1
|
Yang Y, Mou Y, Wan LX, Zhu S, Wang G, Gao H, Liu B. Rethinking therapeutic strategies of dual-target drugs: An update on pharmacological small-molecule compounds in cancer. Med Res Rev 2024; 44:2600-2623. [PMID: 38769656 DOI: 10.1002/med.22057] [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: 02/15/2022] [Revised: 06/06/2023] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Oncogenes and tumor suppressors are well-known to orchestrate several signaling cascades, regulate extracellular and intracellular stimuli, and ultimately control the fate of cancer cells. Accumulating evidence has recently revealed that perturbation of these key modulators by mutations or abnormal protein expressions are closely associated with drug resistance in cancer therapy; however, the inherent drug resistance or compensatory mechanism remains to be clarified for targeted drug discovery. Thus, dual-target drug development has been widely reported to be a promising therapeutic strategy for improving drug efficiency or overcoming resistance mechanisms. In this review, we provide an overview of the therapeutic strategies of dual-target drugs, especially focusing on pharmacological small-molecule compounds in cancer, including small molecules targeting mutation resistance, compensatory mechanisms, synthetic lethality, synergistic effects, and other new emerging strategies. Together, these therapeutic strategies of dual-target drugs would shed light on discovering more novel candidate small-molecule drugs for the future cancer treatment.
Collapse
Affiliation(s)
- Yiren Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yi Mou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Lin-Xi Wan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Shiou Zhu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Guan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Huiyuan Gao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Bo Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Pan J, Zhang Y, He L, Wu Y, Xiao W, Zhang J, Xu Y. STRIP2 is regulated by the transcription factor Sp1 and promotes lung adenocarcinoma progression via activating the PI3K/AKT/mTOR/MYC signaling pathway. Genomics 2024; 116:110923. [PMID: 39191354 DOI: 10.1016/j.ygeno.2024.110923] [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: 02/22/2024] [Revised: 07/26/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Patients with lung adenocarcinoma (LUAD) generally have poor prognosis. The role of striatin-interacting protein 2 (STRIP2) in LUAD remain unclear. METHODS Liquid chromatography-mass spectrometry analyses were used to screen the STRIP2-binding proteins and co-immunoprecipitation verified these interactions. A dual luciferase reporter assay explored the transcription factor activating STRIP2 transcription. Xenograft and lung metastasis models assessed STRIP2's role in tumor growth and metastasis in vivo. RESULTS STRIP2 is highly expressed in LUAD tissues and is linked to poor prognosis. STRIP2 expression in LUAD cells significantly promoted cell proliferation, invasion, and migration in vitro and in vivo. Mechanistically, STRIP2 boosted the PI3K/AKT/mTOR/MYC cascades by binding AKT. In addition, specificity protein 1, potently activated STRIP2 transcription by binding to the STRIP2 promoter. Blocking STRIP2 reduces tumor growth and lung metastasis in xenograft models. CONCLUSIONS Our study identifies STRIP2 is a key driver of LUAD progression and a potential therapeutic target.
Collapse
Affiliation(s)
- Junfan Pan
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Yuan Zhang
- The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Liu He
- School of Basic Medicine, Fujian Medical University, Fuzhou 350004, China
| | - Yue Wu
- School of Basic Medicine, Fujian Medical University, Fuzhou 350004, China
| | - Weijin Xiao
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China.
| | - Jing Zhang
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China.
| | - Yiquan Xu
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China.
| |
Collapse
|
3
|
Huffman KE, Li LS, Carstens R, Park H, Girard L, Avila K, Wei S, Kollipara R, Timmons B, Sudderth J, Bendris N, Kim J, Villalobos P, Fujimoto J, Schmid S, Deberardinis RJ, Wistuba I, Heymach J, Kittler R, Akbay EA, Posner B, Wang Y, Lam S, Kliewer SA, Mangelsdorf DJ, Minna JD. Glucocorticoid mediated inhibition of LKB1 mutant non-small cell lung cancers. Front Oncol 2023; 13:1025443. [PMID: 37035141 PMCID: PMC10078807 DOI: 10.3389/fonc.2023.1025443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/20/2023] [Indexed: 04/11/2023] Open
Abstract
The glucocorticoid receptor (GR) is an important anti-cancer target in lymphoid cancers but has been understudied in solid tumors like lung cancer, although glucocorticoids are often given with chemotherapy regimens to mitigate side effects. Here, we identify a dexamethasone-GR mediated anti-cancer response in a subset of aggressive non-small cell lung cancers (NSCLCs) that harbor Serine/Threonine Kinase 11 (STK11/LKB1) mutations. High tumor expression of carbamoyl phosphate synthase 1 (CPS1) was strongly linked to the presence of LKB1 mutations, was the best predictor of NSCLC dexamethasone (DEX) sensitivity (p < 10-16) but was not mechanistically involved in DEX sensitivity. Subcutaneous, orthotopic and metastatic NSCLC xenografts, biomarker-selected, STK11/LKB1 mutant patient derived xenografts, and genetically engineered mouse models with KRAS/LKB1 mutant lung adenocarcinomas all showed marked in vivo anti-tumor responses with the glucocorticoid dexamethasone as a single agent or in combination with cisplatin. Mechanistically, GR activation triggers G1/S cell cycle arrest in LKB1 mutant NSCLCs by inducing the expression of the cyclin-dependent kinase inhibitor, CDKN1C/p57(Kip2). All findings were confirmed with functional genomic experiments including CRISPR knockouts and exogenous expression. Importantly, DEX-GR mediated cell cycle arrest did not interfere with NSCLC radiotherapy, or platinum response in vitro or with platinum response in vivo. While DEX induced LKB1 mutant NSCLCs in vitro exhibit markers of cellular senescence and demonstrate impaired migration, in vivo DEX treatment of a patient derived xenograft (PDX) STK11/LKB1 mutant model resulted in expression of apoptosis markers. These findings identify a previously unknown GR mediated therapeutic vulnerability in STK11/LKB1 mutant NSCLCs caused by induction of p57(Kip2) expression with both STK11 mutation and high expression of CPS1 as precision medicine biomarkers of this vulnerability.
