1
|
Shankar S, Kumar Y, Sharma N, Chandra R, Kumar S. Disposable Zirconium trisulfide-Reduced graphene oxide modified conducting thread based electrochemical biosensor for lung cancer diagnosis. Bioelectrochemistry 2024; 160:108801. [PMID: 39226732 DOI: 10.1016/j.bioelechem.2024.108801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024]
Abstract
Flexible technology in sensors have received much attention in monitoring of human health through various physiological indicators. Thus, it drawn a lot of interest in the development of flexible substrate for the diagnosis of various diseases via analysis of analytes. Present work focusses on the development of ecofriendly, portable, flexible, conducting thread (Th) and used as smart substrate for fabrication of biosensor towards ultrasensitive detection of the lung cancer biomarker (cytoskeleton-associated protein 4; CKAP4). The zirconium trisulfide-reduced graphene oxide nanocomposite and poly(3,4-ethylenedioxythiophene):poly(styrenesulfonate) (PEDOT:PSS) modified cotton thread based biosensor was fabricated via dip coating method. Next, successive immobilization of monoclonal antibodies of CKAP4 (anti-CKAP4) and bovine serum albumin (BSA) was performed via drop cast approach using fabricated electrode [nZrS3@rGO/PEDOT:PSS/Th]. The response of fabricated electrode (BSA/anti-CKAP4/ZrS3@rGO/PEDOT:PSS/Th) was recorded electrochemically versus CKAP4 concentration via chronoamperometry (CA). The results showed wider linear detection range of 6.25-800 pg mL-1, excellent sensitivity of 85.2 µA[log(pg mL-1)]-1cm-2 with good stability up to 42 days. The response of fabricated biosensor was supported by investigating response of CKAP4 biomarker present in patients of lung cancer (concentration as determined through enzyme-linked immunosorbent assay) and obtained results exhibited excellent correlation with that of standard samples.
Collapse
Affiliation(s)
- Saurav Shankar
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Yogesh Kumar
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Neera Sharma
- Department of Chemistry, Hindu College, University of Delhi, Delhi 110007, India
| | - Ramesh Chandra
- Department of Chemistry, University of Delhi, Delhi 110007, India; Maharaja Surajmal Brij University, Kumher, Bharatpur 321201, India.
| | - Suveen Kumar
- Department of Chemistry, University of Delhi, Delhi 110007, India.
| |
Collapse
|
2
|
Han Q, Fernandez J, Rajczewski AT, Kono TJY, Weirath NA, Rahim A, Lee AS, Seabloom D, Tretyakova NY. A Multi-Omics Study of Epigenetic Changes in Type II Alveolar Cells of A/J Mice Exposed to Environmental Tobacco Smoke. Int J Mol Sci 2024; 25:9365. [PMID: 39273313 PMCID: PMC11394788 DOI: 10.3390/ijms25179365] [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: 07/24/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Lung cancer remains a major contributor to cancer fatalities, with cigarette smoking known to be responsible for up to 80% of cases. Based on the ability of cigarette smoke to induce inflammation in the lungs and increased lung cancer incidence in smokers with inflammatory conditions such as COPD, we hypothesized that inflammation plays an important role in the carcinogenicity of cigarette smoke. To test this hypothesis, we performed multi-omic analyses of Type II pneumocytes of A/J mice exposed to cigarette smoke for various time periods. We found that cigarette smoke exposure resulted in significant changes in DNA methylation and hydroxymethylation, gene expression patterns, and protein abundance that were partially reversible and contributed to an inflammatory and potentially oncogenic phenotype.
Collapse
Affiliation(s)
- Qiyuan Han
- Department of Biochemistry, Biophysics and Molecular Biology, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; (Q.H.); (A.T.R.)
| | - Jenna Fernandez
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; (J.F.); (N.A.W.); (A.R.)
| | - Andrew T. Rajczewski
- Department of Biochemistry, Biophysics and Molecular Biology, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; (Q.H.); (A.T.R.)
| | - Thomas J. Y. Kono
- Minnesota Supercomputing Institute, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA;
| | - Nicholas A. Weirath
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; (J.F.); (N.A.W.); (A.R.)
| | - Abdur Rahim
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; (J.F.); (N.A.W.); (A.R.)
| | - Alexander S. Lee
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Evanston, IL 60611, USA;
| | - Donna Seabloom
- AeroCore Testing Services, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA;
| | - Natalia Y. Tretyakova
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; (J.F.); (N.A.W.); (A.R.)
| |
Collapse
|
3
|
Fasano R, Serratì S, Rafaschieri T, Longo V, Di Fonte R, Porcelli L, Azzariti A. Small-Cell Lung Cancer: Is Liquid Biopsy a New Tool Able to Predict the Efficacy of Immunotherapy? Biomolecules 2024; 14:396. [PMID: 38672414 PMCID: PMC11048475 DOI: 10.3390/biom14040396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Small-cell lung cancer (SCLC) cases represent approximately 15% of all lung cancer cases, remaining a recalcitrant malignancy with poor survival and few treatment options. In the last few years, the addition of immunotherapy to chemotherapy improved clinical outcomes compared to chemotherapy alone, resulting in the current standard of care for SCLC. However, the advantage of immunotherapy only applies to a few SCLC patients, and predictive biomarkers selection are lacking for SCLC. In particular, due to some features of SCLC, such as high heterogeneity, elevated cell plasticity, and low-quality tissue samples, SCLC biopsies cannot be used as biomarkers. Therefore, the characterization of the tumor and, subsequently, the selection of an appropriate therapeutic combination may benefit greatly from liquid biopsy. Soluble factors, circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and extracellular vesicles (EVs) are now useful tools in the characterization of SCLC. This review summarizes the most recent data on biomarkers detectable with liquid biopsy, emphasizing their role in supporting tumor detection and their potential role in SCLC treatment choice.
Collapse
Affiliation(s)
- Rossella Fasano
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.Le O. Flacco, 65, 70124 Bari, Italy; (R.F.); (T.R.); (R.D.F.); (L.P.); (A.A.)
| | - Simona Serratì
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.Le O. Flacco, 65, 70124 Bari, Italy; (R.F.); (T.R.); (R.D.F.); (L.P.); (A.A.)
| | - Tania Rafaschieri
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.Le O. Flacco, 65, 70124 Bari, Italy; (R.F.); (T.R.); (R.D.F.); (L.P.); (A.A.)
| | - Vito Longo
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy;
| | - Roberta Di Fonte
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.Le O. Flacco, 65, 70124 Bari, Italy; (R.F.); (T.R.); (R.D.F.); (L.P.); (A.A.)
| | - Letizia Porcelli
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.Le O. Flacco, 65, 70124 Bari, Italy; (R.F.); (T.R.); (R.D.F.); (L.P.); (A.A.)
| | - Amalia Azzariti
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V.Le O. Flacco, 65, 70124 Bari, Italy; (R.F.); (T.R.); (R.D.F.); (L.P.); (A.A.)
| |
Collapse
|
4
|
Schöne N, Kemper M, Menck K, Evers G, Krekeler C, Schulze AB, Lenz G, Wardelmann E, Binder C, Bleckmann A. PD-L1 on large extracellular vesicles is a predictive biomarker for therapy response in tissue PD-L1-low and -negative patients with non-small cell lung cancer. J Extracell Vesicles 2024; 13:e12418. [PMID: 38453684 PMCID: PMC10920108 DOI: 10.1002/jev2.12418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/24/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
Immunotherapy has revolutionized the treatment of patients with non-small cell lung cancer (NSCLC). High expression of tissue PD-L1 (tPD-L1) is currently the only approved biomarker for predicting treatment response. However, even tPD-L1 low (1-49%) and absent (<1%) patients might benefit from immunotherapy but, to date, there is no reliable biomarker, that can predict response in this particular patient subgroup. This study aimed to test whether tumour-associated extracellular vesicles (EVs) could fill this gap. Using NSCLC cell lines, we identified a panel of tumour-related antigens that were enriched on large EVs (lEVs) compared to smaller EVs. The levels of lEVs carrying these antigens were significantly elevated in plasma of NSCLC patients (n = 108) and discriminated them from controls (n = 77). Among the tested antigens, we focused on programmed cell death ligand 1 (PD-L1), which is a well-known direct target for immunotherapy. In plasma lEVs, PD-L1 was mainly found on a population of CD45- /CD62P+ lEVs and thus seemed to be associated with platelet-derived vesicles. Patients with high baseline levels of PD-L1+ lEVs in blood showed a significantly better response to immunotherapy and prolonged survival. This was particularly true in the subgroup of NSCLC patients with low or absent tPD-L1 expression, thus identifying PD-L1-positive lEVs in plasma as a novel predictive and prognostic marker for immunotherapy.
Collapse
Affiliation(s)
- Nadja Schöne
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| | - Marcel Kemper
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| | - Kerstin Menck
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| | - Georg Evers
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| | - Carolin Krekeler
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| | - Arik Bernard Schulze
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| | - Georg Lenz
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
| | - Eva Wardelmann
- University of Münster, Gerhard‐Domagk‐Institute of PathologyMünsterGermany
| | - Claudia Binder
- University Medicine Göttingen, Clinic for Hematology/Medical OncologyGöttingenGermany
| | - Annalen Bleckmann
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| |
Collapse
|
5
|
Zheng Q, Li M, Qiu Y, Yang J, Cao Y. Overexpression of SLC35F2 is a potential prognostic biomarker for lung adenocarcinoma. Heliyon 2024; 10:e23828. [PMID: 38187235 PMCID: PMC10767229 DOI: 10.1016/j.heliyon.2023.e23828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 01/09/2024] Open
Abstract
Objective To explore the potential clinical and prognostic significance of Homo sapiens solute carrier family 35 member F2 (SLC35F2) in the context of lung adenocarcinoma (LUAD). Methods The expression pattern of SLC35F2 in LUAD tissues and normal tissues was analyzed in The Cancer Genome Atlas (TCGA) datasets and validated in 12 pairs of fresh clinical LUAD tissues and their corresponding adjacent normal tissues using quantitative real-time PCR (qRT-PCR) and western blotting. Immunohistochemistry (IHC) was used to assess the protein expression of SLC35F2 in 60 paraffin-embedded LUAD tissues, and its associations with clinicopathological parameters were further examined. The prognostic significance of SLC35F2 mRNA expression was also evaluated using the Kaplan-Meier method, and Cox regression models in LUAD patients from the TCGA database. The potential utility of SLC35F2 as an indicator of recurrence or metastasis was explored through the follow-up of selected clinical LUAD cases. Lastly, gene set enrichment analysis (GSEA) was conducted to investigate the underlying biological mechanisms and signaling pathways. Results Bioinformatics analysis utilizing the TCGA database indicated that SLC35F2 mRNA exhibited heightened expression in LUAD tissues when compared to normal tissues. These findings were further substantiated through the examination of 12 pairs of clinical LUAD tissues and their corresponding adjacent normal tissues, employing qRT-PCR and western blotting techniques. IHC results from a cohort of 60 LUAD patients demonstrated an up-regulation of SLC35F2 in 38 out of 60 individuals (63.3 %), which exhibited a significant correlation with tumor size, lymph node metastasis, and clinical stage (all P < 0.05). Both the Kaplan-Meier curve and the Cox proportional hazard analyses indicated a strong association between the up-regulation of SLC35F2 mRNA expression and unfavorable overall survival (OS) in patients with LUAD, as observed in the TCGA datasets (P < 0.05). The follow-up findings from select clinical LUAD cases provided evidence that the expression of SLC35F2 could serve as a dependable biomarker for monitoring the recurrence or metastasis. Additionally, the GSEA highlighted the enrichment of apoptosis, adhesion, small cell lung cancer (SCLC), and p53 signaling pathways in the subgroup of LUAD patients with elevated SLC35F2 expression. Conclusion SLC35F2 exhibited an up-regulated in both mRNA and protein expression, rendering it a valuable independent prognostic indicator for patients diagnosed with LUAD.
Collapse
Affiliation(s)
- Qingzhu Zheng
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Mingjie Li
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yingkun Qiu
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jiahao Yang
- Department of Laboratory Medicine, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, 350004, China
| | - Yingping Cao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| |
Collapse
|
6
|
Mehta D, Gupta D, Kafle A, Kaur S, Nagaiah TC. Advances and Challenges in Nanomaterial-Based Electrochemical Immunosensors for Small Cell Lung Cancer Biomarker Neuron-Specific Enolase. ACS OMEGA 2024; 9:33-51. [PMID: 38222505 PMCID: PMC10785636 DOI: 10.1021/acsomega.3c06388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/05/2023] [Accepted: 11/30/2023] [Indexed: 01/16/2024]
Abstract
Early and rapid detection of neuron-specific enolase (NSE) is highly significant, as it is putative biomarker for small-cell lung cancer as well as COVID-19. Electrochemical techniques have attracted substantial attention for the early detection of cancer biomarkers due to the important properties of simplicity, high sensitivity, specificity, low cost, and point-of-care detection. This work reviews the clinically relevant labeled and label-free electrochemical immunosensors developed so far for the analysis of NSE. The prevailing role of nanostructured materials as electrode matrices is thoroughly discussed. Subsequently, the key performances of various immunoassays are critically evaluated in terms of limit of detection, linear ranges, and incubation time for clinical translation. Electrochemical techniques coupled with screen-printed electrodes developing market level commercialization of NSE sensors is also discussed. Finally, the review concludes with the current challenges associated with available methods and provides a future outlook toward commercialization opportunities for easy detection of NSE.
Collapse
Affiliation(s)
- Daisy Mehta
- Department of Chemistry, Indian
Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Divyani Gupta
- Department of Chemistry, Indian
Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Alankar Kafle
- Department of Chemistry, Indian
Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Sukhjot Kaur
- Department of Chemistry, Indian
Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Tharamani C. Nagaiah
- Department of Chemistry, Indian
Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| |
Collapse
|
7
|
Wang J, Wang H, Xu J, Song Q, Zhou B, Shangguan J, Xue M, Wang Y. Identification of protein signatures for lung cancer subtypes based on BPSO method. PLoS One 2023; 18:e0294243. [PMID: 38060494 PMCID: PMC10703216 DOI: 10.1371/journal.pone.0294243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/27/2023] [Indexed: 12/18/2023] Open
Abstract
The objective of this study was to identify protein biomarkers that can distinguish between LUAD and LUSC, critical for personalized treatment plans. The proteomic profiling data of LUAD and LUSC samples from TCPA database, along with phenotype and survival information from TCGA database were downloaded and preprocessed for analysis. We used BPSO feature selection method and identified 10 candidate protein biomarkers that have better classifying performance, as analyzed by t-SNE and PCA algorithms. To explore the causalities among these proteins and their associations with tumor subtypes, we conducted the PCStable algorithm to construct a regulatory network. Results indicated that 4 proteins, MIG6, CD26, NF2, and INPP4B, were directly linked to the lung cancer subtypes and may be useful in guiding therapeutic decision-making. Besides, spearman correlation, Cox proportional hazard model and Kaplan-Meier curve was employed to validate the biological significance of the candidate proteins. In summary, our study highlights the importance of protein biomarkers in the classification of lung cancer subtypes and the potential of computational methods for identifying key biomarkers and understanding their underlying biological mechanisms.
