1
|
Chen HQ, Wang N, Zeng Y, Shi Y, Zhang Z, Li JY, Li YW, Deng SW, Zhou ZY, Liu WB. KCNJ15 inhibits chemical-induced lung carcinogenesis and progression through GNB1 mediated Hippo pathway. Toxicology 2024:154034. [PMID: 39725264 DOI: 10.1016/j.tox.2024.154034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/14/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) have been regarded as important environmental carcinogens that can cause lung cancer. However, the underlying epigenetic mechanism during PAHs-induced lung carcinogenesis has remained largely unknown. Previously, we screened some novel epigenetic regulatory genes during 3-methylcholanthrene (3-MCA)-induced lung carcinogenesis, including the potassium inwardly rectifying channel subfamily J member 15 (KCNJ15) gene. This study aimed to investigate the expression regulation, function, and mechanism of KCNJ15 through database analysis, malignant transformed cell model, and xenograft tumor models. We found that KCNJ15 was remarkably under-expressed during lung carcinogenesis and progression. Elevated levels of DNA methylation were associated with a gradual decrease of KCNJ15 expression in 3-MCA-induced malignantly transformed HBE cells. High expression of KCNJ15 was strongly correlated with good survival prognosis in lung cancer patients. KCNJ15 overexpression significantly inhibited the growth, invasion, and migration of lung cancer cells both in vitro and in vivo. Conversely, knockdown of KCNJ15 resulted in an opposite phenotype. Moreover, KCNJ15 inhibited the Hippo pathway by activating YAP phosphorylation and inhibiting YAP expression. Mechanistically, there was a significant protein-protein interaction between KCNJ15 and the G protein subunit beta 1 (GNB1). GNB1 overexpression partially restored the inhibition of Hippo pathway regulated by KCNJ15. In summary, our data demonstrated that KCNJ15, as a novel epigenetic silencing tumor suppressor, regulates cell growth, invasion, and migration by binding to GNB1 protein mediating the Hippo-YAP signaling pathway during chemical-induced lung carcinogenesis and progression. It provides novel insights into epigenetic regulation mechanism during carcinogenesis induced by environmental pollutants.
Collapse
Affiliation(s)
- Hong-Qiang Chen
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Na Wang
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China
| | - Yong Zeng
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yu Shi
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Zhe Zhang
- Department of Breast and Thyroid Surgery, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Jiang-Ying Li
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Ya-Wen Li
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China
| | - Shuang-Wu Deng
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China
| | - Zi-Yuan Zhou
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wen-Bin Liu
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
2
|
Saqib M, Din ZS, Zafar S, Munawar N, Nawaz R, Ahmed S, Hamdard MH. Lung cancer, platinum analog-based frontline treatment and pharmacogenetic limitations. Per Med 2024; 21:385-400. [PMID: 39560009 DOI: 10.1080/17410541.2024.2391269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 08/08/2024] [Indexed: 11/20/2024]
Abstract
Lung cancer has the highest mortality rate among all the highly prevalent neoplasia globally. The major concern with its frontline treatment-cisplatin, is the rapid progression of chemoresistance and multi-organ-based toxicities including hearing loss and tinnitus, nephrotoxicity, hepatotoxicity and myelosuppression including anemia and neutropenia. In this review, studies concluding the association of single nucleotide polymorphisms (SNP) in disparate genes with aforementioned toxicity points are summarized to observe the pharmacogenomic pattern. Especially, SNPs in ATP7B, ERCC-1, ERCC-2, MATE-1, OCT-2, ABCB-1, ABCC-1, ABCG-2, ABCC-2, SLC22A, ERCC-5, BRCA-1, GSTM-3, GSTM-4 and GSTM-5 genes appear to be associated with the therapeutic response and/or adverse effects of cisplatin. We recommend utilizing this information to minimize the risk of treatment failure due to chemoresistance and adverse effects on other organs.