Collapse
Affiliation(s)
- Kenneth E. Huffman
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Long Shan Li
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ryan Carstens
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Hyunsil Park
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Luc Girard
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Kimberley Avila
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Shuguang Wei
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Rahul Kollipara
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Brenda Timmons
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jessica Sudderth
- Children’s Medical Center Research Institute at University of Texas (UT) Southwestern Medical Center, Dallas, TX, United States
| | - Nawal Bendris
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jiyeon Kim
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States
- Department of Urology, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, United States
| | - Pamela Villalobos
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sandra Schmid
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ralph J. Deberardinis
- Children’s Medical Center Research Institute at University of Texas (UT) Southwestern Medical Center, Dallas, TX, United States
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John Heymach
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Ralf Kittler
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Esra A. Akbay
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Bruce Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yuzhuo Wang
- British Columbia Cancer Center, Vancouver, BC, Canada
| | - Stephen Lam
- British Columbia Cancer Center, Vancouver, BC, Canada
| | - Steven A. Kliewer
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - David J. Mangelsdorf
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - John D. Minna
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
4
|
Nunes Azevedo FF, Freitas de Sousa FJ, Santos de Oliveira FL, Vieira Carletti J, Zanatta G. Binding site hotspot map of PI3Kα and mTOR in the presence of selective and dual ATP-competitive inhibitors. J Biomol Struct Dyn 2023; 41:1085-1097. [PMID: 34913837 DOI: 10.1080/07391102.2021.2016487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The PI3K/Akt/mTOR signaling pathway plays a pivotal role in cellular metabolism, growth and survival. PI3Kα hyperactivation impairs downstream signaling, including mTOR regulation, and are linked to poor prognosis and refractory cancer treatment. To support multi-target drug discovery, we took advantage from existing PI3Kα and mTOR crystallographic structures to map similarities and differences in their ATP-binding pockets in the presence of selective or dual inhibitors. Molecular dynamics and MM/PBSA calculations were employed to study the binding profile and identify the relative contribution of binding site residues. Our analysis showed that while varying parameters of solute and solvent dielectric constant interfered in the absolute binding free energy, it had no effect in the relative per residue contribution. In all complexes, the most important interactions were observed within 3-3.5 Å from inhibitors, responding for ∼75-100% of the total calculated interaction energy. While closest residues are essential for the strength of the binding of all ligands, more distant residues seem to have a larger impact on the binding of the dual inhibitor, as observed for PI3Kα residues Phe934, Lys802 and Asp805 and, mTOR residues Leu2192, Phe2358, Leu2354, Lys2187 and Tyr2225. A detailed description of individual residue contribution in the presence of selective or dual inhibitors is provided as an effort to improve the understanding of molecular mechanisms controlling multi-target inhibition. This work provides key information to support further studies seeking the rational design of potent PI3K/mTOR dual inhibitors for cancer treatment.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | | | | | | | - Geancarlo Zanatta
- Postgraduate Programme in Biochemistry, Department of Biochemistry at Federal, University of Ceará, Fortaleza, Ceará, Brazil.,Department of Physics at Federal, University of Ceará, Fortaleza, Ceará, Brazil
| |
Collapse
|
5
|
