1
|
Toma C, Popa R, Ciobanu L, Baldea I, Amorim I, Bochynska D, Wolfe A, Negoescu A, Gal C, Taulescu M. Overexpression of IL-6 and STAT3 may provide new insights into ovine pulmonary adenocarcinoma development. BMC Vet Res 2025; 21:29. [PMID: 39833798 PMCID: PMC11744984 DOI: 10.1186/s12917-024-04429-6] [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: 11/14/2022] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Ovine pulmonary adenocarcinoma (OPA) is caused by Jaagsiekte sheep retrovirus (JSRV) and is considered an important potential animal model for human lung cancer. The precise mechanisms of OPA oncogenesis are still uncertain. The transcription factor signal transducer and activator of transcription 3 (STAT3) is activated by interleukin-6 (IL-6) in many cancers, but this aspect is unknown in OPA. We therefore aimed to evaluate the expression of IL-6 and STAT3 in OPA for its potential role in pulmonary carcinogenesis. RESULTS Lung tissues from 9 grossly normal and JRSV-negative sheep and 20 cases of JSRV-positive OPA sheep were included in the study. Tissue samples were stained with antibodies against IL-6, STAT3, and JSRV-MA. IL-6 and STAT3 were further quantified in both groups using Western Blot (WB). Immunohistochemically, IL‑6 was expressed in stromal, inflammatory, and epithelial cells in all cases of OPA, while STAT3 immunoexpression was restricted to epithelial cells. In the OPA group, the percentage of immunolabelled cells for STAT3 accounted for a mean value of 96%. Using the H-SCORE method, 95% of cases were considered positive for STAT3 expression. Control tissues showed multifocal and weak immunoexpression for both markers. Using WB analyses, a highly significant amount of both IL-6 (p = 0.0078) and STAT3 (p < 0.0001) proteins were present in lung neoplasms, by comparison to the control lungs. CONCLUSIONS Our data showed overexpression of IL-6 and STAT3 in lung tissues from OPA compared to lungs from JSRV-negative sheep. These results suggest a potential role of IL6-STAT3 in OPA carcinogenesis.
Collapse
Affiliation(s)
- Corina Toma
- Department of Veterinary Pathology, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania.
| | - Roxana Popa
- Department of Veterinary Pathology, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Lidia Ciobanu
- Regional Institute of Gastroenterology and Hepatology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Cluj-Napoca, Romania
| | - Ioana Baldea
- Department of Physiology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj- Napoca, Romania
| | - Irina Amorim
- Department of Pathology and Molecular Immunology of the Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal
| | - Diana Bochynska
- Ross University School of Veterinary Medicine, Basseterre, St. Kitts and Nevis
| | - Alan Wolfe
- Pathobiology Section, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Andrada Negoescu
- Department of Veterinary Pathology, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Claudiu Gal
- Department of Veterinary Pathology, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
- Synevovet laboratory, Bucharest, Romania
| | - Marian Taulescu
- Department of Veterinary Pathology, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| |
Collapse
|
2
|
Bhuia MS, Chowdhury R, Afroz M, Akbor MS, Al Hasan MS, Ferdous J, Hasan R, de Alencar MVOB, Mubarak MS, Islam MT. Therapeutic Efficacy Studies on the Monoterpenoid Hinokitiol in the Treatment of Different Types of Cancer. Chem Biodivers 2025:e202401904. [PMID: 39776341 DOI: 10.1002/cbdv.202401904] [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: 08/06/2024] [Revised: 12/10/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025]
Abstract
Hinokitiol (HK), a monoterpenoid that naturally occurs in plants belonging to the Cupressaceae family, possesses important biological activities, including an anticancer effect. This review summarizes its anticancer potential and draws possible molecular interventions. In addition, it evaluates the biopharmaceutical, toxicological properties, and clinical application of HK to establish its viability for future advancement as a dependable anticancer medication. The assessment is based on the most recent information available from various databases. Findings demonstrate that HK possesses substantial therapeutic advantages against diverse types of cancer (colon, cervical, breast, bone, endometrial, liver, prostate, oral, and skin) through various molecular mechanisms. HK induces oxidative stress, cytotoxicity, apoptosis, cell-cycle arrest at the G and S phases, and autophagy through modulation of phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), p38/ERK/MAPK, nuclear factor kappa B, and c-Jun N-terminal kinase signaling pathways. Furthermore, this compound exhibits good oral bioavailability with excellent plasma clearance. Clinical uses of HK demonstrate therapeutic advantages without any significant negative effects. A thorough study of the pertinent data suggests that HK may serve as a viable candidate for developing novel cancer therapies. Consequently, more extensive studies are necessary to evaluate its cancer treatment efficacy, safety, and possible long-term hazards.
Collapse
Affiliation(s)
- Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Meher Afroz
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
| | - Md Showkot Akbor
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
| | - Md Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Jannatul Ferdous
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
| | - Rubel Hasan
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | | | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, Dhaka, Bangladesh
| |
Collapse
|
3
|
Ahmad S, Bano N, Khanna K, Gupta D, Raza K. Reporting multitargeted potency of Tiaprofenic acid against lung cancer: Molecular fingerprinting, MD simulation, and MTT-based cell viability assay studies. Int J Biol Macromol 2024; 276:133872. [PMID: 39019378 DOI: 10.1016/j.ijbiomac.2024.133872] [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: 05/21/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Lung Cancer (LC) is among the most death-causing cancers, has caused the most destruction and is a gender-neutral cancer, and WHO has kept this cancer on its priority list to find the cure. We have used high-throughput virtual screening, standard precision docking, and extra precise docking for extensive screening of Drug Bank compounds, and the uniqueness of this study is that it considers multiple protein targets of prognosis and metastasis of LC. The docking and MM\GBSA calculation scores for the Tiaprofenic acid (DB01600) against all ten proteins range from -8.422 to -5.727 kcal/mol and - 47.43 to -25.72 kcal/mol, respectively. Also, molecular fingerprinting helped us to understand the interaction pattern of Tiaprofenic acid among all the proteins. Further, we extended our analysis to the molecular dynamic simulation in a neutralised SPC water medium for 100 ns. We analysed the root mean square deviation, fluctuations, and simulative interactions among the protein, ligand, water molecules, and protein-ligand complexes. Most complexes have shown a deviation of <2 Å as cumulative understanding. Also, the fluctuations were lesser, and only a few residues showed the fluctuation with a huge web of interaction between the protein and ligand, providing an edge that supports that the protein and ligand complexes were stable. In the MTT-based Cell Viability Assay, Tiaprofenic Acid exhibited concentration-dependent anti-cancer efficacy against A549 lung cancer cells, significantly reducing viability at 100 μg/mL. These findings highlight its potential as a therapeutic candidate, urging further exploration into the underlying molecular mechanisms for lung cancer treatment.
Collapse
Affiliation(s)
- Shaban Ahmad
- Computational Intelligence and Bioinformatics Lab, Department of Computer Science, Jamia Millia Islamia, New Delhi 110025, India.
| | - Nagmi Bano
- Computational Intelligence and Bioinformatics Lab, Department of Computer Science, Jamia Millia Islamia, New Delhi 110025, India; Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India.
| | - Kushagra Khanna
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia.
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India.
| | - Khalid Raza
- Computational Intelligence and Bioinformatics Lab, Department of Computer Science, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
4
|
Siddiquee T, Bhaskaran NA, Nathani K, Sawarkar SP. Empowering lung cancer treatment: Harnessing the potential of natural phytoconstituent-loaded nanoparticles. Phytother Res 2024. [PMID: 38806412 DOI: 10.1002/ptr.8241] [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: 10/16/2023] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024]
Abstract
Lung cancer, the second leading cause of cancer-related deaths, accounts for a substantial portion, representing 18.4% of all cancer fatalities. Despite advances in treatment modalities such as chemotherapy, surgery, and immunotherapy, significant challenges persist, including chemoresistance, non-specific targeting, and adverse effects. Consequently, there is an urgent need for innovative therapeutic approaches to overcome these limitations. Natural compounds, particularly phytoconstituents, have emerged as promising candidates due to their potent anticancer properties and relatively low incidence of adverse effects compared to conventional treatments. However, inherent challenges such as poor solubility, rapid metabolism, and enzymatic degradation hinder their clinical utility. To address these obstacles, researchers have increasingly turned to nanotechnology-based drug delivery systems (DDS). Nanocarriers offer several advantages, including enhanced drug stability, prolonged circulation time, and targeted delivery to tumor sites, thereby minimizing off-target effects. By encapsulating phytoconstituents within nanocarriers, researchers aim to optimize their bioavailability and therapeutic efficacy while reducing systemic toxicity. Moreover, the integration of nanotechnology with phytoconstituents allows for a nuanced understanding of the intricate molecular pathways involved in lung cancer pathogenesis. This integrated approach holds promise for modulating key cellular processes implicated in tumor growth and progression. Additionally, by leveraging the synergistic effects of phytoconstituents and nanocarriers, researchers seek to develop tailored therapeutic strategies that maximize efficacy while minimizing adverse effects. In conclusion, the integration of phytoconstituents with nanocarriers represents a promising avenue for advancing lung cancer treatment. This synergistic approach has the potential to revolutionize current therapeutic paradigms by offering targeted, efficient, and minimally toxic interventions. Continued research in this field holds the promise of improving patient outcomes and addressing unmet clinical needs in lung cancer management.
Collapse
Affiliation(s)
- Taufique Siddiquee
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | - Navya Ajitkumar Bhaskaran
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | - Khushali Nathani
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | - Sujata P Sawarkar
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| |
Collapse
|
5
|
Lv X, Yang L, Xie Y, Momeni MR. Non-coding RNAs and exosomal non-coding RNAs in lung cancer: insights into their functions. Front Cell Dev Biol 2024; 12:1397788. [PMID: 38859962 PMCID: PMC11163066 DOI: 10.3389/fcell.2024.1397788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/02/2024] [Indexed: 06/12/2024] Open
Abstract
Lung cancer is the second most common form of cancer worldwide Research points to the pivotal role of non-coding RNAs (ncRNAs) in controlling and managing the pathology by controlling essential pathways. ncRNAs have all been identified as being either up- or downregulated among individuals suffering from lung cancer thus hinting that they may play a role in either promoting or suppressing the spread of the disease. Several ncRNAs could be effective non-invasive biomarkers to diagnose or even serve as effective treatment options for those with lung cancer, and several molecules have emerged as potential targets of interest. Given that ncRNAs are contained in exosomes and are implicated in the development and progression of the malady. Herein, we have summarized the role of ncRNAs in lung cancer. Moreover, we highlight the role of exosomal ncRNAs in lung cancer.
Collapse
Affiliation(s)
- Xiaolong Lv
- Department of Cardiothoracic Surgery, The People’s Hospital of Changshou, Chongqing, China
| | - Lei Yang
- Department of Cardiothoracic Surgery, The People’s Hospital of Tongliang District, Chongqing, China
| | - Yunbo Xie
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | |
Collapse
|
6
|
Zhao W, Liu Y, Yang Y, Wang L. New link between RNH1 and E2F1: regulates the development of lung adenocarcinoma. BMC Cancer 2024; 24:635. [PMID: 38783241 PMCID: PMC11118993 DOI: 10.1186/s12885-024-12392-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a non-small cell carcinoma. Ribonuclease/angiogenin inhibitor 1 (RNH1) exerts multiple roles in virous cancers. E2F1 is a critical transcription factor involved in the LUAD development. Here, we analyze the expression of RNH1 in LUAD patients, investigate the biological function of RNH1 in LUAD, and demonstrate its potential mechanisms through E2F1 in LUAD. METHODS In the present study, we presented the expression of RNH1 in LUAD based on the database and confirmed it by western blot detection of RNH1 in human LUAD tissues. Lentiviral infection was constructed to silence or overexpress RNH1 in NCI-H1395 and NCI-H1437 cells. We assess the role of RNH1 on proliferation in LUAD cells by MTT assay, colony formation assays, and cell cycle detection. Hoechst staining and flow cytometry were used to evaluate the effects of RNH1 on apoptosis of LUAD cells. The function of RNH1 in invasion and migration was investigated by Transwell assay. Dual luciferase assay, ChIP detection, and pull-down assay were conducted to explore the association of E2F1 in the maintenance of RNH1 expression and function. The regulation of E2F1 on the functions of RNH1 in LUAD cells was explored. Mouse experiments were performed to confirm the in-vivo role of RNH1 in LUAD. mRNA sequencing indicated that RNH1 overexpression altered the expression profile of LUAD cells. RESULTS RNH1 expression in LUAD tissues of patients was presented in this work. Importantly, RNH1 knockdown improved the proliferation, migration and invasion abilities of cells and RNH1 overexpression produced the opposite effects. Dual luciferase assay proved that E2F1 bound to the RNH1 promoter (-1064 ∼ -1054, -1514 ∼ -1504) to reduce the transcriptional activity of RNH1. ChIP assay indicated that E2F1 DNA was enriched at the RNH1 promoter (-1148 ∼ -943, -1628 ∼ -1423). Pull-down assays also showed the association between E2F1 and RNH1 promoter (-1148 ∼ -943). E2F1 overexpression contributed to the malignant behavior of LUAD cells, while RNH1 overexpression reversed it. High-throughput sequencing showed that RNH1 overexpression induced multiple genes expression changes, thereby modulating LUAD-related processes. CONCLUSION Our study demonstrates that binding of E2F1 to the RNH1 promoter may lead to inhibition of RNH1 expression and thus promoting the development of LUAD.