Collapse
Affiliation(s)
- Maryam Saqib
- Shifa College of Medicine, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Zari Salahud Din
- Shifa College of Medicine, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Sehrish Zafar
- Combined Military Hospital College, Lahore, Pakistan
| | - Nayla Munawar
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, 15551, United Arab Emirates
| | - Rukhsana Nawaz
- Department of Clinical Psychology, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, 15551, United Arab Emirates
| | - Sagheer Ahmed
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | | |
Collapse
|
3
|
Li C, Li Z, Wang L, Zhang K, Li Z, Ji Y, Li J, Zhang Y, Chen L. Conquering dual challenges: A sialic-modified liposome for targeting activated neutrophils to tackle comorbid lung inflammation and cancer metastasis. J Control Release 2024; 376:930-948. [PMID: 39476871 DOI: 10.1016/j.jconrel.2024.10.048] [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: 05/13/2024] [Revised: 09/05/2024] [Accepted: 10/21/2024] [Indexed: 11/09/2024]
Abstract
In clinical settings, cancer frequently coexists with multi-system diseases. Owing to compromised immune systems, patients with cancer exhibit an increased susceptibility to infections and inflammation. Notably, lung inflammation occurs with high incidence among these patients. Furthermore, the inflammatory milieu within the lungs often accelerates the metastasis of cancer, thereby enhancing mortality rates and posing substantial challenges for clinical management. To date, effective strategies addressing both lung inflammation and cancer concurrently are lacking. In this context, we introduce a novel therapeutic approach involving a sialic acid-lipid derivative (SA-PG10-C18) modified doxorubicin-curcumin co-loaded liposome (DOX/CUR-SAL). This formulation effectively targeted activated neutrophils, which are abundantly present in inflammatory and metastatic lung tissues. DOX/CUR-SAL notably inhibited neutrophil-mediated pro-inflammatory and pro-metastatic processes. Utilizing a newly established mouse model of acute lung injury (ALI) and metastasis comorbidity, DOX/CUR-SAL modulated the lung immune microenvironment and arrested the progression of both inflammation and metastasis, without inducing side effects. The treated animals demonstrated favorable survival conditions, persisting beyond 45 days. This innovative therapeutic strategy offers a novel concept and reference for treating comorbid conditions of tumors and inflammation, thus breaking the clinical impasse where lung inflammation and cancer metastasis have been treated separately.
Collapse
Affiliation(s)
- Cong Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China; Liaoning Key Laboratory of New Drug Research & Development, Shenyang 110036, China
| | - Zhihang Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Lihong Wang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Kexin Zhang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Zehao Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Yating Ji
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Jing Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Yifan Zhang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Lijiang Chen
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China; Liaoning Key Laboratory of New Drug Research & Development, Shenyang 110036, China.
| |
Collapse
|
4
|
Wang Y, Li D, Li Q, Basnet A, Efird JT, Seki N. Neutrophil estimation and prognosis analysis based on existing lung squamous cell carcinoma datasets: the development and validation of a prognosis prediction model. Transl Lung Cancer Res 2024; 13:2023-2037. [PMID: 39263021 PMCID: PMC11384491 DOI: 10.21037/tlcr-24-411] [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: 05/08/2024] [Accepted: 08/06/2024] [Indexed: 09/13/2024]
Abstract
Background Notwithstanding the rapid developments in precision medicine in recent years, lung cancer still has a low survival rate, especially lung squamous cell cancer (LUSC). The tumor microenvironment (TME) plays an important role in the progression of lung cancer, in which high neutrophil levels are correlated with poor prognosis, potentially due to their interactions with tumor cells via pro-inflammatory cytokines and chemokines. However, the precise mechanisms of how neutrophils influence lung cancer remain unclear. This study aims to explore these mechanisms and develop a prognosis predictive model in LUSC, addressing the knowledge gap in neutrophil-related cancer pathogenesis. Methods LUSC datasets from the Xena Hub and Gene Expression Omnibus (GEO) databases were used, comprising 473 tumor samples and 195 tumor samples, respectively. Neutrophil contents in these samples were estimated using CIBERSORT, xCell, and microenvironment cell populations (MCP) counter tools. Differentially expressed genes (DEGs) were identified using DEseq2, and a weighted gene co-expression network analysis (WGCNA) was performed to identify neutrophil-related genes. A least absolute shrinkage and selection operator (LASSO) Cox regression model was constructed for prognosis prediction, and the model's accuracy was validated using Kaplan-Meier survival curves and time-dependent receiver operating characteristic (ROC) curves. Additionally, genomic changes, immune correlations, drug sensitivity, and immunotherapy response were analyzed to further validate the model's predictive power. Results Neutrophil content was