Liu Z, Zhou K, Zeng J, Zhou X, Li H, Peng K, Liu X, Feng F, Jiang B, Zhao M, Ma T. Liver kinase B1 in exosomes inhibits immune checkpoint programmed death ligand 1 and metastatic progression of intrahepatic cholangiocarcinoma. Oncol Rep 2022; 48:155. [PMID: 35856436 PMCID: PMC9350976 DOI: 10.3892/or.2022.8367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/27/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Zhuo Liu
- Third Department of General Surgery, The Central Hospital of Xiangtan, Xiangtan, Hunan 411100, P.R. China
| | - Kunyan Zhou
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, D‑30159 Hannover, Germany
| | - Jian Zeng
- Third Department of General Surgery, The Central Hospital of Xiangtan, Xiangtan, Hunan 411100, P.R. China
| | - Xin Zhou
- Third Department of General Surgery, The Central Hospital of Xiangtan, Xiangtan, Hunan 411100, P.R. China
| | - Huanyu Li
- Third Department of General Surgery, The Central Hospital of Xiangtan, Xiangtan, Hunan 411100, P.R. China
| | - Ke Peng
- Scientific Research Department, The Central Hospital of Xiangtan, Xiangtan, Hunan 411100, P.R. China
| | - Xiang Liu
- Third Department of General Surgery, The Central Hospital of Xiangtan, Xiangtan, Hunan 411100, P.R. China
| | - Feng Feng
- Third Department of General Surgery, The Central Hospital of Xiangtan, Xiangtan, Hunan 411100, P.R. China
| | - Bin Jiang
- Third Department of General Surgery, The Central Hospital of Xiangtan, Xiangtan, Hunan 411100, P.R. China
| | - Ming Zhao
- Third Department of General Surgery, The Central Hospital of Xiangtan, Xiangtan, Hunan 411100, P.R. China
| | - Tiexiang Ma
- Third Department of General Surgery, The Central Hospital of Xiangtan, Xiangtan, Hunan 411100, P.R. China
| |
Collapse
|
6
|
Ndembe G, Intini I, Perin E, Marabese M, Caiola E, Mendogni P, Rosso L, Broggini M, Colombo M. LKB1: Can We Target an Hidden Target? Focus on NSCLC. Front Oncol 2022; 12:889826. [PMID: 35646638 PMCID: PMC9131655 DOI: 10.3389/fonc.2022.889826] [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: 03/04/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
LKB1 (liver kinase B1) is a master regulator of several processes such as metabolism, proliferation, cell polarity and immunity. About one third of non-small cell lung cancers (NSCLCs) present LKB1 alterations, which almost invariably lead to protein loss, resulting in the absence of a potential druggable target. In addition, LKB1-null tumors are very aggressive and resistant to chemotherapy, targeted therapies and immune checkpoint inhibitors (ICIs). In this review, we report and comment strategies that exploit peculiar co-vulnerabilities to effectively treat this subgroup of NSCLCs. LKB1 loss leads to an enhanced metabolic avidity, and treatments inducing metabolic stress were successful in inhibiting tumor growth in several preclinical models. Biguanides, by compromising mitochondria and reducing systemic glucose availability, and the glutaminase inhibitor telaglenastat (CB-839), inhibiting glutamate production and reducing carbon intermediates essential for TCA cycle progression, have provided the most interesting results and entered different clinical trials enrolling also LKB1-null NSCLC patients. Nutrient deprivation has been investigated as an alternative therapeutic intervention, giving rise to interesting results exploitable to design specific dietetic regimens able to counteract cancer progression. Other strategies aimed at targeting LKB1-null NSCLCs exploit its pivotal role in modulating cell proliferation and cell invasion. Several inhibitors of LKB1 downstream proteins, such as mTOR, MEK, ERK and SRK/FAK, resulted specifically active on LKB1-mutated preclinical models and, being molecules already in clinical experimentation, could be soon proposed as a specific therapy for these patients. In particular, the rational use in combination of these inhibitors represents a very promising strategy to prevent the activation of collateral pathways and possibly avoid the potential emergence of resistance to these drugs. LKB1-null phenotype has been correlated to ICIs resistance but several studies have already proposed the mechanisms involved and potential interventions. Interestingly, emerging data highlighted that LKB1 alterations represent positive determinants to the new KRAS specific inhibitors response in KRAS co-mutated NSCLCs. In conclusion, the absence of the target did not block the development of treatments able to hit LKB1-mutated NSCLCs acting on several fronts. This will give patients a concrete chance to finally benefit from an effective therapy.
