAC by different KRAS mutations were then investigated in vitro and in vivo. Moreover, the effects of THZ1 on gene transcription and phosphorylation of RNA polymerase II (RNAPOLII) in different KRAS mutant PDACs were assessed, and the effect of THZ1 on super-enhancer activity was evaluated using chromatin immunoprecipitation sequencing. Lastly, the effects of THZ1 on the binding of H3K27ac to PIK3CA and on the PI3K/AKT/mTOR signalling were analysed. RESULTS High CDK7 expression was significantly linked to worse survival within PDAC patients carrying KRAS-G12V mutation but not in those with KRAS-G12D mutation. The apoptosis-inducing effect of THZ1 was markedly stronger in KRAS-G12V PDAC than KRAS-G12D cancer. THZ1 significantly inhibited the growth of xenograft tumour with KRAS-G12V mutation, and the inhibition was markedly stronger than for KRAS-G12D tumour. In mini-cell-derived xenograft (CDX) models, THZ1 significantly suppressed KRAS-G12V PDAC but not KRAS-G12D cancer. THZ1 significantly suppressed the phosphorylation of RNAPOLII, and this effect was stronger in KRAS-G12V PDAC (especially at ser5). KRAS-G12V PDAC had more H3K27ac-binding super-enhancers, and the inhibition of THZ1 on super-enhancer activity was also stronger in KRAS-G12V PDAC. Furthermore, THZ1 significantly weakened the binding of H3K27ac to PIK3CA in KRAS-G12V PDAC. THZ1 significantly suppressed the PI3K/AKT/mTOR pathway and its downstream markers, and this effect was stronger in KRAS-G12V cells. CONCLUSIONS In this hypothesis-generating study, THZ1 might selectively inhibit certain PDACs with KRAS-G12V mutation more potently compared with some other PDACs with KRAS-G12D mutation, which might be associated with its effect on super-enhancer activity and the PI3K/AKT/mTOR signalling. Our findings might offer novel key clues for the precise management of PDAC and important evidence for future targeted trial design. HIGHLIGHTS THZ1 had a stronger effect on PDAC-bearing KRAS-G12V mutation than G12D mutation. Suppressive effect of THZ1 on phosphorylation of RNAPOLII was stronger in KRAS-G12V than KRAS-G12D PDAC. Inhibition of THZ1 on super-enhancer activity and H3K27ac binding to PIK3CA was stronger in KRAS-G12V PDAC. Suppressive effect of THZ1 on PI3K/AKT/mTOR pathway was stronger in KRAS-G12V PDAC.
Collapse
Affiliation(s)
- Lei Huang
- Department of OncologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hui Yang
- Department of OncologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Kaidi Chen
- School of Pharmaceutical ScienceZhejiang Chinese Medical UniversityHangzhouChina
| | - Jing Yuan
- Department of PathologyChinese PLA General HospitalBeijingChina
| | - Jie Li
- Department of PathologyChinese PLA General HospitalBeijingChina
| | - Guanghai Dai
- Department of Medical OncologyChinese PLA General HospitalBeijingChina
| | - Mancang Gu
- School of Pharmaceutical ScienceZhejiang Chinese Medical UniversityHangzhouChina
- Academy of Chinese Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Yan Shi
- Department of General SurgeryShanghai Seventh People's HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
24
|
Arnett E, Pahari S, Leopold Wager CM, Hernandez E, Bonifacio JR, Lumbreras M, Renshaw C, Montoya MJ, Opferman JT, Schlesinger LS. Combination of MCL-1 and BCL-2 inhibitors is a promising approach for a host-directed therapy for tuberculosis. Biomed Pharmacother 2023; 168:115738. [PMID: 37864894 PMCID: PMC10841846 DOI: 10.1016/j.biopha.2023.115738] [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: 06/02/2023] [Revised: 10/04/2023] [Accepted: 10/14/2023] [Indexed: 10/23/2023] Open
Abstract
Tuberculosis (TB) accounts for 1.6 million deaths annually and over 25% of deaths due to antimicrobial resistance. Mycobacterium tuberculosis (M.tb) drives MCL-1 expression (family member of anti-apoptotic BCL-2 proteins) to limit apoptosis and grow intracellularly in human macrophages. The feasibility of re-purposing specific MCL-1 and BCL-2 inhibitors to limit M.tb growth, using inhibitors that are in clinical trials and FDA-approved for cancer treatment has not be tested previously. We show that specifically inhibiting MCL-1 and BCL-2 induces apoptosis of M.tb-infected macrophages, and markedly reduces M.tb growth in human and murine macrophages, and in a pre-clinical model of human granulomas. MCL-1 and BCL-2 inhibitors limit growth of drug resistant and susceptible M.tb in macrophages and act in additive fashion with the antibiotics isoniazid and rifampicin. This exciting work uncovers targeting the intrinsic apoptosis pathway as a promising approach for TB host-directed therapy. Since safety and activity studies are underway in cancer clinics for MCL-1 and BCL-2 inhibitors, we expect that re-purposing them for TB treatment should translate more readily and rapidly to the clinic. Thus, the work supports further development of this host-directed therapy approach to augment current TB treatment.
Collapse
Affiliation(s)
- Eusondia Arnett
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA.
| | - Susanta Pahari
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Chrissy M Leopold Wager
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Elizabeth Hernandez
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Jordan R Bonifacio
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Miranda Lumbreras
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Charles Renshaw
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Maria J Montoya
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | | | - Larry S Schlesinger
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA.