Collapse
Affiliation(s)
- Jihan Wang
- Department of Basic Medicine, School of Medicine, Xi’an International University, Xi’an, 710077, China
| | - Hanping Wang
- Department of Basic Medicine, School of Medicine, Xi’an International University, Xi’an, 710077, China
- Engineering Research Center of Personalized Anti-aging Health Product Development and Transformation, Universities of Shaanxi Province, Xi’an, 710077, China
| | - Jing Xu
- Department of Basic Medicine, School of Medicine, Xi’an International University, Xi’an, 710077, China
| | - Qiying Song
- Department of Basic Medicine, School of Medicine, Xi’an International University, Xi’an, 710077, China
| | - Baozhen Zhou
- Department of Basic Medicine, School of Medicine, Xi’an International University, Xi’an, 710077, China
| | - Jingbo Shangguan
- Department of Basic Medicine, School of Medicine, Xi’an International University, Xi’an, 710077, China
| | - Mengju Xue
- Department of Basic Medicine, School of Medicine, Xi’an International University, Xi’an, 710077, China
| | - Yangyang Wang
- School of Electronics and Information, Northwestern Polytechnical University, Xi’an, 710129, China
| |
Collapse
|
8
|
Bibikova M, Fan J. Liquid biopsy for early detection of lung cancer. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:200-206. [PMID: 39171286 PMCID: PMC11332910 DOI: 10.1016/j.pccm.2023.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Indexed: 08/23/2024]
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. Early cancer detection plays an important role in improving treatment success and patient prognosis. In the past decade, liquid biopsy became an important tool for cancer diagnosis, as well as for treatment selection and response monitoring. Liquid biopsy is a broad term that defines a non-invasive test done on a sample of blood or other body fluid to look for cancer cells or other analytes that can include DNA, RNA, or other molecules released by tumor cells. Liquid biopsies mainly include circulating tumor DNA, circulating RNA, microRNA, proteins, circulating tumor cells, exosomes, and tumor-educated platelets. This review summarizes the progress and clinical application potential of liquid biopsy for early detection of lung cancer.
Collapse
Affiliation(s)
- Marina Bibikova
- AnchorDx, Inc., 46305 Landing Parkway, Fremont, CA 94538, USA
| | - Jianbing Fan
- Department of Pathology, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
9
|
Saputra HA, Jannath KA, Kim KB, Park DS, Shim YB. Conducting polymer composite-based biosensing materials for the diagnosis of lung cancer: A review. Int J Biol Macromol 2023; 252:126149. [PMID: 37582435 DOI: 10.1016/j.ijbiomac.2023.126149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/17/2023]
Abstract
The development of a simple and fast cancer detection method is crucial since early diagnosis is a key factor in increasing survival rates for lung cancer patients. Among several diagnosis methods, the electrochemical sensor is the most promising one due to its outstanding performance, portability, real-time analysis, robustness, amenability, and cost-effectiveness. Conducting polymer (CP) composites have been frequently used to fabricate a robust sensor device, owing to their excellent physical and electrochemical properties as well as biocompatibility with nontoxic effects on the biological system. This review brings up a brief overview of the importance of electrochemical biosensors for the early detection of lung cancer, with a detailed discussion on the design and development of CP composite materials for biosensor applications. The review covers the electrochemical sensing of numerous lung cancer markers employing composite electrodes based on the conducting polyterthiophene, poly(3,4-ethylenedioxythiophene), polyaniline, polypyrrole, molecularly imprinted polymers, and others. In addition, a hybrid of the electrochemical biosensors and other techniques was highlighted. The outlook was also briefly discussed for the development of CP composite-based electrochemical biosensors for POC diagnostic devices.
Collapse
Affiliation(s)
- Heru Agung Saputra
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Republic of Korea
| | - Khatun A Jannath
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Republic of Korea
| | - Kwang Bok Kim
- Digital Health Care R&D Department, Korea Institute of Industrial Technology, Cheonan 31056, Republic of Korea
| | - Deog-Su Park
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Republic of Korea
| | - Yoon-Bo Shim
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
10
|
Stephens EKH, Guayco Sigcha J, Lopez-Loo K, Yang IA, Marshall HM, Fong KM. Biomarkers of lung cancer for screening and in never-smokers-a narrative review. Transl Lung Cancer Res 2023; 12:2129-2145. [PMID: 38025810 PMCID: PMC10654441 DOI: 10.21037/tlcr-23-291] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023]
Abstract
Background and Objective Lung cancer is the leading cause of cancer-related mortality worldwide, partially attributed to late-stage diagnoses. In order to mitigate this, lung cancer screening (LCS) of high-risk patients is performed using low dose computed tomography (CT) scans, however this method is burdened by high false-positive rates and radiation exposure for patients. Further, screening programs focus on individuals with heavy smoking histories, and as such, never-smokers who may otherwise be at risk of lung cancer are often overlooked. To resolve these limitations, biomarkers have been posited as potential supplements or replacements to low-dose CT, and as such, a large body of research in this area has been produced. However, comparatively little information exists on their clinical efficacy and how this compares to current LCS strategies. Methods Here we conduct a search and narrative review of current literature surrounding biomarkers of lung cancer to supplement LCS, and biomarkers of lung cancer in never-smokers (LCINS). Key Content and Findings Many potential biomarkers of lung cancer have been identified with varying levels of sensitivity, specificity, clinical efficacy, and supporting evidence. Of the markers identified, multi-target panels of circulating microRNAs, lipids, and metabolites are likely the most clinically efficacious markers to aid current screening programs, as these provide the highest sensitivity and specificity for lung cancer detection. However, circulating lipid and metabolite levels are known to vary in numerous systemic pathologies, highlighting the need for further validation in large cohort randomised studies. Conclusions Lung cancer biomarkers is a fast-expanding area of research and numerous biomarkers with potential clinical applications have been identified. However, in all cases the level of evidence supporting clinical efficacy is not yet at a level at which it can be translated to clinical practice. The priority now should be to validate existing candidate markers in appropriate clinical contexts and work to integrating these into clinical practice.
Collapse
Affiliation(s)
- Edward K. H. Stephens
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Jazmin Guayco Sigcha
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Kenneth Lopez-Loo
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Ian A. Yang
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
| | - Henry M. Marshall
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
| | - Kwun M. Fong
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
| |
Collapse
|
11
|
McCabe A, Zaheed O, Derlipanska M, Merrin G, Dean K. The copious capabilities of non-coding RNAs in cancer regulation, diagnosis and treatment. Cancer Treat Res Commun 2023; 37:100768. [PMID: 37852123 DOI: 10.1016/j.ctarc.2023.100768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 10/20/2023]
Abstract
Globally, cancer is one of the leading causes of mortality, accounting for 10 million deaths per year. Non-coding RNAs (ncRNAs) play integral and diverse roles in cancer, possessing the ability to both promote oncogenesis and impede tumor formation. This review discusses the various roles of microRNAs, transfer RNA-derived small RNAs, long non-coding RNAs and lncRNA-derived microproteins in cancer progression and prevention. We highlight the diagnostic and therapeutic potential of these ncRNAs, with a particular focus on detection in liquid biopsies and targeting of ncRNAs with small inhibitory molecules. Ultimately, the biological functions of cancer-associated ncRNAs, as well as the development of ncRNA-based technologies, are compelling areas for further research, holding the possibility of revolutionizing cancer treatment and diagnosis.
Collapse
Affiliation(s)
- Aideen McCabe
- School of Biochemistry and Cell Biology, College of Science, Engineering and Food Science, University College Cork, Ireland; The SFI Centre for Research Training in Genomics Data Science, Ireland
| | - Oza Zaheed
- School of Biochemistry and Cell Biology, College of Science, Engineering and Food Science, University College Cork, Ireland; The SFI Centre for Research Training in Genomics Data Science, Ireland
| | - Magdalina Derlipanska
- School of Biochemistry and Cell Biology, College of Science, Engineering and Food Science, University College Cork, Ireland
| | - George Merrin
- School of Biochemistry and Cell Biology, College of Science, Engineering and Food Science, University College Cork, Ireland
| | - Kellie Dean
- School of Biochemistry and Cell Biology, College of Science, Engineering and Food Science, University College Cork, Ireland.
| |
Collapse
|
12
|
Kim H, Lee JK, Oh AC, Kim HR, Hong YJ. The Usefulness of the Ratio of Antigen-Autoantibody Immune Complexes to Their Free Antigens in the Diagnosis of Non-Small Cell Lung Cancer. Diagnostics (Basel) 2023; 13:2999. [PMID: 37761366 PMCID: PMC10529727 DOI: 10.3390/diagnostics13182999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/21/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Autoantibodies against specific lung cancer-associated antigens have been suggested for the performance of lung cancer diagnosis. This study aimed to evaluate the diagnostic performance of the antigen-autoantibody immune complex (AIC) against its free antigens for CYFRA21-1, ProGRP, neutrophil gelatinase-associated lipocalin (NGAL), and neuron-specific enolase (NSE) in non-small cell lung cancer (NSCLC). In total, 85 patients with NSCLC and 120 healthy controls (HCs) were examined using a 9-guanine DNA chip method. The ratios of AICs to their antigens and the combinations of ratios consisting of two to four markers were calculated. The levels of AICs for CYFRA21-1, ProGRP, NGAL, and NSE were higher than those for their free antigens in all participants. The levels of each free antigens distinguished patients with NSCLC from the HCs. The ratios of the AIC to its antigen and seven combinations of two to four ratios were significantly higher in patients with NSCLC than in the HCs. Excellent diagnostic performance was observed for all combination ratios (C4-1), with 85.9% sensitivity and 86.7% specificity at a 3.51 cut-off. Higher sensitivity was observed in the early stages (0-I) and adenocarcinoma than in stages II-IV and other pathological types. Combining all ratios of AICs and their antigens for all four markers was useful when diagnosing NSCLC.
Collapse
Affiliation(s)
- Heyjin Kim
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (J.K.L.)
| | - Jin Kyung Lee
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (J.K.L.)
| | - Ae-Chin Oh
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (J.K.L.)
| | - Hye-Ryoun Kim
- Division of Pulmonology, Department of Internal Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea;
| | - Young Jun Hong
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (J.K.L.)
| |
Collapse
|
13
|
Minamibata A, Kono Y, Arimoto T, Marunaka Y, Takayama K. Variability of serum CYFRA 21 - 1 and its susceptibility to clinical characteristics in individuals without cancer: a 4-year retrospective analysis. BMC Pulm Med 2023; 23:344. [PMID: 37705035 PMCID: PMC10500899 DOI: 10.1186/s12890-023-02650-x] [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: 05/26/2023] [Accepted: 09/11/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND CYFRA 21 - 1 is a useful marker for diagnosing and monitoring lung cancer. However, its stability remains unclear. Moreover, while its applicability to screening is now being investigated, CYFRA 21 - 1 levels in individuals without cancer, who are targets for cancer screening, have not yet been the focus of research. Therefore, the present study investigated variability in and the factors increasing serum CYFRA 21 - 1 levels. METHODS This retrospective study recruited 951 individuals undergoing annual medical examinations for six years. We used data obtained in the first four years. Variability in serum CYFRA 21 - 1 levels over a period of four years were investigated. CYFRA 21 - 1 was categorized as normal (≤ 3.5 ng/ml) or elevated (> 3.5 ng/ml). The rate of an elevated level in one visit and the transition from an elevated to normal level between visits were visualized. A multiple logistic regression model was used to study the relationships between the frequency of elevated CYFRA 21 - 1 levels and clinical characteristics, such as age, sex, body mass index, weight changes, and the smoking status. RESULTS Approximately 5% of subjects had elevated CYFRA 21 - 1 levels once in five tests over four years, while 15% had elevated CYFRA 21 - 1 levels once or more. Among subjects with elevated CYFRA 21 - 1 levels in one blood test, between 63 and 72% had normal levels in the next test. The median CYFRA 21 - 1 level in subjects with elevations in one blood test significantly decreased in the next test at all four time points. The frequency of elevated CYFRA 21 - 1 levels was associated with an older age [odds ratio (OR) = 6.99, 95% confidence interval (CI) = 3.01-16.2], current heavy smoking (OR = 3.46, 95% CI = 1.52-7.9), and weight loss (OR = 1.86, 95% CI = 1.07-3.24). CONCLUSIONS Variability in and the factors increasing serum CYFRA 21 - 1 levels beyond the cut-off value need to be considered when interpretating CYFRA 21 - 1 test results. The future application of CYFRA 21 - 1 to lung cancer screening may require more than a single measurement.
Collapse
Affiliation(s)
- Asami Minamibata
- Medical Research Institute, Kyoto Industrial Health Association, 67 Kita-Tsuboicho Nishinokyo Nakagyo-ku, Kyoto, 604-8472, Japan.
| | - Yoshihito Kono
- Medical Research Institute, Kyoto Industrial Health Association, 67 Kita-Tsuboicho Nishinokyo Nakagyo-ku, Kyoto, 604-8472, Japan
| | - Taichiro Arimoto
- Medical Research Institute, Kyoto Industrial Health Association, 67 Kita-Tsuboicho Nishinokyo Nakagyo-ku, Kyoto, 604-8472, Japan
| | - Yoshinori Marunaka
- Medical Research Institute, Kyoto Industrial Health Association, 67 Kita-Tsuboicho Nishinokyo Nakagyo-ku, Kyoto, 604-8472, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
14
|
Wei F, Yu P, Cheng J, Li F, Chia D, Wong DTW. Single-Droplet Microsensor for Ultra-Short Circulating EFGR Mutation Detection in Lung Cancer Based on Multiplex EFIRM Liquid Biopsy. Int J Mol Sci 2023; 24:10387. [PMID: 37373532 DOI: 10.3390/ijms241210387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/30/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Liquid biopsy is a rapidly emerging field that involves the minimal/non-invasive assessment of signature somatic mutations through the analysis of circulating tumor DNA (ctDNA) shed by tumor cells in bodily fluids. Broadly speaking, the unmet need in liquid biopsy lung cancer detection is the lack of a multiplex platform that can detect a mutation panel of lung cancer genes using a minimum amount of sample, especially for ultra-short ctDNA (usctDNA). Here, we developed a non-PCR and non-NGS-based single-droplet-based multiplexing microsensor technology, "Electric-Field-Induced Released and Measurement (EFIRM) Liquid Biopsy" (m-eLB), for lung cancer-associated usctDNA. The m-eLB provides a multiplexable assessment of usctDNA within a single droplet of biofluid in only one well of micro-electrodes, as each electrode is coated with different probes for the ctDNA. This m-eLB prototype demonstrates accuracy for three tyrosine-kinase-inhibitor-related EGFR target sequences in synthetic nucleotides. The accuracy of the multiplexing assay has an area under the curve (AUC) of 0.98 for L858R, 0.94 for Ex19 deletion, and 0.93 for T790M. In combination, the 3 EGFR assay has an AUC of 0.97 for the multiplexing assay.