Collapse
Affiliation(s)
- Gloriana Ndembe
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ilenia Intini
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elisa Perin
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Mirko Marabese
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elisa Caiola
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Paolo Mendogni
- Thoracic Surgery and Lung Transplantation Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Rosso
- Thoracic Surgery and Lung Transplantation Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Massimo Broggini
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marika Colombo
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
7
|
Zheng Z, Li Y, Lu X, Zhang J, Liu Q, Zhou D, Deng X, Qiu Y, Chen Q, Zheng H, Dai J. A novel mTOR-associated gene signature for predicting prognosis and evaluating tumor immune microenvironment in lung adenocarcinoma. Comput Biol Med 2022; 145:105394. [PMID: 35325730 DOI: 10.1016/j.compbiomed.2022.105394] [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: 01/24/2022] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND The mechanistic target of rapamycin (mTOR) was proven to have great impact on apoptosis, cell proliferation, autophagy, and many other fundamental cellular processes; moreover, it closely correlates with tumor occurrence and development. However, few studies have constructed signatures based on mTOR-associated genes to assess multiple indicators of prognosis in lung adenocarcinoma (LUAD) patients. METHODS mTOR-associated gene sets, whole mRNA expression matrices, and clinical information of LUAD patients in training and validation cohorts were obtained from multiple public databases. Multiple methods were used to screen candidate genes, construct signatures, validate internally and externally, and conduct further studies: differentially expressed gene analysis, LASSO Cox regression analysis, Cox regression analysis, risk factor analysis, nomogram analysis, functional enrichment analysis, analyses in tumor immune microenvironment, and therapy. RESULTS A prognostic signature containing 8 genes (LDHA, SLA, WNT7A, PLK1, CCT6A, BTG2, TXNRD1, and DDIT4) was constructed. It performed well in both internal and external validation. Subsequent analysis found that the prognostic signature was of great significance in evaluating the tumor immune microenvironment and could guide the treatment of patients with LUAD to a certain extent. CONCLUSION The constructed mTOR-associated gene signature accurately predicted the prognostic pattern of patients with LUAD and is expected to be extremely useful in guiding LUAD therapy.
Collapse
Affiliation(s)
- Zhi Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Yanqi Li
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Xiao Lu
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Jiao Zhang
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Quanxing Liu
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Dong Zhou
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Xufeng Deng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Qian Chen
- Cancer Center of Daping Hospital, Third Military Medical University (Army Medical University), ChangJiang Sub-Road, Chongqing, 400042, China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China.
| | - Jigang Dai
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China.
| |
Collapse
|
8
|
Liu H, Guan L, Zhou Q, Huang H, Xu L. LKB1 alleviates high glucose‑ and high fat‑induced inflammation and the expression of GnRH and sexual precocity‑related genes, in mouse hypothalamic cells by activating the AMPK/FOXO1 signaling pathway. Mol Med Rep 2022; 25:143. [PMID: 35234270 PMCID: PMC8915400 DOI: 10.3892/mmr.2022.12659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/23/2021] [Indexed: 11/16/2022] Open
Abstract
Precocious puberty (PP) is a developmental disorder. Hypothalamic cells can produce gonadotropin-releasing hormone (GnRH), the final output of neuroendocrine regulation that occurs during puberty. The aim of the present study was to investigate the role of live kinase B1 (LKB1), also known as serine/threonine kinase, in the progression of PP and identify the underlying mechanisms. First, the levels of LKB1 in peripheral blood and peripheral blood mononuclear cells of children with PP were detected by reverse transcription-quantitative (RT-q) PCR or western blotting. After the GT1-7 mouse hypothalamus cell line was treated with high glucose (HG) and high fat (HF), the expression of LKB1 and GnRH was tested. LKB1 was overexpressed by transfection with a pcDNA3.1 plasmid and the levels of inflammatory factors, GnRH, PP-related factors and proteins in the AMP-activated protein kinase (AMPK)/forkhead box protein O1 (FOXO1) pathway were determined using RT-qPCR or western blot analysis. Subsequently, Compound C, an inhibitor of AMPK/FOXO1 signaling, was used to clarify whether the effects of LKB1 on PP were mediated by the regulation of this pathway. Results indicated that children with PP exhibited a lower LKB1 expression. In addition, HG and HF culture resulted in an enhanced GnRH expression and a reduced LKB1 expression in GT1-7 cells. LKB1 overexpression inhibited the contents of TNF-α, IL-6 and GnRH in in GT1-7 cells exposed to HG and HF and reduced the expression of PP-related proteins, including estrogen receptor-β, cluster of differentiation 36 and G-protein-coupled receptor. In addition, the expression of phosphorylated (p)-AMPK and p-FOXO1 was markedly downregulated following LKBI overexpression. Furthermore, compound C intervention partially diminished the inhibitory effects of LKB1-mediated upregulation on the levels of inflammation and PP-related factors. In conclusion, these results demonstrated that LKB1 alleviated HG- and HF-induced inflammation, as well as the expression of GnRH and sexual precocity-related genes, in GT1-7 cells by activating the AMPK/FOXO1 signaling pathway.