significantly higher in adjacent normal tissue compared to LUSC tissue (P<0.001). High neutrophil content was associated with worse overall survival (OS) (P=0.02), disease-free survival (DFS) (P=0.02), and progression-free survival (PFS) (P=0.03) using different software estimates. Nine gene modules were identified, with blue and yellow modules showing strong correlations with neutrophil prognosis (P<0.001). Eight genes were selected for the prognostic model, which accurately predicted 1-, 3-, and 5-year survival in both the training set [area under the curve (AUC) value =0.60, 0.63, 0.66, respectively] and validation set (AUC value =0.58, 0.58, 0.59, respectively), with significant prognosis differences between high- and low-risk groups (P<0.001). The model's independent prognostic factors included risk group, pathologic M stage, and tumor stage (P<0.05). A further molecular mechanism analysis revealed differences between risk groups were revealed in immune checkpoint and human leukocyte antigen (HLA) gene expression, hallmark pathways, drug sensitivity, and immunotherapy responses. Conclusions This study established a risk-score model that effectively predicts the prognosis of LUSC patients and sheds light on the molecular mechanisms involved. The findings enhance the understanding of neutrophil-tumor interactions, offering potential targets for personalized treatments. However, further experimental validation and clinical studies are required to confirm these findings and address study limitations, including reliance on public databases and focus on a specific lung cancer subtype.
Collapse
Affiliation(s)
- Youyu Wang
- Department of Thoracic Surgery, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Dongfang Li
- Department of Thoracic Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Qiang Li
- Department of Oncology, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Alina Basnet
- Division of Hematology-Oncology, Upstate Cancer Center, Upstate Medical University, Syracuse, NY, USA
| | - Jimmy T Efird
- VA Cooperative Studies Program Coordinating Center, Boston, MA, USA
- Department of Radiation Oncology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Nobuhiko Seki
- Division of Medical Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Xiao X, Zheng L, Zeng H, Chen D, Liu L, Dong C, Zhang Y. Expression and diagnostic value of serum free light chain in lung cancer. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:914-920. [PMID: 39311787 PMCID: PMC11420966 DOI: 10.11817/j.issn.1672-7347.2024.230576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Indexed: 09/26/2024]
Abstract
OBJECTIVES The expression of serum free light chain (FLC) is abnormal in various diseases, but its role in lung cancer remains unclear. This study aims to investigate the expression and diagnostic value of serum FLC in lung cancer. METHODS A total of 80 lung cancer patients treated at Xiangdong Hospital, Hunan Normal University from January to December 2021 were selected as the lung cancer group. Another 80 healthy individuals undergoing routine physical examinations during the same period were chosen as the control group. General information and serum κFLC and λFLC levels were collected for all subjects. Clinical indicators such as serum carcinoembryonic antigen (CEA), cytokeratin fragment antigen 21-1 (CYFRA21-1) levels, tumor diameter, histological type, TNM stage, and lymph node metastasis status were recorded for lung cancer patients. The expression levels of serum FLC [κFLC, λFLC, and FLC (κ+λ)] were compared between the lung cancer group and the control group. Lung cancer patients were grouped based on gender, age, smoking history, tumor diameter, TNM stage, histological type, and lymph node metastasis to compare differences in serum κFLC and λFLC levels. Receiver operating characteristic (ROC) curves were used to evaluate the diagnostic value of serum FLC alone and in combination with other indicators in lung cancer. RESULTS The expression levels of serum FLC (κ+λ) and κFLC were significantly higher in the lung cancer group than those in the control group (both P<0.001), while there was no significant difference in serum λFLC levels between the 2 groups (P>0.05). There were no significant differences in serum κFLC levels among lung cancer patients with different tumor diameters, histological types, or TNM stages (all P>0.05); however, serum κFLC levels were higher in lung cancer patients with lymph node metastasis than in those without, with statistical significance (P=0.033). There were no significant differences in serum λFLC levels based on tumor diameter or histological type (both P>0.05), but serum λFLC levels were higher in stage III+IV and lymph node metastatic lung cancer patients compared to stage I+II and non-metastatic patients, with statistical significance (P=0.033 and P=0.019, respectively). The area under the curve (AUC) for κFLC and CEA in diagnosing lung cancer showed no significant difference (P=0.333). The combination of κFLC+CYFRA21-1 had the highest diagnostic efficacy (AUC=0.875) and sensitivity (71.3%). The AUC for the combined diagnosis of κFLC+λFLC+CEA+CYFRA21-1 was 0.915 (95% CI 0.860 to 0.953, P<0.001). CONCLUSIONS Serum FLC is highly expressed in lung cancer and is associated with its invasion and metastasis. Serum FLC, particularly κFLC, has diagnostic value for lung cancer, and the combined detection of FLC, CEA, and CYFRA21-1 offers the best diagnostic efficacy.