| |
Collapse
|
25
|
Yuda J, Will C, Phillips DC, Abraham L, Alvey C, Avigdor A, Buck W, Besenhofer L, Boghaert E, Cheng D, Cojocari D, Doyle K, Hansen TM, Huang K, Johnson EF, Judd AS, Judge RA, Kalvass JC, Kunzer A, Lam LT, Li R, Martin RL, Mastracchio A, Mitten M, Petrich A, Wang J, Ward JE, Zhang H, Wang X, Wolff JE, Bell-McGuinn KM, Souers AJ. Selective MCL-1 inhibitor ABBV-467 is efficacious in tumor models but is associated with cardiac troponin increases in patients. COMMUNICATIONS MEDICINE 2023; 3:154. [PMID: 37880389 PMCID: PMC10600239 DOI: 10.1038/s43856-023-00380-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 10/09/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND MCL-1 is a prosurvival B-cell lymphoma 2 family protein that plays a critical role in tumor maintenance and survival and can act as a resistance factor to multiple anticancer therapies. Herein, we describe the generation and characterization of the highly potent and selective MCL-1 inhibitor ABBV-467 and present findings from a first-in-human trial that included patients with relapsed/refractory multiple myeloma (NCT04178902). METHODS Binding of ABBV-467 to human MCL-1 was assessed in multiple cell lines. The ability of ABBV-467 to induce tumor growth inhibition was investigated in xenograft models of human multiple myeloma and acute myelogenous leukemia. The first-in-human study was a multicenter, open-label, dose-escalation study assessing safety, pharmacokinetics, and efficacy of ABBV-467 monotherapy. RESULTS Here we show that administration of ABBV-467 to MCL-1-dependent tumor cell lines triggers rapid and mechanism-based apoptosis. In vivo, intermittent dosing of ABBV-467 as monotherapy or in combination with venetoclax inhibits the growth of xenografts from human hematologic cancers. Results from a clinical trial evaluating ABBV-467 in patients with multiple myeloma based on these preclinical data indicate that treatment with ABBV-467 can result in disease control (seen in 1 patient), but may also cause increases in cardiac troponin levels in the plasma in some patients (seen in 4 of 8 patients), without other corresponding cardiac findings. CONCLUSIONS The selectivity of ABBV-467 suggests that treatment-induced troponin release is a consequence of MCL-1 inhibition and therefore may represent a class effect of MCL-1 inhibitors in human patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Abraham Avigdor
- Institute of Hematology, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | - Erwin Boghaert
- AbbVie Inc, North Chicago, IL, USA
- , Pleasant Prairie, WI, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Mike Mitten
- AbbVie Inc, North Chicago, IL, USA
- , Beach Park, IL, USA
| | - Adam Petrich
- AbbVie Inc, North Chicago, IL, USA
- Northwestern University, Chicago, IL, USA
- Daiichi Sankyo, Basking Ridge, NJ, USA
| | - Jin Wang
- AbbVie Inc, North Chicago, IL, USA
| | - James E Ward
- AbbVie Inc, North Chicago, IL, USA
- Seagen Inc., Bothell, WA, USA
| | | | | | | | | | | |
Collapse
|
26
|
Xie X, Yu T, Li X, Zhang N, Foster LJ, Peng C, Huang W, He G. Recent advances in targeting the "undruggable" proteins: from drug discovery to clinical trials. Signal Transduct Target Ther 2023; 8:335. [PMID: 37669923 PMCID: PMC10480221 DOI: 10.1038/s41392-023-01589-z] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/22/2023] [Accepted: 08/02/2023] [Indexed: 09/07/2023] Open
Abstract
Undruggable proteins are a class of proteins that are often characterized by large, complex structures or functions that are difficult to interfere with using conventional drug design strategies. Targeting such undruggable targets has been considered also a great opportunity for treatment of human diseases and has attracted substantial efforts in the field of medicine. Therefore, in this review, we focus on the recent development of drug discovery targeting "undruggable" proteins and their application in clinic. To make this review well organized, we discuss the design strategies targeting the undruggable proteins, including covalent regulation, allosteric inhibition, protein-protein/DNA interaction inhibition, targeted proteins regulation, nucleic acid-based approach, immunotherapy and others.
Collapse
Affiliation(s)
- Xin Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Tingting Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
| | - Xiang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
- Department of Dermatology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Leonard J Foster
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China.
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China.
| | - Gu He
- Department of Dermatology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
27
|
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
|
28
|
Gaur T, Poddutoori R, Khare L, Bagal B, Rashmi S, Patkar N, Tembhare P, Pg S, Shetty D, Dutt A, Zhang Q, Konopleva M, Platzbeckar U, Gupta S, Samajdar S, Ramchandra M, Khattry N, Hasan SK. Novel covalent CDK7 inhibitor potently induces apoptosis in acute myeloid leukemia and synergizes with Venetoclax. J Exp Clin Cancer Res 2023; 42:186. [PMID: 37507802 PMCID: PMC10386772 DOI: 10.1186/s13046-023-02750-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
INTRODUCTION The emergence of resistance to the highly successful BCL2-directed therapy is a major unmet need in acute myeloid leukemia (AML), an aggressive malignancy with poor survival rates. Towards identifying therapeutic options for AML patients who progress on BCL2-directed therapy, we studied a clinical-stage CDK7 inhibitor XL102, which is being evaluated in solid tumors (NCT04726332). MATERIALS AND METHODS To determine the anti-proliferative effects of XL102, we performed experiments including time-resolved fluorescence resonance energy transfer, target occupancy, cell cycle and apoptosis-based assays. We also included genetically characterized primary myeloid blasts from de novo and relapsed/refractory AML patients. For mechanistic studies, CRISPR/Cas9 mediated knockout of CDK7 and c-Myc and immunoblotting were performed. NOD/SCID orthotropic and subcutaneous AML xenografts were used to determine anti-leukemic effects. To assess the synergistic effects of XL102 with Venetoclax, we performed RNA sequencing and gene set enrichment analysis using Venetoclax sensitive and resistant model systems. RESULTS XL102, a highly specific, orally bioavailable covalent inhibitor of CDK7. Inhibitory effect on CDK7 by XL102 in primary myeloid blasts (n = 54) was in nanomolar range (mean = 300 nM; range = 4.0-952 nM). XL102 treated AML cells showed a reduction in phosphorylation levels of Serine 2/5/7 at carboxy-terminal domain of RNA polymerase II. T-loop phosphorylation of CDK1(Thr161) and CDK2(Thr160) was inhibited by XL102 in dose-dependent manner leading to cell-cycle arrest. c-Myc downregulation and enhanced levels of p53 and p21 in XL102 treated cells were observed. Increased levels of p21 and activation of p53 by XL102 were mimicked by genetic ablation of CDK7, which supports that the observed effects of XL102 are due to CDK7 inhibition. XL102 treated AML xenografts showed remarkable reduction in hCD45 + marrow cells (mean = 0.60%; range = 0.04%-3.53%) compared to vehicle control (mean = 38.2%; range = 10.1%-78%), with corresponding increase in p53, p21 and decrease in c-Myc levels. The data suggests XL102 induces apoptosis in AML cells via CDK7/c-Myc/p53 axis. RNA-sequencing from paired Venetoclax-sensitive and Venetoclax-resistant cells treated with XL102 showed downregulation of genes involved in proliferation and apoptosis. CONCLUSION Taken together, XL102 with Venetoclax led to synergistic effects in overcoming resistance and provided a strong rationale for clinical evaluation of XL102 as a single agent and in combination with Venetoclax.
Collapse
Affiliation(s)
- Tarang Gaur
- Hasan Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
| | - Ramulu Poddutoori
- Aurigene Oncology Limited, Electronic City Hosur Road, Bangalore, 560100, India
| | - Leena Khare
- Aurigene Oncology Limited, Electronic City Hosur Road, Bangalore, 560100, India
| | - Bhausaheb Bagal
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, 400014, India
| | - Sonal Rashmi
- Dutt Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
- Present Address: CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Nikhil Patkar
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Hematopathology Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
| | - Prashant Tembhare
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Hematopathology Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
| | - Subramanian Pg
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Hematopathology Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
| | - Dhanlaxmi Shetty
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Department of Cytogenetics, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
| | - Amit Dutt
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Dutt Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
| | - Qi Zhang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | | | - Uwe Platzbeckar
- Medical Clinic and Policlinic I, Hematology and Cellular Therapy, University Hospital Leipzig, Johannisallee 32, 04103, Leipzig, Germany
| | - Sudeep Gupta
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, 400014, India
| | - Susanta Samajdar
- Aurigene Oncology Limited, Electronic City Hosur Road, Bangalore, 560100, India
| | - Murali Ramchandra
- Aurigene Oncology Limited, Electronic City Hosur Road, Bangalore, 560100, India
| | - Navin Khattry
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India.