Collapse
Affiliation(s)
- Hui Liu
- Department of Endocrine Genetics and Metabolism, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Limei Guan
- Department of Endocrine Genetics and Metabolism, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Qing Zhou
- Department of Endocrine Genetics and Metabolism, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Hailong Huang
- Fujian Provincial Key Laboratory for Prenatal Diagnosis and Birth Defect, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Liangpu Xu
- Fujian Provincial Key Laboratory for Prenatal Diagnosis and Birth Defect, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
9
|
Galan-Cobo A, Stellrecht CM, Yilmaz E, Yang C, Qian Y, Qu X, Akhter I, Ayres ML, Fan Y, Tong P, Diao L, Ding J, Giri U, Gudikote J, Nilsson M, Wierda WG, Wang J, Skoulidis F, Minna JD, Gandhi V, Heymach JV. Enhanced Vulnerability of LKB1-Deficient NSCLC to Disruption of ATP Pools and Redox Homeostasis by 8-Cl-Ado. Mol Cancer Res 2022; 20:280-292. [PMID: 34654720 PMCID: PMC8816854 DOI: 10.1158/1541-7786.mcr-21-0448] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/30/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022]
Abstract
Loss-of-function somatic mutations of STK11, a tumor suppressor gene encoding LKB1 that contributes to the altered metabolic phenotype of cancer cells, is the second most common event in lung adenocarcinomas and often co-occurs with activating KRAS mutations. Tumor cells lacking LKB1 display an aggressive phenotype, with uncontrolled cell growth and higher energetic and redox stress due to its failure to balance ATP and NADPH levels in response to cellular stimulus. The identification of effective therapeutic regimens for patients with LKB1-deficient non-small cell lung cancer (NSCLC) remains a major clinical need. Here, we report that LKB1-deficient NSCLC tumor cells displayed reduced basal levels of ATP and to a lesser extent other nucleotides, and markedly enhanced sensitivity to 8-Cl-adenosine (8-Cl-Ado), an energy-depleting nucleoside analog. Treatment with 8-Cl-Ado depleted intracellular ATP levels, raised redox stress, and induced cell death leading to a compensatory suppression of mTOR signaling in LKB1-intact, but not LKB1-deficient, cells. Proteomic analysis revealed that the MAPK/MEK/ERK and PI3K/AKT pathways were activated in response to 8-Cl-Ado treatment and targeting these pathways enhanced the antitumor efficacy of 8-Cl-Ado. IMPLICATIONS: Together, our findings demonstrate that LKB1-deficient tumor cells are selectively sensitive to 8-Cl-Ado and suggest that therapeutic approaches targeting vulnerable energy stores combined with signaling pathway inhibitors merit further investigation for this patient population.
Collapse
Affiliation(s)
- Ana Galan-Cobo
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christine M Stellrecht
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
| | - Emrullah Yilmaz
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Chao Yang
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
| | - Yu Qian
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiao Qu
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Ishita Akhter
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mary L Ayres
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Youhong Fan
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pan Tong
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jie Ding
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Gastrointestinal Surgery, Guizhou Provincial People's Hospital, Guiyang, P.R. China
| | - Uma Giri
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jayanthi Gudikote
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Monique Nilsson
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ferdinandos Skoulidis
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John D Minna
- Hamon Center for Therapeutic Oncology Research and Simmons Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, Texas
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John V Heymach
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
10
|
Middleton G, Robbins H, Andre F, Swanton C. A state-of-the-art review of stratified medicine in cancer: towards a future precision medicine strategy in cancer. Ann Oncol 2022; 33:143-157. [PMID: 34808340 DOI: 10.1016/j.annonc.2021.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Building on the success of targeted therapy in certain well-defined cancer genotypes, three platform studies-NCI-MATCH, LUNG-MAP and The National Lung Matrix Trial (NLMT)-have attempted to discover new genotype-matched therapies for people with cancer. PATIENTS AND METHODS We review the outputs from these platform studies. This review led us to propose a series of recommendations and considerations that we hope will inform future precision medicine programmes in cancer. RESULTS The three studies collectively screened over 13 000 patients. Across 37 genotype-matched cohorts, there have been 66/875 responders, with an overall response rate of 7.5%. Targeting copy number gain yielded 5/199 responses across nine biomarker-drug matched cohorts, with a response rate of 2.5%. CONCLUSIONS The majority of these studies used single-agent targeted therapies. Whilst preclinical data can suggest rational combination treatment to reverse adaptive resistance or block parallel activated pathways, there is an essential need for accurate modelling of the toxicity-activity trade-off of combinations. Agent selection is often suboptimal; dose expansion should only be carried out with agents with clear clinical proof of mechanism and high target selectivity. Targeting copy number change has been disappointing; it is crucial to define the drivers on shared amplicons that include the targeted aberration. Maximising outcomes with currently available targeted therapies requires moving towards a more contextualised stratified medicine acknowledging the criticality of the genomic, transcriptional and immunological context on which the targeted aberration is inscribed. Genomic complexity and instability is likely to be a leading cause of targeted therapy failure in genomically complex cancers. Preclinical models must be developed that more accurately capture the genomic complexity of human disease. The degree of attrition of studies carried out after standard-of-care therapy suggests that serious efforts be made to develop a suite of precision medicine studies in the minimal residual disease setting.
