1
|
Wang L, Kong Y, Zhang Y, Mu C. The impact of PD-L1 expression status on the prognosis of ALK-positive lung cancer patients. Cancer Treat Res Commun 2025; 42:100868. [PMID: 39908681 DOI: 10.1016/j.ctarc.2025.100868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/31/2024] [Accepted: 01/09/2025] [Indexed: 02/07/2025]
Abstract
OBJECTIVE This study analyzes the clinical characteristics of ALK-positive lung cancer patients and the impact of PD-L1 expression on their prognosis, providing insights for diagnosis and treatment. MATERIALS AND METHODS A total of 291 ALK-positive lung cancer patients, tested for PD-L1 expression at the First Affiliated Hospital of Soochow University between January 2019 and November 2021, were included. Clinical data and prognostic information were collected. Statistical analysis was conducted using SPSS 26.0 to explore the relationship between clinical features and prognosis. RESULTS In ALK positive patients, the PD-L1 positivity rate was 59.8 %, with 19.2 % showing strong positivity. PD-L1 positive patients were predominantly male, over 55 years old, and presented more clinical symptoms, higher Ki-67 expression, larger tumor sizes, and more advanced disease stages. The progression-free survival (PFS) and overall survival (OS) were significantly lower in the ALK+PD-L1+ group compared to the ALK+PD-L1- group. Those receiving targeted therapy in stage IIIB-IVB non-squamous non-small cell lung cancer had significantly higher PFS and OS. CONCLUSION PD-L1 positivity is common in ALK-positive lung cancer patients and correlates with poorer prognosis. PD-L1 serves as an independent predictor of adverse outcomes, indicating its potential as a biomarker for assessing lung cancer severity and prognosis.
Collapse
Affiliation(s)
- Li Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yingjun Kong
- Department of Respiratory Medicine, Pingshan District Central Hospital of Shenzhen, Guangdong, 518122, China
| | - Yao Zhang
- Department of Respiratory Medicine, Pingshan District Central Hospital of Shenzhen, Guangdong, 518122, China
| | - Chuanyong Mu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
2
|
Chauhan P, Pandey P, Ramniwas S, Khan F. Review Deciphering the Potential of Nanotherapeutics in Lung Cancer Management. Curr Cancer Drug Targets 2025; 25:539-554. [PMID: 38561624 DOI: 10.2174/0115680096302203240308104740] [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: 01/03/2024] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 04/04/2024]
Abstract
Lung cancer remains a formidable challenge in oncology, necessitating the development of more effective prognostic and diagnostic techniques due to inefficient conventional therapeutic approaches and inadequate methods for early lung cancer diagnosis. Despite immense progress in the development of innovative strategies to alleviate the impact of this devastating disease, the outcomes, unfortunately, remain unsatisfactory, particularly in targeted drug delivery methods. Consequently, nanotechnology has emerged as a revolutionary force in cancer research to develop more effective targeted drug delivery tools due to its extraordinary capacity at the atomic and molecular levels. It has appeared as a beacon of hope in this area of unmet need, providing innovative ways for the prognosis and diagnosis of lung carcinoma. Therefore, this comprehensive review delves into the evolving field of nano-based therapeutics, shedding light on their potential to transform lung cancer treatment. This study meticulously explores the most promising nano-based strategies that have been extensively linked with the treatment of lung carcinoma and mainly emphasizes targeted drug delivery methods and therapies. Additionally, this review encapsulates the favorable results of clinical trials, which support the potential pathways for further development of nanotherapeutics in lung cancer management.
Collapse
Affiliation(s)
- Prashant Chauhan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, Uttar Pradesh, India
| | - Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, Uttar Pradesh, India
| | - Seema Ramniwas
- University Centre of Research and Development, University Institute of Biotechnology, Chandigarh University Gharuan, Mohali, Punjab, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
3
|
Tang L, Wang T. A fatty acid metabolism-related genes model for predicting the prognosis and immunotherapy effect of lung adenocarcinoma. Cancer Biomark 2024; 41:18758592241296285. [PMID: 40095456 DOI: 10.1177/18758592241296285] [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: 03/19/2025]
Abstract
ObjectiveLung adenocarcinoma (LUAD) is a common and highly heterogeneous malignancy cancer with increasing morbidity and mortality. Dysregulation of fatty acid metabolism (FAM) has been identified as a key regulator of LUAD progression. Our purpose was to establish a risk model of FAM-related genes to provide a reference for the prognosis prediction of LUAD.MethodsFirstly, we screened FAM-related differentially expressed genes (DEGs) based on the Cancer Genome Atlas (TCGA) database, and identified the prognostic signatures by Cox-regression analysis. The least absolute shrinkage and selection operator algorithm (LASSO) was used to obtain the formula for risk model. And the analysis of Gene Expression Omnibus (GEO) dataset used to verify. Nomogram was produced for individualized prediction in clinical treatment. Immune cell function and drug sensitivity analysis used to screen potential therapeutic drugs.ResultsPatients in low-risk had better overall survival (OS). High-risk patients exhibit higher TMB and lower TIDE scores, and they are more likely to benefit from immunotherapy. The analysis of GEO verified that risk model has a high prediction accuracy.ConclusionThe risk model based on 17 FAM-related DEGs is of great value in predicting the prognosis of LUAD, and these prognostic signatures may be potential therapeutic targets for LUAD.