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, 400014, India.
| | - Syed K Hasan
- Hasan Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India.
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India.
| |
Collapse
|
29
|
Wei Y, Zhang L, Wang C, Li Z, Luo M, Xie G, Yang X, Li M, Ren S, Zhao D, Gao R, Gong J. Anti-apoptotic protein BCL-XL as a therapeutic vulnerability in gastric cancer. Animal Model Exp Med 2023; 6:245-254. [PMID: 37271936 PMCID: PMC10272913 DOI: 10.1002/ame2.12330] [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: 03/30/2023] [Accepted: 05/08/2023] [Indexed: 06/06/2023] Open
Abstract
BACKGROUND New therapeutic targets are needed to improve the outcomes for gastric cancer (GC) patients with advanced disease. Evasion of programmed cell death (apoptosis) is a hallmark of cancer cells and direct induction of apoptosis by targeting the pro-survival BCL2 family proteins represents a promising therapeutic strategy for cancer treatment. Therefore, understanding the molecular mechanisms underpinning cancer cell survival could provide a molecular basis for potential therapeutic interventions. METHOD Here we explored the role of BCL2L1 and the encoded anti-apoptotic BCL-XL in GC. Using Droplet Digital PCR (ddPCR) technology to investigate the DNA amplification of BCL2L1 in GC samples and GC cell lines, the sensitivity of GC cell lines to selective BCL-XL inhibitors A1155463 and A1331852, pan-inhibitor ABT-263, and VHL-based PROTAC-BCL-XL was analyzed using (CellTiter-Glo) CTG assay in vitro. Western Blot (WB) was used to detect the protein expression of BCL2 family members in GC cell lines and the manner in which PROTAC-BCL-XL kills GC cells. Co-immunoprecipitation (Co-IP) was used to investigate the mechanism of A1331852 and ABT-263 kills GC cell lines. DDPCR, WB, and real-time PCR (RTPCR) were used to investigate the correlation between DNA, RNA, protein levels, and drug activity. RESULTS The functional assay showed that a subset of GC cell lines relies on BCL-XL for survival. In gastric cancer cell lines, BCL-XL inhibitors A1155463 and A1331852 are more sensitive than the pan BCL2 family inhibitor ABT-263, indicating that ABT-263 is not an optimal inhibitor of BCL-XL. VHL-based PROTAC-BCL-XL DT2216 appears to be active in GC cells. DT2216 induces apoptosis of gastric cancer cells in a time- and dose-dependent manner through the proteasome pathway. Statistical analysis showed that the BCL-XL protein level predicts the response of GC cells to BCL-XL targeting therapy and BCL2L1 gene CNVs do not reliably predict BCL-XL expression. CONCLUSION We identified BCL-XL as a promising therapeutic target in a subset of GC cases with high levels of BCL-XL protein expression. Functionally, we demonstrated that both selective BCL-XL inhibitors and VHL-based PROTAC BCL-XL can potently kill GC cells that are reliant on BCL-XL for survival. However, we found that BCL2L1 copy number variations (CNVs) cannot reliably predict BCL-XL expression, but the BCL-XL protein level serves as a useful biomarker for predicting the sensitivity of GC cells to BCL-XL-targeting compounds. Taken together, our study pinpointed BCL-XL as potential druggable target for specific subsets of GC.
Collapse
Affiliation(s)
- Yumin Wei
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Liping Zhang
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Chao Wang
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Zefeng Li
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Mingjie Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen UniversityGuangzhouChina
| | - Guomin Xie
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Xingjiu Yang
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Mengyuan Li
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Shuyue Ren
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Dongbing Zhao
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ran Gao
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Jia‐Nan Gong
- National Human Diseases Animal Model Resource Center, The Institute of Laboratory Animal ScienceChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| |
Collapse
|
30
|
Romanov-Michailidis F, Hsiao CC, Urner LM, Jerhaoui S, Surkyn M, Miller B, Vos A, Dominguez Blanco M, Bueters R, Vinken P, Bekkers M, Walker D, Pietrak B, Eyckmans W, Dores-Sousa JL, Joo Koo S, Lento W, Bauser M, Philippar U, Rombouts FJR. Discovery of an Oral, Beyond-Rule-of-Five Mcl-1 Protein-Protein Interaction Modulator with the Potential of Treating Hematological Malignancies. J Med Chem 2023; 66:6122-6148. [PMID: 37114951 DOI: 10.1021/acs.jmedchem.2c01953] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Avoidance of apoptosis is critical for the development and sustained growth of tumors. The pro-survival protein myeloid cell leukemia 1 (Mcl-1) is an anti-apoptotic member of the Bcl-2 family of proteins which is overexpressed in many cancers. Upregulation of Mcl-1 in human cancers is associated with high tumor grade, poor survival, and resistance to chemotherapy. Therefore, pharmacological inhibition of Mcl-1 is regarded as an attractive approach to treating relapsed or refractory malignancies. Herein, we disclose the design, synthesis, optimization, and early preclinical evaluation of a potent and selective small-molecule inhibitor of Mcl-1. Our exploratory design tactics focused on structural modifications which improve the potency and physicochemical properties of the inhibitor while minimizing the risk of functional cardiotoxicity. Despite being in the "non-Lipinski" beyond-Rule-of-Five property space, the developed compound benefits from exquisite oral bioavailability in vivo and induces potent pharmacodynamic inhibition of Mcl-1 in a mouse xenograft model.