Collapse
Affiliation(s)
- Xi Xiao
- Department of Respiratory Medicine, Xiangdong Hospital, Hunan Normal University, Liling Hunan 412200.
| | - Li Zheng
- Department of Respiratory Medicine, Xiangdong Hospital, Hunan Normal University, Liling Hunan 412200.
| | - Hua Zeng
- Department of Neurology, Xiangdong Hospital, Hunan Normal University, Liling Hunan 412200
| | - Dan Chen
- Department of Respiratory Medicine, Xiangdong Hospital, Hunan Normal University, Liling Hunan 412200.
| | - Liqin Liu
- Department of Radiology, Xiangdong Hospital, Hunan Normal University, Liling Hunan 412200, China
| | - Caimei Dong
- Department of Respiratory Medicine, Xiangdong Hospital, Hunan Normal University, Liling Hunan 412200
| | - Yanping Zhang
- Department of Respiratory Medicine, Xiangdong Hospital, Hunan Normal University, Liling Hunan 412200
| |
Collapse
|
6
|
Roshan-Zamir M, Khademolhosseini A, Rajalingam K, Ghaderi A, Rajalingam R. The genomic landscape of the immune system in lung cancer: present insights and continuing investigations. Front Genet 2024; 15:1414487. [PMID: 38983267 PMCID: PMC11231382 DOI: 10.3389/fgene.2024.1414487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
Lung cancer is one of the most prevalent malignancies worldwide, contributing to over a million cancer-related deaths annually. Despite extensive research investigating the genetic factors associated with lung cancer susceptibility and prognosis, few studies have explored genetic predispositions regarding the immune system. This review discusses the most recent genomic findings related to the susceptibility to or protection against lung cancer, patient survival, and therapeutic responses. The results demonstrated the effect of immunogenetic variations in immune system-related genes associated with innate and adaptive immune responses, cytokine, and chemokine secretions, and signaling pathways. These genetic diversities may affect the crosstalk between tumor and immune cells within the tumor microenvironment, influencing cancer progression, invasion, and prognosis. Given the considerable variability in the individual immunegenomics profiles, future studies should prioritize large-scale analyses to identify potential genetic variations associated with lung cancer using highthroughput technologies across different populations. This approach will provide further information for predicting response to targeted therapy and promotes the development of new measures for individualized cancer treatment.