Collapse
Affiliation(s)
- Lingxue Tang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Tong Wang
- Department of General Practice, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
4
|
Zanini U, Faverio P, Bonfanti V, Falzone M, Cortinovis D, Arcangeli S, Petrella F, Ferrara G, Mura M, Luppi F. The 'Liaisons dangereuses' Between Lung Cancer and Interstitial Lung Diseases: A Focus on Acute Exacerbation. J Clin Med 2024; 13:7085. [PMID: 39685543 DOI: 10.3390/jcm13237085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Patients with interstitial lung disease (ILD) are about five times more likely to develop lung cancer than those without ILD. The presence of ILD in lung cancer patients complicates diagnosis and management, resulting in lower survival rates. Diagnostic and treatment procedures needed for cancer can increase the risk of acute exacerbation (AE), one of the most severe complications for these patients. Bronchoscopic techniques are generally considered safe, but they can trigger AE-ILD, particularly after cryoprobe biopsies. Surgical procedures for lung cancer, including lung biopsies and resections, carry an elevated risk of AE-ILD. Postoperative complications and mortality rates highlight the importance of meticulous surgical planning and postoperative care. Furthermore, cancer treatments, such as chemotherapy, are all burdened by a risk of AE-ILD occurrence. Radiotherapy is important for managing both early-stage and advanced lung cancer, but it also poses risks. Stereotactic body radiation and particle beam therapies have varying degrees of safety, with the latter potentially offering a lower risk of AE. Percutaneous ablation techniques can help patients who are not eligible for surgery. However, these procedures may complicate ILD, and their associated risks still need to be fully understood, necessitating further research for improved safety. Overall, while advancements in lung cancer treatment have improved outcomes for many patients, the complexity of managing patients with concomitant ILD needs careful consideration and multidisciplinary assessment. This review provides a detailed evaluation of these risks, emphasizing the need for personalized treatment approaches and monitoring to improve patient outcomes in this challenging population.
Collapse
Affiliation(s)
- Umberto Zanini
- Department of Medicine and Surgery, University of Milano-Bicocca, SC Pneumologia, Fondazione IRCCS "San Gerardo dei Tintori", 20900 Monza, Italy
| | - Paola Faverio
- Department of Medicine and Surgery, University of Milano-Bicocca, SC Pneumologia, Fondazione IRCCS "San Gerardo dei Tintori", 20900 Monza, Italy
| | - Valentina Bonfanti
- Department of Medicine and Surgery, University of Milano-Bicocca, SC Pneumologia, Fondazione IRCCS "San Gerardo dei Tintori", 20900 Monza, Italy
| | - Maria Falzone
- Department of Medicine and Surgery, University of Milano-Bicocca, SC Pneumologia, Fondazione IRCCS "San Gerardo dei Tintori", 20900 Monza, Italy
| | - Diego Cortinovis
- Department of Medicine and Surgery, University of Milano-Bicocca, SC Oncologia, Fondazione IRCCS "San Gerardo dei Tintori", 20900 Monza, Italy
| | - Stefano Arcangeli
- Department of Medicine and Surgery, University of Milano-Bicocca, SC Radioterapia, Fondazione IRCCS "San Gerardo dei Tintori", 20900 Monza, Italy
| | - Francesco Petrella
- Department of Medicine and Surgery, University of Milano-Bicocca, SC Chirurgia Toracica, Fondazione IRCCS "San Gerardo dei Tintori", 20900 Monza, Italy
| | - Giovanni Ferrara
- Division of Pulmonary Medicine, University of Alberta, and Alberta Health Services, Edmonton, AB T6G 2B7, Canada
| | - Marco Mura
- Division of Respirology, Western University, London, ON N6A 3K7, Canada
| | - Fabrizio Luppi
- Department of Medicine and Surgery, University of Milano-Bicocca, SC Pneumologia, Fondazione IRCCS "San Gerardo dei Tintori", 20900 Monza, Italy
| |
Collapse
|
5
|
Mehta A, Priya V P L, Teja L, Pillai K. The Prevalence of Programmed Death Ligand-1 (PD-L1) Expression in Non-Small Cell Lung Cancer: An Experience From Tertiary Care Hospital. Cureus 2024; 16:e72291. [PMID: 39583517 PMCID: PMC11585287 DOI: 10.7759/cureus.72291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND The prevalence of programmed cell death ligand-1(PD-L1) expression in non-small cell lung cancer (NSCLC) within unselected populations remains a research topic, though PD-L1 expression is important in guiding treatment decisions. OBJECTIVES The study objective was to ascertain the prevalence of PD-L1 expression in patients of NSCLC and its association with clinicopathological characteristics and other gene mutations. METHODS Samples from patients with NSCLCs were analyzed in the study to determine the expression of PD-L1 through immunohistochemistry (IHC) using rabbit anti-human PDL-1/CD274 monoclonal antibody. Correlation analysis was done using Pearson's correlation coefficient (r). The study analyzed the association between PD-L1 expression and clinicopathological features. RESULTS A total of 245 consecutive patients (154 men with 62.9%) with NSCLCs were subjected to PD-L1 testing, and 30.6% (n=75) were identified to be positive. It was twice as prevalent in men than women, with 154 men and 91 females, respectively. The PD-L1 expression failed to demonstrate any statistical significance with age, gender, smoking status, type of materials, location of biopsy, Eastern Cooperative Oncology Group Performance Status, and Epidermal Growth Factor Receptor mutation. High PD-L1 expression with tumor proportion score (TPS ≥ 50) was observed in 38.8% of patients, and it was more prevalent in female patients 29 (38.7%), smokers 59 (78.7%), stage IV tumors 44 (58.7%), and stage III tumors 28 (37.3%). CONCLUSIONS Our study in Kerala showed PD-L1 expression in NSCLC patients, correlated with clinicopathologic factors. Males, COPD, ALK+, and advanced-stage tumors had higher expression. Females, smokers, poorly differentiated tumors, and stage IV tumors had high PD-L1.