Collapse
Affiliation(s)
| | - Chien-Chi Hsiao
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Lorenz M Urner
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Soufyan Jerhaoui
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Michel Surkyn
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Bradley Miller
- Janssen Research & Development LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Ann Vos
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | | | - Ruud Bueters
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Petra Vinken
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Mariette Bekkers
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - David Walker
- Janssen Research & Development LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Beth Pietrak
- Janssen Research & Development LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Werner Eyckmans
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | | | - Seong Joo Koo
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - William Lento
- Janssen Research & Development LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Marcus Bauser
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Ulrike Philippar
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | | |
Collapse
|
31
|
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: 16] [Impact Index Per Article: 8.0] [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
|
32
|
Andreozzi F, Dragani M, Quivoron C, Le Bras F, Assi T, Danu A, Belhadj K, Lazarovici J, Cotteret S, Bernard OA, Ribrag V, Michot JM. Precision Medicine Approach Based on Molecular Alterations for Patients with Relapsed or Refractory Multiple Myeloma: Results from the MM-EP1 Study. Cancers (Basel) 2023; 15:cancers15051508. [PMID: 36900299 PMCID: PMC10001403 DOI: 10.3390/cancers15051508] [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: 12/03/2022] [Revised: 02/18/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Despite that cytogenetic and molecular analysis of tumor cells can rapidly identify recurring molecular abnormalities, no personalized therapy is currently available in the setting of relapsed/refractory multiple myeloma (r/r MM). METHODS MM-EP1 is a retrospective study aimed at comparing a personalized molecular-oriented (MO) versus a non-molecular-oriented (no-MO) approach in r/r MM. Actionable molecular targets and their associated therapies were the BRAF V600E mutation and BRAF inhibitors; t(11;14)(q13;q32) and BCL2 inhibitors; and t(4;14)(p16;q32) with FGFR3 fusion/rearrangements and FGFR3 inhibitors. RESULTS One hundred three highly pretreated r/r MM patients with a median age of 67 years (range 44-85) were included. Seventeen (17%) patients were treated using an MO approach with BRAF inhibitors (vemurafenib or dabrafenib, n = 6), BCL2 inhibitor (venetoclax, n = 9), or FGFR3 inhibitor (erdafitinib, n = 2). Eighty-six (86%) patients received non-MO therapies. Overall response rate was 65% in MO patients versus 58% in the non-MO group (p = 0.053). Median PFS and OS were 9 and 6 months (HR = 0.96; CI95 = 0.51-1.78; p = 0.88) and 26 and 28 months (HR = 0.98; CI95 = 0.46-2.12; p = 0.98), respectively, in MO and no-MO patients. CONCLUSION Despite the low number of patients treated with an MO approach, this study highlights the strengths and weakness of a molecular-targeted approach for the treatment of multiple myeloma. Widespread biomolecular techniques and improvement of precision medicine treatment algorithms could improve selection for precision medicine in myeloma.
Collapse
Affiliation(s)
- Fabio Andreozzi
- Département d’Innovation Thérapeutique et d’Essais Précoces, Gustave Roussy, 94805 Villejuif, France
- Hematology Department, Institute Jules Bordet, 1070 Bruxelles, Belgium
- Correspondence:
| | - Matteo Dragani
- Hematology Department, Gustave Roussy, 94805 Villejuif, France
| | - Cyril Quivoron
- Translational Research Hematological Laboratory, Gustave Roussy, 94805 Villejuif, France
- INSERM U1170, Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France
| | - Fabien Le Bras
- Hematology Department, Assistance Publique Hôpitaux de Paris, Hôpital Henri Mondor, 94000 Créteil, France
| | - Tarek Assi
- Hematology Department, Gustave Roussy, 94805 Villejuif, France
| | - Alina Danu
- Hematology Department, Gustave Roussy, 94805 Villejuif, France
| | - Karim Belhadj
- Hematology Department, Assistance Publique Hôpitaux de Paris, Hôpital Henri Mondor, 94000 Créteil, France
| | | | - Sophie Cotteret
- Department of Medical Biology and Pathology, Gustave Roussy, 94805 Villejuif, France
| | - Olivier A. Bernard
- INSERM U1170, Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France
| | - Vincent Ribrag
- Département d’Innovation Thérapeutique et d’Essais Précoces, Gustave Roussy, 94805 Villejuif, France
- Hematology Department, Gustave Roussy, 94805 Villejuif, France
- Translational Research Hematological Laboratory, Gustave Roussy, 94805 Villejuif, France
- INSERM U1170, Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France
| | - Jean-Marie Michot
- Département d’Innovation Thérapeutique et d’Essais Précoces, Gustave Roussy, 94805 Villejuif, France
- Hematology Department, Gustave Roussy, 94805 Villejuif, France
- INSERM U1170, Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France
| |
Collapse
|
33
|
Hoogenboezem EN, Patel SS, Cavnar AB, Lo JH, Babb LM, Francini N, Patil P, Colazo JM, Michell DL, Sanchez VM, McCune JT, Ma J, DeJulius CR, Lee LH, Rosch JC, Allen RM, Stokes LD, Hill JL, Vickers KC, Cook RS, Duvall CL. Structural Optimization of siRNA Conjugates for Albumin Binding Achieves Effective MCL1-Targeted Cancer Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.14.528574. [PMID: 36824780 PMCID: PMC9948981 DOI: 10.1101/2023.02.14.528574] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
The high potential for therapeutic application of siRNAs to silence traditionally undruggable oncogenic drivers remains largely untapped due to the challenges of tumor cell delivery. Here, siRNAs were optimized for in situ binding to albumin through C18 lipid modifications to improve pharmacokinetics and tumor delivery. Systematic variation of siRNA conjugates revealed a lead structure with divalent C18 lipids each linked through three repeats of hexaethylene glycol connected by phosphorothioate bonds. Importantly, we discovered that locating the branch site of the divalent lipid structure proximally (adjacent to the RNA) rather than at a more distal site (after the linker segment) promotes association with albumin, while minimizing self-assembly and lipoprotein association. Comparison to higher albumin affinity (diacid) lipid variants and siRNA directly conjugated to albumin underscored the importance of conjugate hydrophobicity and reversibility of albumin binding for siRNA delivery and bioactivity in tumors. The lead conjugate increased tumor siRNA accumulation 12-fold in orthotopic mouse models of triple negative breast cancer over the parent siRNA. When applied for silencing of the anti-apoptotic oncogene MCL-1, this structure achieved approximately 80% MCL1 silencing in orthotopic breast tumors. Furthermore, application of the lead conjugate structure to target MCL1 yielded better survival outcomes in three independent, orthotopic, triple negative breast cancer models than an MCL1 small molecule inhibitor. These studies provide new structure-function insights on optimally leveraging siRNA-lipid conjugate structures that associate in situ with plasma albumin for molecular-targeted cancer therapy.