Collapse
Affiliation(s)
- Wenyue Zhao
- Department of Thoracic Surgery, The First Hospital of China Medical University, 155# Nanjing North Street, Shenyang, Liaoning, China
| | - Yang Liu
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ying Yang
- Department of Operating Room, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liming Wang
- Department of Thoracic Surgery, The First Hospital of China Medical University, 155# Nanjing North Street, Shenyang, Liaoning, China.
| |
Collapse
|
7
|
Gomatou G, Masaoutis C, Vamvakaris I, Kotteas E, Bouros E, Tzilas V, Bouros D. Differential immunohistochemical expression of hTERT in lung cancer patients with and without idiopathic pulmonary fibrosis. Pulmonology 2024; 30:214-221. [PMID: 35153179 DOI: 10.1016/j.pulmoe.2021.12.001] [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: 10/04/2021] [Revised: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Human telomerase reverse transcriptase (hTERT) is the catalytic subunit of telomerase enzyme, which adds nucleotides to telomeres and counteracts their length shortening. The development of a telomere maintenance mechanism represents a hallmark of cancer. On the other hand, idiopathic pulmonary fibrosis (IPF) is associated with mutations in telomerase genes and shorter telomeres. IPF is frequently complicated with lung cancer. AIM To investigate the expression of hTERT in lung cancer with co-existing IPF and to compare with lung cancer without fibrosis. METHODS Diagnostic lung cancerous biopsies were retrieved from 18 patients with lung cancer and concomitant IPF, as well as 18 age and gender matched controls with lung cancer without pulmonary fibrosis. The expression of hTERT was studied with immunohistochemistry. ImajeJ software was used to quantitate subcellular stain intensity. Immunohistochemical investigation of two senescence-associated markers, p16 and p21, was also performed in all 36 cases. RESULTS Both groups highly expressed hTERT, without significant difference (100% vs 95%, p = 0.521). Evaluation of p16 and p21 immunostaining revealed negative to minimal immunoreactivity in both groups. hTERT localization exhibited higher median nuclear intensity in the group of lung cancer with IPF (0.62 vs 0.45, p = 0.016), while cytoplasmic intensity did not differ significantly (0.17 vs 0.15, p = 0.463). Higher median nuclear intensity was also correlated with small cell lung cancer subtype in the whole study sample (0.69 vs 0.45, p = 0.09). CONCLUSION hTERT is highly expressed in lung cancer with concomitant IPF, but with differential localization compared to lung cancer without IPF, implying differences in pathogenicity and requiring further investigation.
Collapse
Affiliation(s)
- G Gomatou
- Interstitial Lung Diseases Unit, 1st Department of Respiratory Medicine, "Sotiria" Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, Athens, Greece; Oncology Unit, Third Department of Medicine, "Sotiria" Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, Athens, Greece.
| | - C Masaoutis
- 1st Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - I Vamvakaris
- Department of Pathology, "Sotiria" Hospital for Diseases of the Chest, Athens, Greece
| | - E Kotteas
- Oncology Unit, Third Department of Medicine, "Sotiria" Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, Athens, Greece
| | - E Bouros
- Interstitial Lung Diseases Unit, 1st Department of Respiratory Medicine, "Sotiria" Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, Athens, Greece
| | - V Tzilas
- Center for Diseases of the Chest, Athens Medical Center, Athens, Greece
| | - D Bouros
- Interstitial Lung Diseases Unit, 1st Department of Respiratory Medicine, "Sotiria" Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, Athens, Greece; Center for Diseases of the Chest, Athens Medical Center, Athens, Greece
| |
Collapse
|
8
|
Lou J, Chu X, Yang X, Jamil M, Zhu H. Deciphering DNA repair gene mutational landscape in uterine corpus endometrial carcinoma patients using next generation sequencing. Am J Cancer Res 2024; 14:210-226. [PMID: 38323278 PMCID: PMC10839304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/25/2023] [Indexed: 02/08/2024] Open
Abstract
Uterine Corpus Endometrial Carcinoma (UCEC) is a significant health concern with a complex genetic landscape impacting disease susceptibility and progression. This study aimed to unravel the spectrum of DNA repair gene mutations in Pakistani UCEC patients through Next Generation Sequencing (NGS) and explore their potential functional consequences via downstream analyses. NGS analysis of genomic DNA from 30 UCEC patients was conducted to identify clinically significant pathogenic mutations in DNA repair genes. This analysis revealed mutations in 4 key DNA repair genes: BRCA1, BRCA2, APC, and CDH1. Kaplan-Meier (KM) analysis was employed to assess the prognostic value of these mutations on patient overall survival (OS) in UCEC. To delve into the functional impact of these mutations, we performed RT-qPCR, immunohistochemistry (IHC), and western blot analyses on the mutated UCEC samples compared to their non-mutated counterparts. These results unveiled the up-regulation in the expression of the mutated genes, suggesting a potential association between the identified mutations and enhanced gene activity. Additionally, targeted bisulfite sequencing analysis was utilized to evaluate DNA methylation patterns in the promoters of the mutated genes. Strikingly, hypomethylation in the promoters of BRCA1, BRCA2, APC, and CDH1 was observed in the mutated UCEC samples relative to the non-mutated, indicating the involvement of epigenetic mechanisms in the altered gene expression. In conclusion, this study offers insights into the genetic landscape of DNA repair gene mutations in Pakistani UCEC patients. The presence of pathogenic mutations in BRCA1, BRCA2, APC, and CDH1, coupled with their down-regulation and hypermethylation, suggests a convergence of genetic and epigenetic factors contributing to genomic instability in UCEC cells. These findings enhance our understanding of UCEC susceptibility and provide potential avenues for targeted therapeutic interventions in Pakistani UCEC patients.
Collapse
Affiliation(s)
- Jun Lou
- Department of Gynecological Oncology, Jiangxi Cancer HospitalNanchang 330029, Jiangxi, China
- The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Key Laboratory of Translational Research for CancerNanchang 330029, Jiangxi, China
| | - Xiaoyan Chu
- Department of Gynecological Oncology, Jiangxi Cancer HospitalNanchang 330029, Jiangxi, China
- The Second Affiliated Hospital of Nanchang Medical CollegeNanchang 330029, Jiangxi, China
| | - Xiaorong Yang
- Department of Gynecological Oncology, Jiangxi Cancer HospitalNanchang 330029, Jiangxi, China
- The Second Affiliated Hospital of Nanchang Medical CollegeNanchang 330029, Jiangxi, China
| | - Muhammad Jamil
- PARC Arid Zone Research CenterDera Ismail Khan 29050, Pakistan
| | - Hong Zhu
- Department of Gynecological Oncology, Jiangxi Cancer HospitalNanchang 330029, Jiangxi, China
- The Second Affiliated Hospital of Nanchang Medical CollegeNanchang 330029, Jiangxi, China
| |
Collapse
|
9
|
Srivastava S, Jayaswal N, Kumar S, Sharma PK, Behl T, Khalid A, Mohan S, Najmi A, Zoghebi K, Alhazmi HA. Unveiling the potential of proteomic and genetic signatures for precision therapeutics in lung cancer management. Cell Signal 2024; 113:110932. [PMID: 37866667 DOI: 10.1016/j.cellsig.2023.110932] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/15/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
Lung cancer's enduring global significance necessitates ongoing advancements in diagnostics and therapeutics. Recent spotlight on proteomic and genetic biomarker research offers a promising avenue for understanding lung cancer biology and guiding treatments. This review elucidates genetic and proteomic lung cancer biomarker progress and their treatment implications. Technological strides in mass spectrometry-based proteomics and next-generation sequencing enable pinpointing of genetic abnormalities and abnormal protein expressions, furnishing vital data for precise diagnosis, patient classification, and customized treatments. Biomarker-driven personalized medicine yields substantial treatment improvements, elevating survival rates and minimizing adverse effects. Integrating omics data (genomics, proteomics, etc.) enhances understanding of lung cancer's intricate biological milieu, identifying novel treatment targets and biomarkers, fostering precision medicine. Liquid biopsies, non-invasive tools for real-time treatment monitoring and early resistance detection, gain popularity, promising enhanced management and personalized therapy. Despite advancements, biomarker repeatability and validation challenges persist, necessitating interdisciplinary efforts and large-scale clinical trials. Integrating artificial intelligence and machine learning aids analyzing vast omics datasets and predicting treatment responses. Single-cell omics reveal cellular connections and intratumoral heterogeneity, valuable for combination treatments. Biomarkers enable accurate diagnosis, tailored medicines, and treatment response tracking, significantly impacting personalized lung cancer care. This approach spurs patient-centered trials, empowering active patient engagement. Lung cancer proteomic and genetic biomarkers illuminate disease biology and treatment prospects. Progressing towards individualized efficient therapies is imminent, alleviating lung cancer's burden through ongoing research, omics integration, and technological strides.
Collapse
Affiliation(s)
- Shriyansh Srivastava
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi 110017, India; Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India
| | - Nandani Jayaswal
- Accurate College of Pharmacy, 49, Knowledge Park-III, Greater Noida, UP, India
| | - Sachin Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi 110017, India
| | - Pramod Kumar Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Sahibzada Ajit Singh Nagar, Punjab, India.
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; Medicinal and Aromatic Plants Research Institute, National Center for Research, P.O. Box: 2424, Khartoum 11111, Sudan
| | - Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; Center for Global Health Research, Saveetha Medical College, and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India.
| | - Asim Najmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan, Saudi Arabia
| | - Khalid Zoghebi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan, Saudi Arabia
| | - Hassan A Alhazmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan, Saudi Arabia
| |
Collapse
|
10
|
Letelier P, Saldías R, Loren P, Riquelme I, Guzmán N. MicroRNAs as Potential Biomarkers of Environmental Exposure to Polycyclic Aromatic Hydrocarbons and Their Link with Inflammation and Lung Cancer. Int J Mol Sci 2023; 24:16984. [PMID: 38069307 PMCID: PMC10707120 DOI: 10.3390/ijms242316984] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 12/18/2023] Open
Abstract
Exposure to atmospheric air pollution containing volatile organic compounds such as polycyclic aromatic hydrocarbons (PAHs) has been shown to be a risk factor in the induction of lung inflammation and the initiation and progression of lung cancer. MicroRNAs (miRNAs) are small single-stranded non-coding RNA molecules of ~20-22 nucleotides that regulate different physiological processes, and their altered expression is implicated in various pathophysiological conditions. Recent studies have shown that the regulation of gene expression of miRNAs can be affected in diseases associated with outdoor air pollution, meaning they could also be useful as biomarkers of exposure to environmental pollution. In this article, we review the published evidence on miRNAs in relation to exposure to PAH pollution and discuss the possible mechanisms that may link these compounds with the expression of miRNAs.
Collapse
Affiliation(s)
- Pablo Letelier
- Laboratorio de Investigación en Salud de Precisión, Departamento de Procesos Diagnósticos y Evaluación, Facultad de Ciencias de la Salud, Universidad Católica de Temuco, Temuco 4813302, Chile; (R.S.); (N.G.)
| | - Rolando Saldías
- Laboratorio de Investigación en Salud de Precisión, Departamento de Procesos Diagnósticos y Evaluación, Facultad de Ciencias de la Salud, Universidad Católica de Temuco, Temuco 4813302, Chile; (R.S.); (N.G.)
| | - Pía Loren
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile;
| | - Ismael Riquelme
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Temuco 4810101, Chile;
| | - Neftalí Guzmán
- Laboratorio de Investigación en Salud de Precisión, Departamento de Procesos Diagnósticos y Evaluación, Facultad de Ciencias de la Salud, Universidad Católica de Temuco, Temuco 4813302, Chile; (R.S.); (N.G.)
| |
Collapse
|
11
|
Almalki WH. Beyond the genome: lncRNAs as regulators of the PI3K/AKT pathway in lung cancer. Pathol Res Pract 2023; 251:154852. [PMID: 37837857 DOI: 10.1016/j.prp.2023.154852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/18/2023] [Accepted: 10/02/2023] [Indexed: 10/16/2023]
Abstract
Lung cancer is a prevalent and devastating disease, representing a significant global health burden. Despite advancements in therapeutic strategies, the molecular mechanisms underlying its pathogenesis remain incompletely understood. Lung cancer typically displays the deregulated activity of the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) pathway, which is vital for cell proliferation, survival, and metastasis. Emerging evidence suggests that long non-coding RNA (lncRNAs) can modulate the PI3K/AKT pathway, offering new insights into lung cancer biology and potential therapeutic opportunities. These lncRNA act as either oncogenes, promoting pathway activation, or tumour suppressors, attenuating pathway signalling. The dysregulation of lncRNA is associated with various cellular processes, including apoptosis, cell cycle control, epithelial-mesenchymal transition (EMT), and angiogenesis, ultimately influencing lung cancer growth and metastasis. The development of novel therapeutic strategies, such as small interfering RNAs (siRNAs), antisense oligonucleotides, and CRISPR/Cas9-mediated gene editing, holds promise for restoring lncRNAs dysregulation and re-establishing the equilibrium of the PI3K/AKT pathway. The emerging role of lncRNAs as regulators of the PI3K/AKT pathway sheds new light on the complex molecular landscape of lung cancer. Understanding the interplay between lncRNA and the PI3K/AKT pathway could lead to the identification of novel biomarkers for prognosis and therapeutic targets for precision medicine. The potential of lncRNAs-based therapeutics may pave the way for more effective and personalized treatment approaches in lung cancer and potentially other malignancies with dysregulated PI3K/AKT signalling. This review aims to explore the emerging role of lncRNAs as key regulators of the PI3K/AKT pathway in lung cancer.