Collapse
Affiliation(s)
- Fang Wei
- School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Peter Yu
- Department of Physics, University of California, Los Angeles, CA 90095, USA
| | - Jordan Cheng
- School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Feng Li
- School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - David Chia
- Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - David T W Wong
- School of Dentistry, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
15
|
Genet SAAM, Wolfs JRE, Vu CBAK, Wolter M, Broeren MAC, van Dongen J, Brunsveld L, Scharnhorst V, van de Kerkhof D. Analysis of Neuron-Specific enolase isozymes in human serum using immunoaffinity purification and liquid chromatography-tandem mass spectrometry quantification. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1223:123701. [PMID: 37086508 DOI: 10.1016/j.jchromb.2023.123701] [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: 01/25/2023] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/24/2023]
Abstract
Neuron-specific enolase (NSE) is a promising small-cell lung cancer (SCLC) biomarker composed of αγ and γγ isozyme dimers. As the conventional immunoassays are prone to interferences and cannot differentiate between the isozymes, we developed a multiplex immunoaffinity (IA) liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay for the quantification of NSEα and NSEγ in human serum. A calibrator was prepared by performing cold denaturation of recombinantly expressed αα and γγ enolase dimers to induce a new dimer equilibrium that was determined to be approximately 1αγ:1γγ:1αα. Selective sample purification was achieved by performing IA extraction using an antibody specific towards NSEγ. The isolated αγ and γγ dimers were denatured and trypsin digested to allow quantification of the selected signature peptides and their corresponding isotopically labelled peptide internal standard. The obtained linear dynamic ranges were determined to be 1.5-56 ng/mL and 0.64-167 ng/mL for NSEα and NSEγ (R2 = 0.88 and 0.97 respectively). Validation of the assay showed acceptable accuracy and precision for NSEα and NSEγ. The method was successfully applied to patient serum in which both isozymes were detected. Compared to the conventional immunoassay, substantially lower total NSE concentrations were measured in IA LC-MS/MS. With this multiplex IA LC-MS/MS assay, the clinical value of quantifying the individual isozymes can be explored. In addition, together with the calibrator described here, it may be applied to standardize NSE immunoassays across different platforms.
Collapse
Affiliation(s)
- Sylvia A A M Genet
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands; Catharina Hospital Eindhoven, Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands
| | - Jur R E Wolfs
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Chris B A K Vu
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Madita Wolter
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Maarten A C Broeren
- Máxima Medical Center, Eindhoven/Veldhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands
| | - Joost van Dongen
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands
| | - Volkher Scharnhorst
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands; Catharina Hospital Eindhoven, Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands
| | - Daan van de Kerkhof
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands; Catharina Hospital Eindhoven, Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands.
| |
Collapse
|
16
|
RGD-decorated PLGA nanoparticles improved effectiveness and safety of cisplatin for lung cancer therapy. Int J Pharm 2023; 633:122587. [PMID: 36623741 DOI: 10.1016/j.ijpharm.2023.122587] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/18/2022] [Accepted: 01/04/2023] [Indexed: 01/08/2023]
Abstract
Upon extensive pharmaceutical and biomedical research to treat lung cancer indicates that lung cancer remains one of the deadliest diseases and the leading cause of death in men and women worldwide. Lung cancer remains untreated and has a high mortality rate due to the limited potential for effective treatment with existing therapies. This highlights the urgent need to develop an effective, precise and sustainable solutions to treat lung cancer. In this study, we developed RGD receptor-targeted PLGA nanoparticles for the controlled and targeted co-delivery of cisplatin (CDDP) and upconversion nanoparticles (UCNP) in lung cancer therapy. Pluronic F127-RGD conjugate was synthesized by carbodiimide chemistry method and the conjugation was confirmed by FTIR and 1HNMR spectroscopy techniques. PLGA nanoparticles were developed by the double emulsification method, then the surface of the prepared nanoparticles was decorated with Pluronic F127-RGD conjugate. The prepared formulations were characterized for their particle size, polydispersity index, zeta potential, surface morphology, drug encapsulation efficiency, and in vitro drug release and haemolysis studies. Pharmacokinetic studies and safety parameters in BAL fluid were assessed in rats. Histopathology of rat lung tissue was performed. The obtained results of particle sizes of the nanoparticle formulations were found 100-200 nm, indicating the homogeneity of dispersed colloidal nanoparticles formulations. Transmission Electron Microscopy (TEM) revealed the spherical shape of the prepared nanoparticles. The drug encapsulation efficiency of PLGA nanoparticles was found to range from 60% to 80% with different nanoparticles counterparts. RGD receptor-targeted PLGA nanoparticles showed controlled drug release for up to 72 h. Further, RGD receptor-targeted PLGA nanoparticles achieved higher cytotoxicity in compared to CFT, CFT, and Ciszest-50 (marketed CDDP injection). The pharmacokinetic study revealed that RGD receptor-targeted PLGA nanoparticles were 4.6-fold more effective than Ciszest-50. Furthermore, RGD receptor-targeted PLGA nanoparticles exhibited negligible damage to lung tissue, low systemic toxicity, and high biocompatible and safety in lung tissue. The results of RGD receptor-targeted PLGA nanoparticles indicated that it is a promising anticancer system that could further exploited as a potent therapeutic approach for lung cancer.
Collapse
|
17
|
Visser E, Genet SAAM, de Kock RPPA, van den Borne BEEM, Youssef-El Soud M, Belderbos HNA, Stege G, de Saegher MEA, van 't Westeinde SC, Brunsveld L, Broeren MAC, van de Kerkhof D, Deiman BALM, Eduati F, Scharnhorst V. Liquid biopsy-based decision support algorithms for diagnosis and subtyping of lung cancer. Lung Cancer 2023; 178:28-36. [PMID: 36773458 DOI: 10.1016/j.lungcan.2023.01.014] [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: 11/04/2022] [Revised: 01/11/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Pathologic subtyping of tissue biopsies is the gold standard for the diagnosis of lung cancer (LC), which could be complicated in cases of e.g. inconclusive tissue biopsies or unreachable tumors. The diagnosis of LC could be supported in a minimally invasive manner using protein tumor markers (TMs) and circulating tumor DNA (ctDNA) measured in liquid biopsies (LBx). This study evaluates the performance of LBx-based decision-support algorithms for the diagnosis of LC and subtyping into small- and non-small-cell lung cancer (SCLC and NSCLC) aiming to directly impact clinical practice. MATERIALS AND METHODS In this multicenter prospective study (NL9146), eight protein TMs (CA125, CA15.3, CEA, CYFRA 21-1, HE4, NSE, proGRP and SCCA) and ctDNA mutations in EGFR, KRAS and BRAF were analyzed in blood of 1096 patients suspected of LC. The performance of individual and combined TMs to identify LC, NSCLC or SCLC was established by evaluating logistic regression models at pre-specified positive predictive values (PPV) of ≥95% or ≥98%. The most informative protein TMs included in the multi-parametric models were selected by recursive feature elimination. RESULTS Single TMs could identify LC, NSCLC and SCLC patients with 46%, 25% and 40% sensitivity, respectively, at pre-specified PPVs. Multi-parametric models combining TMs and ctDNA significantly improved sensitivities to 65%, 67% and 50%, respectively. CONCLUSION In patients suspected of LC, the LBx-based decision-support algorithms allowed identification of about two-thirds of all LC and NSCLC patients and half of SCLC patients. These models therefore show clinical value and may support LC diagnostics, especially in patients for whom pathologic subtyping is impossible or incomplete.
Collapse
Affiliation(s)
- Esther Visser
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Catharina Hospital Eindhoven, Eindhoven, the Netherlands; Máxima Medical Center, Eindhoven/Veldhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands.
| | - Sylvia A A M Genet
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Catharina Hospital Eindhoven, Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Remco P P A de Kock
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Catharina Hospital Eindhoven, Eindhoven, the Netherlands; Máxima Medical Center, Eindhoven/Veldhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands
| | | | | | | | | | | | | | - Luc Brunsveld
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Maarten A C Broeren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Máxima Medical Center, Eindhoven/Veldhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands
| | - Daan van de Kerkhof
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Catharina Hospital Eindhoven, Eindhoven, the Netherlands
| | - Birgit A L M Deiman
- Catharina Hospital Eindhoven, Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands
| | - Federica Eduati
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands; Eindhoven Artificial Intelligence Systems Institute, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Volkher Scharnhorst
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Catharina Hospital Eindhoven, Eindhoven, the Netherlands; Expert Center Clinical Chemistry Eindhoven, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands; Eindhoven Artificial Intelligence Systems Institute, Eindhoven University of Technology, Eindhoven, the Netherlands
| |
Collapse
|
18
|
Casagrande GMS, Silva MDO, Reis RM, Leal LF. Liquid Biopsy for Lung Cancer: Up-to-Date and Perspectives for Screening Programs. Int J Mol Sci 2023; 24:2505. [PMID: 36768828 PMCID: PMC9917347 DOI: 10.3390/ijms24032505] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/09/2022] [Accepted: 12/19/2022] [Indexed: 01/31/2023] Open
Abstract
Lung cancer is the deadliest cancer worldwide. Tissue biopsy is currently employed for the diagnosis and molecular stratification of lung cancer. Liquid biopsy is a minimally invasive approach to determine biomarkers from body fluids, such as blood, urine, sputum, and saliva. Tumor cells release cfDNA, ctDNA, exosomes, miRNAs, circRNAs, CTCs, and DNA methylated fragments, among others, which can be successfully used as biomarkers for diagnosis, prognosis, and prediction of treatment response. Predictive biomarkers are well-established for managing lung cancer, and liquid biopsy options have emerged in the last few years. Currently, detecting EGFR p.(Tyr790Met) mutation in plasma samples from lung cancer patients has been used for predicting response and monitoring tyrosine kinase inhibitors (TKi)-treated patients with lung cancer. In addition, many efforts continue to bring more sensitive technologies to improve the detection of clinically relevant biomarkers for lung cancer. Moreover, liquid biopsy can dramatically decrease the turnaround time for laboratory reports, accelerating the beginning of treatment and improving the overall survival of lung cancer patients. Herein, we summarized all available and emerging approaches of liquid biopsy-techniques, molecules, and sample type-for lung cancer.
Collapse
Affiliation(s)
| | - Marcela de Oliveira Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331 Rua Antenor Duarte Vilela, Barretos 14784-400, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331 Rua Antenor Duarte Vilela, Barretos 14784-400, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Letícia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331 Rua Antenor Duarte Vilela, Barretos 14784-400, Brazil
- Barretos School of Medicine Dr. Paulo Prata—FACISB, Barretos 14785-002, Brazil
| |
Collapse
|
19
|
Liu B, Li Y, Suo L, Zhang W, Cao H, Wang R, Luan J, Yu X, Dong L, Wang W, Xu S, Lu S, Shi M. Characterizing microbiota and metabolomics analysis to identify candidate biomarkers in lung cancer. Front Oncol 2022; 12:1058436. [PMID: 36457513 PMCID: PMC9705781 DOI: 10.3389/fonc.2022.1058436] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/01/2022] [Indexed: 09/21/2023] Open
Abstract
Background Lung cancer is the leading malignant disease and cause of cancer-related death worldwide. Most patients with lung cancer had insignificant early symptoms so that most of them were diagnosed at an advanced stage. In addition to factors such as smoking, pollution, lung microbiome and its metabolites play vital roles in the development of lung cancer. However, the interaction between lung microbiota and carcinogenesis is lack of systematically characterized and controversial. Therefore, the purpose of this study was to excavate the features of the lung microbiota and metabolites in patients and verify potential biomarkers for lung cancer diagnosis. Methods Lung tissue flushing solutions and bronchoalveolar lavage fluid samples came from patients with lung cancer and non-lung cancer. The composition and variations of the microbiota and metabolites in samples were explored using muti-omics technologies including 16S rRNA amplicon sequencing, metagenomics and metabolomics. Results The metabolomics analysis indicated that 40 different metabolites, such as 9,10-DHOME, sphingosine, and cysteinyl-valine, were statistically significant between two groups (VIP > 1 and P < 0.05). These metabolites were significantly enriched into 11 signal pathways including sphingolipid, autophagy and apoptosis signaling pathway (P < 0.05). The analysis of lung microbiota showed that significant changes reflected the decrease of microbial diversity, changes of distribution of microbial taxa, and variability of the correlation networks of lung microbiota in lung cancer patients. In particular, we found that oral commensal microbiota and multiple probiotics might be connected with the occurrence and progression of lung cancer. Moreover, our study found 3 metabolites and 9 species with significantly differences, which might be regarded as the potential clinical diagnostic markers associated with lung cancer. Conclusions Lung microbiota and metabolites might play important roles in the pathogenesis of lung cancer, and the altered metabolites and microbiota might have the potential to be clinical diagnostic markers and therapeutic targets associated with lung cancer.
Collapse
Affiliation(s)
- Bo Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
- Department of Pulmonary and Critical Care Medicine, Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, China
- Department of Pulmonary and Critical Care Medicine, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yige Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Lijun Suo
- Department of Pulmonary and Critical Care Medicine, Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, China
| | - Wei Zhang
- Department of Thoracic Surgery, Zibo Municipal Hospital, Zibo, China
| | - Hongyun Cao
- Department of Pulmonary and Critical Care Medicine, Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, China
| | - Ruicai Wang
- Department of Pathology, Zibo Municipal Hospital, Zibo, China
| | - Jiahui Luan
- Department of Pulmonary and Critical Care Medicine, Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, China
| | - Xiaofeng Yu
- Department of Pulmonary and Critical Care Medicine, Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, China
| | - Liang Dong
- Department of Pulmonary and Critical Care Medicine, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Wenjing Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Shiyang Xu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, China
| | - Shiyong Lu
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, China
| | - Mei Shi
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, China
| |
Collapse
|
20
|
Tahanovich AD, Kauhanka NN, Murashka DI, Kolb AV, Prokhorova VI, Got'ko OV, Derzhavets LA. Preoperative blood markers for prediction of recurrence-free survival after surgical treatment of patients with stage III lung adenocarcinoma. Klin Lab Diagn 2022; 67:640-646. [PMID: 36398772 DOI: 10.51620/0869-2084-2022-67-11-640-646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The possibility of the preoperative level of 42 indicators characterizing the cellular composition and metabolism in blood of patients with stage III lung adenocarcinoma (AC) to predict their relapse-free survival was studied. Blood samples of 451 patients with newly diagnosed AK stage III after their surgical treatment (resection volume - R0) have been investigated. The duration of the relapse-free period (period of observation - 1 year), cellular composition of the blood, concentration of C-RP, albumin, Cyfra 21-1 antigens, SCC, TPA, chemokines CXCL5, CXCL8, pyruvate kinase TuM2 PK isoenzyme, HIF-1α and hyaluronic acid in blood serum so as the proportion of blood cells with CXCR1 and CXCR2, CD44V6 receptors in blood serum were measured. To determine the dependence of the duration of the relapse-free period after the treatment on the observation time, Kaplan-Meier graphs were built. The relationship between the determined parameters and survival was judged using single- and multi-factor Cox proportional hazard models. Comparison of groups with different risk of AK recurrence was performed using the Log Rank test and χ2. The assessment of the predictive information content of laboratory tests was carried out using ROC analysis. It was shown that the concentration of monocytes, eosinophilic leukocytes, the relative quantity of lymphocytes with CXCR1 receptor, the level of Cyfra 21-1 before surgical treatment were associated with the duration of the relapse-free period. A regression equation was compiled, which included the level of Cyfra 21-1, relative content of lymphocytes with CXCR1, and the eosinophilic leukocytes / monocytes ratio. Based on the threshold value Y=0,597, a Kaplan-Meier plot of patient survival was built and the results of it correspond to the TNM stratification. The prognostic sensitivity of the results of the equation - 85,7%, the specificity - 94,7%.