Collapse
Affiliation(s)
| | - Shrusti S. Patel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Ashley B. Cavnar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Justin H. Lo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Lauren M. Babb
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Nora Francini
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Prarthana Patil
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Juan M. Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN
| | | | - Violeta M. Sanchez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Joshua T. McCune
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Jinqi Ma
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | | | | | - Jonah C. Rosch
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
| | - Ryan M. Allen
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Larry D. Stokes
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Jordan L. Hill
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Kasey C. Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Rebecca S. Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| |
Collapse
|
34
|
Chen L, Chauhan J, Yap JL, Goodis CC, Wilder PT, Fletcher S. Discovery of N-sulfonylated aminosalicylic acids as dual MCL-1/BCL-xL inhibitors. RSC Med Chem 2023; 14:103-112. [PMID: 36760746 PMCID: PMC9890589 DOI: 10.1039/d2md00277a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
The anti-apoptotic protein MCL-1, which is overexpressed in multiple cancers, is presently a focus for the development of targeted drugs in oncology. We previously discovered inhibitors of MCL-1 based on 1-sulfonylated 1,2,3,4-tetrahydroquinoline-6-carboxylic acids ("1,6-THQs"). However, with the nitrogen atom constrained in the bicyclic ring, we were unable to modify the alkyl portion of the tertiary sulfonamide functionality. Moreover, the introduction of additional functional groups onto the benzene ring portion of the THQ bicycle would not be trivial. Therefore, we elected to deconstruct the piperidine-type ring of the 6-carboxy-THQ lead to create a new 4-aminobenzoic acid scaffold. Given its simplicity, this permitted us to introduce diversity at the sulfonamide nitrogen, as well as vary the positions and substituents of the benzene ring. One of our most potent MCL-1 inhibitors, 6e-OH, exhibited a K i of 0.778 μM. Heteronuclear single quantum coherence experiments suggested 6e-OH bound in the canonical BH3-binding groove, with significant perturbations of R263, which forms a salt bridge with MCL-1's pro-apoptotic binding partners, as well as residues in the p2 pocket. Selectivity studies indicated that our compounds are dual inhibitors of MCL-1 and BCL-xL, with 17cd the most potent dual inhibitor: K i = 0.629 μM (MCL-1), 1.67 μM (BCL-xL). Whilst selective inhibitors may be more desirable in certain instances, polypharmacological agents whose additional target(s) address other pathways associated with the disease state, or serve to counter resistance mechanisms to the primary target, may prove particularly effective therapeutics. Since selective MCL-1 inhibition may be thwarted by overexpression of sister anti-apoptotic proteins, including BCL-xL and BCL-2, we believe our work lays a solid foundation towards the development of multi-targeting anti-cancer drugs.
Collapse
Affiliation(s)
- Lijia Chen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy 20 N. Pine St. Baltimore MD 21201 USA
| | - Jay Chauhan
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy 20 N. Pine St. Baltimore MD 21201 USA
| | - Jeremy L Yap
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy 20 N. Pine St. Baltimore MD 21201 USA
| | - Christopher C Goodis
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy 20 N. Pine St. Baltimore MD 21201 USA
| | - Paul T Wilder
- University of Maryland School of Medicine 20 S. Greene St. Baltimore MD 21201 USA
- University of Maryland Greenebaum Cancer Center 20 S. Greene St. Baltimore MD 21201 USA
| | - Steven Fletcher
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy 20 N. Pine St. Baltimore MD 21201 USA
- University of Maryland Greenebaum Cancer Center 20 S. Greene St. Baltimore MD 21201 USA
| |
Collapse
|
35
|
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
|
36
|
Kaloni D, Diepstraten ST, Strasser A, Kelly GL. BCL-2 protein family: attractive targets for cancer therapy. Apoptosis 2023; 28:20-38. [PMID: 36342579 PMCID: PMC9950219 DOI: 10.1007/s10495-022-01780-7] [Citation(s) in RCA: 173] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2022] [Indexed: 11/09/2022]
Abstract
Acquired resistance to cell death is a hallmark of cancer. The BCL-2 protein family members play important roles in controlling apoptotic cell death. Abnormal over-expression of pro-survival BCL-2 family members or abnormal reduction of pro-apoptotic BCL-2 family proteins, both resulting in the inhibition of apoptosis, are frequently detected in diverse malignancies. The critical role of the pro-survival and pro-apoptotic BCL-2 family proteins in the regulation of apoptosis makes them attractive targets for the development of agents for the treatment of cancer. This review describes the roles of the various pro-survival and pro-apoptotic members of the BCL-2 protein family in normal development and organismal function and how defects in the control of apoptosis promote the development and therapy resistance of cancer. Finally, we discuss the development of inhibitors of pro-survival BCL-2 proteins, termed BH3-mimetic drugs, as novel agents for cancer therapy.
Collapse
Affiliation(s)
- Deeksha Kaloni
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC Australia ,Department of Medical Biology, University of Melbourne, Melbourne, VIC Australia
| | - Sarah T Diepstraten
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC Australia
| | - Andreas Strasser
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC Australia ,Department of Medical Biology, University of Melbourne, Melbourne, VIC Australia
| | - Gemma L Kelly
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia. .,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
37
|
Targeting anti-apoptotic pathways eliminates senescent melanocytes and leads to nevi regression. Nat Commun 2022; 13:7923. [PMID: 36564381 PMCID: PMC9789033 DOI: 10.1038/s41467-022-35657-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Human melanocytic nevi (moles) result from a brief period of clonal expansion of melanocytes. As a cellular defensive mechanism against oncogene-induced hyperplasia, nevus-resident melanocytes enter a senescent state of stable cell cycle arrest. Senescent melanocytes can persist for months in mice and years in humans with a risk to escape the senescent state and progress to melanoma. The mechanisms providing prolonged survival of senescent melanocytes remain poorly understood. Here, we show that senescent melanocytes in culture and in nevi express high level of the anti-apoptotic BCL-2 family member BCL-W but remain insensitive to the pan-BCL-2 inhibitor ABT-263. We demonstrate that resistance to ABT-263 is driven by mTOR-mediated enhanced translation of another anti-apoptotic member, MCL-1. Strikingly, the combination of ABT-263 and MCL-1 inhibitors results in synthetic lethality to senescent melanocytes, and its topical application sufficient to eliminate nevi in male mice. These data highlight the important role of redundant anti-apoptotic mechanisms for the survival advantage of senescent melanocytes, and the proof-of-concept for a non-invasive combination therapy for nevi removal.
Collapse
|
38
|
Krawiec K, Strzałka P, Czemerska M, Wiśnik A, Zawlik I, Wierzbowska A, Pluta A. Targeting Apoptosis in AML: Where Do We Stand? Cancers (Basel) 2022; 14:cancers14204995. [PMID: 36291779 PMCID: PMC9600036 DOI: 10.3390/cancers14204995] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/01/2022] [Accepted: 10/08/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary In patients with acute myeloid leukemia (AML), genetic mutations can cause cells to evade regulated cell death (RCD), resulting in excessive cell proliferation. The best-known form of RCD is apoptosis, which prevents the emergence of cancer cells; disturbances in this process are an important factor in the development and progression of AML. Clearly, it is essential to understand the mechanisms of apoptosis to establish a personalized, patient-specific approach in AML therapy. Therefore, this paper comprehensively reviews the current range of AML treatment approaches related to apoptosis and highlights other promising concepts such as neddylation. Abstract More than 97% of patients with acute myeloid leukemia (AML) demonstrate genetic mutations leading to excessive proliferation combined with the evasion of regulated cell death (RCD). The most prominent and well-defined form of RCD is apoptosis, which serves as a defense mechanism against the emergence of cancer cells. Apoptosis is regulated in part by the BCL-2 family of pro- and anti-apoptotic proteins, whose balance can significantly determine cell survival. Apoptosis evasion plays a key role in tumorigenesis and drug resistance, and thus in the development and progression of AML. Research on the structural and biochemical aspects of apoptosis proteins and their regulators offers promise for new classes of targeted therapies and strategies for therapeutic intervention. This review provides a comprehensive overview of current AML treatment options related to the mechanism of apoptosis, particularly its mitochondrial pathway, and other promising concepts such as neddylation. It pays particular attention to clinically-relevant aspects of current and future AML treatment approaches, highlighting the molecular basis of individual therapies.