Collapse
Affiliation(s)
- Asmita Mehta
- Respiratory Medicine, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, IND
| | - Lakshmi Priya V P
- Respiratory Medicine, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, IND
| | - Lakshmi Teja
- Respiratory Medicine, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, IND
| | - Krishna Pillai
- Respiratory Medicine, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, IND
| |
Collapse
|
6
|
Na SW, Yi JM, Yeo H, Park SM, Jeong M, Chun J, Jeong MK. Bojungikki-Tang Augments Pembrolizumab Efficacy in Human PBMC-Injected H460 Tumor-Bearing Mice. Life (Basel) 2024; 14:1246. [PMID: 39459546 PMCID: PMC11508561 DOI: 10.3390/life14101246] [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: 06/10/2024] [Revised: 09/02/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Bojungikki-Tang (BJIKT) is traditionally used to enhance digestive function and immunity. It has gained attention as a supplement to chemotherapy or targeted therapy owing to its immune-boosting properties. This study aimed to evaluate the synergistic anti-tumor effects of BJIKT in combination with pembrolizumab in a preclinical model. MHC I/II double knockout NSG mice were humanized with peripheral blood mononuclear cells (PBMCs) and injected subcutaneously with H460 lung tumor cells to establish a humanized tumor model. Both agents were administered to evaluate their impact on tumor growth and immune cell behavior. Immunohistochemistry showed decreased exhaustion markers in CD8(+) and CD4(+) T cells within the tumor, indicating enhanced T cell activity. Additionally, RNA sequencing, transcriptome analysis, and quantitative PCR analysis were performed on tumor tissues to investigate the molecular mechanisms underlying the observed effects. The results confirmed that BJIKT improved T cell function and tumor necrosis factor signaling while suppressing transforming growth factor-β signaling. This modulation led to cell cycle arrest and apoptosis. These findings demonstrate that BJIKT, when combined with pembrolizumab, produces significant anti-tumor effects by altering immune pathways and enhancing the anti-tumor immune response. This study provides valuable insights into the role of BJIKT in the tumor microenvironment and its potential to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Se Won Na
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; (S.W.N.); (J.-M.Y.); (M.J.)
| | - Jin-Mu Yi
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; (S.W.N.); (J.-M.Y.); (M.J.)
| | - Heerim Yeo
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea; (H.Y.); (S.-M.P.)
| | - Sang-Min Park
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea; (H.Y.); (S.-M.P.)
| | - Mibae Jeong
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; (S.W.N.); (J.-M.Y.); (M.J.)
| | - Jaemoo Chun
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; (S.W.N.); (J.-M.Y.); (M.J.)
| | - Mi-Kyung Jeong
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; (S.W.N.); (J.-M.Y.); (M.J.)
| |
Collapse
|
7
|
Gentzler RD, Guittar J, Mitra A, Iams WT, Driessen T, Schwind R, Stein MM, Kaneva K, Hyun SW, Liu Y, Dugan AJ, Vibat CRT, Sangli C, Freaney J, Rivers Z, Feliciano JL, Lo C, Sasser K, Ben-Shachar R, Nimeiri H, Patel JD, Chaudhuri AA. Dynamic Changes in Circulating Tumor Fraction as a Predictor of Real-World Clinical Outcomes in Solid Tumor Malignancy Patients Treated with Immunotherapy. Oncol Ther 2024; 12:509-524. [PMID: 39037536 PMCID: PMC11333675 DOI: 10.1007/s40487-024-00287-2] [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: 04/11/2024] [Accepted: 06/10/2024] [Indexed: 07/23/2024] Open
Abstract
INTRODUCTION A dynamic molecular biomarker that can identify early efficacy of immune checkpoint inhibitor (ICI) therapy remains an unmet clinical need. Here we evaluate if a novel circulating tumor DNA (ctDNA) assay, xM, used for treatment response monitoring (TRM), that quantifies changes in ctDNA tumor fraction (TF), can predict outcome benefits in patients treated with ICI alone or in combination with chemotherapy in a real-world (RW) cohort. METHODS This retrospective study consisted of patients with advanced cancer from the Tempus de-identified clinical genomic database who received longitudinal liquid-based next-generation sequencing. Eligible patients had a blood sample ≤ 40 days prior to the start of ICI initiation and an on-treatment blood sample 15-180 days post ICI initiation. TF was calculated via an ensemble algorithm that utilizes TF estimates derived from variants and copy number information. Patients with molecular response (MR) were defined as patients with a ≥ 50% decrease in TF between tests. In the subset of patients with rw-imaging data between 2 and 18 weeks of ICI initiation, the predictive value of MR in addition to rw-imaging was compared to a model of rw-imaging alone. RESULTS The evaluable cohort (N = 86) was composed of 14 solid cancer types. Patients received either ICI monotherapy (38.4%, N = 33) or ICI in combination with chemotherapy (61.6%, N = 53). Patients with MR had significantly longer rw-overall survival (rwOS) (hazard ratio (HR) 0.4, P = 0.004) and rw-progression free survival (rwPFS) (HR 0.4, P = 0.005) than patients with molecular non-response (nMR). Similar results were seen in the ICI monotherapy subcohort; HR 0.2, P = 0.02 for rwOS and HR 0.2, P = 0.01 for rwPFS. In the subset of patients with matched rw-imaging data (N = 51), a model incorporating both MR and rw-imaging was superior in predicting rwOS than rw-imaging alone (P = 0.02). CONCLUSIONS xM used for TRM is a novel serial quantitative TF algorithm that can be used clinically to evaluate ICI therapy efficacy.