Collapse
Affiliation(s)
| | | | | | - A V Kolb
- Belarusian State Medical University
| | | | - O V Got'ko
- National Centre of oncology and medical radiology
| | | |
Collapse
|
21
|
Davies M, Davey MG, Miller N. The Potential of MicroRNAs as Clinical Biomarkers to Aid Ovarian Cancer Diagnosis and Treatment. Genes (Basel) 2022; 13:2054. [PMID: 36360295 PMCID: PMC9690044 DOI: 10.3390/genes13112054] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/24/2022] [Accepted: 11/04/2022] [Indexed: 07/30/2023] Open
Abstract
Ovarian cancer is a commonly diagnosed malignancy in women. When diagnosed at an early stage, survival outcomes are favourable for the vast majority, with up to 90% of ovarian cancer patients being free of disease at 5 years follow-up. Unfortunately, ovarian cancer is typically diagnosed at an advanced stage due to the majority of patients remaining asymptomatic until the cancer has metastasised, resulting in poor outcomes for the majority. While the molecular era has facilitated the subclassification of the disease into distinct clinical subtypes, ovarian cancer remains managed and treated as a single disease entity. MicroRNAs (miRNAs) are small (19-25 nucleotides), endogenous molecules which are integral to regulating gene expression. Aberrant miRNA expression profiles have been described in several cancers, and have been implicated to be useful biomarkers which may aid cancer diagnostics and treatment. Several preliminary studies have identified candidate tumour suppressor and oncogenic miRNAs which may be involved in the development and progression of ovarian cancer, highlighting their candidacy as oncological biomarkers; understanding the mechanisms by which these miRNAs regulate the key processes involved in oncogenesis can improve our overall understanding of cancer development and identify novel biomarkers and therapeutic targets. This review highlights the potential role of miRNAs which may be utilised to aid diagnosis, estimate prognosis and enhance therapeutic strategies in the management of primary ovarian cancer.
Collapse
|
22
|
Bracht T, Kleefisch D, Schork K, Witzke KE, Chen W, Bayer M, Hovanec J, Johnen G, Meier S, Ko YD, Behrens T, Brüning T, Fassunke J, Buettner R, Uszkoreit J, Adamzik M, Eisenacher M, Sitek B. Plasma Proteomics Enable Differentiation of Lung Adenocarcinoma from Chronic Obstructive Pulmonary Disease (COPD). Int J Mol Sci 2022; 23:ijms231911242. [PMID: 36232544 PMCID: PMC9569607 DOI: 10.3390/ijms231911242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major risk factor for the development of lung adenocarcinoma (AC). AC often develops on underlying COPD; thus, the differentiation of both entities by biomarker is challenging. Although survival of AC patients strongly depends on early diagnosis, a biomarker panel for AC detection and differentiation from COPD is still missing. Plasma samples from 176 patients with AC with or without underlying COPD, COPD patients, and hospital controls were analyzed using mass-spectrometry-based proteomics. We performed univariate statistics and additionally evaluated machine learning algorithms regarding the differentiation of AC vs. COPD and AC with COPD vs. COPD. Univariate statistics revealed significantly regulated proteins that were significantly regulated between the patient groups. Furthermore, random forest classification yielded the best performance for differentiation of AC vs. COPD (area under the curve (AUC) 0.935) and AC with COPD vs. COPD (AUC 0.916). The most influential proteins were identified by permutation feature importance and compared to those identified by univariate testing. We demonstrate the great potential of machine learning for differentiation of highly similar disease entities and present a panel of biomarker candidates that should be considered for the development of a future biomarker panel.
Collapse
Affiliation(s)
- Thilo Bracht
- Clinic for Anesthesiology, Intensive Care and Pain Therapy, University Medical Center Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Correspondence: (T.B.); (B.S.); Tel.: +49-234-32-29985 (T.B.); +49-234-32-24362 (B.S.)
| | - Daniel Kleefisch
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Karin Schork
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Kathrin E. Witzke
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Weiqiang Chen
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Malte Bayer
- Clinic for Anesthesiology, Intensive Care and Pain Therapy, University Medical Center Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Jan Hovanec
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA), 44789 Bochum, Germany
| | - Georg Johnen
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA), 44789 Bochum, Germany
| | - Swetlana Meier
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA), 44789 Bochum, Germany
| | - Yon-Dschun Ko
- Department of Internal Medicine, Johanniter-Kliniken Bonn GmbH, Johanniter Krankenhaus, 53113 Bonn, Germany
| | - Thomas Behrens
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA), 44789 Bochum, Germany
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr University Bochum (IPA), 44789 Bochum, Germany
| | - Jana Fassunke
- Institute of Pathology, Medical Faculty and Center for Molecular Medicine (CMMC), University of Cologne, 50924 Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, Medical Faculty and Center for Molecular Medicine (CMMC), University of Cologne, 50924 Cologne, Germany
| | - Julian Uszkoreit
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Michael Adamzik
- Clinic for Anesthesiology, Intensive Care and Pain Therapy, University Medical Center Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
| | - Martin Eisenacher
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Barbara Sitek
- Clinic for Anesthesiology, Intensive Care and Pain Therapy, University Medical Center Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
- Medizinisches Proteom-Center, Ruhr-University Bochum, 44801 Bochum, Germany
- Correspondence: (T.B.); (B.S.); Tel.: +49-234-32-29985 (T.B.); +49-234-32-24362 (B.S.)
| |
Collapse
|
23
|
Joshi S, Kallappa S, Kumar P, Shukla S, Ghosh R. Simple diagnosis of cancer by detecting CEA and CYFRA 21-1 in saliva using electronic sensors. Sci Rep 2022; 12:15315. [PMID: 36097151 PMCID: PMC9468134 DOI: 10.1038/s41598-022-19593-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/31/2022] [Indexed: 11/10/2022] Open
Abstract
One way of early diagnosis of cancer is by detecting the biomarkers that get introduced into easily accessible body fluids. We report the development of portable and rapid electronic biosensors for quantitative detection of two secretive cancer biomarkers-Carcinoembryonic antigen (CEA) and Cytokeratin fragment 19 (CYFRA 21-1). The reduced graphene oxide (rGO)/ melamine (MEL)/antibodies/ bovine serum albumin (BSA) based devices were tested for 1 pg/mL to 800 ng/mL of CEA and CYFRA 21-1. The responses of the sensors ranged from 7.14 to 59.1% and from 6.18 to 64% for 1 pg/mL to 800 ng/mL CEA and CYFRA 21-1 respectively. A read-out circuit was assembled to develop a portable prototype which was used to assess the concentrations of the two antigens present in saliva samples of 14 subjects. The prototype could accurately discriminate between 9 oral squamous cell carcinoma patients and 5 healthy controls.
Collapse
Affiliation(s)
- Sowmya Joshi
- Department of Electrical Engineering, Indian Institute of Technology Dharwad, Dharwad, 580011, Karnataka, India
| | - Shashidhar Kallappa
- Department of Surgical Oncology, Karnataka Institute of Medical Sciences, Hubli, 580029, Karnataka, India
| | - Pranjal Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Dharwad, Dharwad, 580011, Karnataka, India
| | - Sudhanshu Shukla
- Department of Biosciences and Bioengineering, Indian Institute of Technology Dharwad, Dharwad, 580011, Karnataka, India
| | - Ruma Ghosh
- Department of Electrical Engineering, Indian Institute of Technology Dharwad, Dharwad, 580011, Karnataka, India.
| |
Collapse
|
24
|
Zahedi S, Carvalho AS, Ejtehadifar M, Beck HC, Rei N, Luis A, Borralho P, Bugalho A, Matthiesen R. Assessment of a Large-Scale Unbiased Malignant Pleural Effusion Proteomics Study of a Real-Life Cohort. Cancers (Basel) 2022; 14:cancers14184366. [PMID: 36139528 PMCID: PMC9496668 DOI: 10.3390/cancers14184366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Pleural effusion (PE) occurs as a consequence of various pathologies. Malignant effusion due to lung cancer is one of the most frequent causes. A method for accurate differentiation of malignant from benign PE is an unmet clinical need. Proteomics profiling of PE has shown promising results. However, mass spectrometry (MS) analysis typically involves the tedious elimination of abundant proteins before analysis, and clinical annotation of proteomics profiled cohorts is limited. This study compares the proteomes of malignant PE and nonmalignant PE, identifies lung cancer malignant markers in agreement with other studies, and identifies markers strongly associated with patient survival. Abstract Background: Pleural effusion (PE) is common in advanced-stage lung cancer patients and is related to poor prognosis. Identification of cancer cells is the standard method for the diagnosis of a malignant PE (MPE). However, it only has moderate sensitivity. Thus, more sensitive diagnostic tools are urgently needed. Methods: The present study aimed to discover potential protein targets to distinguish malignant pleural effusion (MPE) from other non-malignant pathologies. We have collected PE from 97 patients to explore PE proteomes by applying state-of-the-art liquid chromatography-mass spectrometry (LC-MS) to identify potential biomarkers that correlate with immunohistochemistry assessment of tumor biopsy or with survival data. Functional analyses were performed to elucidate functional differences in PE proteins in malignant and benign samples. Results were integrated into a clinical risk prediction model to identify likely malignant cases. Sensitivity, specificity, and negative predictive value were calculated. Results: In total, 1689 individual proteins were identified by MS-based proteomics analysis of the 97 PE samples, of which 35 were diagnosed as malignant. A comparison between MPE and benign PE (BPE) identified 58 differential regulated proteins after correction of the p-values for multiple testing. Furthermore, functional analysis revealed an up-regulation of matrix intermediate filaments and cellular movement-related proteins. Additionally, gene ontology analysis identified the involvement of metabolic pathways such as glycolysis/gluconeogenesis, pyruvate metabolism and cysteine and methionine metabolism. Conclusion: This study demonstrated a partial least squares regression model with an area under the curve of 98 and an accuracy of 0.92 when evaluated on the holdout test data set. Furthermore, highly significant survival markers were identified (e.g., PSME1 with a log-rank of 1.68 × 10−6).
Collapse
Affiliation(s)
- Sara Zahedi
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal
| | - Ana Sofia Carvalho
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal
| | - Mostafa Ejtehadifar
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal
| | - Hans C. Beck
- Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark
| | - Nádia Rei
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal
| | - Ana Luis
- Hospital CUF Descobertas, CUF Oncologia, 1998-018 Lisbon, Portugal
| | - Paula Borralho
- Hospital CUF Descobertas, CUF Oncologia, 1998-018 Lisbon, Portugal
| | - António Bugalho
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal
- Hospital CUF Descobertas, CUF Oncologia, 1998-018 Lisbon, Portugal
- Correspondence: (A.B.); (R.M.)
| | - Rune Matthiesen
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal
- Correspondence: (A.B.); (R.M.)
| |
Collapse
|
25
|
Anju, Kumar A, Yadav P, Navik U, Jaitak V. Chemical composition , in vitro and in silico evaluation of essential oil from Eucalyptus tereticornis leaves for lung cancer. Nat Prod Res 2022; 37:1656-1661. [PMID: 35938316 DOI: 10.1080/14786419.2022.2107642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Chemical composition of the essential oil (EO) of Eucalyptus tereticornis leaves was studied by gas chromatography-mass spectrometry. Forty-five constituents were identified in the oil hydrodistilled from the sample collected from Ghudda Village, Bathinda (Pb), India of which eucalyptol (34.39%) and ledol (9.92%) were the major constituents. In vitro antioxidant and anticancer potential of EO was analysed by DPPH 2,2-diphenylpicrylhydrazyl (DPPH) and MTT assay. The percentage free radical scavenging activity was found to be 63.77%. The antiproliferative activity was analysed using MTT assay in adenocarcinomic human alveolar basal epithelial A549 cancer cell line and showed IC50 value of 47.14 µg/ml. In silico study of EO, constituents were performed using Maestro 12.9 against EGFR (PDB ID-2RGP). Five constituents from EO showed high dockscore as compared to standard Mobicertinib which indicated the effectiveness of oil constituents against lung cancer.
Collapse
Affiliation(s)
- Anju
- Laboratory of Natural Product Chemistry, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda, India
| | - Amit Kumar
- Laboratory of Natural Product Chemistry, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda, India
| | - Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Vikas Jaitak
- Laboratory of Natural Product Chemistry, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda, India
| |
Collapse
|
26
|
Cheng TM, Chu HY, Huang HM, Li ZL, Chen CY, Shih YJ, Whang-Peng J, Cheng RH, Mo JK, Lin HY, Wang K. Toxicologic Concerns with Current Medical Nanoparticles. Int J Mol Sci 2022; 23:7597. [PMID: 35886945 PMCID: PMC9322368 DOI: 10.3390/ijms23147597] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/28/2022] [Accepted: 07/05/2022] [Indexed: 02/05/2023] Open
Abstract
Nanotechnology is one of the scientific advances in technology. Nanoparticles (NPs) are small materials ranging from 1 to 100 nm. When the shape of the supplied nanoparticles changes, the physiological response of the cells can be very different. Several characteristics of NPs such as the composition, surface chemistry, surface charge, and shape are also important parameters affecting the toxicity of nanomaterials. This review covered specific topics that address the effects of NPs on nanomedicine. Furthermore, mechanisms of different types of nanomaterial-induced cytotoxicities were described. The distributions of different NPs in organs and their adverse effects were also emphasized. This review provides insight into the scientific community interested in nano(bio)technology, nanomedicine, and nanotoxicology. The content may also be of interest to a broad range of scientists.
Collapse
Affiliation(s)
- Tsai-Mu Cheng
- Graduate Institute for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (T.-M.C.); (H.-Y.C.)