Collapse
Affiliation(s)
- G Middleton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| | - H Robbins
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - F Andre
- Institut Gustave Roussy, INSERM Unité 981, Université Paris-Sud, Villejuif, France; PRISM Center for Precision Medicine
| | - C Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| |
Collapse
|
11
|
Zhou Y, Zhang Y, Li Y, Liu L, Li Z, Liu Y, Xiao Y. MicroRNA-106a-5p promotes the proliferation, autophagy and migration of lung adenocarcinoma cells by targeting LKB1/AMPK. Exp Ther Med 2021; 22:1422. [PMID: 34707704 PMCID: PMC8543179 DOI: 10.3892/etm.2021.10857] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/07/2021] [Indexed: 12/31/2022] Open
Abstract
It has previously been reported that lung cancer has the highest morbidity and mortality rate worldwide; however, the pathogenesis underlying lung cancer has not been fully elucidated. The aim of the present was primarily to assess the influence of microRNA (miR)-106a-5p on the biological behaviors of lung cancer cells. In the present study, bioinformatics analysis was used to analyze the expression characteristics of miR-106a-5p and its relationship with the prognosis of patients with lung adenocarcinoma (LUAD) in The Cancer Genome Atlas. A dual luciferase reporter assay was performed to verify the binding of miR-106a-5p and liver kinase B1 (LKB1). The Cell Counting Kit-8, colony formation and Transwell assays were utilized to detect cell viability, proliferation and migration, respectively. Protein and RNA expression levels were examined by western blotting and reverse transcription-quantitative PCR analysis, respectively. It was observed that miR-106a-5p was highly expressed in LUAD and associated with poor prognosis. miR-106a-5p promoted the proliferation and migration of LUAD cells, and inhibited autophagy. By contrast, LKB1 inhibited cell proliferation and migration, promoted autophagy and blocked the cancer-promoting effects of miR-106a-5p. Overexpression of miR-106a-5p inhibited the phosphorylation of AMP-activated protein kinase (AMPK) and tuberin (TSC2), and promoted the phosphorylation of mTOR. By contrast, overexpression of LKB1 blocked the promotion of mTOR phosphorylation, and the inhibition of AMPK and TSC2 phosphorylation caused by miR-106a-5p. In summary, the results of the present study indicated that miR-106a-5p regulated the phosphorylation of the AMPK pathway by targeting LKB1, and was involved in the proliferation, migration and autophagy of LUAD cells.
Collapse
Affiliation(s)
- Yushan Zhou
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Yuxuan Zhang
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Yanli Li
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Liqiong Liu
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Zhidong Li
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Yanhong Liu
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Yi Xiao
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| |
Collapse
|
12
|
Sun CY, Li YZ, Cao D, Zhou YF, Zhang MY, Wang HY. Rapamycin and trametinib: a rational combination for treatment of NSCLC. Int J Biol Sci 2021; 17:3211-3223. [PMID: 34421360 PMCID: PMC8375233 DOI: 10.7150/ijbs.62752] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/11/2021] [Indexed: 02/02/2023] Open
Abstract
Mammalian target of rapamycin (mTOR) is one of the most commonly activated pathways in human cancers, including lung cancer. Targeting mTOR with molecule inhibitors is considered as a useful therapeutic strategy. However, the results obtained from the clinical trials with the inhibitors so far have not met the original expectations, largely because of the drug resistance. Thus, combined or multiple drug therapy can bring about more favorable clinical outcomes. Here, we found that activation of ERK pathway was responsible for rapamycin drug resistance in non-small-cell lung cancer (NSCLC) cells. Accordingly, rapamycin-resistant NSCLC cells were more sensitive to ERK inhibitor (ERKi), trametinib, and in turn, trametinib-resistant NSCLC cells were also susceptible to rapamycin. Combining rapamycin with trametinib led to a potent synergistic antitumor efficacy, which induced G1-phase cycle arrest and apoptosis. In addition, rapamycin synergized with another ERKi, MEK162, and in turn, trametinib synergized with other mTORi, Torin1 and OSI-027. Mechanistically, rapamycin in combination with trametinib resulted in a greater decrease of phosphorylation of AKT, ERK, mTOR and 4EBP1. In xenograft mouse model, co-administration of rapamycin and trametinib caused a substantial suppression in tumor growth without obvious drug toxicity. Overall, our study identifies a reasonable combined strategy for treatment of NSCLC.
Collapse
Affiliation(s)
- Chao-Yue Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, China 510060
| | - Yi-Zhuo Li
- Department of Medical Imaging, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, China 510060
| | - Di Cao
- Department of Medical Imaging, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, China 510060
| | - Yu-Feng Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, China 510060
| | - Mei-Yin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, China 510060
| | - Hui-Yun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, China 510060
| |
Collapse
|
13
|
Zhang T, Zhang L, Payne PRO, Li F. Synergistic Drug Combination Prediction by Integrating Multiomics Data in Deep Learning Models. Methods Mol Biol 2021; 2194:223-238. [PMID: 32926369 DOI: 10.1007/978-1-0716-0849-4_12] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Intrinsic and acquired drug resistance is a major challenge in cancer therapy. Synergistic drug combinations could help to overcome drug resistance. However, the number of possible drug combinations is enormous, and it is infeasible to experimentally screen all drug combinations with limited resources. Therefore, computational models to predict and prioritize effective drug combinations are important for combination therapy discovery. Compared with existing models, we propose a novel deep learning model, AuDNNsynergy, to predict the synergy of pairwise drug combinations by integrating multiomics data. Specifically, three autoencoders are trained using the gene expression, copy number, and genetic mutation data of tumor samples from The Cancer Genome Atlas (TCGA). Then the gene expression, copy number, and mutation of individual cancer cell lines are coded using the three trained autoencoders. The physicochemical features of individual drugs and the encoded omics data of individual cancer cell lines are used as the input features of a deep neural network that predicts the synergy score of given pairwise drug combinations against the specific cancer cell lines. The comparison results showed the proposed AuDNNsynergy model outperforms, specifically in terms of rank correlation metric, four state-of-the-art approaches, namely, DeepSynergy, Gradient Boosting Machines, Random Forests, and Elastic Nets.