Collapse
Affiliation(s)
- Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| |
Collapse
|
12
|
Malik P, Rani R, Solanki R, Patel VH, Mukherjee TK. Understanding the feasibility of chemotherapeutic and immunotherapeutic targets against non-small cell lung cancers: an update of resistant responses and recent combinatorial therapies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:850-895. [PMID: 37970206 PMCID: PMC10645466 DOI: 10.37349/etat.2023.00171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 05/17/2023] [Indexed: 11/17/2023] Open
Abstract
Despite consistent progress in prompt diagnosis and curative therapies in the last decade, lung cancer (LC) continues to threaten mankind, accounting for nearly twice the casualties compared to prostate, breast, and other cancers. Statistics associate ~25% of 2021 cancer-related deaths with LC, more than 80% of which are explicitly caused by tobacco smoking. Prevailing as small and non-small cell pathologies, with respective occurring frequency of nearly 15% and 80-85%, non-small cell LCs (NSCLCs) are prominently distinguished into lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), subtypes. Since the first use of epidermal growth factor receptor (EGFR) inhibitor gefitinib for NSCLC treatment in 2002, immense progress has been made for targeted therapies with the next generation of drugs spanning across the chronological generations of small molecule inhibitors. The last two years have overseen the clinical approval of more than 10 therapeutic agents as first-line NSCLC medications. However, uncertain mutational aberrations as well as systemic resistant responses, and abysmal overall survival curtail the combating efficacies. Of late, immune checkpoint inhibitors (ICIs) against various molecules including programmed cell death-1 (PD-1) and its ligand (PD-L1) have been demonstrated as reliable LC treatment targets. Keeping these aspects in mind, this review article discusses the success of NSCLC chemo and immunotherapies with their characteristic effectiveness and future perspectives.
Collapse
Affiliation(s)
- Parth Malik
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Ruma Rani
- Indian Council of Agricultural Research (ICAR)-National Research Centre on Equines, Hisar 125001, Haryana, India
| | - Raghu Solanki
- School of Life Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | | | | |
Collapse
|
13
|
Karger A, Mansouri S, Leisegang MS, Weigert A, Günther S, Kuenne C, Wittig I, Zukunft S, Klatt S, Aliraj B, Klotz LV, Winter H, Mahavadi P, Fleming I, Ruppert C, Witte B, Alkoudmani I, Gattenlöhner S, Grimminger F, Seeger W, Pullamsetti SS, Savai R. ADPGK-AS1 long noncoding RNA switches macrophage metabolic and phenotypic state to promote lung cancer growth. EMBO J 2023; 42:e111620. [PMID: 37545364 PMCID: PMC10505917 DOI: 10.15252/embj.2022111620] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/06/2023] [Accepted: 07/08/2023] [Indexed: 08/08/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) influence the transcription of gene networks in many cell types, but their role in tumor-associated macrophages (TAMs) is still largely unknown. We found that the lncRNA ADPGK-AS1 was substantially upregulated in artificially induced M2-like human macrophages, macrophages exposed to lung cancer cells in vitro, and TAMs from human lung cancer tissue. ADPGK-AS1 is partly located within mitochondria and binds to the mitochondrial ribosomal protein MRPL35. Overexpression of ADPGK-AS1 in macrophages upregulates the tricarboxylic acid cycle and promotes mitochondrial fission, suggesting a phenotypic switch toward an M2-like, tumor-promoting cytokine release profile. Macrophage-specific knockdown of ADPGK-AS1 induces a metabolic and phenotypic switch (as judged by cytokine profile and production of reactive oxygen species) to a pro-inflammatory tumor-suppressive M1-like state, inhibiting lung tumor growth in vitro in tumor cell-macrophage cocultures, ex vivo in human tumor precision-cut lung slices, and in vivo in mice. Silencing ADPGK-AS1 in TAMs may thus offer a novel therapeutic strategy for lung cancer.
Collapse
Affiliation(s)
- Annika Karger
- Max Planck Institute for Heart and Lung ResearchMember of the German Center for Lung Research (DZL), Member of the Cardio‐Pulmonary Institute (CPI)Bad NauheimGermany
- Institute for Lung Health (ILH)Justus Liebig UniversityGiessenGermany
| | - Siavash Mansouri
- Max Planck Institute for Heart and Lung ResearchMember of the German Center for Lung Research (DZL), Member of the Cardio‐Pulmonary Institute (CPI)Bad NauheimGermany
- Institute for Lung Health (ILH)Justus Liebig UniversityGiessenGermany
| | - Matthias S Leisegang
- Institute for Cardiovascular Physiology, Medical FacultyGoethe University FrankfurtFrankfurtGermany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of MedicineGoethe University FrankfurtFrankfurtGermany
- Frankfurt Cancer Institute (FCI)Goethe University FrankfurtFrankfurtGermany
| | - Stefan Günther
- Max Planck Institute for Heart and Lung ResearchMember of the German Center for Lung Research (DZL), Member of the Cardio‐Pulmonary Institute (CPI)Bad NauheimGermany
| | - Carsten Kuenne
- Max Planck Institute for Heart and Lung ResearchMember of the German Center for Lung Research (DZL), Member of the Cardio‐Pulmonary Institute (CPI)Bad NauheimGermany
| | - Ilka Wittig
- Functional Proteomics, Medical SchoolGoethe University FrankfurtFrankfurtGermany
| | - Sven Zukunft
- Institute for Vascular Signalling, Centre for Molecular MedicineGoethe UniversityFrankfurtGermany
| | - Stephan Klatt
- Institute for Vascular Signalling, Centre for Molecular MedicineGoethe UniversityFrankfurtGermany
| | - Blerina Aliraj
- Institute of Biochemistry I, Faculty of MedicineGoethe University FrankfurtFrankfurtGermany
| | - Laura V Klotz
- Translational Lung Research Center (TLRC), Member of the DZLHeidelbergGermany
- Department of Thoracic SurgeryThoraxklinik at the University Hospital HeidelbergHeidelbergGermany
| | - Hauke Winter
- Translational Lung Research Center (TLRC), Member of the DZLHeidelbergGermany
- Department of Thoracic SurgeryThoraxklinik at the University Hospital HeidelbergHeidelbergGermany
| | - Poornima Mahavadi
- Department of Internal MedicineMember of the DZL, Member of CPI, Justus Liebig UniversityGiessenGermany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular MedicineGoethe UniversityFrankfurtGermany
| | - Clemens Ruppert
- Department of Internal MedicineMember of the DZL, Member of CPI, Justus Liebig UniversityGiessenGermany
| | - Biruta Witte
- Department of General and Thoracic SurgeryUniversity Hospital GiessenGiessenGermany
| | - Ibrahim Alkoudmani
- Department of General and Thoracic SurgeryUniversity Hospital GiessenGiessenGermany
| | | | - Friedrich Grimminger
- Institute for Lung Health (ILH)Justus Liebig UniversityGiessenGermany
- Department of Internal MedicineMember of the DZL, Member of CPI, Justus Liebig UniversityGiessenGermany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung ResearchMember of the German Center for Lung Research (DZL), Member of the Cardio‐Pulmonary Institute (CPI)Bad NauheimGermany
- Institute for Lung Health (ILH)Justus Liebig UniversityGiessenGermany
- Department of Internal MedicineMember of the DZL, Member of CPI, Justus Liebig UniversityGiessenGermany
| | - Soni Savai Pullamsetti
- Max Planck Institute for Heart and Lung ResearchMember of the German Center for Lung Research (DZL), Member of the Cardio‐Pulmonary Institute (CPI)Bad NauheimGermany
- Institute for Lung Health (ILH)Justus Liebig UniversityGiessenGermany
- Department of Internal MedicineMember of the DZL, Member of CPI, Justus Liebig UniversityGiessenGermany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung ResearchMember of the German Center for Lung Research (DZL), Member of the Cardio‐Pulmonary Institute (CPI)Bad NauheimGermany
- Institute for Lung Health (ILH)Justus Liebig UniversityGiessenGermany
- Frankfurt Cancer Institute (FCI)Goethe University FrankfurtFrankfurtGermany
- Department of Internal MedicineMember of the DZL, Member of CPI, Justus Liebig UniversityGiessenGermany
| |
Collapse
|
14
|
Hamouz M, Hammouz RY, Bajwa MA, Alsayed AW, Orzechowska M, Bednarek AK. A Functional Genomics Review of Non-Small-Cell Lung Cancer in Never Smokers. Int J Mol Sci 2023; 24:13314. [PMID: 37686122 PMCID: PMC10488233 DOI: 10.3390/ijms241713314] [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: 08/04/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
There is currently a dearth of information regarding lung cancer in never smokers (LCINS). Additionally, there is a difference in somatic mutations, tumour mutational burden, and chromosomal aberrations between smokers and never smokers (NS), insinuating a different disease entity in LCINS. A better understanding of actionable driver alterations prevalent in LCINS and the genomic landscape will contribute to identifying new molecular targets of relevance for NS that will drastically improve outcomes. Differences in treatment outcomes between NS and smokers, as well as sexes, with NSCLC suggest unique tumour characteristics. Epidermal growth factor receptor (EGFR) tyrosine kinase mutations and echinoderm microtubule-associated protein-like 4 anaplastic lymphoma kinase (EML4-ALK) gene rearrangements are more common in NS and have been associated with chemotherapy resistance. Moreover, NS are less likely to benefit from immune mediators including PD-L1. Unravelling the genomic and epigenomic underpinnings of LCINS will aid in the development of not only novel targeted therapies but also more refined approaches. This review encompasses driver genes and pathways involved in the pathogenesis of LCINS and a deeper exploration of the genomic landscape and tumour microenvironment. We highlight the dire need to define the genetic and environmental aspects entailing the development of lung cancer in NS.
Collapse
|
15
|
Schlensog M, Ruehlmann AC, Haeberle L, Opitz F, Becher AK, Goering W, Buth J, Knoefel WT, Ladage D, Meyer A, Esposito I. Tenascin-C affects invasiveness of EGFR-mutated lung adenocarcinoma through a putative paracrine loop. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166684. [PMID: 36878305 DOI: 10.1016/j.bbadis.2023.166684] [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: 09/12/2022] [Revised: 01/26/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
Tenascin C (TNC) is an extracellular matrix (ECM) protein and a potential biomarker affecting progression of different tumor types, such as pancreatic and lung cancer. Alternative splicing variants of TNC are known to have an impact on interaction partners like other ECM proteins or cell surface receptors, including epidermal growth factor receptor (EGFR), leading to numerous and sometimes opposite roles of TNC in tumor cell dissemination and proliferation. Only little is known about the impact of TNC on biologic characteristics of lung cancer, such as invasion and metastatic potential. In the present study, we could link an increased expression of TNC in lung adenocarcinoma (LUAD) tissues with an unfavorable clinical outcome of patients. Furthermore, we investigated the functional role of TNC in LUAD. Immunohistochemical staining of TNC revealed a significant increase of TNC levels in primary tumours and metastases compared to normal lung tissue. Additionally, a significant correlation between TNC mRNA expression and EGFR copy number and protein expression levels has been determined. Moreover, inhibition of TNC in lung fibroblasts led to reduced invasiveness of LUAD cells harboring EGFR-activating mutations and to a shorter lamellipodia perimeter and a reduced lamellipodia area on the surface of LUAD cells. This study provides the evidence that TNC expression might be a biological relevant factor in LUAD progression in an EGFR-dependent manner and that it regulates tumor cell invasion by rearrangement of the actin cytoskeleton, especially affecting lamellipodia formation.
Collapse
Affiliation(s)
- Martin Schlensog
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Ann-Cathrin Ruehlmann
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Lena Haeberle
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Friederike Opitz
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Ann-Kathrin Becher
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Wolfgang Goering
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Juliane Buth
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Wolfram Trudo Knoefel
- Department of General, Visceral and Pediatric Surgery, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Dennis Ladage
- Department of Pneumology, Kliniken Maria Hilf GmbH, Moenchengladbach, Germany
| | - Andreas Meyer
- Department of Pneumology, Kliniken Maria Hilf GmbH, Moenchengladbach, Germany
| | - Irene Esposito
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany.
| |
Collapse
|
16
|
Smok-Kalwat J, Mertowska P, Mertowski S, Smolak K, Kozińska A, Koszałka F, Kwaśniewski W, Grywalska E, Góźdź S. The Importance of the Immune System and Molecular Cell Signaling Pathways in the Pathogenesis and Progression of Lung Cancer. Int J Mol Sci 2023; 24:1506. [PMID: 36675020 PMCID: PMC9861992 DOI: 10.3390/ijms24021506] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/13/2023] Open
Abstract
Lung cancer is a disease that in recent years has become one of the greatest threats to modern society. Every year there are more and more new cases and the percentage of deaths caused by this type of cancer increases. Despite many studies, scientists are still looking for answers regarding the mechanisms of lung cancer development and progression, with particular emphasis on the role of the immune system. The aim of this literature review was to present the importance of disorders of the immune system and the accompanying changes at the level of cell signaling in the pathogenesis of lung cancer. The collected results showed that in the process of immunopathogenesis of almost all subtypes of lung cancer, changes in the tumor microenvironment, deregulation of immune checkpoints and abnormalities in cell signaling pathways are involved, which contribute to the multistage and multifaceted carcinogenesis of this type of cancer. We, therefore, suggest that in future studies, researchers should focus on a detailed analysis of tumor microenvironmental immune checkpoints, and to validate their validity, perform genetic polymorphism analyses in a wide range of patients and healthy individuals to determine the genetic susceptibility to lung cancer development. In addition, further research related to the analysis of the tumor microenvironment; immune system disorders, with a particular emphasis on immunological checkpoints and genetic differences may contribute to the development of new personalized therapies that improve the prognosis of patients.