Collapse
Affiliation(s)
- Ryan D Gentzler
- Department of Hematology and Oncology, University of Virginia, Charlottesville, VA, USA
| | - John Guittar
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Akash Mitra
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Wade T Iams
- Division of Hematology/Oncology, Vanderbuilt-Ingram Cancer Center, Nashville, TN, USA
| | - Terri Driessen
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Regina Schwind
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Michelle M Stein
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Kristiyana Kaneva
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Seung Won Hyun
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Yan Liu
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Adam J Dugan
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Cecile Rose T Vibat
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Chithra Sangli
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Jonathan Freaney
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Zachary Rivers
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | | | - Christine Lo
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Kate Sasser
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Rotem Ben-Shachar
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA.
| | - Halla Nimeiri
- Tempus AI, Inc., 600 West Chicago Avenue, Suite 510, Chicago, IL, 60654, USA
| | - Jyoti D Patel
- Department of Medicine, Northwestern University, Evanston, IL, USA
| | - Aadel A Chaudhuri
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
8
|
Ma S, Li F, Wang L. The Construction of a Nomogram Using the Pan-Immune-Inflammation Value Combined with a PILE Score for Immunotherapy Prediction Prognosis in Advanced NSCLC. Cancer Manag Res 2024; 16:741-751. [PMID: 38974092 PMCID: PMC11227331 DOI: 10.2147/cmar.s461964] [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: 01/30/2024] [Accepted: 06/18/2024] [Indexed: 07/09/2024] Open
Abstract
Purpose The purpose of this study was to investigate the predictive value of Pan-Immune-Inflammation Value (PIV) combined with the PILE score for immunotherapy in patients with advanced non-small cell lung cancer (NSCLC) and to construct a nomogram prediction model to provide reference for clinical work. Patients and Methods Patients with advanced NSCLC who received ICIs treatment in Qingdao Municipal Hospital from January 2019 to December 2021 were selected as the study subjects. The chi-square test, Kaplan-Meier survival analysis, and Cox proportional risk regression analysis were used to evaluate the prognosis. The results were visualized by a nomogram, and the performance of the model was judged by indicators such as the area under the subject operating characteristic curve (AUC) and C-index. The patients were divided into high- and low-risk groups by PILE score, and the prognosis of patients in different risk groups was evaluated. Results Multivariate Cox regression analysis showed that immune-related adverse events (irAEs) were prognostic factors for overall survival (OS) improvement, and ECOG PS score ≥2, bone metastases before treatment, and high PIV expression were independent risk factors for OS. The C index of OS predicted by the nomogram model is 0.750 (95% CI: 0.677-0.823), and the Calibration and ROC curves show that the model has good prediction performance. Compared with the low-risk group, patients in the high-risk group of PILE were associated with a higher inflammatory state and poorer physical condition, which often resulted in a poorer prognosis. Conclusion PIV can be used as a prognostic indicator for patients with advanced NSCLC treated with ICIs, and a nomogram prediction model can be constructed to evaluate the survival prediction of patients, thus contributing to better clinical decision-making and prognosis assessment.
Collapse
Affiliation(s)
- Shixin Ma
- Graduate School, Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
- Department of Thoracic Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, 266071, People’s Republic of China
| | - Fei Li
- Department of Thoracic Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, 266071, People’s Republic of China
| | - Lunqing Wang
- Department of Thoracic Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, 266071, People’s Republic of China
| |
Collapse
|
9
|
Wang JN, Zhang Y, Huang CY, Li K, Yu XB. Development of Vogt-Koyanagi-Harada disease-like uveitis during treatment by anti-programmed death-1 antibody: a case report. BMC Ophthalmol 2024; 24:240. [PMID: 38849786 PMCID: PMC11157912 DOI: 10.1186/s12886-024-03484-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 05/15/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Several immune checkpoint inhibitors (ICIs) have been linked to the occurrence of Vogt-Koyanagi-Harada disease (VKHD)-like uveitis. Among the ICIs, there has been no report of immune-related adverse events (irAEs) caused by a new programmed death protein-1(PD-1) monoclonal antibody (Toripalimab). CASE PRESENTATION This paper presents a case of VKHD-like uveitis that arose following Toripalimab therapy for urothelial cancer of the bladder, and the patient experienced symptoms 10 days after the final dosage of 20 months of medication treatment. This patient with bladder uroepithelial carcinoma had severe binocular acute panuveitis with exudative retinal detachment after receiving Toripalimab therapy. Binocular VKHD-like uveitis was suggested as a diagnosis. Both eyes recovered after discontinuing immune checkpoint inhibitors and local and systemic corticosteroid treatment. CONCLUSIONS This report suggests that VKHD-like uveitis can also occur in patients receiving novel PD-1 antibodies and the importance of paying attention to eye complications in patients receiving treatment over a long period.