Collapse
Affiliation(s)
- Kinga Krawiec
- Department of Hematology, Medical University of Lodz, 93-513 Lodz, Poland
- Copernicus Multi-Specialist Oncology and Traumatology Center, 93-513 Lodz, Poland
| | - Piotr Strzałka
- Department of Hematology, Medical University of Lodz, 93-513 Lodz, Poland
- Copernicus Multi-Specialist Oncology and Traumatology Center, 93-513 Lodz, Poland
| | - Magdalena Czemerska
- Department of Hematology, Medical University of Lodz, 93-513 Lodz, Poland
- Copernicus Multi-Specialist Oncology and Traumatology Center, 93-513 Lodz, Poland
| | - Aneta Wiśnik
- Copernicus Multi-Specialist Oncology and Traumatology Center, 93-513 Lodz, Poland
| | - Izabela Zawlik
- Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland
- Laboratory of Molecular Biology, Centre for Innovative Research in Medical and Natural Sciences, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland
| | - Agnieszka Wierzbowska
- Department of Hematology, Medical University of Lodz, 93-513 Lodz, Poland
- Copernicus Multi-Specialist Oncology and Traumatology Center, 93-513 Lodz, Poland
| | - Agnieszka Pluta
- Department of Hematology, Medical University of Lodz, 93-513 Lodz, Poland
- Copernicus Multi-Specialist Oncology and Traumatology Center, 93-513 Lodz, Poland
- Correspondence:
| |
Collapse
|
39
|
Fraser CS, Spetz JKE, Qin X, Presser A, Choiniere J, Li C, Yu S, Blevins F, Hata AN, Miller JW, Bradshaw GA, Kalocsay M, Sanchorawala V, Sarosiek S, Sarosiek KA. Exploiting endogenous and therapy-induced apoptotic vulnerabilities in immunoglobulin light chain amyloidosis with BH3 mimetics. Nat Commun 2022; 13:5789. [PMID: 36184661 PMCID: PMC9527241 DOI: 10.1038/s41467-022-33461-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 09/16/2022] [Indexed: 01/11/2023] Open
Abstract
Immunoglobulin light chain (AL) amyloidosis is an incurable hematologic disorder typically characterized by the production of amyloidogenic light chains by clonal plasma cells. These light chains misfold and aggregate in healthy tissues as amyloid fibrils, leading to life-threatening multi-organ dysfunction. Here we show that the clonal plasma cells in AL amyloidosis are highly primed to undergo apoptosis and dependent on pro-survival proteins MCL-1 and BCL-2. Notably, this MCL-1 dependency is indirectly targeted by the proteasome inhibitor bortezomib, currently the standard of care for this disease and the related plasma cell disorder multiple myeloma, due to upregulation of pro-apoptotic Noxa and its inhibitory binding to MCL-1. BCL-2 inhibitors sensitize clonal plasma cells to multiple front-line therapies including bortezomib, dexamethasone and lenalidomide. Strikingly, in mice bearing AL amyloidosis cell line xenografts, single agent treatment with the BCL-2 inhibitor ABT-199 (venetoclax) produces deeper remissions than bortezomib and triples median survival. Mass spectrometry-based proteomic analysis reveals rewiring of signaling pathways regulating apoptosis, proliferation and mitochondrial metabolism between isogenic AL amyloidosis and multiple myeloma cells that divergently alter their sensitivity to therapies. These findings provide a roadmap for the use of BH3 mimetics to exploit endogenous and induced apoptotic vulnerabilities in AL amyloidosis.
Collapse
Affiliation(s)
- Cameron S Fraser
- John B. Little Center for Radiation Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
- Program in Molecular and Integrative Physiological Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, 02115, USA
| | - Johan K E Spetz
- John B. Little Center for Radiation Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
- Program in Molecular and Integrative Physiological Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, 02115, USA
| | - Xingping Qin
- John B. Little Center for Radiation Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
- Program in Molecular and Integrative Physiological Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, 02115, USA
| | - Adam Presser
- John B. Little Center for Radiation Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
- Program in Molecular and Integrative Physiological Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, 02115, USA
| | - Jonathan Choiniere
- John B. Little Center for Radiation Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
- Program in Molecular and Integrative Physiological Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, 02115, USA
| | - Chendi Li
- Massachusetts General Hospital Cancer Center, Charlestown, MA, 02129, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Stacey Yu
- John B. Little Center for Radiation Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
- Program in Molecular and Integrative Physiological Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, 02115, USA
| | - Frances Blevins
- Section of Hematology & Medical Oncology, Boston Medical Center, Boston, MA, 02118, USA
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Aaron N Hata
- Massachusetts General Hospital Cancer Center, Charlestown, MA, 02129, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Jeffrey W Miller
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
| | - Gary A Bradshaw
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, 02115, USA
| | - Marian Kalocsay
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, 02115, USA
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vaishali Sanchorawala
- Section of Hematology & Medical Oncology, Boston Medical Center, Boston, MA, 02118, USA
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Shayna Sarosiek
- Section of Hematology & Medical Oncology, Boston Medical Center, Boston, MA, 02118, USA.
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, 02118, USA.
- Dana-Farber Cancer Institute, Harvard Cancer Center, Boston, 02215, USA.
| | - Kristopher A Sarosiek
- John B. Little Center for Radiation Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA.
- Program in Molecular and Integrative Physiological Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA.
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, 02115, USA.
| |
Collapse
|
40
|
Systematic profiling of conditional degron tag technologies for target validation studies. Nat Commun 2022; 13:5495. [PMID: 36127368 PMCID: PMC9489723 DOI: 10.1038/s41467-022-33246-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 09/08/2022] [Indexed: 12/03/2022] Open
Abstract
Conditional degron tags (CDTs) are a powerful tool for target validation that combines the kinetics and reversible action of pharmacological agents with the generalizability of genetic manipulation. However, successful design of a CDT fusion protein often requires a prolonged, ad hoc cycle of construct design, failure, and re-design. To address this limitation, we report here a system to rapidly compare the activity of five unique CDTs: AID/AID2, IKZF3d, dTAG, HaloTag, and SMASh. We demonstrate the utility of this system against 16 unique protein targets. We find that expression and degradation are highly dependent on the specific CDT, the construct design, and the target. None of the CDTs leads to efficient expression and/or degradation across all targets; however, our systematic approach enables the identification of at least one optimal CDT fusion for each target. To enable the adoption of CDT strategies more broadly, we have made these reagents, and a detailed protocol, available as a community resource. Conditional Degron Tags are a valuable tool to validate and study novel therapeutic targets. Here, the authors compared 5 orthogonal tags across 16 unique proteins and provide a panel of vectors for users to systematically screen the tags with their own protein of interest.