Collapse
Affiliation(s)
- Jolanta Smok-Kalwat
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Konrad Smolak
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Aleksandra Kozińska
- Student Research Group of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Filip Koszałka
- Student Research Group of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Wojciech Kwaśniewski
- Department of Gynecologic Oncology and Gynecology, Medical University of Lublin, 20-081 Lublin, Poland
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Stanisław Góźdź
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| |
Collapse
|
17
|
Verma A, Yadav P, Rajput S, Verma S, Arora S, Kumar R, Bhatti JS, Khurana A, Navik U. ALK and ERBB2 Protein Inhibition is Involved in the Prevention of Lung Cancer Development by Vincamine. Anticancer Agents Med Chem 2023; 23:1587-1595. [PMID: 37046197 DOI: 10.2174/1871520623666230412102532] [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: 11/15/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND According to the WHO report of 2022, 2.21 million new cases and 1.80 million deaths were reported for lung cancer in the year 2020. Therefore, there is an urgent need to explore novel, safe, and effective therapeutic interventions for lung cancer. OBJECTIVE To find the potential targets of vincamine using a network pharmacology approach and docking studies and to evaluate the anti-cancer effect of vincamine on A549 cell line. METHODS Hence, in the present study, we explored the anti-cancer potential of vincamine by using network pharmacology, molecular docking, and in vitro approaches. Network pharmacology demonstrated that the most common targets of vincamine are G-protein coupled receptors, cytosolic proteins, and enzymes. Among these targets, two targets, ALK and ERBB2 protein, were common between vincamine and non-small cell lung cancer. RESULTS We discovered a link between these two targets and their companion proteins, as well as cancer-related pathways. In addition, a docking investigation between the ligand for vincamine and two targeted genes revealed a strong affinity toward these targeted proteins. Further, the in vitro study demonstrated that vincamine treatment for 72 h led to dosedependent (0-500 μM) cytotoxicity on the A549 lung cancer cell line with an IC50 value of 291.7 μΜ. The wound-healing assay showed that vincamine treatment (150 and 300 μM) significantly inhibited cell migration and invasion. Interestingly, acridine orange/ethidium bromide dual staining demonstrated that vincamine treatment induces apoptosis in A549 cells. Additionally, the dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay showed an increased level of reactive oxygen species (ROS) after the vincamine treatment, indicating ROS-mediated apoptosis in A549 cells. CONCLUSION Altogether, based on our findings, we hypothesize that vincamine-induced apoptosis of lung cancer cells via ALK and ERBB2 protein modulation may be an attractive futuristic strategy for managing lung cancer in combination with chemotherapeutic agents to obtain synergistic effects with reduced side effects.
Collapse
Affiliation(s)
- Aarti Verma
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - Sonu Rajput
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - Saloni Verma
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - Sahil Arora
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - Raj Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - Jasvinder Singh Bhatti
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Ghudda, Bathinda, 151401, India
| | - Amit Khurana
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelstrasse 30, D-52074, Aachen, Germany
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, India
| |
Collapse
|
18
|
Wei QY, Lau ATY, Mo HY, Zhong QH, Zhao XY, Yu FY, Han J, Wu YY, Xu YM. Effects of CYP3A43 Expression on Cell Proliferation and Migration of Lung Adenocarcinoma and Its Clinical Significance. Int J Mol Sci 2022; 24:ijms24010113. [PMID: 36613552 PMCID: PMC9820144 DOI: 10.3390/ijms24010113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/13/2022] [Accepted: 11/26/2022] [Indexed: 12/24/2022] Open
Abstract
The cytochrome P450s (CYP450s) include key oxidative enzymes involved in the metabolism of various carcinogens and anticancer drugs. Bioinformatic studies have demonstrated the association of CYP3A43 with liver cancer and ovarian cancer. However, the biological function of CYP3A43 in tumor progression remains unclear. To further reveal the role of CYP3A43 in tumor progression, we first analyzed the data from the UALCAN database and found that CYP3A43 was negatively correlated to the cancer staging and lymph node metastasis of lung adenocarcinoma (LUAD). We established stable CYP3A43-knockdown LUAD H1299 cell line and found that its knockdown enhanced cell proliferation, colony formation, and migration in vitro, and promoted the growth of tumor xenograft in vivo. Interestingly, when CYP3A43 was ectopically-expressed in the LUAD cell lines, decreased cell proliferation and ERK1/2 phosphorylation level were observed. Lastly, we also identified CYP3A43 co-expressed genes in LUAD from LinkedOmics database followed by GO and KEGG analyses. In conclusion, our results indicate the unprecedented role of CYP3A43 in the suppression of LUAD and provide new possibilities for targeted therapy of this life-threatening disease.
Collapse
Affiliation(s)
- Qi-Yao Wei
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Andy T. Y. Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Hai-Ying Mo
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Qiu-Hua Zhong
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Xiao-Yun Zhao
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Fei-Yuan Yu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Jin Han
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Yu-Yao Wu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
19
|
Alnajeebi AM, Alharbi HFH, Alelwani W, Babteen NA, Alansari WS, Shamlan G, Eskandrani AA. COVID-19 Candidate Genes and Pathways Potentially Share the Association with Lung Cancer. Comb Chem High Throughput Screen 2022; 25:2463-2472. [PMID: 34254909 DOI: 10.2174/1386207324666210712092649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 01/27/2023]
Abstract
COVID-19 is considered as the most challenging in the current situation but lung cancer is also the leading cause of death in the global population. These two malignancies are among the leading human diseases and are highly complex in terms of diagnostic and therapeutic approaches as well as the most frequent and highly complex and heterogeneous in nature. Based on the latest update, it is known that the patients suffering from lung cancer, are considered to be significantly at higher risk of COVID-19 infection in terms of survival and there are a number of evidences which support the hypothesis that these diseases may share the same functions and functional components. Multi-level unwanted alterations such as (epi-)genetic alterations, changes at the transcriptional level, and altered signaling pathways (receptor, cytoplasmic, and nuclear level) are the major sources which promote a number of complex diseases and such heterogeneous level of complexities are considered as the major barrier in the development of therapeutics. With so many challenges, it is critical to understand the relationships and the common shared aberrations between them which is difficult to unravel and understand. A simple approach has been applied for this study where differential gene expression analysis, pathway enrichment, and network level understanding are carried out. Since, gene expression changes and genomic alterations are related to the COVID-19 and lung cancer but their pattern varies significantly. Based on the recent studies, it appears that the patients suffering from lung cancer and and simultaneously infected with COVID-19, then survival chance is lessened. So, we have designed our goal to understand the genes commonly overexpressed and commonly enriched pathways in case of COVID-19 and lung cancer. For this purpose, we have presented the summarized review of the previous works where the pathogenesis of lung cancer and COVID-19 infection have been focused and we have also presented the new finding of our analysis. So, this work not only presents the review work but also the research work. This review and research study leads to the conclusion that growth promoting pathways (EGFR, Ras, and PI3K), growth inhibitory pathways (p53 and STK11), apoptotic pathways (Bcl- 2/Bax/Fas), and DDR pathways and genes are commonly and dominantly altered in both the cases COVID-19 and lung cancer.
Collapse
Affiliation(s)
- Afnan M Alnajeebi
- College of Science, Department of Biochemistry, University of Jeddah, Jeddah, Saudi Arabia
| | - Hend F H Alharbi
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, KSA
| | - Walla Alelwani
- College of Science, Department of Biochemistry, University of Jeddah, Jeddah, Saudi Arabia
| | - Nouf A Babteen
- College of Science, Department of Biochemistry, University of Jeddah, Jeddah, Saudi Arabia
| | - Wafa S Alansari
- College of Science, Department of Biochemistry, University of Jeddah, Jeddah, Saudi Arabia
| | - Ghalia Shamlan
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Areej A Eskandrani
- Chemistry Department, Faculty of Science, Taibah University, Medina, Saudi Arabia
| |
Collapse
|
20
|
Vikas, Mehata AK, Suseela MNL, Behera C, Kumari P, Mahto SK, Muthu MS. Chitosan-alginate nanoparticles of cabazitaxel: Design, dual-receptor targeting and efficacy in lung cancer model. Int J Biol Macromol 2022; 221:874-890. [PMID: 36089091 DOI: 10.1016/j.ijbiomac.2022.09.053] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 11/05/2022]
Abstract
Cabazitaxel (CZT) loaded chitosan-alginate based (CSA) nanoparticles were developed with dual targeting functions of both folate receptor and epidermal growth factor receptor (EGFR) using ionic gelation technique. The chitosan-folate conjugate was synthesized, and characterized by using FTIR, NMR and Mass spectroscopy. The physicochemical parameters and morphology of all CSA nanoparticles were examined. The degree of conjugation of folic acid and cetuximab (CTXmab) was determined by UV-Visible spectroscopy and Bradford assay, respectively. Moreover, XPS analysis also supported the presence of the ligands on nanoparticles. The cellular-uptake study performed on A-549 cells demonstrated a significant enhancement in the uptake of dual-receptor targeted CSA nanoparticles than non-targeted and single-receptor targeted CSA nanoparticles. Further, CZT-loaded dual receptors targeted CSA nanoparticles also showed significantly lower IC50 values (~38 folds) than the CZT control against A-549 cells. Further, in-vivo histopathological evaluations of dual receptor-targeted CSA nanoparticles have demonstrated better safety in Wistar rats. Moreover, its treatment on the Benzo(a)pyrene (B(a)P) induced lung cancer mice model has showed the enhanced anticancer efficacy of CZT with a prolonged survival rate.
Collapse
Affiliation(s)
- Vikas
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - M Nikitha Lakshmi Suseela
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Chittaranjan Behera
- PK-PD Tox & Formulation Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Pooja Kumari
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Sanjeev Kumar Mahto
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, UP, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, UP, India.
| |
Collapse
|
21
|
Zhang X, Zhang Y, Qiu X, Cai J, Yang Z, Song F. Extracellular Vesicles Derived from Lung Cancer Cells Induce Transformation of Normal Fibroblasts into Lung Cancer-Associated Fibroblasts and Promote Metastasis of Lung Cancer by Delivering lncRNA HOTAIR. Stem Cells Int 2022; 2022:3805013. [PMID: 36267242 PMCID: PMC9578906 DOI: 10.1155/2022/3805013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/11/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
Human lung cancer (LC) cells A549/H358, normal lung epithelial cells BEAS-2B, and lung normal fibroblasts (NFs) were cultured, followed by transfection of H358 cells with HOTAIR shRNA. Extracellular vesicles (EVs) extracted from H358 cells were identified. The internalization of Dil-labeled-EVs by NFs was tested, and protein levels of cancer-associated fibroblast (CAF) surface markers, inflammatory cytokines, cell proliferation, invasion, and migration, and lncRNA HOTAIR levels were determined. A549 cells were cultured in an H358-EVs-treated conditioned medium of NFs (NFCM), followed by intravenous injection of A549 cells into nude mice. The lesions and Ki-67-positive cells in lung tissues were measured. The results showed that tumor cell-derived EVs (T-EVs) motivated the transformation of NFs into CAFs. Specifically, EVs can be internalized by NFs, and the protein levels of CAF surface markers and inflammation levels were elevated in H358-EVs-treated NFs. The proliferation, invasion, and migration of A549 cells cultured in T-EVs-treated NFCM were increased. H358-EVs carried HOTAIR into NFs and promoted the transformation of NFs into CAFs. Inhibition of HOTAIR partially reversed the promoting effect of H358-EVs on the transformation of NFs into CAFs and invasion and migration of LC cells. T-EVs promoted metastasis of LC in vivo by transforming NFs into CAFs.
Collapse
Affiliation(s)
- Xiaoxuan Zhang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Yan Zhang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
| | - Xin Qiu
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
| | - Jing Cai
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
| | - Zhenzhou Yang
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Fangzhou Song
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
22
|
Kennedy JJ, Woodcock A, Ivey RG, Lin C, Corral G, Hooper E, Martin G, Longman G, Stancik B, Cromwell EA, Whiteaker JR, Zhao L, Lorentzen TD, Thielman S, Paulovich AG. Preserving the Phosphoproteome of Clinical Biopsies Using a Quick-Freeze Collection Device. Biopreserv Biobank 2022; 20:436-445. [PMID: 36301140 PMCID: PMC9603275 DOI: 10.1089/bio.2022.0068] [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] [Indexed: 11/12/2022] Open
Abstract
There is growing interest in proteomic analyses of tissue biopsies to reveal pathophysiology and identify biomarkers. The current gold standard for collecting tissue biopsies for preserving the proteome and post-translational modifications is flash freezing in liquid nitrogen (LN2). However, in many clinical settings, this is not an option due to unavailability of LN2 nor trained personnel for rapid biospecimen processing. To address this need, we developed a proof-of-concept quick-freeze prototype device to rapidly freeze biospecimens at the point-of-care to preserve the phosphoproteome without the need for LN2. Our objectives were to develop the device, demonstrate the ease of use, confirm the ability to ship through existing cold chain logistics, and evaluate the cooling performance (i.e., cool a tissue sample to <0°C in <60 seconds, below -8°C in <120 seconds, and maintain temperature <0°C for >60 minutes) in the context of preserving the proteome in a tissue biospecimen. To demonstrate feasibility, the performance of the prototype was benchmarked against flash freezing in LN2 using a murine melanoma patient-derived xenograft model subjected to total body irradiation to elicit phosphosignaling in the DNA damage response network. Tumors were harvested and quadrisected, with two parts of the tumor being snap frozen in LN2, and the remaining two parts being rapidly cooled in the prototype quick-freeze biospecimen containers. Phosphoproteins were profiled by liquid chromatography tandem mass spectrometry and quantified by targeted multiple reaction monitoring MS. Overall, the phosphoproteome was equivalent in biospecimens processed using the quick-freeze containers to those using the LN2 gold standard, although the measurements of a subset of phosphopeptides in the device-frozen specimens were more variable than LN2-frozen specimens. The prototype device forms the framework for development of a commercial device that will improve tissue biopsy preservation for measurement of important phosphosignaling molecules.