Collapse
Affiliation(s)
- Tianyu Zhang
- Institute for Informatics (I2), Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.,School of Mathematical Science, Dalian University of Technology, Dalian, Liaoning, China
| | - Liwei Zhang
- School of Mathematical Science, Dalian University of Technology, Dalian, Liaoning, China
| | - Philip R O Payne
- Institute for Informatics (I2), Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Fuhai Li
- Institute for Informatics (I2), Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA. .,Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
14
|
Zhang H, Feng J, Zeng A, Payne P, Li F. Predicting Tumor Cell Response to Synergistic Drug Combinations Using a Novel Simplified Deep Learning Model. AMIA ... ANNUAL SYMPOSIUM PROCEEDINGS. AMIA SYMPOSIUM 2021; 2020:1364-1372. [PMID: 33936513 PMCID: PMC8075535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Drug combinations targeting multiple targets/pathways are believed to be able to reduce drug resistance. Computational models are essential for novel drug combination discovery. In this study, we proposed a new simplified deep learning model, DeepSignalingSynergy, for drug combination prediction. Compared with existing models that use a large number of chemical-structure and genomics features in densely connected layers, we built the model on a small set of cancer signaling pathways, which can mimic the integration of multi-omics data and drug target/mechanism in a more biological meaningful and explainable manner. The evaluation results of the model using the NCI ALMANAC drug combination screening data indicated the feasibility of drug combination prediction using a small set of signaling pathways. Interestingly, the model analysis suggested the importance of heterogeneity of the 46 signaling pathways, which indicates that some new signaling pathways should be targeted to discover novel synergistic drug combinations.
Collapse
Affiliation(s)
- Heming Zhang
- Institute for Informatics (I2), Washington University School of Medicine
- Computer Science
| | - Jiarui Feng
- Institute for Informatics (I2), Washington University School of Medicine
- Data science
| | - Amanda Zeng
- Institute for Informatics (I2), Washington University School of Medicine
- Data science
| | - Philip Payne
- Institute for Informatics (I2), Washington University School of Medicine
| | - Fuhai Li
- Institute for Informatics (I2), Washington University School of Medicine
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Correspondence:
| |
Collapse
|
15
|
Yang C, Ding H, Yang Y, Yang L, Yang Y, Fang M, Ren J, Hu R, Wang C, Geng W. BAP1 regulates AMPK-mTOR signalling pathway through deubiquitinating and stabilizing tumour-suppressor LKB1. Biochem Biophys Res Commun 2020; 529:1025-1032. [PMID: 32819560 DOI: 10.1016/j.bbrc.2020.05.223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 05/31/2020] [Indexed: 12/11/2022]
Abstract
Liver kinase B1 (LKB1), a tumour suppressor, participates in many cellular processes, including cell survival, growth, apoptosis, transformation, and metabolism. Upon performing yeast two-hybrid screening, co-immunoprecipitation, and GST pull-down, we identified that BRCA1-associated protein 1 (BAP1), a deubiquitinase, interacts with LKB1. Immunoblotting was performed to examine the effect of BAP1 on the activation of 5' AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR), downstream of LKB1. The relationship between BAP1 deficiency and cancer cell proliferation was examined using cell survival assay and soft agar assay. qRT-PCR and oil red O staining were performed to evaluate lipid synthesis. Our findings reveal that BAP1 deubiquitinates LKB1, inhibits its degradation, and stabilises it, thereby affecting AMPK activation and downstream mTOR activity. BAP1 deficiency may enhance cellular proliferation as well as lipid synthesis.