Collapse
Affiliation(s)
- Jia-Ning Wang
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Peking University Fifth School of Clinical Medicine, Beijing, China
| | - Yue Zhang
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Chen-Ye Huang
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Kang Li
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao-Bing Yu
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
10
|
Asakawa A, Yoshimoto R, Kobayashi M, Izumi N, Maejima T, Deguchi T, Kubota K, Takahashi H, Yamada M, Ishibashi S, Onishi I, Kinowaki Y, Kurata M, Kobayashi M, Ishibashi H, Okubo K, Ohashi K, Kitagawa M, Yamamoto K. The Comprehensive Characterization of B7-H3 Expression in the Tumor Microenvironment of Lung Squamous Cell Carcinoma: A Retrospective Study. Cancers (Basel) 2024; 16:2140. [PMID: 38893259 PMCID: PMC11171371 DOI: 10.3390/cancers16112140] [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: 04/26/2024] [Revised: 05/19/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Lung squamous cell carcinoma (LSCC) is refractory to various therapies for non-small cell cancer; therefore, new therapeutic approaches are required to improve the prognosis of LSCC. Although immunotherapies targeting B7 family molecules were explored as treatments for several cancer types, the expression and significance of B7-H3 in the tumor microenvironment (TME) and its relationship with other immune checkpoint molecules have not yet been investigated in detail. We used high-throughput quantitative multiplex immunohistochemistry to examine B7-H3 expression in the TME. We investigated the relationship between B7-H3 expression and prognosis as well as changes in the TME with B7-H3 expression using 110 surgically resected pathological specimens retrospectively. We examined the correlation between B7-H3 and programmed cell death-ligand 1 (PD-L1) expression in single cells. High B7-H3 expression in tumor cells was associated with a better prognosis and a significant increase in the number of CD163+PD-L1+ macrophages. Quantitative analysis revealed that there is a positive correlation between B7-H3 and PD-L1 expression in tumor and stromal cells, as well as in intratumoral tumor-infiltrating lymphocytes and tumor-associated macrophages in the same cells. CD68+, CD163+, and CK+ cells with PD-L1+ phenotypes had higher B7-H3 expression compared to PD-L1- cells. Our findings demonstrate a correlation between B7-H3 and PD-L1 expression in the same cells, indicating that therapies targeting B7-H3 could provide additional efficacy in patients refractory to PD-L1-targeting therapies.
Collapse
Affiliation(s)
- Ayaka Asakawa
- Department of Thoracic Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; (A.A.); (M.K.); (H.I.); (K.O.)
| | - Ryoto Yoshimoto
- Molecular Pathology Group, Translational Research Department, Daiichi Sankyo RD Novare, 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan; (R.Y.); (M.K.); (H.T.); (M.Y.)
| | - Maki Kobayashi
- Molecular Pathology Group, Translational Research Department, Daiichi Sankyo RD Novare, 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan; (R.Y.); (M.K.); (H.T.); (M.Y.)
| | - Nanae Izumi
- Translational Science Department, Daiichi Sankyo, Inc., Basking Ridge, NJ 07920, USA; (N.I.); (K.K.)
| | - Takanori Maejima
- Translational Science Department I, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan; (T.M.); (T.D.)
| | - Tsuneo Deguchi
- Translational Science Department I, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan; (T.M.); (T.D.)
| | - Kazuishi Kubota
- Translational Science Department, Daiichi Sankyo, Inc., Basking Ridge, NJ 07920, USA; (N.I.); (K.K.)
| | - Hisashi Takahashi
- Molecular Pathology Group, Translational Research Department, Daiichi Sankyo RD Novare, 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan; (R.Y.); (M.K.); (H.T.); (M.Y.)
| | - Miyuki Yamada
- Molecular Pathology Group, Translational Research Department, Daiichi Sankyo RD Novare, 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan; (R.Y.); (M.K.); (H.T.); (M.Y.)
| | - Sachiko Ishibashi
- Department of Comprehensive Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; (S.I.); (I.O.); (Y.K.); (M.K.); (M.K.)
| | - Iichiroh Onishi
- Department of Comprehensive Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; (S.I.); (I.O.); (Y.K.); (M.K.); (M.K.)
| | - Yuko Kinowaki
- Department of Comprehensive Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; (S.I.); (I.O.); (Y.K.); (M.K.); (M.K.)
| | - Morito Kurata
- Department of Comprehensive Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; (S.I.); (I.O.); (Y.K.); (M.K.); (M.K.)
| | - Masashi Kobayashi
- Department of Thoracic Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; (A.A.); (M.K.); (H.I.); (K.O.)
| | - Hironori Ishibashi
- Department of Thoracic Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; (A.A.); (M.K.); (H.I.); (K.O.)
| | - Kenichi Okubo
- Department of Thoracic Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; (A.A.); (M.K.); (H.I.); (K.O.)
| | - Kenichi Ohashi
- Department of Human Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan;
| | - Masanobu Kitagawa
- Department of Comprehensive Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; (S.I.); (I.O.); (Y.K.); (M.K.); (M.K.)
| | - Kouhei Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; (S.I.); (I.O.); (Y.K.); (M.K.); (M.K.)