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsiu-Yi Chu
- Graduate Institute for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (T.-M.C.); (H.-Y.C.)
| | - Haw-Ming Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Zi-Lin Li
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (Z.-L.L.); (C.-Y.C.); (Y.-J.S.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Chiang-Ying Chen
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (Z.-L.L.); (C.-Y.C.); (Y.-J.S.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Ya-Jung Shih
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (Z.-L.L.); (C.-Y.C.); (Y.-J.S.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | | | - R. Holland Cheng
- Department of Molecular & Cellular Biology, University of California, Davis, CA 95616, USA;
| | - Ju-Ku Mo
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Hung-Yun Lin
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Molecular & Cellular Biology, University of California, Davis, CA 95616, USA;
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | - Kuan Wang
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (Z.-L.L.); (C.-Y.C.); (Y.-J.S.)
| |
Collapse
|
27
|
Ma C, Zhao J, Wu Y, Wang J, Wang H. Diagnostic value of abnormal chromosome 3p genes in small‑cell lung cancer. Oncol Lett 2022; 24:209. [PMID: 35720498 PMCID: PMC9185142 DOI: 10.3892/ol.2022.13330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/20/2022] [Indexed: 12/24/2022] Open
Abstract
The diagnosis of small cell lung carcinoma (SCLC) remains a great challenge. Changes in chromosome 3p (chr3) genes are usually observed in the pathogenesis of lung cancer, which suggests that these chr3 genes may be a diagnostic marker in the early stage of SCLC. The present study explored the diagnostic value of the chr3 gene in SCLC using Bioinformatics. Furthermore, reverse transcription-quantitative PCR (RT-qPCR) was used to reveal the expression patterns of diagnostic biomarkers in human pulmonary alveolar epithelial cells and in the SCLC cell line NCI-H146. A total of 33 differentially expressed (DE) chr3 genes and 1,156 module genes associated with clinical features of patients with SCLC were identified and functional enrichment analysis indicated that all these genes were significantly enriched in cell cycle terms. The area under the receiver operating characteristic curve demonstrated that the overlapping genes of the DE-chr3 and module genes, namely cell division cycle 25 A (CDC25A), FYVE and coiled-coil domain autophagy adaptor 1 (FYCO1) and lipid raft linker 1 (RFTN1), were relatively accurate in distinguishing normal from SCLC samples, and may thus be considered diagnostic biomarkers. CDC25A was overexpressed in SCLC samples, while FYCO1 and RFTN1 were highly expressed in normal samples, as evidenced by the RT-qPCR results. Single-gene gene set enrichment analysis suggested that the diagnostic biomarkers were significantly associated with cell cycle, ATP-binding cassette transporter, immune cell differentiation, immune response and multiple respiratory disease pathways. Furthermore, a total of 141 drugs were predicted by The Comparative Toxicogenomics Database to be able to modulate the expression of the diagnostic biomarkers, of which 8 drugs were shared among the three aforementioned diagnostic biomarkers. The present study identified three novel and powerful diagnostic biomarkers for SCLC based on chr3 genes. Suggestions for the development and selection of drugs for clinical treatment based on diagnostic biomarkers were also provided.
Collapse
Affiliation(s)
- Chunxu Ma
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Jihua Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Ying Wu
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Jun Wang
- College of Physical Education, Yunnan Agricultural University, Kunming, Yunnan 650201, P.R. China
| | - Hao Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| |
Collapse
|
28
|
Wang Y, Gao Y, Yin Y, Pan Y, Wang Y, Song Y. Nanomaterial-assisted microfluidics for multiplex assays. Mikrochim Acta 2022; 189:139. [PMID: 35275267 DOI: 10.1007/s00604-022-05226-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/10/2022] [Indexed: 02/07/2023]
Abstract
Simultaneous detection of different biomarkers from a single specimen in a single test, allowing more rapid, efficient, and low-cost analysis, is of great significance for accurate diagnosis of disease and efficient monitoring of therapy. Recently, developments in microfabrication and nanotechnology have advanced the integration of nanomaterials in microfluidic devices toward multiplex assays of biomarkers, combining both the advantages of microfluidics and the unique properties of nanomaterials. In this review, we focus on the state of the art in multiplexed detection of biomarkers based on nanomaterial-assisted microfluidics. Following an overview of the typical microfluidic analytical techniques and the most commonly used nanomaterials for biochemistry analysis, we highlight in detail the nanomaterial-assisted microfluidic strategies for different biomarkers. These highly integrated platforms with minimum sample consumption, high sensitivity and specificity, low detection limit, enhanced signals, and reduced detection time have been extensively applied in various domains and show great potential in future point-of-care testing and clinical diagnostics.
Collapse
Affiliation(s)
- Yanping Wang
- Sino-French Engineer School, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Yanfeng Gao
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| | - Yi Yin
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yongchun Pan
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| | - Yuzhen Wang
- Key Laboratory of Flexible Electronics & Institute of Advanced Materials, Jiangsu National Synergistic Innovation Center for Advanced Materials, Nanjing Tech University, Nanjing, 211816, China
| | - Yujun Song
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
29
|
Insights into the Unique Lung Microbiota Profile of Pulmonary Tuberculosis Patients Using Metagenomic Next-Generation Sequencing. Microbiol Spectr 2022; 10:e0190121. [PMID: 35196800 PMCID: PMC8865484 DOI: 10.1128/spectrum.01901-21] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The microbiota plays an important role in human health and disease development. The lung microbiota profile in pulmonary tuberculosis (TB) patients and the effects of anti-TB treatment on the profile need to be determined thoroughly and comprehensively. This study primarily aimed to determine the lung microbiota profile associated with pulmonary TB and characterize the longitudinal changes during anti-TB treatment. A total of 53 participants, comprising 8 healthy individuals, 12 untreated pulmonary TB patients, 15 treated pulmonary TB patients, 11 cured pulmonary TB patients, and 7 lung cancer patients, were recruited in the present study. Bronchioalveolar lavage fluid (BALF) samples were collected from the above participants, and throat swabs were taken from healthy individuals. Microbiomes in the samples were examined using metagenomic next-generation sequencing (mNGS). Differences in microbiota profiles were determined through a comparison of the indicated groups. Our findings indicated that the BALF samples displayed decreased richness and diversity of the microbiota compared to those of the throat swab samples, and these two kinds of samples exhibited obvious separation on principal-coordinate analysis (PCoA) plots. Untreated pulmonary TB patients displayed a unique lung microbiota signature distinct from that of healthy individuals and lung cancer patients. Our data first demonstrated that anti-TB treatment with first-line drugs increases alpha diversity and significantly affects the beta diversity of the lung microbiota, while it also induces antibiotic resistance genes (ARGs). IMPORTANCE Characterization of the lung microbiota could lead to a better understanding of the pathogenesis of pulmonary TB. Here, we applied the metagenomic shotgun sequencing instead of 16S rRNA sequencing method to characterize the lung microbiota using the BALF samples instead of sputum. We found that alterations in the lung microbiota are associated with TB infection and that anti-TB treatment significantly affects the alpha and beta diversity of the lung microbiota in pulmonary TB patients. These findings could help us better understand TB pathogenesis.
Collapse
|
30
|
Boyero L, Martin-Padron J, Fárez-Vidal ME, Rodriguez MI, Andrades Á, Peinado P, Arenas AM, Ritoré-Salazar F, Alvarez-Perez JC, Cuadros M, Medina PP. PKP1 and MYC create a feedforward loop linking transcription and translation in squamous cell lung cancer. Cell Oncol (Dordr) 2022; 45:323-332. [PMID: 35182388 DOI: 10.1007/s13402-022-00660-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2022] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Plakophilin 1 (PKP1) is well-known as an important component of the desmosome, a cell structure specialized in spot-like cell-to-cell adhesion. Although desmosomes have generally been associated with tumor suppressor functions, we recently found that PKP1 is recurrently overexpressed in squamous cell lung cancer (SqCLC) to exert an oncogenic role by enhancing the translation of MYC (c-Myc), a major oncogene. In this study, we aim to further characterize the functional relationship between PKP1 and MYC. METHODS To determine the functional relationship between PKP1 and MYC, we performed correlation analyses between PKP1 and MYC mRNA expression levels, gain/loss of function models, chromatin immunoprecipitation (ChIP) and promoter mutagenesis followed by luciferase assays. RESULTS We found a significant correlation between the mRNA levels of MYC and PKP1 in SqCLC primary tumor samples. In addition, we found that MYC is a direct transcription factor of PKP1 and binds to specific sequences within its promoter. In agreement with this, we found that MYC knockdown reduced PKP1 protein expression in different SqCLC models, which may explain the PKP1-MYC correlation that we found. Conversely, we found that PKP1 knockdown reduced MYC protein expression, while PKP1 overexpression enhanced MYC expression in these models. CONCLUSIONS Based on these results, we propose a feedforward functional relationship in which PKP1 enhances MYC translation in conjunction with the translation initiation complex by binding to the 5'-UTR of MYC mRNA, whereas MYC promotes PKP1 transcription by binding to its promoter. These results suggest that PKP1 may serve as a therapeutic target for SqCLC.
Collapse
Affiliation(s)
- Laura Boyero
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
- Department of Biochemistry and Molecular Biology III, University of Granada, Granada, Spain
- Institute of Biomedicine of Seville (IBiS) (HUVR, CSIC, University of Seville), Seville, Spain
| | - Joel Martin-Padron
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
- Department of Biochemistry and Molecular Biology III, University of Granada, Granada, Spain
| | - María Esther Fárez-Vidal
- Department of Biochemistry and Molecular Biology III, University of Granada, Granada, Spain
- Institute for Biomedical Research Ibs. Granada, University Hospital Complex of Granada/University of Granada, Granada, Spain
| | - Maria Isabel Rodriguez
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
- Department of Biochemistry and Molecular Biology III, University of Granada, Granada, Spain
- Institute for Biomedical Research Ibs. Granada, University Hospital Complex of Granada/University of Granada, Granada, Spain
| | - Álvaro Andrades
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
- Institute for Biomedical Research Ibs. Granada, University Hospital Complex of Granada/University of Granada, Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Granada, Spain
| | - Paola Peinado
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
- Institute of Biomedicine of Seville (IBiS) (HUVR, CSIC, University of Seville), Seville, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Granada, Spain
| | - Alberto M Arenas
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Granada, Spain
| | - Félix Ritoré-Salazar
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Granada, Spain
| | - Juan Carlos Alvarez-Perez
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
- Institute for Biomedical Research Ibs. Granada, University Hospital Complex of Granada/University of Granada, Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Granada, Spain
| | - Marta Cuadros
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
- Department of Biochemistry and Molecular Biology III, University of Granada, Granada, Spain
- Institute for Biomedical Research Ibs. Granada, University Hospital Complex of Granada/University of Granada, Granada, Spain
| | - Pedro P Medina
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain.
- Institute for Biomedical Research Ibs. Granada, University Hospital Complex of Granada/University of Granada, Granada, Spain.
- Department of Biochemistry and Molecular Biology I, University of Granada, Granada, Spain.
| |
Collapse
|
31
|
Haince JF, Joubert P, Bach H, Ahmed Bux R, Tappia PS, Ramjiawan B. Metabolomic Fingerprinting for the Detection of Early-Stage Lung Cancer: From the Genome to the Metabolome. Int J Mol Sci 2022; 23:ijms23031215. [PMID: 35163138 PMCID: PMC8835988 DOI: 10.3390/ijms23031215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
The five-year survival rate of lung cancer patients is very low, mainly because most newly diagnosed patients present with locally advanced or metastatic disease. Therefore, early diagnosis is key to the successful treatment and management of lung cancer. Unfortunately, early detection methods of lung cancer are not ideal. In this brief review, we described early detection methods such as chest X-rays followed by bronchoscopy, sputum analysis followed by cytological analysis, and low-dose computed tomography (LDCT). In addition, we discussed the potential of metabolomic fingerprinting, compared to that of other biomarkers, including molecular targets, as a low-cost, high-throughput blood-based test that is both feasible and affordable for early-stage lung cancer screening of at-risk populations. Accordingly, we proposed a paradigm shift to metabolomics as an alternative to molecular and proteomic-based markers in lung cancer screening, which will enable blood-based routine testing and be accessible to those patients at the highest risk for lung cancer.
Collapse
Affiliation(s)
| | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Pathology, Laval University, Quebec, QC G1V 4G5, Canada;
| | - Horacio Bach
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC V6H 3Z6, Canada;
| | - Rashid Ahmed Bux
- BioMark Diagnostics Inc., Richmond, BC V6X 2W8, Canada; (J.-F.H.); (R.A.B.)
| | - Paramjit S. Tappia
- Asper Clinical Research Institute, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
- Correspondence: ; Tel.: +1-204-258-1230
| | - Bram Ramjiawan
- Asper Clinical Research Institute, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
- Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| |
Collapse
|
32
|
Yang M, Ding J, Luo Q, Chen X, Chen F. Improving the diagnostic efficacy of squamous cell carcinoma antigen for oral squamous cell carcinoma via saponin disruption of serum extracellular vesicles. Clin Chim Acta 2021; 525:40-45. [PMID: 34921893 DOI: 10.1016/j.cca.2021.12.012] [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] [Received: 05/06/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The diagnostic value of squamous cell carcinoma antigen (SCCA) for oral squamous cell carcinoma (OSCC) is insufficient. Recently, extracellular vesicles (EVs) have displayed great potential for improving diagnostic efficacy. However, one of the main challenges that restricts the application of EVs is the lack of a clinically suitable separation method for the intra-vesicular protein detection. METHODS Saponin was used to destroy serum EVs membranes for releasing the intra-vesicular SCCA into the serum, circumventing the purification process of EVs. The concentrations of SCCA were measured and compared in 113 healthy people and 73 OSCC patients pre- and post-saponin treatment. RESULTS The concentration of serum SCCA significantly increased after saponin destroyed the membrane of EVs. The area under the curve (AUC) of serum SCCA for OSCC diagnosis was 0.6444 (95% CI, 0.5595 to 0.7293). The diagnostic AUC of serum EVs-derived SCCA reached 0.7969 (95% CI, 0.735 to 0.8588). CONCLUSIONS The results suggested that serum EVs disrupted by saponin could improve the diagnostic efficacy of SCCA for OSCC, which provides a simple, rapid, and high-throughput method to detect the intra-vesicular proteins of EVs and holds great potential for clinical application.