Collapse
Affiliation(s)
- Jacob J. Kennedy
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington, USA
| | | | - Richard G. Ivey
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington, USA
| | - ChenWei Lin
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington, USA
| | - Guy Corral
- Product Creation Studio, Seattle, Washington, USA
| | - Eli Hooper
- Product Creation Studio, Seattle, Washington, USA
| | | | - Gina Longman
- Product Creation Studio, Seattle, Washington, USA
| | | | - Elizabeth A. Cromwell
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington, USA
| | - Jeffrey R. Whiteaker
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington, USA
| | - Lei Zhao
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington, USA
| | - Travis D. Lorentzen
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington, USA
| | | | - Amanda G. Paulovich
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington, USA
| |
Collapse
|
23
|
Keogh A, Finn S, Radonic T. Emerging Biomarkers and the Changing Landscape of Small Cell Lung Cancer. Cancers (Basel) 2022; 14:cancers14153772. [PMID: 35954436 PMCID: PMC9367597 DOI: 10.3390/cancers14153772] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Small cell lung cancer (SCLC) is an aggressive cancer representing 15% of all lung cancers. Unlike other types of lung cancer, treatments for SCLC have changed very little in the past 20 years and therefore, the survival rate remains low. This is due, in part, to the lack of understanding of the biological basis of this disease and the previous idea that all SCLCs are the same. Multiple recent studies have identified that SCLCs have varying biological activity and can be divided into four different groups. The advantage of this is that each of these four groups responds differently to new treatments, which hopefully will dramatically improve survival. Additionally, the aim of these new treatments is to specifically target these biological differences in SCLC so normal/non cancer cells are unaffected, leading to decreased side effects and a better quality of life. There is still a lot unknown about SCLC, but these new findings offer a glimmer of hope for patients in the future. Abstract Small cell lung cancer (SCLC) is a high-grade neuroendocrine malignancy with an aggressive behavior and dismal prognosis. 5-year overall survival remains a disappointing 7%. Genomically, SCLCs are homogeneous compared to non-small cell lung cancers and are characterized almost always by functional inactivation of RB1 and TP53 with no actionable mutations. Additionally, SCLCs histologically appear uniform. Thus, SCLCs are currently managed as a single disease with platinum-based chemotherapy remaining the cornerstone of treatment. Recent studies have identified expression of dominant transcriptional signatures which may permit classification of SCLCs into four biologically distinct subtypes, namely, SCLC-A, SCLC-N, SCLC-P, and SCLC-I. These groups are readily detectable by immunohistochemistry and also have potential predictive utility for emerging therapies, including PARPi, immune checkpoint inhibitors, and DLL3 targeted therapies. In contrast with their histology, studies have identified that SCLCs display both inter- and intra-tumoral heterogeneity. Identification of subpopulations of cells with high expression of PLCG2 has been linked with risk of metastasis. SCLCs also display subtype switching under therapy pressure which may contribute furthermore to metastatic ability and chemoresistance. In this review, we summarize the recent developments in the understanding of the biology of SCLCs, and discuss the potential diagnostic, prognostic, and treatment opportunities the four proposed subtypes may present for the future. We also discuss the emerging evidence of tumor heterogeneity and plasticity in SCLCs which have been implicated in metastasis and acquired therapeutic resistance seen in these aggressive tumors.
Collapse
Affiliation(s)
- Anna Keogh
- Department of Histopathology, St. James’s Hospital, D08 NHY1 Dublin, Ireland;
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 HD53 Dublin, Ireland
- Correspondence:
| | - Stephen Finn
- Department of Histopathology, St. James’s Hospital, D08 NHY1 Dublin, Ireland;
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 HD53 Dublin, Ireland
| | - Teodora Radonic
- Department of Pathology, Amsterdam University Medical Center, VUMC, University Amsterdam, 1081 HV Amsterdam, The Netherlands;
| |
Collapse
|
24
|
Johnson M, Chiara Garassino M, Mok T, Mitsudomi T. Treatment Strategies and Outcomes for Patients with EGFR-mutant Non-Small Cell Lung Cancer Resistant to EGFR Tyrosine Kinase Inhibitors: Focus on Novel Therapies. Lung Cancer 2022; 170:41-51. [DOI: 10.1016/j.lungcan.2022.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 12/16/2022]
|
25
|
Hidayat S, Ibrahim FM, Suhandi C, Muchtaridi M. A systematic review: Molecular docking simulation of small molecules as anticancer non-small cell lung carcinoma drug candidates. J Adv Pharm Technol Res 2022; 13:141-147. [PMID: 35935689 PMCID: PMC9355054 DOI: 10.4103/japtr.japtr_311_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/15/2022] [Indexed: 11/04/2022] Open
Abstract
Non-small cell lung carcinoma (NSCLC) is a type of lung cancer with the highest prevalence and mortality rate worldwide. Many cases of this type of cancer are overexpression on epidermal growth factor receptor (EGFR). The use of currently available EGFR inhibitors as one of the treatment options for NSCLC still shows various shortcomings, especially the high failure rate of therapy due to resistance. It is important to find NSCLC drug candidates with EGFR inhibitory activity. There are various published articles and it is prominent to draw evidence-based scientific conclusions as a basis of decision-making to select potential compounds for further research. Polymer matrix composites and ScienceDirect are used as a database for article screening. Research using molecular docking method targeted to EGFR with parameters of Gibbs energy and amino acid interactions between ligands and drug targets are included in inclusion criteria. Compounds that achieve docking parameters and have comparable activity to NSCLC guideline drugs are conscientiously ranked. There are only 11 compounds that achieved the docking parameters and had comparable EGFR inhibitory potential. Top-rated compounds include 1,3,5-trisubstituted pyrazoline (3c), 1,3,5-trisubstituted pyrazoline (6c), 1,3,5-trisubstituted pyrazoline (8d), N-(3,4-Dimethylphenyl)-2-[(4-oxo-3-(4-sulfamoylphenyl)-3,4-dihydrobenzo[g] quinazolin-2-yl) thio] acetamide. The top-rated compounds can be used and considered for further research processes.
Collapse
Affiliation(s)
- Syahrul Hidayat
- Apothecary Study Program, Faculty of Pharmacy, Universitas Padjadjaran, West Java Province, Indonesia
| | - Faisal Maulana Ibrahim
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, West Java Province, Indonesia
| | - Cecep Suhandi
- Apothecary Study Program, Faculty of Pharmacy, Universitas Padjadjaran, West Java Province, Indonesia
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, West Java Province, Indonesia,Address for correspondence: Prof. Muchtaridi Muchtaridi, Jl. Bandung-Sumedang KM 21, Jatinangor, West Java 45363, Indonesia. E-mail:
| |
Collapse
|
26
|
Md S, Alhakamy NA, Karim S, Gabr GA, Iqubal MK, Murshid SSA. Signaling Pathway Inhibitors, miRNA, and Nanocarrier-Based Pharmacotherapeutics for the Treatment of Lung Cancer: A Review. Pharmaceutics 2021; 13:2120. [PMID: 34959401 PMCID: PMC8708027 DOI: 10.3390/pharmaceutics13122120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is one of the most commonly diagnosed cancers and is responsible for a large number of deaths worldwide. The pathogenic mechanism of lung cancer is complex and multifactorial in origin. Thus, various signaling pathways as targets for therapy are being examined, and many new drugs are in the pipeline. However, both conventional and target-based drugs have been reported to present significant adverse effects, and both types of drugs can affect the clinical outcome in addition to patient quality of life. Recently, miRNA has been identified as a promising target for lung cancer treatment. Therefore, miRNA mimics, oncomiRs, or miRNA suppressors have been developed and studied for possible anticancer effects. However, these miRNAs also suffer from the limitations of low stability, biodegradation, thermal instability, and other issues. Thus, nanocarrier-based drug delivery for the chemotherapeutic drug delivery in addition to miRNA-based systems have been developed so that existing limitations can be resolved, and enhanced therapeutic outcomes can be achieved. Thus, this review discusses lung cancer's molecular mechanism, currently approved drugs, and their adverse effects. We also discuss miRNA biosynthesis and pathogenetic role, highlight pre-clinical and clinical evidence for use of miRNA in cancer therapy, and discussed limitations of this therapy. Furthermore, nanocarrier-based drug delivery systems to deliver chemotherapeutic drugs and miRNAs are described in detail. In brief, the present review describes the mechanism and up-to-date possible therapeutic approaches for lung cancer treatment and emphasizes future prospects to bring these novel approaches from bench to bedside.
Collapse
Affiliation(s)
- Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Shahid Karim
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Gamal A Gabr
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Satam Bin Abdulaziz University, Al-Kharj 16278, Saudi Arabia;
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India;
- Sentiss Research Centre, Product Development Department, Sentiss Pharma Pvt Ltd., Gurugram 122001, India
| | - Samar S. A. Murshid
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| |
Collapse
|
27
|
Maddah SM, Mostafavi G, Amin Malek M, Anbarestani M, Sharif Y, Mir Hassani Z. Combined application of cisplatin and salicylic acid suppresses cell growth and promotes apoptosis in human lung cancer cell lines. Biologia (Bratisl) 2021. [DOI: 10.1007/s11756-021-00920-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
28
|
Pelosi G. KEAP1 and TP53 (Co)mutation in Lung Adenocarcinoma: Another Bullet for Immunotherapy? J Thorac Oncol 2021; 16:1979-1983. [PMID: 34809800 DOI: 10.1016/j.jtho.2021.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 01/21/2023]
Affiliation(s)
- Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Inter-Hospital Pathology Division, Istituto di Ricovero e Cura a Carattere Scientifico MultiMedica, Milan, Italy.
| |
Collapse
|
29
|
Wiest N, Majeed U, Seegobin K, Zhao Y, Lou Y, Manochakian R. Role of Immune Checkpoint Inhibitor Therapy in Advanced EGFR-Mutant Non-Small Cell Lung Cancer. Front Oncol 2021; 11:751209. [PMID: 34868953 PMCID: PMC8634952 DOI: 10.3389/fonc.2021.751209] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/28/2021] [Indexed: 12/25/2022] Open
Abstract
Over the last decade, the treatment of advanced non-small cell lung cancer (NSCLC) has undergone rapid changes with innovations in oncogene-directed therapy and immune checkpoint inhibitors. In patients with epidermal growth factor receptor (EGFR) gene mutant (EGFRm) NSCLC, newer-generation tyrosine kinase inhibitors (TKIs) are providing unparalleled survival benefit and tolerability. Unfortunately, most patients will experience disease progression and thus an urgent need exists for improved subsequent lines of therapies. The concurrent revolution in immune checkpoint inhibitor (ICI) therapy is providing novel treatment options with improved clinical outcomes in wild-type EGFR (EGFRwt) NSCLC; however, the application of ICI therapy to advanced EGFRm NSCLC patients is controversial. Early studies demonstrated the inferiority of ICI monotherapy to EGFR TKI therapy in the first line setting and inferiority to chemotherapy in the second line setting. Additionally, combination ICI and EGFR TKI therapies have demonstrated increased toxicities, and EGFR TKI therapy given after first-line ICI therapy has been correlated with severe adverse events. Nonetheless, combination therapies including dual-ICI blockade and ICI, chemotherapy, and angiogenesis inhibitor combinations are areas of active study with some intriguing signals in preliminary studies. Here, we review previous and ongoing clinical studies of ICI therapy in advanced EGFRm NSCLC. We discuss advances in understanding the differences in the tumor biology and tumor microenvironment (TME) of EGFRm NSCLC tumors that may lead to novel approaches to enhance ICI efficacy. It is our goal to equip the reader with a knowledge of current therapies, past and current clinical trials, and active avenues of research that provide the promise of novel approaches and improved outcomes for patients with advanced EGFRm NSCLC.
Collapse
Affiliation(s)
- Nathaniel Wiest
- Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Umair Majeed
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Karan Seegobin
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Yujie Zhao
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Yanyan Lou
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Rami Manochakian
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
30
|
Fathinavid A, Ghobadi MZ, Najafi A, Masoudi-Nejad A. Identification of common microRNA between COPD and non-small cell lung cancer through pathway enrichment analysis. BMC Genom Data 2021; 22:41. [PMID: 34635059 PMCID: PMC8507163 DOI: 10.1186/s12863-021-00986-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Different factors have been introduced which influence the pathogenesis of chronic obstructive pulmonary disease (COPD) and non-small cell lung cancer (NSCLC). COPD as an independent factor is involved in the development of lung cancer. Moreover, there are certain resemblances between NSCLC and COPD, such as growth factors, activation of intracellular pathways, as well as epigenetic factors. One of the best approaches to understand the possible shared pathogenesis routes between COPD and NSCLC is to study the biological pathways that are activated. MicroRNAs (miRNAs) are critical biomolecules that implicate the regulation of several biological and cellular processes. As such, the main goal of this study was to use a systems biology approach to discover common dysregulated miRNAs between COPD and NSCLC, one that targets most genes within common enriched pathways. RESULTS To reconstruct the miRNA-pathways for each disease, we used the microarray miRNA expression data. Then, we employed "miRNA set enrichment analysis" (MiRSEA) to identify the most significant joint miRNAs between COPD and NSCLC based on the enrichment scores. Overall, our study revealed the involvement of the targets of miRNAs (such as has-miR-15b, hsa-miR-106a, has-miR-17, has-miR-103, and has-miR-107) in the most important common biological pathways. CONCLUSIONS According to the promising results of the pathway analysis, the identified miRNAs can be utilized as the new potential signatures for therapy through understanding the molecular mechanisms of both diseases.