- Department of Human Pathology, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan;
| |
Collapse
|
11
|
Gao F, You X, Yang L, Zou X, Sui B. Boosting immune responses in lung tumor immune microenvironment: A comprehensive review of strategies and adjuvants. Int Rev Immunol 2024; 43:280-308. [PMID: 38525925 DOI: 10.1080/08830185.2024.2333275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/12/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024]
Abstract
The immune system has a substantial impact on the growth and expansion of lung malignancies. Immune cells are encompassed by a stroma comprising an extracellular matrix (ECM) and different cells like stromal cells, which are known as the tumor immune microenvironment (TIME). TME is marked by the presence of immunosuppressive factors, which inhibit the function of immune cells and expand tumor growth. In recent years, numerous strategies and adjuvants have been developed to extend immune responses in the TIME, to improve the efficacy of immunotherapy. In this comprehensive review, we outline the present knowledge of immune evasion mechanisms in lung TIME, explain the biology of immune cells and diverse effectors on these components, and discuss various approaches for overcoming suppressive barriers. We highlight the potential of novel adjuvants, including toll-like receptor (TLR) agonists, cytokines, phytochemicals, nanocarriers, and oncolytic viruses, for enhancing immune responses in the TME. Ultimately, we provide a summary of ongoing clinical trials investigating these strategies and adjuvants in lung cancer patients. This review also provides a broad overview of the current state-of-the-art in boosting immune responses in the TIME and highlights the potential of these approaches for improving outcomes in lung cancer patients.
Collapse
Affiliation(s)
- Fei Gao
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Xiaoqing You
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Liu Yang
- Department of Oncology, Da Qing Long Nan Hospital, Daqing, Heilongjiang Province, China
| | - Xiangni Zou
- Department of Nursing, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Bowen Sui
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| |
Collapse
|
12
|
Ting Z, Wu Z, Yang C, Li Z, Huang H, Gan J, Li N, Li X, Lyu J, Wu Y, Qin S. lncRNA CERS6-AS1 upregulates the expression of ANLN by sponging miR-424-5p to promote the progression and drug resistance of lung adenocarcinoma. Noncoding RNA Res 2024; 9:221-235. [PMID: 38094657 PMCID: PMC10716711 DOI: 10.1016/j.ncrna.2023.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 10/17/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) play a crucial role in tumor generation and progression. However, the exact functional significance and underlying molecular mechanism by which lncRNA CERS6-AS1 operates in the context of lung adenocarcinoma (LUAD) remain unknown. We aimed to evaluate the potential role of the CERS6-AS1/miR-424-5p/ANLN axis in the progression of LUAD through bioinformatics and cytobehavioral experiments, and to provide a new insight into the combined treatment of LUAD. Based on the TCGA database, the expression of CERS6-AS1 in pan-cancer was evaluated, and its prognostic performance in LUAD was evaluated by ROC curve, survival curve and COX analysis. In addition, quantification of CERS6-AS1 expression levels in LUAD patients and lung cancer cells using quantitative real-time polymerase chain reaction (RT-qPCR), and further validate the functional significance of CERS6-AS1 in promoting the proliferation, migration, and invasion abilities of lung cancer cells. The competitive endogenous RNA (ceRNA) network was constructed, and miR-424-5p inhibitors were applied to CERS6-AS1 knockdown cells. The potential downstream genes associated with the regulatory axis of CERS6-AS1/miR-424-5p were analyzed by PPI network and gene enrichment analysis (KEGG). Finally, we evaluated the prognostic value of high expression of ANLN in LUAD and its effects on immune cell infiltration, tumor mutation burden, chemotherapy response, and immunotherapy. CERS6-AS1 expression was significantly elevated in both LUAD patients and lung cancer cells. In the CERS6-AS1 knockdown assay, the proliferation, invasion, migration and epithelial-mesenchymal transformation (EMT) of cancer cells were significantly inhibited. Notably, there was a prominent upregulation of miR-424-5p expression in cells where CERS6-AS1 was knocked down. Co-transfection of siRNA and miR-424-5p inhibitors into lung cancer cells restored the restriction on lung cancer cells. Anillin (ANLN) has been identified as a potential target gene for miR-424-5p and as a prognostic and immune biomarker associated with immune cell infiltration and tumor mutational burden in LUAD. Additionally, ANLN impacts the efficacy of chemotherapy and immunotherapy in LUAD patients. This study reveals a novel regulatory mechanism in which CERS6-AS1 may contribute to the progression of LUAD by influencing the expression of ANLN as a competitive sponge for miR-424-5p.