Collapse
Affiliation(s)
- Mina Roshan-Zamir
- School of Medicine, Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Khademolhosseini
- School of Medicine, Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kavi Rajalingam
- Cowell College, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Abbas Ghaderi
- School of Medicine, Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Raja Rajalingam
- Immunogenetics and Transplantation Laboratory, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
7
|
Torres-Martínez S, Calabuig-Fariñas S, Gallach S, Mosqueda M, Munera-Maravilla E, Sirera R, Navarro L, Blasco A, Camps C, Jantus-Lewintre E. Circulating Immune Proteins: Improving the Diagnosis and Clinical Outcome in Advanced Non-Small Cell Lung Cancer. Int J Mol Sci 2023; 24:17587. [PMID: 38139416 PMCID: PMC10743468 DOI: 10.3390/ijms242417587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023] Open
Abstract
Immunotherapy has been proven a viable treatment option for non-small cell lung cancer (NSCLC) treatment in patients. However, some patients still do not benefit. Finding new predictive biomarkers for immunocheckpoint inhibitor (ICI) response will improve treatment management in the clinical routine. In this regard, liquid biopsy is a useful and noninvasive alternative to surgical biopsies. In the present study, we evaluated the potential diagnostic, prognostic, and predictive value of seven different soluble mediators involved in immunoregulation. Fifty-two plasma samples from advanced NSCLC treated in first-line with pembrolizumab at baseline (PRE) and at first response assessment (FR) were analyzed. In terms of diagnostic value, our results revealed that sFGL1, sGAL-3, and sGAL-1 allowed for optimal diagnostic efficacy for cancer patients. Additionally, the combination of sFGL1 and sGAL-3 significantly improved diagnostic accuracy. Regarding the predictive value to assess patients' immune response, sCD276 levels at PRE were significantly higher in patients without tumor response (p = 0.035). Moreover, we observed that high levels of sMICB at PRE were associated with absence of clinical benefit (pembrolizumab treatment less than 6 months) (p = 0.049), and high levels of sMICB and sGAL-3 at FR are also related to a lack of clinical benefit (p = 0.027 and p = 0.03, respectively). Finally, in relation to prognosis significance, at PRE and FR, sMICB levels above the 75th percentile are related to poor progression-free survival (PFS) (p = 0.013 and p = 0.023, respectively) and overall survival (OS) (p = 0.001 and p = 0.011, respectively). An increase in sGAL3 levels at FR was associated with worse PFS (p = 0.037). Interestingly, high sGAL-3 at PRE was independently associated with PFS and OS with a hazard ratio (HR) of 2.45 (95% CI 1.14-5.25; p = 0.021) and 4.915 (95% CI 1.89-12.73; p = 0.001). In conclusion, plasma levels of sFGL1, sGAL-3, and sGAL-1 could serve as diagnostic indicators and sMICB, sCD276, and sGAL3 were linked to outcomes, suggesting their potential in assessing NSCLC under pembrolizumab treatment. Our results highlight the value of employing soluble immune biomarkers in advanced lung cancer patients treated with pembrolizumab at first-line.
Collapse
Affiliation(s)
- Susana Torres-Martínez
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Silvia Calabuig-Fariñas
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
- Department of Pathology, Universitat de València, 46010 Valencia, Spain
| | - Sandra Gallach
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Marais Mosqueda
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
| | - Ester Munera-Maravilla
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Rafael Sirera
- Department of Biotechnology, Universitat Politècnica de València, 46022 Valencia, Spain;
| | - Lara Navarro
- Department of Pathology, Hospital General Universitario de Valencia, 46014 Valencia, Spain;
| | - Ana Blasco
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
- Department of Medical Oncology, Hospital General Universitario de Valencia, 46014 Valencia, Spain
| | - Carlos Camps
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
- Department of Medical Oncology, Hospital General Universitario de Valencia, 46014 Valencia, Spain
- Department of Medicine, Universitat de València, 46010 Valencia, Spain
| | - Eloisa Jantus-Lewintre
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
- Department of Biotechnology, Universitat Politècnica de València, 46022 Valencia, Spain;
- Nanomedicine, Centro Investigación Príncipe Felipe—Universitat Politècnica de Valencia, 46022 Valencia, Spain
| |
Collapse
|
8
|
Wang Y, Zhu J, Zhou N, Wang Y, Zhang X. Changes in T lymphocyte subsets predict the efficacy of atezolizumab in advanced non-small cell lung cancer: a retrospective study. J Thorac Dis 2023; 15:5669-5679. [PMID: 37969284 PMCID: PMC10636460 DOI: 10.21037/jtd-23-1169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/01/2023] [Indexed: 11/17/2023]
Abstract