Collapse
|
41
|
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
|
42
|
Drennen B, Goodis CC, Bowen N, Yu W, Vickers G, Wilder PT, MacKerell AD, Fletcher S. Scaffold hopping from indoles to indazoles yields dual MCL-1/BCL-2 inhibitors from MCL-1 selective leads. RSC Med Chem 2022; 13:963-969. [PMID: 36092148 PMCID: PMC9384788 DOI: 10.1039/d2md00095d] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/30/2022] [Indexed: 11/21/2022] Open
Abstract
Overexpression of the anti-apoptotic BCL-2 proteins is associated with the development and progression of a range of cancers. Venetoclax, an FDA-approved BCL-2 inhibitor, is fast becoming the standard-of-care for acute myeloid leukemia and chronic lymphocytic leukemia. However, the median survival offered by venetoclax is only 18 months (as part of a combination therapy regimen), and one of the primary culprits for this is the concomitant upregulation of sister anti-apoptotic proteins, in particular MCL-1 (and BCL-xL), which provides an escape route that manifests as venetoclax resistance. Since inhibition of BCL-xL leads to thrombocytopenia, we believe that a dual MCL-1/BCL-2 inhibitor may provide an enhanced therapeutic effect relative to a selective BCL-2 inhibitor. Beginning with a carboxylic acid-containing literature compound that is a potent inhibitor of MCL-1 and a moderate inhibitor of BCL-2, we herein describe our efforts to develop dual inhibitors of MCL-1 and BCL-2 by scaffold hopping from an indole core to an indazole framework. Subsequently, further elaboration of our novel N2-substituted, indazole-3-carboxylic acid lead into a family of indazole-3-acylsulfonamides resulted in improved inhibition of both MCL-1 and BCL-2, possibly through occupation of the p4 pocket, with minimal or no inhibition of BCL-xL.
Collapse
Affiliation(s)
- Brandon Drennen
- 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, Department of Pharmaceutical Sciences20 N. Pine St.BaltimoreMD 21201USA
| | - Nathan Bowen
- Department of Chemistry, Cardiff UniversityCF10 3ATUK
| | - Wenbo Yu
- University of Maryland School of Pharmacy, Department of Pharmaceutical Sciences 20 N. Pine St. Baltimore MD 21201 USA
| | | | - Paul T. Wilder
- University of Maryland School of Medicine20 S. Greene St.BaltimoreMD 21201USA
| | - Alexander D. MacKerell
- University of Maryland School of Pharmacy, Department of Pharmaceutical Sciences20 N. Pine St.BaltimoreMD 21201USA,University of Maryland Greenebaum Cancer Center20 S. Greene St.BaltimoreMD 21201USA
| | - 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
|
43
|
The FGFR2c/PKCε Axis Controls MCL-1-Mediated Invasion in Pancreatic Ductal Adenocarcinoma Cells: Perspectives for Innovative Target Therapies. Biomedicines 2022; 10:biomedicines10071652. [PMID: 35884957 PMCID: PMC9312859 DOI: 10.3390/biomedicines10071652] [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: 06/01/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 11/23/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy whose main characterizations are Kirsten Rat Sarcoma-activating mutations (KRAS) and a highly aggressive phenotype. Based on our recent findings demonstrating that the highly aberrant expression of the mesenchymal isoform of Fibroblast Growth Factor Receptor 2 (FGFR2c) in PDAC cells activates Protein-Kinase C Epsilon (PKCε), which in turn controls receptor-mediated epithelial to mesenchymal transition (EMT), here we investigated the involvement of these signaling events in the establishment of additional tumorigenic features. Using PDAC cell lines expressing divergent levels of the FGFR2c and stable protein depletion approaches by short hairpin RNA (shRNA), we found that FGFR2c expression and its PKCε downstream signaling are responsible for the invasive response to Fibroblast Growth Factor 2 (FGF2) and for anchorage-independent growth. In addition, in vitro clonogenic assays, coupled with the check of the amount of cleaved Poly Adenosine Diphosphate-Ribose Polymerase 1 (PARP1) by Western blot, highlighted the involvement of both FGFR2c and PKCε in cell viability. Finally, monitoring of Myeloid Cell Leukemia 1 (MCL-1) expression and Sarcoma kinase family (SRC) phosphorylation suggested that the FGFR2c/PKCε axis could control cell migration/invasion possibly via MCL-1/SRC-mediated reorganization of the actin cytoskeleton. Being PKCs RAS-independent substrates, the identification of PKCε as a hub molecule downstream FGFR2c at the crossroad of signaling networks governing the main malignant tumor hallmarks could represent an important advance towards innovative target therapies overcoming RAS.
Collapse
|
44
|
Chiou JT, Lee YC, Wang LJ, Chang LS. BCL2 inhibitor ABT-199 and BCL2L1 inhibitor WEHI-539 coordinately promote NOXA-mediated degradation of MCL1 in human leukemia cells. Chem Biol Interact 2022; 361:109978. [PMID: 35561756 DOI: 10.1016/j.cbi.2022.109978] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/05/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023]
Abstract
Human leukemia U937 cells that were continuously treated with hydroquinone (HQ) were transformed into U937/HQ cells with increased MCL1 and BCL2L1 expression. Compared with their parental cells, U937/HQ cells were less sensitive to ABT-263 (BCL2/BCL2L1 inhibitor)/ABT-199 (BCL2 inhibitor) cytotoxicity. The combination of WEHI-539 (BCL2L1 inhibitor) with either ABT-199 or ABT-263 showed synergistic cytotoxicity to U937 and U937/HQ cells. Therefore, we further investigated the cytotoxic mechanism induced by the combination of WEHI-539 and ABT-199. The combined treatment of WEHI-539 and ABT-199 induced NOX4/ROS/p38 MAPK axis-mediated autophagy, which in turn accelerated β-TrCP mRNA turnover. Downregulation of β-TrCP increased Sp1 expression, thereby promoting Sp1-mediated NOXA transcription, which in turn induced NOXA-dependent MCL1 degradation. Enforced expression of MCL1 alleviated the cytotoxicity of WEHI-539 plus ABT-199 to induce the loss of mitochondrial membrane potential and cell viability. WEHI-539 alone induced Sp1/NOXA axis-mediated MCL1 downregulation, while ABT-199 significantly decreased the dose of WEHI-539 by approximately 350- and 50-fold to induce MCL1 suppression in parental and HQ-selected cells, respectively. Furthermore, WEHI-539 sensitized ABT-199-resistant U937 cells to ABT-199 cytotoxicity by inducing NOXA-mediated degradation of MCL1. Collectively, the data in this study indicate that ABT-199 and WEHI-539 cooperatively induce NOXA-dependent MCL1 degradation, and the inhibition of MCL1 mainly explains their combined cytotoxicity in parental, HQ-selected, and ABT-199-resistant U937 cells.