Collapse
Affiliation(s)
- Meng Yang
- Department of Clinical Laboratory Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Jieying Ding
- Department of Clinical Laboratory Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Qingqiong Luo
- Department of Clinical Laboratory Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Xu Chen
- Department of Clinical Laboratory Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Fuxiang Chen
- Department of Clinical Laboratory Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Faculty of Medical Laboratory Science, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, PR China.
| |
Collapse
|
33
|
Taheri N, Khoshsafar H, Ghanei M, Ghazvini A, Bagheri H. Dual-template rectangular nanotube molecularly imprinted polypyrrole for label-free impedimetric sensing of AFP and CEA as lung cancer biomarkers. Talanta 2021; 239:123146. [PMID: 34942484 DOI: 10.1016/j.talanta.2021.123146] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 12/06/2021] [Accepted: 12/11/2021] [Indexed: 02/04/2023]
Abstract
A high-performance sensing layer based on dual-template molecularly imprinted polymer (DMIP) was fabricated and successfully applied for one-by-one detection of carcinoembryonic antigen (CEA) and alpha-fetoprotein (AFP) as lung cancer biomarkers. The plastic antibodies of AFP and CEA were created into the electropolymerized polypyrrole (PPy) on a fluorine-doped tin oxide (FTO) electrode. Raman spectroscopy, field emission scanning electron microscopy (FE-SEM), cyclic voltammetry (CV), and electrochemical impedance spectroscopy (EIS) tests were performed to pursue the formation and characterization of the sensing layer. Methyl orange (MO) increased the conductivity of PPy and induced the formation of MO doped PPy (PPy-MO) rectangular-shaped nanotubes. Using impedimetric detection, the rebinding of the template antigens was evaluated, the charge transfer resistance increased as the concentration of AFP and CEA increased. The linear dynamic ranges of 5-104 and 10-104 pg mL-1 and detection limits of 1.6 and 3.3 pg mL-1 were obtained for CEA and AFP, respectively. Given satisfactory results in the determination of AFP and CEA in the human serum samples, high sensitivity, and good stability of DMIP sensor made it a promising method for sensing of AFP and CEA in serum samples.
Collapse
Affiliation(s)
- Navid Taheri
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hosein Khoshsafar
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Ghazvini
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hasan Bagheri
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Skurikhin E, Pershina O, Zhukova M, Widera D, Ermakova N, Pan E, Pakhomova A, Morozov S, Kubatiev A, Dygai A. Potential of Stem Cells and CART as a Potential Polytherapy for Small Cell Lung Cancer. Front Cell Dev Biol 2021; 9:778020. [PMID: 34926461 PMCID: PMC8678572 DOI: 10.3389/fcell.2021.778020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/18/2021] [Indexed: 12/15/2022] Open
Abstract
Despite the increasing urgency of the problem of treating small cell lung cancer (SCLC), information on the causes of its development is fragmentary. There is no complete understanding of the features of antitumor immunity and the role of the microenvironment in the development of SCLC resistance. This impedes the development of new methods for the diagnosis and treatment of SCLC. Lung cancer and chronic obstructive pulmonary disease (COPD) have common pathogenetic factors. COPD is a risk factor for lung cancer including SCLC. Therefore, the search for effective approaches to prevention, diagnosis, and treatment of SCLC in patients with COPD is an urgent task. This review provides information on the etiology and pathogenesis of SCLC, analyses the effectiveness of current treatment options, and critically evaluates the potential of chimeric antigen receptor T cells therapy (CART therapy) in SCLC. Moreover, we discuss potential links between lung cancer and COPD and the role of endothelium in the development of COPD. Finally, we propose a new approach for increasing the efficacy of CART therapy in SCLC.
Collapse
Affiliation(s)
- Evgenii Skurikhin
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Olga Pershina
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Mariia Zhukova
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Reading, United Kingdom
| | - Natalia Ermakova
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Edgar Pan
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Angelina Pakhomova
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Sergey Morozov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Aslan Kubatiev
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander Dygai
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
35
|
Lupu LM, Wiegand P, Holdschick D, Mihoc D, Maeser S, Rawer S, Völklein F, Malek E, Barka F, Knauer S, Uth C, Hennermann J, Kleinekofort W, Hahn A, Barka G, Przybylski M. Identification and Affinity Determination of Protein-Antibody and Protein-Aptamer Epitopes by Biosensor-Mass Spectrometry Combination. Int J Mol Sci 2021; 22:12832. [PMID: 34884636 PMCID: PMC8657952 DOI: 10.3390/ijms222312832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022] Open
Abstract
Analytical methods for molecular characterization of diagnostic or therapeutic targets have recently gained high interest. This review summarizes the combination of mass spectrometry and surface plasmon resonance (SPR) biosensor analysis for identification and affinity determination of protein interactions with antibodies and DNA-aptamers. The binding constant (KD) of a protein-antibody complex is first determined by immobilizing an antibody or DNA-aptamer on an SPR chip. A proteolytic peptide mixture is then applied to the chip, and following removal of unbound material by washing, the epitope(s) peptide(s) are eluted and identified by MALDI-MS. The SPR-MS combination was applied to a wide range of affinity pairs. Distinct epitope peptides were identified for the cardiac biomarker myoglobin (MG) both from monoclonal and polyclonal antibodies, and binding constants determined for equine and human MG provided molecular assessment of cross immunoreactivities. Mass spectrometric epitope identifications were obtained for linear, as well as for assembled ("conformational") antibody epitopes, e.g., for the polypeptide chemokine Interleukin-8. Immobilization using protein G substantially improved surface fixation and antibody stabilities for epitope identification and affinity determination. Moreover, epitopes were successfully determined for polyclonal antibodies from biological material, such as from patient antisera upon enzyme replacement therapy of lysosomal diseases. The SPR-MS combination was also successfully applied to identify linear and assembled epitopes for DNA-aptamer interaction complexes of the tumor diagnostic protein C-Met. In summary, the SPR-MS combination has been established as a powerful molecular tool for identification of protein interaction epitopes.
Collapse
Affiliation(s)
- Loredana-Mirela Lupu
- Centre for Analytical Biochemistry and Biomedical Mass Spectrometry (AffyMSLifeChem), and Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Marktstrasse 29, 65428 Rüsselsheim am Main, Germany; (L.-M.L.); (P.W.); (D.H.); (D.M.); (S.M.); (S.R.); (E.M.); (W.K.)
| | - Pascal Wiegand
- Centre for Analytical Biochemistry and Biomedical Mass Spectrometry (AffyMSLifeChem), and Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Marktstrasse 29, 65428 Rüsselsheim am Main, Germany; (L.-M.L.); (P.W.); (D.H.); (D.M.); (S.M.); (S.R.); (E.M.); (W.K.)
| | - Daria Holdschick
- Centre for Analytical Biochemistry and Biomedical Mass Spectrometry (AffyMSLifeChem), and Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Marktstrasse 29, 65428 Rüsselsheim am Main, Germany; (L.-M.L.); (P.W.); (D.H.); (D.M.); (S.M.); (S.R.); (E.M.); (W.K.)
- Department of Engineering & Institute for Microtechnologies (IMTECH), RheinMain University, 65428 Rüsselsheim am Main, Germany;
| | - Delia Mihoc
- Centre for Analytical Biochemistry and Biomedical Mass Spectrometry (AffyMSLifeChem), and Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Marktstrasse 29, 65428 Rüsselsheim am Main, Germany; (L.-M.L.); (P.W.); (D.H.); (D.M.); (S.M.); (S.R.); (E.M.); (W.K.)
| | - Stefan Maeser
- Centre for Analytical Biochemistry and Biomedical Mass Spectrometry (AffyMSLifeChem), and Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Marktstrasse 29, 65428 Rüsselsheim am Main, Germany; (L.-M.L.); (P.W.); (D.H.); (D.M.); (S.M.); (S.R.); (E.M.); (W.K.)
| | - Stephan Rawer
- Centre for Analytical Biochemistry and Biomedical Mass Spectrometry (AffyMSLifeChem), and Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Marktstrasse 29, 65428 Rüsselsheim am Main, Germany; (L.-M.L.); (P.W.); (D.H.); (D.M.); (S.M.); (S.R.); (E.M.); (W.K.)
| | - Friedemann Völklein
- Department of Engineering & Institute for Microtechnologies (IMTECH), RheinMain University, 65428 Rüsselsheim am Main, Germany;
| | - Ebrahim Malek
- Centre for Analytical Biochemistry and Biomedical Mass Spectrometry (AffyMSLifeChem), and Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Marktstrasse 29, 65428 Rüsselsheim am Main, Germany; (L.-M.L.); (P.W.); (D.H.); (D.M.); (S.M.); (S.R.); (E.M.); (W.K.)
- Department of Engineering & Institute for Microtechnologies (IMTECH), RheinMain University, 65428 Rüsselsheim am Main, Germany;
| | - Frederik Barka
- Sunchrom GmbH, Industriestr. 18, 61381 Friedrichsdorf, Germany; (F.B.); (G.B.)
| | - Sascha Knauer
- Sulfotools GmbH, Bahnhofsplatz 1, 65428 Rüsselsheim am Main, Germany; (S.K.); (C.U.)
| | - Christina Uth
- Sulfotools GmbH, Bahnhofsplatz 1, 65428 Rüsselsheim am Main, Germany; (S.K.); (C.U.)
| | - Julia Hennermann
- Department of Pediatrics, Universitätsmedizin Mainz, 55130 Mainz, Germany;
| | - Wolfgang Kleinekofort
- Centre for Analytical Biochemistry and Biomedical Mass Spectrometry (AffyMSLifeChem), and Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Marktstrasse 29, 65428 Rüsselsheim am Main, Germany; (L.-M.L.); (P.W.); (D.H.); (D.M.); (S.M.); (S.R.); (E.M.); (W.K.)
- Department of Engineering & Institute for Microtechnologies (IMTECH), RheinMain University, 65428 Rüsselsheim am Main, Germany;
| | - Andreas Hahn
- Department of Child Neurology, Justus-Liebig-University Giessen, Feulgenstraße 10-12, 35389 Giessen, Germany;
| | - Günes Barka
- Sunchrom GmbH, Industriestr. 18, 61381 Friedrichsdorf, Germany; (F.B.); (G.B.)
| | - Michael Przybylski
- Centre for Analytical Biochemistry and Biomedical Mass Spectrometry (AffyMSLifeChem), and Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry, Marktstrasse 29, 65428 Rüsselsheim am Main, Germany; (L.-M.L.); (P.W.); (D.H.); (D.M.); (S.M.); (S.R.); (E.M.); (W.K.)
- Department of Engineering & Institute for Microtechnologies (IMTECH), RheinMain University, 65428 Rüsselsheim am Main, Germany;
| |
Collapse
|
36
|
Tahanovich AD, Kauhanka NN, Prohorova VI, Murashka DI, Gotko OV. [Predicting the risk of tumor progression in patients with early stages of adenocarcinoma and squamous cell lung carcinoma based on laboratory parameters]. BIOMEDITSINSKAIA KHIMIIA 2021; 67:507-517. [PMID: 34964445 DOI: 10.18097/pbmc20216706507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Non-small cell carcinoma (NSCLC) prevails in the structure of the incidence of lung cancer. In patients with I stage NSCLC, only 60-70% overcome the 5-year survival barrier, and at II stage it decreases to 35-40%. The reason for such a high mortality rate is almost always a relapse of the disease. The main histological forms of NSCLC - adenocarcinoma (AC) and squamous cell carcinoma (SCLC) - differ in the course, protocols and effectiveness of the treatment. Comparative survival data for AK and PCLC are controversial, and reliable biomarkers for determining the risk of tumor progression are lacking. In thus study we have investigated the possibility of using laboratory parameters characterizing the level of some blood proteins involved in carcinogenesis in patients with early stages of AC and SCLC to determine the risk of disease progression. We retrospectively analyzed the duration of the relapse-free period after surgical treatment for one year in 1250 patients (816 with stages I and II of adenocarcinoma, G1-3 and 434 with early stages of SCLC, G1-3). In 81 patients with AC and 36 - with SCLC (stages I-II, G1-3) the level of CYFRA 21-1 and SCC by electrochemiluminescent method, chemokines CXCL5, CXCL8, TPA, pyruvate kinase M2, HIF-1α and hyaluronic acid by enzyme immunoassay, receptors CXCR1, CXCR2, CD44v6 by flow cytometry were determined. Using the Kaplan-Meier graphical analysis, groups of low (stage I G1-2 + stage II G1) and high (stage I G3 + stage II G2-3) risk of tumor progression were identified. In the case of the one-year survival rate of patients with AC was higher than with SCLC. In patients with AC and a high risk of tumor recurrence, compared with a low one, the level of CYFRA 21-1, the mean intensity of fluorescence (MFI) of the CXCR1 receptor in granulocytes, and the relative content of the CXCR2 receptor in lymphocytes were higher. In the case of rapid progression of SCLC in patients, the relative content of the CXCR2 receptor in lymphocytes, the proportion of monocytes equipped with the CD44v6 receptor, and the SCC level were higher than with slow progression. Regression equations, including combinations of the above parameters (threshold value for AC - 0,512, for SCLC - 0,409, sensitivity - 91,9% and 90,0%, specificity - 90,0% and 87,5%, respectively), allow to predict the probability of tumor recurrence.
Collapse
Affiliation(s)
| | - N N Kauhanka
- Belarusian State Medical University, Minsk, Belarus
| | - V I Prohorova
- N.N. Alexandrov Republican Scientific and Practical Center of Oncology and Medical Radiology, Lesnoy, Belarus
| | - D I Murashka
- Belarusian State Medical University, Minsk, Belarus
| | - O V Gotko
- N.N. Alexandrov Republican Scientific and Practical Center of Oncology and Medical Radiology, Lesnoy, Belarus
| |
Collapse
|
37
|
Liu Y, Si S, Dong S, Ji B, Li H, Liu S. Ultrasensitive electrochemical immunosensor for ProGRP detection based on 3D-rGO@Au nanocomposite. Microchem J 2021. [DOI: 10.1016/j.microc.2021.106644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
38
|
Morozov D, Mironov V, Moryachkov RV, Shchugoreva IA, Artyushenko PV, Zamay GS, Kolovskaya OS, Zamay TN, Krat AV, Molodenskiy DS, Zabluda VN, Veprintsev DV, Sokolov AE, Zukov RA, Berezovski MV, Tomilin FN, Fedorov DG, Alexeev Y, Kichkailo AS. The role of SAXS and molecular simulations in 3D structure elucidation of a DNA aptamer against lung cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:316-327. [PMID: 34458013 PMCID: PMC8379633 DOI: 10.1016/j.omtn.2021.07.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022]
Abstract
Aptamers are short, single-stranded DNA or RNA oligonucleotide molecules that function as synthetic analogs of antibodies and bind to a target molecule with high specificity. Aptamer affinity entirely depends on its tertiary structure and charge distribution. Therefore, length and structure optimization are essential for increasing aptamer specificity and affinity. Here, we present a general optimization procedure for finding the most populated atomistic structures of DNA aptamers. Based on the existed aptamer LC-18 for lung adenocarcinoma, a new truncated LC-18 (LC-18t) aptamer LC-18t was developed. A three-dimensional (3D) shape of LC-18t was reported based on small-angle X-ray scattering (SAXS) experiments and molecular modeling by fragment molecular orbital or molecular dynamic methods. Molecular simulations revealed an ensemble of possible aptamer conformations in solution that were in close agreement with measured SAXS data. The aptamer LC-18t had stronger binding to cancerous cells in lung tumor tissues and shared the binding site with the original larger aptamer. The suggested approach reveals 3D shapes of aptamers and helps in designing better affinity probes.