Collapse
Affiliation(s)
- Zhuo Ting
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zuotao Wu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Chuyi Yang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zihao Li
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Hongyu Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jinyan Gan
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Nijiao Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xiaohong Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jueqi Lyu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yanbin Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Shouming Qin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
13
|
Miao K, Zhang L. Pathogenesis, pathological characteristics and individualized therapy for immune-related adverse effects. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:215-222. [PMID: 39171279 PMCID: PMC11332905 DOI: 10.1016/j.pccm.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Indexed: 08/23/2024]
Abstract
Immune checkpoint inhibitors (ICIs) are a class of antitumor medications that target immune checkpoints, which induce the activation of lymphocytes. These treatments effectively prolong the survival of patients with advanced tumors, especially lung cancer. However, in addition to tumor killing effects, ICIs may also cause an imbalance between immune tolerance and immunity. Over-activated lymphocytes may cause various types of damage to multiple organs throughout the body, called immune-related adverse events. In this review, we summarize the pathogenesis, pathological characteristics, biomarkers, and therapeutic agents for immune-related adverse events.
Collapse
Affiliation(s)
- Kang Miao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Dongcheng District, Beijing 100005, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Dongcheng District, Beijing 100005, China
| |
Collapse
|
14
|
Liu J, Gao J. Efficacy of immunotherapy as second-line or later-line therapy and prognostic significance of KRAS or TP53 mutations in advanced non-small cell lung cancer patients. Eur J Cancer Prev 2023; 32:590-599. [PMID: 37038985 DOI: 10.1097/cej.0000000000000799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
OBJECTIVE In this retrospective study, we aimed to assess the relationship between mutations in the Kirsten rats sarcoma viral oncogene (KRAS )/ tumor protein p53 (TP53 ) genes and the efficacy of immune checkpoint inhibitors (ICIs) therapy as a second-line or later-line treatment for patients with stage IIIB/IV non-small cell lung cancer (NSCLC). METHODS We retrospectively analyzed the clinical data of 143 patients with stage IIIB/IV NSCLC who were admitted to the Cancer Hospital of Harbin Medical University between January 2019 and September 2022. Kaplan-Meier survival curve analysis was performed to analyze the survival outcomes. Univariate and multivariate Cox proportional risk models were used to analyze the factors associated with the progression-free survival (PFS) and overall survival (OS) of advanced-stage NSCLC patients who received ICIs as second-line or later-line therapy. RESULTS NSCLC patients with KRAS or TP53 mutations treated with ICIs showed significantly higher objective response rate, disease control rate, PFS, and OS compared to NSCLC patients with wild-type KRAS / TP53 (P < 0.05). Multivariate Cox regression analysis showed that a combined treatment regimen of ICIs plus chemotherapy was significantly associated with prolonged PFS [hazard ratio = 0.192; 95% confidence interval (CI), 0.094-0.392; P < 0.001] and OS (hazard ratio = 0.414; 95% CI, 0.281-0.612; P < 0.001). CONCLUSION KRAS or TP53 mutations were associated with improved PFS of advanced NSCLC patients treated with ICIs as second-line or later-line therapy. KRAS or TP53 mutations show great potential as clinical biomarkers to predict the efficacy of ICIs therapy.
Collapse
Affiliation(s)
| | - Jianing Gao
- Department of Urology Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
15
|
Proulx-Rocray F, Routy B, Nassabein R, Belkaid W, Tran-Thanh D, Malo J, Tonneau M, Ouarzadi OE, Florescu M, Tehfe M, Blais N. The prognostic impact of KRAS, TP53, STK11 and KEAP1 mutations and their influence on the NLR in NSCLC patients treated with immunotherapy. Cancer Treat Res Commun 2023; 37:100767. [PMID: 37832364 DOI: 10.1016/j.ctarc.2023.100767] [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: 09/25/2022] [Revised: 08/23/2023] [Accepted: 10/08/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND PD-L1 expression is used to predict NSCLC response to ICIs, but its performance is suboptimal. The impact of KRAS mutations in these patients is unclear. Studies evaluating co-mutations in TP53, STK11 and KEAP1 as well as the NLR showed that they may predict the benefit of ICIs. PATIENTS & METHODS This is a retrospective study of patients with NSCLC treated with ICIs at the CHUM between July 2015 and June 2020. OS and PFS were compared using Kaplan-Meier and logrank methods. Co-mutations in TP53, STK11 and KEAP1 as well as the NLR were accounted for. ORR and safety were compared using Wald method. RESULTS From 100 patients with known KRAS status, 50 were mutated (KRASMut). Mutation in TP53, STK11 and KEAP1 were present, and their status known in, respectively, 19/40 (47.5 %), 8/39 (20.5 %) and 4/38 (10.5 %) patients. STK11Mut and KEAP1Mut were associated with shorter overall survival when compared with wild type tumors (respectively median OS of 3.3 vs 20.4, p = 0.0001 and 10.1 vs 17.7, p = 0.24). When KRAS status was compounded with STK11/KEAP1, KRASMut trended to a better prognosis in STK11+KEAP1WT tumors (median OS 21.1 vs 15.8 for KRASWT, p = 0.15), but not for STK11+/-KEAP1Mut tumors. The NLR was strongly impacted by STK11 (6.0Mutvs 3.6WT, p = 0.014) and TP53 (3.2Mutvs 4.8WT, p = 0.048), but not by KEAP1 or KRAS mutations. CONCLUSION STK11Mut and KEAP1Mut are adverse predictors of ICI therapy benefit. The NLR is strongly impacted by STK11Mut but not by KEAP1Mut, suggesting differences in their resistance mechanism. In STK11-KEAP1WT tumors, KRASMut seem associated with improved survival in NSCLC patients treated with ICIs. MICROABSTRACT Response of NSCLC to immunotherapy is not easily predictable. We conducted a retrospective study in 100 patients with NSCLC and a known KRAS status. By accounting for different co-mutations, KRAS mutation was found to be associated with a better median overall survival in STK11 and KEAP1 wild-type tumors (21.1 vs 15.8, p = 0.15). NLR was impacted by STK11, but not KEAP1 mutation, suggesting a difference in their resistance mechanism.