Collapse
Affiliation(s)
- Amirhossein Fathinavid
- Laboratory of Systems Biology and Bioinformatics (LBB), Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran
| | - Mohadeseh Zarei Ghobadi
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Ali Najafi
- Molecular Biology Research Center, System Biology and Poisoning Institute, Tehran, Iran
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
31
|
Velagacherla V, Suresh A, Mehta CH, Nayak UY. Advances and challenges in nintedanib drug delivery. Expert Opin Drug Deliv 2021; 18:1687-1706. [PMID: 34556001 DOI: 10.1080/17425247.2021.1985460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Nintedanib (N.T.B) is an orally administered tyrosine kinase inhibitor that has been approved recently by U.S.F.D.A for idiopathic pulmonary fibrosis (I.P.F) and systemic sclerosis-associated interstitial lung disease (S.Sc-I.L.D). N.T.B is also prescribed in COVID-19 patients associated with I.P.F. However, it has an extremely low bioavailability of around 4.7%, and hence, researchers are attempting to address this drawback by different approaches. AREAS COVERED This review article focuses on enlisting all the formulation attempts explored by researchers to increase the bioavailability of N.T.B while also providing meaningful insight into the unexplored areas in formulation development, such as targeting of the lymphatic system and transdermal delivery. All the patents on the formulation development of N.T.B have also been summarized. EXPERT OPINION N.T.B has the potential to act on multiple diseases that are still being discovered, but its extremely low bioavailability is a challenge that is to be dealt with for obtaining the full benefit. Few studies have been performed aiming at improving the bioavailability, but there are unexplored areas that can be used, a few of which are explained in this article. However, the ability to reproduce laboratory results when scaling up to the industry level is the only factor to be taken into consideration.
Collapse
Affiliation(s)
- Varalakshmi Velagacherla
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Akhil Suresh
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Chetan H Mehta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Usha Y Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
32
|
Jin JO, Puranik N, Bui QT, Yadav D, Lee PCW. The Ubiquitin System: An Emerging Therapeutic Target for Lung Cancer. Int J Mol Sci 2021; 22:9629. [PMID: 34502538 PMCID: PMC8431782 DOI: 10.3390/ijms22179629] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 12/25/2022] Open
Abstract
The ubiquitin system, present in all eukaryotes, contributes to regulating multiple types of cellular protein processes such as cell signaling, cell cycle, and receptor trafficking, and it affects the immune response. In most types of cancer, unusual events in ubiquitin-mediated signaling pathway modulation can lead to a variety of clinical outcomes, including tumor formation and metastasis. Similarly, ubiquitination acts as a core component, which contributes to the alteration of cell signaling activity, dictating biosignal turnover and protein fates. As lung cancer acquires the most commonly mutated proteins, changes in the ubiquitination of the proteins contribute to the development of lung cancer. Various inhibitors targeting the ubiquitin system have been developed for clinical applications in lung cancer treatment. In this review, we summarize the current research advances in therapeutics for lung cancer by targeting the ubiquitin system.
Collapse
Affiliation(s)
- Jun-O Jin
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 201508, China
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea
| | - Nidhi Puranik
- Biological Sciences Department, Bharathiar University, Coimbatore 641046, Tamil Nadu, India;
| | - Quyen Thu Bui
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea;
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea
| | - Peter Chang-Whan Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea;
| |
Collapse
|
33
|
Liguori NR, Lee Y, Borges W, Zhou L, Azzoli C, El-Deiry WS. Absence of Biomarker-Driven Treatment Options in Small Cell Lung Cancer, and Selected Preclinical Candidates for Next Generation Combination Therapies. Front Pharmacol 2021; 12:747180. [PMID: 34531756 PMCID: PMC8438120 DOI: 10.3389/fphar.2021.747180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 08/09/2021] [Indexed: 12/27/2022] Open
Abstract
Lung cancer is the second most common cancer in the United States, and small cell lung cancer (SCLC) accounts for about 15% of all lung cancers. In SCLC, more than other malignancies, the standard of care is based on clinical demonstration of efficacy, and less on a mechanistic understanding of why certain treatments work better than others. This is in large part due to the virulence of the disease, and lack of clinically or biologically relevant biomarkers beyond routine histopathology. While first line therapies work in the majority of patients with extensive stage disease, development of resistance is nearly universal. Although neuroendocrine features, Rb and p53 mutations are common, the current lack of actionable biomarkers has made it difficult to develop more effective treatments. Some progress has been made with the application of immune checkpoint inhibitors. There are new agents, such as lurbinectedin, that have completed late-phase clinical testing while other agents are still in the pre-clinical phase. ONC201/TIC10 is an imipridone with strong in vivo and in vitro antitumor properties and activity against neuroendocrine tumors in phase 1 clinical testing. ONC201 activates the cellular integrated stress response and induces the TRAIL pro-apoptotic pathway. Combination treatment of lurbinectedin with ONC201 are currently being investigated in preclinical studies that may facilitate translation into clinical trials for SCLC patients.
Collapse
Affiliation(s)
- Nicholas R. Liguori
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Young Lee
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - William Borges
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Cancer Center at Brown University, Thoracic Oncology, Providence, RI, United States
| | - Christopher Azzoli
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Cancer Center at Brown University, Thoracic Oncology, Providence, RI, United States
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, RI, United States
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI, United States
- Cancer Center at Brown University, Thoracic Oncology, Providence, RI, United States
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, RI, United States
| |
Collapse
|
34
|
Sonkar A, Kumar P, Gautam A, Maity B, Saha S. New Scope of Targeted Therapies in Lung Carcinoma. Mini Rev Med Chem 2021; 22:629-639. [PMID: 34353252 DOI: 10.2174/1389557521666210805104714] [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: 08/29/2020] [Revised: 12/30/2020] [Accepted: 04/27/2021] [Indexed: 11/22/2022]
Abstract
Lung cancer (LC) is the leading cause of cancer deaths worldwide. Recent research has also shown LC as a genomic disease, causing somatic mutations in patients. Tests related to mutational analysis and genome profiles have lately expanded significantly in the genetics/genomics field of LC. This review summarizes the current knowledge about different signalling pathways of LC based on the clinical impact of molecular targets. It describes the main molecular pathways and changes involved in the development, progression, and cellular breakdown of LC and the molecular changes. This review focuses on approved and targeted experimental therapies such as immunotherapy and clinical trials that examine the different targeted approaches to treating LC. We aimto clarify the differences in the extent of various genetic mutations in several areas for LC patients. Targeted molecular therapies for LC can be continued with advanced racial differences in genetic changes, which have a significant impact on the choice of drug treatment and our understanding of the profile of drug susceptibility/resistance. The most relevant genes described in this review are EGFR, KRAS, MET, BRAF, PIK3CA, STK11, ERBB3, PTEN, and RB1. Combined research efforts in this field are required to understand the genetic difference in LC outcomes in the future.
Collapse
Affiliation(s)
- Archana Sonkar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raibareli Road, Lucknow 226025. India
| | - Pranesh Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raibareli Road, Lucknow 226025. India
| | - Anurag Gautam
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raibareli Road, Lucknow 226025. India
| | - Biswanath Maity
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh. India
| | - Sudipta Saha
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raibareli Road, Lucknow 226025. India
| |
Collapse
|
35
|
Shen J, Su Z. Vanillin oxime inhibits lung cancer cell proliferation and activates apoptosis through JNK/ERK-CHOP pathway. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2021; 25:273-280. [PMID: 34187946 PMCID: PMC8255118 DOI: 10.4196/kjpp.2021.25.4.273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/16/2020] [Accepted: 09/26/2020] [Indexed: 11/29/2022]
Abstract
Lung cancer despite advancement in the medical field continues to be a major threat to human lives and accounts for a high proportion of fatalities caused by cancers globally. The current study investigated vanillin oxime, a derivative of vanillin, against lung cancer cells for development of treatment and explored the mechanism. Cell viability changes by vanillin oxime were measured using MTT assay. Vanillin oxime-mediated apoptosis was detected in A549 and NCI-H2170 cells at 48 h of exposure by flow cytometry. The CEBP homologous protein (CHOP) and death receptor 5 (DR5) levels were analysed by RT-PCR and protein levels by Western blotting. Vanillin oxime in concentration-dependent way suppressed A549 and NCI-H2170 cell viabilities. On exposure to 12.5 and 15 μM concentrations of vanillin oxime elevated Bax, caspase-3, and -9 levels in A549 and NCI-H2170 cells were observed. Vanillin oxime exposure suppressed levels of Bcl-2, survivin, Bcl-xL, cFLIP, and IAPs proteins in A549 and NCI-H2170 cells. It stimulated significant elevation in DR4 and DR5 levels in A549 and NCI-H2170 cells. In A549 and NCI-H2170 cells vanillin oxime exposure caused significant (p < 0.05) enhancement in CHOP and DR5 mRNA expression. Vanillin oxime exposure of A549 and NCI-H2170 cells led to significant (p < 0.05) enhancement in levels of phosphorylated extracellular-signal-regulated kinase and c-Jun N-terminal kinase. Thus, vanillin oxime inhibits pulmonary cell proliferation via induction of apoptosis through tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) mediated pathway. Therefore, vanillin oxime may be studied further to develop a treatment for lung cancer.
Collapse
Affiliation(s)
- Jie Shen
- Department of Respiratory, Yan'an People's Hospital, Yan'an, Shaanxi 716000, China
| | - Zhixiang Su
- Department of Medical Oncology Hospital Unit 3, Shaanxi Provincial Cancer Hospital, Xian 710061, China
| |
Collapse
|
36
|
Mukhopadhyay D, AlSawaftah N, Husseini GA. Identification of Novel MicroRNAs Targeting SARS-CoV-2 through the Regulation of TMPRSS2/PI3K/AKT/PTEN Alignment in Lung Cancer: An in Silico Analysis. ACS Pharmacol Transl Sci 2021; 4:1075-1078. [PMID: 34151202 DOI: 10.1021/acsptsci.1c00040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Indexed: 12/23/2022]
Abstract
In this study, we investigated the interactions between SARS-CoV-2 and miRNAs associated with lung cancer using bioinformatic approaches. A special focus was placed on TMPRSS2 and lung cancer progression pathways involving AKT/PI3K/PTEN genes.
Collapse
Affiliation(s)
- Debasmita Mukhopadhyay
- Chemical Engineering Department, American University of Sharjah, Sharjah, United Arab Emirates
| | - Nour AlSawaftah
- Chemical Engineering Department, American University of Sharjah, Sharjah, United Arab Emirates
| | - Ghaleb A Husseini
- Chemical Engineering Department, American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
37
|
Etu SF, Hossain MA, Rouf ASS, Alqahtani A, Qais N. Molecular docking and anticancer activity determination of 5,10-dihydro-7,8-dimethyl alloxazine derived from lumichrome of riboflavin. MAIN GROUP CHEMISTRY 2021. [DOI: 10.3233/mgc-210025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cancer is accountable for the demise of numerous lives worldwide annually. In this research a derivative of lumichrome, 5,10-dihydro-7,8-dimethyl alloxazine was assessed for its anticancer property through docking study. It was appraised after performing molecular docking study of the 5,10-dihydro-7,8-dimethyl alloxazine, there was a strong interaction between multiple oncogenic target proteins like CDK2/CCNE2 (–8.5 kcal/mol), TDP2 (–8 kcal/mol), NAD-SIRT2 (–10.9 kcal/mol) and lung cancer and acute lymphoblastic leukemia (ALL). Additionally, according to ADMET analysis, the synthesized compound 5,10-dihydro-7,8-dimethyl alloxazine also has good physicochemical characteristics to be a drug candidate. Consequently, these verdicts will assist the development of a novel anti-lung cancer and anti-leukemic agent which will eventually improve the endurance of cancer patients.
Collapse
Affiliation(s)
| | - M. Alamgir Hossain
- Department of Pharmacy, Jagannath University, Chittaranjan Ave., Dhaka, Bangladesh
| | | | - Ali Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Guraiger, Abha, Saudi Arabia
| | - Nazmul Qais
- Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
38
|
Mehta M, Dhanjal DS, Satija S, Wadhwa R, Paudel KR, Chellappan DK, Mohammad S, Haghi M, Hansbro PM, Dua K. Advancing of Cellular Signaling Pathways in Respiratory Diseases Using Nanocarrier Based Drug Delivery Systems. Curr Pharm Des 2021; 26:5380-5392. [PMID: 33198611 DOI: 10.2174/1381612826999201116161143] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 10/01/2020] [Indexed: 11/22/2022]
Abstract
Cell Signaling pathways form an integral part of our existence that allows the cells to comprehend a stimulus and respond back. Such reactions to external cues from the environment are required and are essential to regulate the normal functioning of our body. Abnormalities in the system arise when there are errors developed in these signals, resulting in a complication or a disease. Presently, respiratory diseases contribute to being the third leading cause of morbidity worldwide. According to the current statistics, over 339 million people are asthmatic, 65 million are suffering from COPD, 2.3 million are lung cancer patients and 10 million are tuberculosis patients. This toll of statistics with chronic respiratory diseases leaves a heavy burden on society and the nation's annual health expenditure. Hence, a better understanding of the processes governing these cellular pathways will enable us to treat and manage these deadly respiratory diseases effectively. Moreover, it is important to comprehend the synergy and interplay of the cellular signaling pathways in respiratory diseases, which will enable us to explore and develop suitable strategies for targeted drug delivery. This review, in particular, focuses on the major respiratory diseases and further provides an in-depth discussion on the various cell signaling pathways that are involved in the pathophysiology of respiratory diseases. Moreover, the review also analyses the defining concepts about advanced nano-drug delivery systems involving various nanocarriers and propose newer prospects to minimize the current challenges faced by researchers and formulation scientists.