Background It has remained unclear how programmed cell death ligand 1 (PD-L1) inhibitors affect peripheral blood lymphocyte (PBL) subsets in patients with advanced non-small cell lung cancer (NSCLC). This study assessed the predictive and prognostic value of PBL subsets in patients with advanced NSCLC who were treated with atezolizumab. Methods A total of 30 patients with advanced NSCLC treated with atezolizumab were selected as the observation group, and 30 healthy individuals were chosen as the control group during same period. Flow cytometry was used to detect lymphocyte subsets before and after treatment. The relationship between the changes of lymphocyte subsets and atezolizumab in the treatment of NSCLC was analyzed and calculated. Results Before treatment, compared with the control group, the number of CD3+, CD4+ T, and CD4+/CD8+ indexes in the observation group were significantly decreased, whereas the level of CD8+ was significantly increased. The number of CD3+, CD4+ T, and CD4+/CD8+ indexes gradually increased with the process of atezolizumab treatment, whereas the number of CD8+ T gradually decreased. After the 4 cycles, the number of CD3+, CD4+ T, and CD4+/CD8+ indexes were significantly increased, and the number of CD8+ was significantly decreased. In the observation group, 22 patients achieved partial response (PR)/stable disease (SD) and 8 patients achieved progressive disease (PD) after 4 cycles of atezolizumab treatment. Before treatment, there were no significant differences in the level of lymphocyte subsets between those who achieved PR/SD or PD. However, a significant difference in the level of lymphocyte subsets appeared after 4 cycles of atezolizumab treatment. Among the 22 patients who achieved PR/SD, the number of CD3+, CD4+ T, and CD4+/CD8+ indexes were significantly increased, whereas the number of CD8+ T lymphocytes was significantly decreased. Meanwhile, the 8 patients who achieved PD displayed different results. In addition, ROC curve combined detection of CD3+, CD4+, and CD8+ T [area under the curve (AUC) =0.9018, P<0.0001] showed good predictive ability for the efficacy of atezolizumab in advanced NSCLC. Conclusions Atezolizumab may alter the level of lymphocyte subsets in patients with advanced NSCLC, and the changes in lymphocyte subsets may predict the efficacy of atezolizumab for these patients.
Collapse
Affiliation(s)
- Yuyang Wang
- Qingdao Medical College, Qingdao University, Qingdao, China
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingjuan Zhu
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Na Zhou
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yun Wang
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xiaochun Zhang
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Shehata SA, Toraih EA, Ismail EA, Hagras AM, Elmorsy E, Fawzy MS. Vaping, Environmental Toxicants Exposure, and Lung Cancer Risk. Cancers (Basel) 2023; 15:4525. [PMID: 37760496 PMCID: PMC10526315 DOI: 10.3390/cancers15184525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/18/2023] [Accepted: 06/22/2023] [Indexed: 09/29/2023] Open
Abstract
Lung cancer (LC) is the second-most prevalent tumor worldwide. According to the most recent GLOBOCAN data, over 2.2 million LC cases were reported in 2020, with an estimated new death incident of 1,796,144 lung cancer cases. Genetic, lifestyle, and environmental exposure play an important role as risk factors for LC. E-cigarette, or vaping, products (EVPs) use has been dramatically increasing world-wide. There is growing concern that EVPs consumption may increase the risk of LC because EVPs contain several proven carcinogenic compounds. However, the relationship between EVPs and LC is not well established. E-cigarette contains nicotine derivatives (e.g., nitrosnornicotine, nitrosamine ketone), heavy metals (including organometal compounds), polycyclic aromatic hydrocarbons, and flavorings (aldehydes and complex organics). Several environmental toxicants have been proven to contribute to LC. Proven and plausible environmental carcinogens could be physical (ionizing and non-ionizing radiation), chemicals (such as asbestos, formaldehyde, and dioxins), and heavy metals (such as cobalt, arsenic, cadmium, chromium, and nickel). Air pollution, especially particulate matter (PM) emitted from vehicles and industrial exhausts, is linked with LC. Although extensive environmental exposure prevention policies and smoking reduction strategies have been adopted globally, the dangers remain. Combined, both EVPs and toxic environmental exposures may demonstrate significant synergistic oncogenicity. This review aims to analyze the current publications on the importance of the relationship between EVPs consumption and environmental toxicants in the pathogenesis of LC.
Collapse
Affiliation(s)
- Shaimaa A. Shehata
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; (S.A.S.); (A.M.H.)
| | - Eman A. Toraih
- Division of Endocrine and Oncologic Surgery, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA;
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ezzat A. Ismail
- Department of Urology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Abeer M. Hagras
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; (S.A.S.); (A.M.H.)
| | - Ekramy Elmorsy
- Department of Pathology, Faculty of Medicine, Northern Border University, Arar 73213, Saudi Arabia;
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Manal S. Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 73213, Saudi Arabia
| |
Collapse
|