Collapse
Affiliation(s)
- Dmitry Morozov
- Nanoscience Center and Department of Chemistry, University of Jyväskylä, P.O. Box 35, 40014 Jyväskylä, Finland
| | - Vladimir Mironov
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Roman V. Moryachkov
- Laboratory of Physics of Magnetic Phenomena, Kirensky Institute of Physics, 50/38 Akademgorodok, Krasnoyarsk 660036, Russia
- Laboratory for Digital Controlled Drugs and Theranostics, Federal Research Center “Krasnoyarsk Science Center SB RAS,” 50 Akademgorodok, Krasnoyarsk 660036, Russia
| | - Irina A. Shchugoreva
- Laboratory for Digital Controlled Drugs and Theranostics, Federal Research Center “Krasnoyarsk Science Center SB RAS,” 50 Akademgorodok, Krasnoyarsk 660036, Russia
- Krasnoyarsk State Medical University, 1 Partizana Zheleznyaka, Krasnoyarsk 660022, Russia
- Department of Chemistry, Siberian Federal University, 79 Svobodny pr., Krasnoyarsk 660041, Russia
| | - Polina V. Artyushenko
- Laboratory for Digital Controlled Drugs and Theranostics, Federal Research Center “Krasnoyarsk Science Center SB RAS,” 50 Akademgorodok, Krasnoyarsk 660036, Russia
- Krasnoyarsk State Medical University, 1 Partizana Zheleznyaka, Krasnoyarsk 660022, Russia
- Department of Chemistry, Siberian Federal University, 79 Svobodny pr., Krasnoyarsk 660041, Russia
| | - Galina S. Zamay
- Laboratory for Digital Controlled Drugs and Theranostics, Federal Research Center “Krasnoyarsk Science Center SB RAS,” 50 Akademgorodok, Krasnoyarsk 660036, Russia
- Krasnoyarsk State Medical University, 1 Partizana Zheleznyaka, Krasnoyarsk 660022, Russia
| | - Olga S. Kolovskaya
- Laboratory for Digital Controlled Drugs and Theranostics, Federal Research Center “Krasnoyarsk Science Center SB RAS,” 50 Akademgorodok, Krasnoyarsk 660036, Russia
- Krasnoyarsk State Medical University, 1 Partizana Zheleznyaka, Krasnoyarsk 660022, Russia
| | - Tatiana N. Zamay
- Krasnoyarsk State Medical University, 1 Partizana Zheleznyaka, Krasnoyarsk 660022, Russia
| | - Alexey V. Krat
- Krasnoyarsk State Medical University, 1 Partizana Zheleznyaka, Krasnoyarsk 660022, Russia
| | - Dmitry S. Molodenskiy
- European Molecular Biology Laboratory, Hamburg Outstation, Notkestrasse 85, 22603 Hamburg, Germany
| | - Vladimir N. Zabluda
- Laboratory of Physics of Magnetic Phenomena, Kirensky Institute of Physics, 50/38 Akademgorodok, Krasnoyarsk 660036, Russia
| | - Dmitry V. Veprintsev
- Krasnoyarsk State Medical University, 1 Partizana Zheleznyaka, Krasnoyarsk 660022, Russia
| | - Alexey E. Sokolov
- Laboratory of Physics of Magnetic Phenomena, Kirensky Institute of Physics, 50/38 Akademgorodok, Krasnoyarsk 660036, Russia
- Laboratory for Digital Controlled Drugs and Theranostics, Federal Research Center “Krasnoyarsk Science Center SB RAS,” 50 Akademgorodok, Krasnoyarsk 660036, Russia
| | - Ruslan A. Zukov
- Krasnoyarsk State Medical University, 1 Partizana Zheleznyaka, Krasnoyarsk 660022, Russia
| | - Maxim V. Berezovski
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie-Curie, Ottawa, ON K1N 6N5, Canada
| | - Felix N. Tomilin
- Laboratory of Physics of Magnetic Phenomena, Kirensky Institute of Physics, 50/38 Akademgorodok, Krasnoyarsk 660036, Russia
- Department of Chemistry, Siberian Federal University, 79 Svobodny pr., Krasnoyarsk 660041, Russia
| | - Dmitri G. Fedorov
- Research Center for Computational Design of Advanced Functional Materials, National Institute of Advanced Industrial Science and Technology, Tsukuba 305-8568, Japan
| | - Yuri Alexeev
- Computational Science Division, Argonne National Laboratory, Lemont, IL, USA
| | - Anna S. Kichkailo
- Laboratory for Digital Controlled Drugs and Theranostics, Federal Research Center “Krasnoyarsk Science Center SB RAS,” 50 Akademgorodok, Krasnoyarsk 660036, Russia
- Krasnoyarsk State Medical University, 1 Partizana Zheleznyaka, Krasnoyarsk 660022, Russia
| |
Collapse
|
39
|
Voronina L, Leonardo C, Mueller‐Reif JB, Geyer PE, Huber M, Trubetskov M, Kepesidis KV, Behr J, Mann M, Krausz F, Žigman M. Molecular Origin of Blood‐Based Infrared Spectroscopic Fingerprints**. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202103272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Liudmila Voronina
- Department of Physics Ludwig Maximilian University of Munich 85748 Garching Germany
- Max Planck Institute of Quantum Optics 85748 Garching Germany
| | - Cristina Leonardo
- Department of Physics Ludwig Maximilian University of Munich 85748 Garching Germany
- Max Planck Institute of Quantum Optics 85748 Garching Germany
| | - Johannes B. Mueller‐Reif
- Department of Proteomics and Signal Transduction Max Planck Institute of Biochemistry 82152 Martinsried Germany
- OmicEra Diagnostics GmbH 82152 Planegg Germany
| | - Philipp E. Geyer
- Department of Proteomics and Signal Transduction Max Planck Institute of Biochemistry 82152 Martinsried Germany
- Novo Nordisk Foundation Center for Protein Research Faculty of Health Sciences University of Copenhagen 2200 Copenhagen Denmark
- OmicEra Diagnostics GmbH 82152 Planegg Germany
| | - Marinus Huber
- Department of Physics Ludwig Maximilian University of Munich 85748 Garching Germany
- Max Planck Institute of Quantum Optics 85748 Garching Germany
| | | | - Kosmas V. Kepesidis
- Department of Physics Ludwig Maximilian University of Munich 85748 Garching Germany
| | - Jürgen Behr
- Comprehensive Pneumology Center Department of Internal Medicine V Clinic of the Ludwig Maximilians University Munich (LMU), Member of the German Center for Lung Research Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction Max Planck Institute of Biochemistry 82152 Martinsried Germany
- Novo Nordisk Foundation Center for Protein Research Faculty of Health Sciences University of Copenhagen 2200 Copenhagen Denmark
| | - Ferenc Krausz
- Department of Physics Ludwig Maximilian University of Munich 85748 Garching Germany
- Max Planck Institute of Quantum Optics 85748 Garching Germany
| | - Mihaela Žigman
- Department of Physics Ludwig Maximilian University of Munich 85748 Garching Germany
- Max Planck Institute of Quantum Optics 85748 Garching Germany
| |
Collapse
|
40
|
Voronina L, Leonardo C, Mueller‐Reif JB, Geyer PE, Huber M, Trubetskov M, Kepesidis KV, Behr J, Mann M, Krausz F, Žigman M. Molecular Origin of Blood-Based Infrared Spectroscopic Fingerprints*. Angew Chem Int Ed Engl 2021; 60:17060-17069. [PMID: 33881784 PMCID: PMC8361728 DOI: 10.1002/anie.202103272] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/30/2021] [Indexed: 12/17/2022]
Abstract
Infrared spectroscopy of liquid biopsies is a time- and cost-effective approach that may advance biomedical diagnostics. However, the molecular nature of disease-related changes of infrared molecular fingerprints (IMFs) remains poorly understood, impeding the method's applicability. Here we probe 148 human blood sera and reveal the origin of the variations in their IMFs. To that end, we supplemented infrared spectroscopy with biochemical fractionation and proteomic profiling, providing molecular information about serum composition. Using lung cancer as an example of a medical condition, we demonstrate that the disease-related differences in IMFs are dominated by contributions from twelve highly abundant proteins-that, if used as a pattern, may be instrumental for detecting malignancy. Tying proteomic to spectral information and machine learning advances our understanding of the infrared spectra of liquid biopsies, a framework that could be applied to probing of any disease.
Collapse
Affiliation(s)
- Liudmila Voronina
- Department of PhysicsLudwig Maximilian University of Munich85748GarchingGermany
- Max Planck Institute of Quantum Optics85748GarchingGermany
| | - Cristina Leonardo
- Department of PhysicsLudwig Maximilian University of Munich85748GarchingGermany
- Max Planck Institute of Quantum Optics85748GarchingGermany
| | - Johannes B. Mueller‐Reif
- Department of Proteomics and Signal TransductionMax Planck Institute of Biochemistry82152MartinsriedGermany
- OmicEra Diagnostics GmbH82152PlaneggGermany
| | - Philipp E. Geyer
- Department of Proteomics and Signal TransductionMax Planck Institute of Biochemistry82152MartinsriedGermany
- Novo Nordisk Foundation Center for Protein ResearchFaculty of Health SciencesUniversity of Copenhagen2200CopenhagenDenmark
- OmicEra Diagnostics GmbH82152PlaneggGermany
| | - Marinus Huber
- Department of PhysicsLudwig Maximilian University of Munich85748GarchingGermany
- Max Planck Institute of Quantum Optics85748GarchingGermany
| | | | - Kosmas V. Kepesidis
- Department of PhysicsLudwig Maximilian University of Munich85748GarchingGermany
| | - Jürgen Behr
- Comprehensive Pneumology CenterDepartment of Internal Medicine VClinic of the Ludwig Maximilians University Munich (LMU), Member of the German Center for Lung ResearchGermany
| | - Matthias Mann
- Department of Proteomics and Signal TransductionMax Planck Institute of Biochemistry82152MartinsriedGermany
- Novo Nordisk Foundation Center for Protein ResearchFaculty of Health SciencesUniversity of Copenhagen2200CopenhagenDenmark
| | - Ferenc Krausz
- Department of PhysicsLudwig Maximilian University of Munich85748GarchingGermany
- Max Planck Institute of Quantum Optics85748GarchingGermany
| | - Mihaela Žigman
- Department of PhysicsLudwig Maximilian University of Munich85748GarchingGermany
- Max Planck Institute of Quantum Optics85748GarchingGermany
| |
Collapse
|
41
|
Application across species of a one health approach to liquid sample handling for respiratory based -omics analysis. Sci Rep 2021; 11:14292. [PMID: 34253818 PMCID: PMC8275668 DOI: 10.1038/s41598-021-93839-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/25/2021] [Indexed: 01/19/2023] Open
Abstract
Airway inflammation is highly prevalent in horses, with the majority of non-infectious cases being defined as equine asthma. Currently, cytological analysis of airway derived samples is the principal method of assessing lower airway inflammation. Samples can be obtained by tracheal wash (TW) or by lavage of the lower respiratory tract (bronchoalveolar lavage (BAL) fluid; BALF). Although BALF cytology carries significant diagnostic advantages over TW cytology for the diagnosis of equine asthma, sample acquisition is invasive, making it prohibitive for routine and sequential screening of airway health. However, recent technological advances in sample collection and processing have made it possible to determine whether a wider range of analyses might be applied to TW samples. Considering that TW samples are relatively simple to collect, minimally invasive and readily available in the horse, it was considered appropriate to investigate whether, equine tracheal secretions represent a rich source of cells and both transcriptomic and proteomic data. Similar approaches have already been applied to a comparable sample set in humans; namely, induced sputum. Sputum represents a readily available source of airway biofluids enriched in proteins, changes in the expression of which may reveal novel mechanisms in the pathogenesis of respiratory diseases, such as asthma and chronic obstructive pulmonary disease. The aim of this study was to establish a robust protocol to isolate macrophages, protein and RNA for molecular characterization of TW samples and demonstrate the applicability of sample handling to rodent and human pediatric bronchoalveolar lavage fluid isolates. TW samples provided a good quality and yield of both RNA and protein for downstream transcriptomic/proteomic analyses. The sample handling methodologies were successfully applicable to BALF for rodent and human research. TW samples represent a rich source of airway cells, and molecular analysis to facilitate and study airway inflammation, based on both transcriptomic and proteomic analysis. This study provides a necessary methodological platform for future transcriptomic and/or proteomic studies on equine lower respiratory tract secretions and BALF samples from humans and mice.
Collapse
|
42
|
Usman M, Ilyas A, Syed B, Hashim Z, Ahmed A, Zarina S. Serum HSP90-Alpha and Oral Squamous Cell Carcinoma : A Prospective Biomarker. Protein Pept Lett 2021; 28:1157-1163. [PMID: 34137356 DOI: 10.2174/0929866528666210616112539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/10/2021] [Accepted: 05/10/2021] [Indexed: 12/24/2022]
Abstract
AIM Current study aims to perform differential protein expression analysis of serum samples from Oral squamous cell carcinoma (OSCC) patients and healthy controls in search of potential diagnostic and/or prognostic biomarker(s). OBJECTIVE OSCC is diagnosed late, resulting in poor survival and high mortality. Identification of non-invasive prognostic biomarker is of utmost importance for early diagnosis and proper management of the disease; hence we used proteomic approach to identify potential biomarkers from serum. METHODS Serum samples (OSCC n=45 and control n=30) were depleted and proteins were separated using 2-D gel electrophoresis followed by identification by mass spectrometric analysis. Gene expression analysis of identified proteins in malignant and normal tissue was also performed to complement proteomics studies. RESULTS Among differentially expressed proteins, a noteworthy observation was up regulation of Heat shock protein alpha (HSP90α) from serum of oral cancer patients. We also observed elevated levels of Haptoglobin (HP) along with down regulation of Type II keratin cytoskeletal 1(KRT1) and serum Albumin (ALB) in oral cancer patients. Gene expression studies of identified proteins in malignant and normal tissue revealed a similar pattern with the exception of KRT1. We believe that elevated levels of serum HSP90 alpha might be used as a potential biomarker. CONCLUSION Our findings suggest the contribution of HSP90 alpha and other identified proteins in oral pathology as pro/anti apoptotic modulators, thus they are being considered as predictive biomarkers.