Collapse
Affiliation(s)
- Meenu Mehta
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Daljeet Singh Dhanjal
- School of Biosciences and Bioengineering, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Saurabh Satija
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Ridhima Wadhwa
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Keshav Raj Paudel
- School of Life Sciences, Faculty of Science, University of Technology Sydney (UTS), Ultimo, NSW, 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Shiva Mohammad
- School of Life Sciences, Faculty of Science, University of Technology Sydney (UTS), Ultimo, NSW, 2007, Australia
| | - Mehra Haghi
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Philip M Hansbro
- School of Life Sciences, Faculty of Science, University of Technology Sydney (UTS), Ultimo, NSW, 2007, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| |
Collapse
|
39
|
Kulkarni NS, Vaidya B, Gupta V. Nano-synergistic combination of Erlotinib and Quinacrine for non-small cell lung cancer (NSCLC) therapeutics - Evaluation in biologically relevant in-vitro models. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 121:111891. [PMID: 33579503 DOI: 10.1016/j.msec.2021.111891] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 01/10/2023]
Abstract
Non-small cell lung cancer (NSCLC), pre-dominant subtype of lung cancer, is a global disorder affecting millions worldwide. One of the early treatments for NSCLC was use of a first-generation tyrosine kinase inhibitor, Erlotinib (Erlo). However, chronic exposure to Erlo led to development of acquired drug resistance (ADR) in NSCLC, limiting the clinical use of Erlo. A potential approach to overcome development of ADR is a multi-drug therapy. It has been previously reported that Erlo and Quinacrine (QA), an anti-malarial drug, can work synergistically to inhibit tumor progression in NSCLC. However, the combination failed at clinical stages, citing lack of efficacy. In this study, an effort has been made to improve the efficacy of Erlo-QA combination via development of nanoformulations, known to enhance therapeutic efficacy of potent chemotherapies. Synergy between Erlo and QA was measured via estimating the combination indices (CI). It was seen that established combination of nanoformulations (CI: 0.25) had better synergy than plain drug solutions (CI: 0.85) in combination. Following extensive in-vitro testing, data were simulated in biologically relevant 3D tumor models. Two tumor models were developed for extensive in-vitro testing, 3D-Spheroids grown in ultra-low attachment culture plates for efficacy evaluation and a 5D-spheroid model in 5D-sphericalplate with capability of growing 750 spheroids/well for protein expression analysis. Extensive studies on these models revealed that combination of Erlo and QA nanoformulations overall had a better effect in terms of synergy enhancement as compared to plain drug combination. Further, effect of combinatorial therapy on molecular markers was evaluated on 5D-Sphericalplate leading to similar effects on synergy enhancement. Results from present study suggests that combination of nanoformulations can improve the synergy between Erlo and QA while reducing the overall therapeutic dose.
Collapse
Affiliation(s)
- Nishant S Kulkarni
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | | | - Vivek Gupta
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA; School of Pharmacy, Keck Graduate Institute, Claremont, CA, USA.
| |
Collapse
|
40
|
Hussen BM, Ahmadi G, Marzban H, Fard Azar ME, Sorayyayi S, Karampour R, Nahand JS, Hidayat HJ, Moghoofei M. The role of HPV gene expression and selected cellular MiRNAs in lung cancer development. Microb Pathog 2020; 150:104692. [PMID: 33301856 DOI: 10.1016/j.micpath.2020.104692] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/04/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The high mortality rate of lung cancer can be justified that strong need to explore new aspect of tumor biology. Human papillomavirus (HPV) has been detected as risk factor for the development of lung cancer. The aim of this study was to determine the role of HPV and cellular/miRNAs genes expression in the epithelial-mesenchymal transition (EMT) and development of lung cancer. METHODS In this case-control study, 109 lung cancer tissue and 52 controls were included. We analyzed the presence of HPV infection, its genotypes (in positive samples) and the expression of viral genes (E2, E6 and E7). Also, We examined the expression of celluar factors including (a) p53 and retinoblastoma (Rb) (as anti-carcinogenic genes), (b) EMT related genes, (c) selected miRNAs. RESULTS Our results reported 51.4% and 23.1% of HPV genome in tumor tissues and control tissues samples, respectively. There was a significant association between the HPV positive status and lung cancer (OR = 3.26, 95% C.I = 1.47-7.02, P = 0.001). HPV type 16 was the most prevalent genotype in tissues. The expression of p53, RB, TIMP1, CCNG-1, E-cad and PTPN13 were decreased while MMP-2 and N-cad were increased in HPV-positive tumor/control tissues compared to HPV-negative tissues. Also, among miRNAs, let-7, miR-23, miR-34, miR-125, miR-146 were downregulated and miR-20, miR-424 were upregulated in HPV-positve tissues compared to HPV-negative tissues. CONCLUSION This study demonstrated that HPV infection and interaction with cellular genes and miRNAs promote EMT which involved in the lung cancer development.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Gelavizh Ahmadi
- Department of Biotechnology, Faculty of Sciences, University of Maragheh, Maragheh, Iran
| | - Havva Marzban
- Department of Pathology and Experimental Animals, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | | | - Saba Sorayyayi
- Department of Clinical Biochemistry, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Romina Karampour
- Department of Pathobiology, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Javid Sadri Nahand
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University, Erbil, Iraq
| | - Mohsen Moghoofei
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
41
|
Mokhlesi A, Talkhabi M. Comprehensive transcriptomic analysis identifies novel regulators of lung adenocarcinoma. J Cell Commun Signal 2020; 14:453-465. [PMID: 32415511 PMCID: PMC7642016 DOI: 10.1007/s12079-020-00565-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022] Open
Abstract
Lung adenocarcinoma (LA) is a subtype of lung cancer that accounts for about 40% of all lung cancers. Analysis of molecular mechanisms controlling this cancer can help scientists to detect, control and treat LA. Here, a microarray dataset (GSE118370) containing six normal lung (NL) and six LA samples was screened using GEO2R to find differentially expressed genes (DEGs). Then, DAVID, KEGG and ChEA were used to analyze DEGs-related gene ontology, pathways and transcription factors (TFs), respectively. The Protein-protein interaction network for DEGs and TFs was constructed by STRING and Cytoscape. To find microRNAs and metabolites associated with DEGs, miRTarBase and HMDB were used, respectively. It was found that 350 genes were upregulated and 608 genes were downregulated in LA. The upregulated genes or LA-related gens were enriched in biological process and pathways such as extracellular matrix disassembly and p53 signaling pathway, whereas the downregulated genes or NL-related genes were enriched in cell adhesion and cell-surface receptor signaling pathway. ESR1, KIF18B, BIRC5, CHEK1, CCNB1 and AURKA were determined as hub genes for LA. FOXA1 and TFAP2A had the highest number of connectivity in LA-related TFs. hsa-miR-192-5p and hsa-miR-215-5p could target the highest number of LA-related genes. Metabolite analysis showed that Estrone and NADPH were among the top ten metabolites associated with LA-related genes. Taken together, LA-related genes, especially the hub genes, TFs, and metabolites might be used as novel markers for LA, as well as for diagnosis and guiding therapeutic strategies of LA.
Collapse
Affiliation(s)
- Amir Mokhlesi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mahmood Talkhabi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
42
|
Mustachio LM, Roszik J. Current Targeted Therapies for the Fight against Non-Small Cell Lung Cancer. Pharmaceuticals (Basel) 2020; 13:ph13110374. [PMID: 33182254 PMCID: PMC7695293 DOI: 10.3390/ph13110374] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/01/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
Lung cancers contribute to the greatest number of cancer-related deaths worldwide and still pose challenges in response to current treatment strategies. Non-small cell lung cancer (NSCLC) accounts for over 85% of lung cancers diagnosed in the United States and novel therapeutics are needed for the treatment of this disease. First and second generation targeted therapies against specific mutated or rearranged oncogenes in NSCLCs show anti-tumor activity and also increase survival. However, many NSCLC patients eventually develop resistance to these therapies or do not properly respond if they have central nervous system metastases. Thus, this review summarizes recent developments and findings related to the generation of novel targeted therapies recently or currently being developed to tackle hurdles that prior therapies were not able to overcome.
Collapse
Affiliation(s)
- Lisa Maria Mustachio
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (L.M.M.); (J.R.); Tel.: +1-832-750-4367 (L.M.M.); +1-713-745-2641 (J.R.)
| | - Jason Roszik
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (L.M.M.); (J.R.); Tel.: +1-832-750-4367 (L.M.M.); +1-713-745-2641 (J.R.)
| |
Collapse
|
43
|
Heffner K, Hizal DB, Majewska NI, Kumar S, Dhara VG, Zhu J, Bowen M, Hatton D, Yerganian G, Yerganian A, O'Meally R, Cole R, Betenbaugh M. Expanded Chinese hamster organ and cell line proteomics profiling reveals tissue-specific functionalities. Sci Rep 2020; 10:15841. [PMID: 32985598 PMCID: PMC7522264 DOI: 10.1038/s41598-020-72959-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/31/2020] [Indexed: 02/07/2023] Open
Abstract
Chinese hamster ovary (CHO) cells are the predominant production vehicle for biotherapeutics. Quantitative proteomics data were obtained from two CHO cell lines (CHO-S and CHO DG44) and compared with seven Chinese hamster (Cricetulus griseus) tissues (brain, heart, kidney, liver, lung, ovary and spleen) by tandem mass tag (TMT) labeling followed by mass spectrometry, providing a comprehensive hamster tissue and cell line proteomics atlas. Of the 8470 unique proteins identified, high similarity was observed between CHO-S and CHO DG44 and included increases in proteins involved in DNA replication, cell cycle, RNA processing, and chromosome processing. Alternatively, gene ontology and pathway analysis in tissues indicated increased protein intensities related to important tissue functionalities. Proteins enriched in the brain included those involved in acidic amino acid metabolism, Golgi apparatus, and ion and phospholipid transport. The lung showed enrichment in proteins involved in BCAA catabolism, ROS metabolism, vesicle trafficking, and lipid synthesis while the ovary exhibited enrichments in extracellular matrix and adhesion proteins. The heart proteome included vasoconstriction, complement activation, and lipoprotein metabolism enrichments. These detailed comparisons of CHO cell lines and hamster tissues will enhance understanding of the relationship between proteins and tissue function and pinpoint potential pathways of biotechnological relevance for future cell engineering.
Collapse
Affiliation(s)
- Kelley Heffner
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,AstraZeneca, Cell Culture and Fermentation Sciences, Gaithersburg, MD, USA
| | - Deniz Baycin Hizal
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Natalia I Majewska
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,AstraZeneca, Cell Culture and Fermentation Sciences, Gaithersburg, MD, USA
| | - Swetha Kumar
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Venkata Gayatri Dhara
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jie Zhu
- AstraZeneca, Cell Culture and Fermentation Sciences, Gaithersburg, MD, USA
| | - Michael Bowen
- Allogene Therapeutics, Product and Process Development, South San Francisco, CA, USA
| | - Diane Hatton
- AstraZeneca, Cell Culture and Fermentation Sciences, Gaithersburg, MD, USA
| | | | | | - Robert O'Meally
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Robert Cole
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Michael Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
44
|
Taylor B, Rice A, Nicholson AG, Hind M, Dean CH. Mechanism of lung development in the aetiology of adult congenital pulmonary airway malformations. Thorax 2020; 75:1001-1003. [PMID: 32732323 PMCID: PMC7569368 DOI: 10.1136/thoraxjnl-2020-214752] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/27/2020] [Accepted: 07/02/2020] [Indexed: 12/04/2022]
Abstract
Congenital pulmonary airway malformations (CPAMs) are rare lung abnormalities that result in cyst formation and are associated with respiratory distress in infants and malignant potential in adults. The pathogenesis of CPAMs remains unknown but data suggest disruption of the normal proximo-distal programme of airway branching and differentiation. Here, we demonstrate that adult human CPAM are lined with epithelium that retains SOX-2 and thyroid transcription factor-1 immunohistochemical markers, characteristic of the developing lung. However, RALDH-1, another key marker, is absent. This suggests a more complex aetiology for CPAM than complete focal arrest of lung development and may provide insight to the associated risk of malignancy.