Collapse
Affiliation(s)
- Cong Yang
- Guizhou University School of Medicine, Guiyang, 550025, China
| | - Hongyu Ding
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yang Yang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Long Yang
- Guizhou University School of Medicine, Guiyang, 550025, China
| | - Yun Yang
- Guizhou University School of Medicine, Guiyang, 550025, China
| | - Meimiao Fang
- Guizhou University School of Medicine, Guiyang, 550025, China
| | - Jin Ren
- Guizhou University School of Medicine, Guiyang, 550025, China
| | - Ronggui Hu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Chengcheng Wang
- Guizhou University School of Medicine, Guiyang, 550025, China; Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Wujun Geng
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
16
|
Caiola E, Iezzi A, Tomanelli M, Bonaldi E, Scagliotti A, Colombo M, Guffanti F, Micotti E, Garassino MC, Minoli L, Scanziani E, Broggini M, Marabese M. LKB1 Deficiency Renders NSCLC Cells Sensitive to ERK Inhibitors. J Thorac Oncol 2019; 15:360-370. [PMID: 31634668 DOI: 10.1016/j.jtho.2019.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 10/07/2019] [Accepted: 10/12/2019] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Serine/threonine kinase 11 (LKB1/STK11) is one of the most mutated genes in NSCLC accounting for approximately one-third of cases and its activity is impaired in approximately half of KRAS-mutated NSCLC. At present, these patients cannot benefit from any specific therapy. METHODS Through CRISPR/Cas9 technology, we systematically deleted LKB1 in both wild-type (WT) and KRAS-mutated human NSCLC cells. By using these isogenic systems together with genetically engineered mouse models we investigated the cell response to ERK inhibitors both in vitro and in vivo. RESULTS In all the systems used here, the loss of LKB1 creates vulnerability and renders these cells particularly sensitive to ERK inhibitors both in vitro and in vivo. The same cells expressing a WT LKB1 poorly respond to these drugs. At the molecular level, in the absence of LKB1, ERK inhibitors induced a marked inhibition of p90 ribosomal S6 kinase activation, which in turn abolished S6 protein activation, promoting the cytotoxic effect. CONCLUSIONS This work shows that ERK inhibitors are effective in LKB1 and LKB1/KRAS-mutated tumors, thus offering a therapeutic strategy for this prognostically unfavorable subgroup of patients. Because ERK inhibitors are already in clinical development, our findings could be easily translatable to the clinic. Importantly, the lack of effect in cells expressing WT LKB1, predicts that treatment of LKB1-mutated tumors with ERK inhibitors should have a favorable toxicity profile.
Collapse
Affiliation(s)
- Elisa Caiola
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Alice Iezzi
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Michele Tomanelli
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elisa Bonaldi
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Arianna Scagliotti
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marika Colombo
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Federica Guffanti
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Edoardo Micotti
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Lucia Minoli
- Mouse & Animal Pathology Lab, Fondazione Filarete, Milan, Italy; Department of Veterinary Medicine, University of Milan, Milan, Italy
| | - Eugenio Scanziani
- Mouse & Animal Pathology Lab, Fondazione Filarete, Milan, Italy; Department of Veterinary Medicine, University of Milan, Milan, Italy
| | - Massimo Broggini
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - Mirko Marabese
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
17
|
Li X, Tang X, Su J, Xu G, Zhao L, Qi Q. Involvement of E-cadherin/AMPK/mTOR axis in LKB1-induced sensitivity of non-small cell lung cancer to gambogic acid. Biochem Pharmacol 2019; 169:113635. [PMID: 31513784 DOI: 10.1016/j.bcp.2019.113635] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/05/2019] [Indexed: 12/24/2022]
Abstract
Liver kinase B1 (LKB1) is a tumor suppressor that functions as master regulator of cell growth, metabolism, survival, and polarity. Patients with NSCLC possessing mutated LKB1 respond to chemotherapy differently from those with wild-type LKB1. Gambogic acid (GA), a small molecule from natural product, has been established as an anti-tumor agent due to its potent activity and low toxicity. Here, we find out that NSCLC cells with wild-type LKB1 are more sensitive to GA in vitro and in vivo. Mechanistic studies pinpoint that the selective inhibition of mTOR signaling confers the stronger suppression of NSCLC in presence of wild-type LKB1, which is involved in the enhancement of p-AMPK. Further studies reveal that GA increases p-AMPK levels through up-regulation of E-cadherin associated with LKB1. In addition, induction of E-cadherin by GA may be through down-regulation of ZEB1, which is independent with LKB1 status. Hence, our findings support that enhanced E-cadherin by GA cooperates LKB1, leading to up-regulation of p-AMPK, and thus blocking of mTOR signaling pathway, which provide theoretical foundation for utilization of GA as a potential targeted drug against NSCLC harboring wild-type LKB1.
Collapse
Affiliation(s)
- Xiaosu Li
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, PR China
| | - Xueyi Tang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, PR China
| | - Jin Su
- Department of Pharmacology, Clinical Translational Center for Targeted Drug, School of Medicine, Jinan University, Guangzhou 510632, Guangdong Province, PR China
| | - Guofang Xu
- Zhengzhou People's Hospital, Zhengzhou 450003, Henan Province, PR China
| | - Limin Zhao
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, PR China.
| | - Qi Qi
- Department of Pharmacology, Clinical Translational Center for Targeted Drug, School of Medicine, Jinan University, Guangzhou 510632, Guangdong Province, PR China.
| |
Collapse
|