Collapse
Affiliation(s)
- Muhammad Usman
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270, Pakistan
| | - Amber Ilyas
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270, Pakistan
| | - Basir Syed
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Orange, CA 92866, United States
| | - Zehra Hashim
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270, Pakistan
| | - Aftab Ahmed
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Orange, CA 92866, United States
| | - Shamshad Zarina
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
43
|
Guo Y, Li K, Gao Y, Zhao S, Shi M, Li J, Liu Z, Wang Z, He L. CLEC3B Identified as a Potential Lung Cancer Biomarker in Serum by Aptamer‐Capture Technology. ChemistrySelect 2021. [DOI: 10.1002/slct.202100605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yuanbin Guo
- College of Environment & Chemical Engineering Yanshan University Qinhuangdao Hebei Province 066004 China
| | - Kun Li
- College of Environment & Chemical Engineering Yanshan University Qinhuangdao Hebei Province 066004 China
| | - Yue Gao
- College of Environment & Chemical Engineering Yanshan University Qinhuangdao Hebei Province 066004 China
| | - Shuhua Zhao
- College of Environment & Chemical Engineering Yanshan University Qinhuangdao Hebei Province 066004 China
| | - Ming Shi
- College of Environment & Chemical Engineering Yanshan University Qinhuangdao Hebei Province 066004 China
| | - Jian Li
- College of Environment & Chemical Engineering Yanshan University Qinhuangdao Hebei Province 066004 China
| | - Zhiwei Liu
- College of Environment & Chemical Engineering Yanshan University Qinhuangdao Hebei Province 066004 China
| | - Zhaoxia Wang
- College of Environment & Chemical Engineering Yanshan University Qinhuangdao Hebei Province 066004 China
| | - Lei He
- College of Environment & Chemical Engineering Yanshan University Qinhuangdao Hebei Province 066004 China
| |
Collapse
|
44
|
Rawal S, Patel M. Bio-Nanocarriers for Lung Cancer Management: Befriending the Barriers. NANO-MICRO LETTERS 2021; 13:142. [PMID: 34138386 PMCID: PMC8196938 DOI: 10.1007/s40820-021-00630-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/23/2021] [Indexed: 05/03/2023]
Abstract
Lung cancer is a complex thoracic malignancy developing consequential to aberrations in a myriad of molecular and biomolecular signaling pathways. It is one of the most lethal forms of cancers accounting to almost 1.8 million new annual incidences, bearing overall mortality to incidence ratio of 0.87. The dismal prognostic scenario at advanced stages of the disease and metastatic/resistant tumor cell populations stresses the requisite of advanced translational interdisciplinary interventions such as bionanotechnology. This review article deliberates insights and apprehensions on the recent prologue of nanobioengineering and bionanotechnology as an approach for the clinical management of lung cancer. The role of nanobioengineered (bio-nano) tools like bio-nanocarriers and nanobiodevices in secondary prophylaxis, diagnosis, therapeutics, and theranostics for lung cancer management has been discussed. Bioengineered, bioinspired, and biomimetic bio-nanotools of considerate translational value have been reviewed. Perspectives on existent oncostrategies, their critical comparison with bio-nanocarriers, and issues hampering their clinical bench side to bed transformation have also been summarized.
Collapse
Affiliation(s)
- Shruti Rawal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382 481, India
| | - Mayur Patel
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382 481, India.
| |
Collapse
|
45
|
Wang Z, Liu Y, Wang Z, Huang X, Huang W. Hydrogel‐based composites: Unlimited platforms for biosensors and diagnostics. VIEW 2021. [DOI: 10.1002/viw.20200165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Zeyi Wang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing China
| | - Yanlei Liu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing China
| | - Zhiwei Wang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing China
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering Northwestern Polytechnical University Xi'an China
| | - Xiao Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing China
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering Northwestern Polytechnical University Xi'an China
| |
Collapse
|
46
|
Monedeiro F, Monedeiro-Milanowski M, Ratiu IA, Brożek B, Ligor T, Buszewski B. Needle Trap Device-GC-MS for Characterization of Lung Diseases Based on Breath VOC Profiles. Molecules 2021; 26:molecules26061789. [PMID: 33810121 PMCID: PMC8004837 DOI: 10.3390/molecules26061789] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 01/08/2023] Open
Abstract
Volatile organic compounds (VOCs) have been assessed in breath samples as possible indicators of diseases. The present study aimed to quantify 29 VOCs (previously reported as potential biomarkers of lung diseases) in breath samples collected from controls and individuals with lung cancer, chronic obstructive pulmonary disease and asthma. Besides that, global VOC profiles were investigated. A needle trap device (NTD) was used as pre-concentration technique, associated to gas chromatography-mass spectrometry (GC-MS) analysis. Univariate and multivariate approaches were applied to assess VOC distributions according to the studied diseases. Limits of quantitation ranged from 0.003 to 6.21 ppbv and calculated relative standard deviations did not exceed 10%. At least 15 of the quantified targets presented themselves as discriminating features. A random forest (RF) method was performed in order to classify enrolled conditions according to VOCs' latent patterns, considering VOCs responses in global profiles. The developed model was based on 12 discriminating features and provided overall balanced accuracy of 85.7%. Ultimately, multinomial logistic regression (MLR) analysis was conducted using the concentration of the nine most discriminative targets (2-propanol, 3-methylpentane, (E)-ocimene, limonene, m-cymene, benzonitrile, undecane, terpineol, phenol) as input and provided an average overall accuracy of 95.5% for multiclass prediction.
Collapse
Affiliation(s)
- Fernanda Monedeiro
- Interdisciplinary Centre of Modern Technologies, Nicolaus Copernicus University in Toruń, 4 Wileńska St., 87-100 Toruń, Poland; (F.M.); (M.M.-M.); (I.-A.R.); (B.B.)
| | - Maciej Monedeiro-Milanowski
- Interdisciplinary Centre of Modern Technologies, Nicolaus Copernicus University in Toruń, 4 Wileńska St., 87-100 Toruń, Poland; (F.M.); (M.M.-M.); (I.-A.R.); (B.B.)
| | - Ileana-Andreea Ratiu
- Interdisciplinary Centre of Modern Technologies, Nicolaus Copernicus University in Toruń, 4 Wileńska St., 87-100 Toruń, Poland; (F.M.); (M.M.-M.); (I.-A.R.); (B.B.)
- “Raluca Ripan” Institute for Research in Chemistry, Babeş-Bolyai University, 30 Fântânele St., RO-400294 Cluj-Napoca, Romania
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, 7 Gagarina St., 87-100 Toruń, Poland
| | - Beata Brożek
- Department of Lung Diseases, Provincial Polyclinic Hospital in Toruń, 4 Krasińskiego St., 87-100 Toruń, Poland;
| | - Tomasz Ligor
- Interdisciplinary Centre of Modern Technologies, Nicolaus Copernicus University in Toruń, 4 Wileńska St., 87-100 Toruń, Poland; (F.M.); (M.M.-M.); (I.-A.R.); (B.B.)
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, 7 Gagarina St., 87-100 Toruń, Poland
- Correspondence: ; Tel.: +48-(56)-665-60-58
| | - Bogusław Buszewski
- Interdisciplinary Centre of Modern Technologies, Nicolaus Copernicus University in Toruń, 4 Wileńska St., 87-100 Toruń, Poland; (F.M.); (M.M.-M.); (I.-A.R.); (B.B.)
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, 7 Gagarina St., 87-100 Toruń, Poland
| |
Collapse
|
47
|
Smolarz M, Widlak P. Serum Exosomes and Their miRNA Load-A Potential Biomarker of Lung Cancer. Cancers (Basel) 2021; 13:cancers13061373. [PMID: 33803617 PMCID: PMC8002857 DOI: 10.3390/cancers13061373] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022] Open
Abstract
Early detection of lung cancer in screening programs is a rational way to reduce mortality associated with this malignancy. Low-dose computed tomography, a diagnostic tool used in lung cancer screening, generates a relatively large number of false-positive results, and its complementation with molecular biomarkers would greatly improve the effectiveness of such programs. Several biomarkers of lung cancer based on different components of blood, including miRNA signatures, were proposed. However, only a few of them have been positively validated in the context of early cancer detection yet, which imposes a constant need for new biomarker candidates. An emerging source of cancer biomarkers are exosomes and other types of extracellular vesicles circulating in body fluids. Hence, different molecular components of serum/plasma-derived exosomes were tested and showed different levels in lung cancer patients and healthy individuals. Several studies focused on the miRNA component of these vesicles. Proposed signatures of exosome miRNA had promising diagnostic value, though none of them have yet been clinically validated. These signatures involved a few dozen miRNA species overall, including a few species that recurred in different signatures. It is worth noting that all these miRNA species have cancer-related functions and have been associated with lung cancer progression. Moreover, a few of them, including known oncomirs miR-17, miR-19, miR-21, and miR-221, appeared in multiple miRNA signatures of lung cancer based on both the whole serum/plasma and serum/plasma-derived exosomes.
Collapse
|
48
|
Kuntamung K, Sangthong P, Jakmunee J, Ounnunkad K. A label-free immunosensor for the detection of a new lung cancer biomarker, GM2 activator protein, using a phosphomolybdic acid/polyethyleneimine coated gold nanoparticle composite. Analyst 2021; 146:2203-2211. [DOI: 10.1039/d0an02149k] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
A novel electrochemical immunosensor for the detection of a new lung cancer biomarker based on a polyoxometalate-adsorbed poly(ethylenimine)-coated gold nanoparticle modified electrode.
Collapse
Affiliation(s)
- Kulrisa Kuntamung
- Department of Chemistry
- Faculty of Science
- Chiang Mai University
- Chiang Mai 50200
- Thailand
| | - Padchanee Sangthong
- Department of Chemistry
- Faculty of Science
- Chiang Mai University
- Chiang Mai 50200
- Thailand
| | - Jaroon Jakmunee
- Department of Chemistry
- Faculty of Science
- Chiang Mai University
- Chiang Mai 50200
- Thailand
| | - Kontad Ounnunkad
- Department of Chemistry
- Faculty of Science
- Chiang Mai University
- Chiang Mai 50200
- Thailand
| |
Collapse
|
49
|
Ratan C, Cicily K D D, Nair B, Nath LR. MUC Glycoproteins: Potential Biomarkers and Molecular Targets for Cancer Therapy. Curr Cancer Drug Targets 2021; 21:132-152. [PMID: 33200711 DOI: 10.2174/1568009620666201116113334] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/02/2020] [Accepted: 10/04/2020] [Indexed: 02/08/2023]
Abstract
MUC proteins have great significance as prognostic and diagnostic markers as well as a potential target for therapeutic interventions in most cancers of glandular epithelial origin. These are high molecular weight glycosylated proteins located in the epithelial lining of several tissues and ducts. Mucins belong to a heterogeneous group of large O-glycoproteins that can be either secreted or membrane-bound. Glycosylation, a post-translational modification affects the biophysical, functional and biochemical properties and provides structural complexity for these proteins. Aberrant expression and glycosylation of mucins contribute to tumour survival and proliferation in many cancers, which in turn activates numerous signalling pathways such as NF-kB, ERα, HIF, MAPK, p53, c-Src, Wnt and JAK-STAT, etc. This subsequently induces cancer cell growth, proliferation and metastasis. The present review mainly demonstrates the functional aspects of MUC glycoproteins along with its unique signalling mechanism and role of aberrant glycosylation in cancer progression and therapeutics. The importance of MUC proteins and its subtypes in a wide spectrum of cancers including but not limited to breast cancer, colorectal cancer, endometrial and cervical cancer, lung cancer, primary liver cancer, pancreatic cancer, prostate cancer and ovarian cancer has been exemplified with significance in targeting the same. Several patents associated with the MUC proteins in the field of cancer therapy are also emphasized in the current review.
Collapse
Affiliation(s)
- Chameli Ratan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala 682041, India
| | - Dalia Cicily K D
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala 682041, India
| | - Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala 682041, India
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala 682041, India
| |
Collapse
|
50
|
Choe W, Chae JD, Lee BH, Kim SH, Park SY, Nimse SB, Kim J, Warkad SD, Song KS, Oh AC, Hong YJ, Kim T. 9G Test TM Cancer/Lung: A Desirable Companion to LDCT for Lung Cancer Screening. Cancers (Basel) 2020; 12:cancers12113192. [PMID: 33143045 PMCID: PMC7692999 DOI: 10.3390/cancers12113192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/26/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Lung cancer is the most common cause of cancer-related deaths globally. Patients diagnosed at early-stage (0–I) have a higher survival rate than the metastasized stages (III–IV). Thus, there is great potential to reduce mortality by diagnosing lung cancer at stage 0~I through community screening. LDCT is a promising method, but it has a high false-positive rate. Therefore, a biomarker test that can be used in combination with LDCT for lung cancer screening to reduce false-positive rates is highly awaited. The present study evaluated the applicability of 9G testTM Cancer/Lung test to detect stage 0~IV lung cancer. 9G testTM Cancer/Lung test detects stage I, stage II, stage III, and stage IV cancers with the sensitivities of 77.5%, 78.1%, 67.4%, and 33.3%, respectively, at the specificity of 97.3%. These results indicate that the 9G testTM Cancer/Lung can be used in conjunction with LDCT to screen lung cancer. Abstract A complimentary biomarker test that can be used in combination with LDCT for lung cancer screening is highly desirable to improve the diagnostic capacity of LDCT and reduce the false-positive rates. Most importantly, the stage I lung cancer detection rate can be dramatically increased by the simultaneous use of a biomarker test with LDCT. The present study was conducted to evaluate 9G testTM Cancer/Lung’s sensitivity and specificity in detecting Stage 0~IV lung cancer. The obtained results indicate that the 9G testTM Cancer/Lung can detect lung cancer with overall sensitivity and specificity of 75.0% (69.1~80.3) and 97.3% (95.0~98.8), respectively. The detection of stage I, stage II, stage III, and stage IV cancers with sensitivities of 77.5%, 78.1%, 67.4%, and 33.3%, respectively, at the specificity of 97.3% have never been reported before. The receiver operating characteristic curve analysis allowed us to determine the population-weighted AUC of 0.93 (95% CI, 0.91–0.95). These results indicate that the 9G testTM Cancer/Lung can be used in conjunction with LDCT to screen lung cancer. Furthermore, obtained results indicate that the use of 9G testTM Cancer/Lung with LDCT for lung cancer screening can increase stage I cancer detection, which is crucial to improve the currently low 5-year survival rates.
Collapse
Affiliation(s)
- Wonho Choe
- Nowon Eulji Medical Center, Department of Laboratory Medicine, Eulji University, Seoul 01830, Korea
| | - Jeong Don Chae
- Nowon Eulji Medical Center, Department of Laboratory Medicine, Eulji University, Seoul 01830, Korea
| | - Byoung-Hoon Lee
- Nowon Eulji Medical Center, Department of Pulmonology and Allergy, Eulji University, Seoul 01830, Korea
| | - Sang-Hoon Kim
- Nowon Eulji Medical Center, Department of Pulmonology and Allergy, Eulji University, Seoul 01830, Korea
| | - So Young Park
- Nowon Eulji Medical Center, Department of Pulmonology and Allergy, Eulji University, Seoul 01830, Korea
| | - Satish Balasaheb Nimse
- Institute of Applied Chemistry and Department of Chemistry, Hallym University, Chuncheon 24252, Korea
| | - Junghoon Kim
- Institute of Applied Chemistry and Department of Chemistry, Hallym University, Chuncheon 24252, Korea
| | | | - Keum-Soo Song
- Biometrix Technology, Inc. 2-2 Bio Venture Plaza 56, Chuncheon 24232, Korea
| | - Ae-Chin Oh
- Departments of Laboratory Medicine, Korea Cancer Center Hospital, Seoul 01812, Korea
| | - Young Jun Hong
- Departments of Laboratory Medicine, Korea Cancer Center Hospital, Seoul 01812, Korea
| | - Taisun Kim
- Institute of Applied Chemistry and Department of Chemistry, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|