Collapse
Affiliation(s)
- Jing-Ting Chiou
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Yuan-Chin Lee
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Liang-Jun Wang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
45
|
Jouffroy M, Neufeld K. Synthesis of Atropisomeric Biaryls via Chiral Suzuki–Miyaura/Enzymatic Kinetic Resolution. ACS Catal 2022. [DOI: 10.1021/acscatal.2c02090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Matthieu Jouffroy
- Chemical Process R&D, Discovery Process Research, Janssen Pharmarceutcia N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Katharina Neufeld
- Chemical Process R&D, High Throughput Experimentation, Janssen Pharmarceutcia N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
46
|
Greil C, Engelhardt M, Wäsch R. The Role of the APC/C and Its Coactivators Cdh1 and Cdc20 in Cancer Development and Therapy. Front Genet 2022; 13:941565. [PMID: 35832196 PMCID: PMC9273091 DOI: 10.3389/fgene.2022.941565] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/08/2022] [Indexed: 12/03/2022] Open
Abstract
To sustain genomic stability by correct DNA replication and mitosis, cell cycle progression is tightly controlled by the cyclic activity of cyclin-dependent kinases, their binding to cyclins in the respective phase and the regulation of cyclin levels by ubiquitin-dependent proteolysis. The spindle assembly checkpoint plays an important role at the metaphase-anaphase transition to ensure a correct separation of sister chromatids before cytokinesis and to initiate mitotic exit, as an incorrect chromosome distribution may lead to genetically unstable cells and tumorigenesis. The ubiquitin ligase anaphase-promoting complex or cyclosome (APC/C) is essential for these processes by mediating the proteasomal destruction of cyclins and other important cell cycle regulators. To this end, it interacts with the two regulatory subunits Cdh1 and Cdc20. Both play a role in tumorigenesis with Cdh1 being a tumor suppressor and Cdc20 an oncogene. In this review, we summarize the current knowledge about the APC/C-regulators Cdh1 and Cdc20 in tumorigenesis and potential targeted therapeutic approaches.
Collapse
|
47
|
Ji T, Margulis BA, Wang Z, Song T, Guo Y, Pan H, Zhang Z. Structure-Based Design and Structure-Activity Relationship Analysis of Small Molecules Inhibiting Bcl-2 Family Members. Pharm Chem J 2022. [DOI: 10.1007/s11094-022-02639-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
48
|
Montero J, Haq R. Adapted to Survive: Targeting Cancer Cells with BH3 Mimetics. Cancer Discov 2022; 12:1217-1232. [PMID: 35491624 PMCID: PMC9306285 DOI: 10.1158/2159-8290.cd-21-1334] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/11/2022] [Accepted: 02/10/2022] [Indexed: 01/07/2023]
Abstract
A hallmark of cancer is cell death evasion, underlying suboptimal responses to chemotherapy, targeted agents, and immunotherapies. The approval of the antiapoptotic BCL2 antagonist venetoclax has finally validated the potential of targeting apoptotic pathways in patients with cancer. Nevertheless, pharmacologic modulators of cell death have shown markedly varied responses in preclinical and clinical studies. Here, we review emerging concepts in the use of this class of therapies. Building on these observations, we propose that treatment-induced changes in apoptotic dependency, rather than pretreatment dependencies, will need to be recognized and targeted to realize the precise deployment of these new pharmacologic agents. SIGNIFICANCE Targeting antiapoptotic family members has proven efficacious and tolerable in some cancers, but responses are infrequent, particularly for patients with solid tumors. Biomarkers to aid patient selection have been lacking. Precision functional approaches that overcome adaptive resistance to these compounds could drive durable responses to chemotherapy, targeted therapy, and immunotherapies.
Collapse
Affiliation(s)
- Joan Montero
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Corresponding Authors: Rizwan Haq, Department of Medical Oncology M423A, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215. Phone: 617-632-6168; E-mail: ; and Joan Montero, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 15-21, Barcelona 08028, Spain. Phone: 34-93-403-9956; E-mail:
| | - Rizwan Haq
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Corresponding Authors: Rizwan Haq, Department of Medical Oncology M423A, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215. Phone: 617-632-6168; E-mail: ; and Joan Montero, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 15-21, Barcelona 08028, Spain. Phone: 34-93-403-9956; E-mail:
| |
Collapse
|
49
|
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: 56] [Impact Index Per Article: 18.7] [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
|
50
|
The BCL-2 inhibitor ABT-199/venetoclax synergizes with proteasome inhibition via transactivation of the MCL-1 antagonist NOXA. Cell Death Dis 2022; 8:215. [PMID: 35443750 PMCID: PMC9021261 DOI: 10.1038/s41420-022-01009-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 12/31/2022]
Abstract
Enhanced expression of anti-apoptotic B-cell lymphoma 2 (BCL-2) protein is frequent in cancer. Targeting of BCL-2 with the specific inhibitor ABT-199 (Venetoclax) has significant clinical activity in malignant diseases such as chronic lymphocytic leukemia and multiple myeloma. The small molecule drug ABT-199 mimics the pro-apoptotic BCL-2 homology domain 3 of BH3-only proteins and blocks the hydrophobic BC-groove in BCL-2. We have previously shown that ABT-199 synergizes with the proteasome inhibitor (PI) bortezomib in soft tissue sarcoma derived cells and cell lines to induce apoptosis. Synergistic apoptosis induction relies on the pore-forming effector BAX and expression of the pro-apoptotic BH3-only protein NOXA. Bortezomib augments expression of NOXA by blocking its proteasomal degradation. Interestingly, shown here for the first time, expression of NOXA is strongly enhanced by ABT-199 induced integrated stress response (ISR). ISR transcription factors ATF3 & ATF4 mediate transactivation of the BH3-only protein NOXA which specifically inhibits the anti-apoptotic MCL-1. Thus, NOXA potentiates the efficacy of the BCL-2 inhibitor ABT-199 by simultaneous inhibition of MCL-1. Hence, ABT-199 has a double impact by directly blocking anti-apoptotic BCL-2 and inhibiting MCL-1 via transactivated NOXA. By preventing degradation of NOXA PIs synergize with ABT-199. Synergism of ABT-199 and PIs therefore occurs on several, previously unexpected levels. This finding should prompt clinical evaluation of combinatorial regimens in further malignancies.
Collapse
|