Collapse
Affiliation(s)
- Bethany Taylor
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Alexandra Rice
- Department of Histopathology, Royal Brompton and Harefield NHS Foundation Trust, London, UK
| | - Andrew G Nicholson
- Department of Histopathology, Royal Brompton and Harefield NHS Foundation Trust, London, UK
| | - Matthew Hind
- National Heart and Lung Institute, Imperial College London, London, UK.,Respiratory Medicine, Department of Respiratory Medicine and National Institute for Health research Respiratory Biomedical Research Unit at the Royal Brompton NHS Foundation Trust and Imperial College, London, UK
| | - Charlotte H Dean
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
45
|
Contreras LM, Gonzalez-Rivera JC, Baldridge KC, Wang DS, Chuvalo-Abraham J, Ruiz LH. Understanding the Functional Impact of VOC-Ozone Mixtures on the Chemistry of RNA in Epithelial Lung Cells. Res Rep Health Eff Inst 2020; 2020:Res Rep Health Eff Inst. 2020 Jul;(201):3-43.. [PMID: 32845096 PMCID: PMC7448316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023] Open
Abstract
Introduction Ambient air pollution is associated with premature death caused by heart disease, stroke, chronic obstructive pulmonary disease (COPD), and lung cancer. Recent studies have suggested that ribonucleic acid (RNA) oxidation is a sensitive environment-related biomarker that is implicated in pathogenesis. Aims and Methods We used a novel approach that integrated RNA-Seq analysis with detection by immunoprecipitation techniques of the prominent RNA oxidative modification 8-oxo-7,8-dihydroguanine (8-oxoG). Our goal was to uncover specific messenger RNA (mRNA) oxidation induced by mixtures of volatile organic compounds (VOCs) and ozone in healthy human epithelial lung cells. To this end, we exposed the BEAS-2B human epithelial lung cell line to the gas- and particle-phase products formed from reactions of 790 ppb acrolein (ACR) and 670 ppb methacrolein (MACR) with 4 ppm ozone. Results Using this approach, we identified 222 potential direct targets of oxidation belonging to previously described pathways, as well as uncharacterized pathways, after air pollution exposures. We demonstrated the effect of our VOC-ozone mixtures on the morphology and actin cytoskeleton of lung cells, suggesting the influence of selective mRNA oxidation in members of pathways regulating physical components of the cells. In addition, we observed the influence of the VOC-ozone mixtures on metabolic cholesterol synthesis, likely implicated as a result of the incidence of mRNA oxidation and the deregulation of protein levels of squalene synthase (farnesyl-diphosphate farnesyltransferase 1 [FDFT1]), a key enzyme in endogenous cholesterol biosynthesis. Conclusions Overall, our findings indicate that air pollution influences the accumulation of 8-oxoG in transcripts of epithelial lung cells that largely belong to stress-induced signaling and metabolic and structural pathways. A strength of the study was that it combined traditional transcriptome analysis with transcriptome-wide 8-oxoG mapping to facilitate the discovery of underlying processes not characterized by earlier approaches. Investigation of the processes mediated by air pollution oxidation of RNA molecules in primary cells and animal models needs to be explored in future studies. Our research has thus opened new avenues to further inform the relationship between atmospheric agents on the one hand and cellular responses on the other that are implicated in diseases.
Collapse
Affiliation(s)
- L M Contreras
- McKetta Department of Chemical Engineering, University of Texas, Austin
| | | | - K C Baldridge
- McKetta Department of Chemical Engineering, University of Texas, Austin
| | - D S Wang
- McKetta Department of Chemical Engineering, University of Texas, Austin
| | | | - L H Ruiz
- McKetta Department of Chemical Engineering, University of Texas, Austin
| |
Collapse
|
46
|
Liu X, Huang S, Guan Y, Zhang Q. Long noncoding RNA OSER1‑AS1 promotes the malignant properties of non‑small cell lung cancer by sponging microRNA‑433‑3p and thereby increasing Smad2 expression. Oncol Rep 2020; 44:599-610. [PMID: 32627026 PMCID: PMC7336450 DOI: 10.3892/or.2020.7645] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023] Open
Abstract
OSER1 antisense RNA 1 (OSER1-AS1), a long noncoding RNA, has been well studied in the context of hepatocellular carcinoma. However, its expression status, specific functions, and tumorigenic mechanism in non-small cell lung cancer (NSCLC) remain uninvestigated. Hence, this study aimed to assess OSER1-AS1 expression, test the malignancy-related biological functions of OSER1-AS1, and illustrate how they affect NSCLC progression. OSER1-AS1 expression in NSCLC was measured by reverse transcription-quantitative polymerase chain reaction. Cell Counting Kit-8 assay, flow cytometry, cell migration and invasion assay, and tumor xenograft assay were performed to analyze the effects of OSER1-AS1 on the malignant phenotypes of NSCLC cells. Bioinformatics prediction with luciferase reporter and RNA immunoprecipitation assays were performed to determine the interaction between OSER1-AS1 and microRNA-433-3p (miR-433-3p). OSER1-AS1 was strongly expressed in NSCLC tissues and cell lines. Enhanced OSER1-AS1 expression was significantly correlated with tumor size, TNM stage, and lymph node metastasis in patients with NSCLC. Patients with NSCLC exhibiting high OSER1-AS1 expression had shorter overall survival than those exhibiting low OSER1-AS1 expression. Functionally, a reduction in OSER1-AS1 expression led to significant decreases in NSCLC cell proliferation, migration, and invasion as well as an increase in cell apoptosis in vivo. OSER1-AS1 knockdown suppressed the tumorigenic ability of NSCLC cells in vivo. Mechanistically, OSER1-AS1 acts as a competing endogenous RNA (ceRNA) in NSCLC cells by sponging miR-433-3p and thereby increasing the expression of mothers against decapentaplegic homolog 2 (Smad2). Finally, restoration experiments revealed that the suppression of miR-433-3p and restoration of Smad2 both counteracted the suppressive effects of OSER1-AS1 depletion in NSCLC cells. Our findings illustrate the biological importance of the OSER1-AS1/miR-433-3p/Smad2 pathway in NSCLC progression and offer a novel perspective regarding the identification of effective therapeutic and diagnostic targets.
Collapse
Affiliation(s)
- Xinmei Liu
- Department of Respiratory Disease, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Shasha Huang
- Department of General Surgery, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Yun Guan
- Department of Respiratory Disease, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Qing Zhang
- Department of Respiratory Disease, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
47
|
Li B, Dai C, Wang L, Deng H, Li Y, Guan Z, Ni H. A novel drug repurposing approach for non-small cell lung cancer using deep learning. PLoS One 2020; 15:e0233112. [PMID: 32525938 PMCID: PMC7289363 DOI: 10.1371/journal.pone.0233112] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 04/28/2020] [Indexed: 01/02/2023] Open
Abstract
Drug repurposing is an attractive and pragmatic way offering reduced risks and development time in the complicated process of drug discovery. In the past, drug repurposing has been largely accidental and serendipitous. The most successful examples so far have not involved a systematic approach. Nowadays, remarkable advances in drugs, diseases and bioinformatic knowledge are offering great opportunities for designing novel drug repurposing approach through comprehensive understanding of drug information. In this study, we introduced a novel drug repurposing approach based on transcriptomic data and chemical structures using deep learning. One strong candidate for repurposing has been identified. Pimozide is an anti-dyskinesia agent that is used for the suppression of motor and phonic tics in patients with Tourette's Disorder. However, our pipeline proposed it as a strong candidate for treating non-small cell lung cancer. The cytotoxicity of pimozide against A549 cell lines has been validated.
Collapse
Affiliation(s)
- Bingrui Li
- Beijing Deep Intelligent Pharma Technologies Co., Ltd, Beijing, China
| | - Chan Dai
- Beijing Deep Intelligent Pharma Technologies Co., Ltd, Beijing, China
| | - Lijun Wang
- Beijing Deep Intelligent Pharma Technologies Co., Ltd, Beijing, China
| | - Hailong Deng
- Beijing Deep Intelligent Pharma Technologies Co., Ltd, Beijing, China
| | - Yingying Li
- Beijing Deep Intelligent Pharma Technologies Co., Ltd, Beijing, China
- * E-mail: (YL); (ZG); (HN)
| | - Zheng Guan
- Beijing Deep Intelligent Pharma Technologies Co., Ltd, Beijing, China
- * E-mail: (YL); (ZG); (HN)
| | - Haihong Ni
- Beijing Deep Intelligent Pharma Technologies Co., Ltd, Beijing, China
- * E-mail: (YL); (ZG); (HN)
| |
Collapse
|
48
|
Ashraf-Uz-Zaman M, Bhalerao A, Mikelis CM, Cucullo L, German NA. Assessing the Current State of Lung Cancer Chemoprevention: A Comprehensive Overview. Cancers (Basel) 2020; 12:E1265. [PMID: 32429547 PMCID: PMC7281533 DOI: 10.3390/cancers12051265] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/06/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
Chemoprevention of lung cancer is thought to significantly reduce the risk of acquiring these conditions in the subpopulation of patients with underlying health issues, such as chronic obstructive pulmonary disorder and smoking-associated lung problems. Many strategies have been tested in the previous decades, with very few translating to successful clinical trials in specific subpopulations of patients. In this review, we analyze these strategies, as well as new approaches that have emerged throughout the last few years, including synthetic lethality concept and microbiome-induced regulation of lung carcinogenesis. Overall, the continuous effort in the area of lung chemoprevention is required to develop practical therapeutical approaches. Given the inconsistency of results obtained in clinical trials targeting lung cancer chemoprevention in various subgroups of patients that differ in the underlying health condition, race, and gender, we believe that individualized approaches will have more promise than generalized treatments.
Collapse
Affiliation(s)
- Md Ashraf-Uz-Zaman
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (M.A.-U.-Z.); (A.B.); (C.M.M.); (L.C.)
| | - Aditya Bhalerao
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (M.A.-U.-Z.); (A.B.); (C.M.M.); (L.C.)
| | - Constantinos M. Mikelis
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (M.A.-U.-Z.); (A.B.); (C.M.M.); (L.C.)
- Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Luca Cucullo
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (M.A.-U.-Z.); (A.B.); (C.M.M.); (L.C.)
- Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Nadezhda A. German
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (M.A.-U.-Z.); (A.B.); (C.M.M.); (L.C.)
- Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
49
|
Non-Coding RNAs in Lung Tumor Initiation and Progression. Int J Mol Sci 2020; 21:ijms21082774. [PMID: 32316322 PMCID: PMC7215285 DOI: 10.3390/ijms21082774] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/07/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
Lung cancer is one of the deadliest forms of cancer affecting society today. Non-coding RNAs, such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), through the transcriptional, post-transcriptional, and epigenetic changes they impose, have been found to be dysregulated to affect lung cancer tumorigenesis and metastasis. This review will briefly summarize hallmarks involved in lung cancer initiation and progression. For initiation, these hallmarks include tumor initiating cells, immortalization, activation of oncogenes and inactivation of tumor suppressors. Hallmarks involved in lung cancer progression include metastasis and drug tolerance and resistance. The targeting of these hallmarks with non-coding RNAs can affect vital metabolic and cell signaling pathways, which as a result can potentially have a role in cancerous and pathological processes. By further understanding non-coding RNAs, researchers can work towards diagnoses and treatments to improve early detection and clinical response.
Collapse
|
50
|
Li W, Li X, Li X, Li M, Yang P, Wang X, Li L, Yang B. Lamin B1 Overexpresses in Lung Adenocarcinoma and Promotes Proliferation in Lung Cancer Cells via AKT Pathway. Onco Targets Ther 2020; 13:3129-3139. [PMID: 32346296 PMCID: PMC7167283 DOI: 10.2147/ott.s229997] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 02/16/2020] [Indexed: 12/19/2022] Open
Abstract
PURPOSE This study aims to investigate the biological effect and molecular mechanism of Lamin B1(LMNB1) in lung cancer cells and its significance for the prognosis of lung adenocarcinoma(LUAD) patients. METHODS In this study, Bioinformatics was performed to analyze the expression at mRNA level and prognosis effect of LMNB1 in LUAD from TCGA dataset. The immunohistochemistry(IHC) assay was conducted to analyzed the expression of LMNB1 at the protein level in LUAD tissues. The correlation between the expression of LMNB1 and the clinical factors in patients with LUAD was analyzed. Next, LMNB1 transfected into LUAD cell lines (A549 and PC-9) which was proved by Western blot. CCK8 assay, cloning formation assay, and xenograft assay were conducted to explore the effect and mechanism of LMNB1 on the proliferation of LUAD cell lines in vitro and in vivo. RESULTS The results of the present study demonstrated that LMNB1 was highly expressed in LUAD tissues and related to tumor stage. High LMNB1 expression was related with more advanced clinicopathological factors such as low degree of differentiation (P=0.02), large tumor size (P<0.01), lymph node metastasis (P<0.01) and higher tumor stage (P<0.01). After knocking down LMNB1, the cell growth rate (P<0.01) and the number of colonies (P<0.01) were significantly reduced, and the level of the proliferating marker Ki67 (P<0.01) was significantly decreased. At the same time, in vivo experiments showed that the tumor volume and tumor of the mice were significantly reduced (P<0.01). Moreover, we found that knockdown LMNB1 can inhibit the proliferation of lung cancer cells by inhibiting AKT phosphorylation by Western blot. CONCLUSION In summary, LMNB1 play an of vital roles in the growth of LUAD cells, highlighting its potential as a therapeutic target for the treatment of LUAD patients.
Collapse
Affiliation(s)
- Wei Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin300192, People’s Republic of China
| | - Xiaoqing Li
- Phase I Clinical Trial Department, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Centre for Cancer, Tianjin300052, People’s Republic of China
| | - Xiaoping Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin300192, People’s Republic of China
| | - Mingjiang Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin300192, People’s Republic of China
| | - Pan Yang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin300192, People’s Republic of China
| | - Xuhui Wang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin300192, People’s Republic of China
| | - Lei Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin300192, People’s Republic of China
| | - Bo Yang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin300192, People’s Republic of China
| |
Collapse
|