Collapse
Affiliation(s)
- Francis Proulx-Rocray
- Medical Oncology Department, Centre Hospitalier de l'Université de Montréal (CHUM), 1051 Sanguinet Street, Montreal, QC, Canada
| | - Bertrand Routy
- Medical Oncology Department, Centre Hospitalier de l'Université de Montréal (CHUM), 1051 Sanguinet Street, Montreal, QC, Canada; Department of Medicine, Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, 900 Saint-Denis Street, Montreal, QC, Canada
| | - Rami Nassabein
- Medical Oncology Department, Centre Hospitalier de l'Université de Montréal (CHUM), 1051 Sanguinet Street, Montreal, QC, Canada
| | - Wiam Belkaid
- Department of Medicine, Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, 900 Saint-Denis Street, Montreal, QC, Canada
| | - Danh Tran-Thanh
- Pathology Department, Centre Hospitalier de l'Université de Montréal, 1051 Sanguinet Street, Montreal, QC, Canada
| | - Julie Malo
- Department of Medicine, Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, 900 Saint-Denis Street, Montreal, QC, Canada
| | - Marion Tonneau
- Department of Medicine, Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, 900 Saint-Denis Street, Montreal, QC, Canada
| | - Omar El Ouarzadi
- Department of Medicine, Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, 900 Saint-Denis Street, Montreal, QC, Canada
| | - Marie Florescu
- Medical Oncology Department, Centre Hospitalier de l'Université de Montréal (CHUM), 1051 Sanguinet Street, Montreal, QC, Canada; Department of Medicine, Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, 900 Saint-Denis Street, Montreal, QC, Canada
| | - Mustapha Tehfe
- Medical Oncology Department, Centre Hospitalier de l'Université de Montréal (CHUM), 1051 Sanguinet Street, Montreal, QC, Canada; Department of Medicine, Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, 900 Saint-Denis Street, Montreal, QC, Canada
| | - Normand Blais
- Medical Oncology Department, Centre Hospitalier de l'Université de Montréal (CHUM), 1051 Sanguinet Street, Montreal, QC, Canada; Department of Medicine, Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, 900 Saint-Denis Street, Montreal, QC, Canada.
| |
Collapse
|
16
|
Meira DD, de Castro e Caetano MC, Casotti MC, Zetum ASS, Gonçalves AFM, Moreira AR, de Oliveira AH, Pesente F, Santana GM, de Almeida Duque D, Pereira GSC, de Castro GDSC, Pavan IP, Chagas JPS, Bourguignon JHB, de Oliveira JR, Barbosa KRM, Altoé LSC, Louro LS, Merigueti LP, Alves LNR, Machado MRR, Roque MLRO, Prates PS, de Paula Segáua SH, dos Santos Uchiya T, Louro TES, Daleprane VE, Guaitolini YM, Vicente CR, dos Reis Trabach RS, de Araújo BC, dos Santos EDVW, de Paula F, Lopes TJS, de Carvalho EF, Louro ID. Prognostic Factors and Markers in Non-Small Cell Lung Cancer: Recent Progress and Future Challenges. Genes (Basel) 2023; 14:1906. [PMID: 37895255 PMCID: PMC10606762 DOI: 10.3390/genes14101906] [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: 09/01/2023] [Revised: 09/29/2023] [Accepted: 10/01/2023] [Indexed: 10/29/2023] Open
Abstract
Lung cancer is a highly aggressive neoplasm and, despite the development of recent therapies, tumor progression and recurrence following the initial response remains unsolved. Several questions remain unanswered about non-small cell lung cancer (NSCLC): (1) Which patients will actually benefit from therapy? (2) What are the predictive factors of response to MAbs and TKIs? (3) What are the best combination strategies with conventional treatments or new antineoplastic drugs? To answer these questions, an integrative literature review was carried out, searching articles in PUBMED, NCBI-PMC, Google Academic, and others. Here, we will examine the molecular genetics of lung cancer, emphasizing NSCLC, and delineate the primary categories of inhibitors based on their molecular targets, alongside the main treatment alternatives depending on the type of acquired resistance. We highlighted new therapies based on epigenetic information and a single-cell approach as a potential source of new biomarkers. The current and future of NSCLC management hinges upon genotyping correct prognostic markers, as well as on the evolution of precision medicine, which guarantees a tailored drug combination with precise targeting.
Collapse
Affiliation(s)
- Débora Dummer Meira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Maria Clara de Castro e Caetano
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Matheus Correia Casotti
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Aléxia Stefani Siqueira Zetum
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - André Felipe Monteiro Gonçalves
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - André Rodrigues Moreira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Augusto Henrique de Oliveira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Fellipe Pesente
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Gabriel Mendonça Santana
- Centro de Ciências da Saúde, Curso de Medicina, Universidade Federal do Espírito Santo (UFES), Vitória 29090-040, Brazil
| | - Daniel de Almeida Duque
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Gierleson Santos Cangussu Pereira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Giulia de Souza Cupertino de Castro
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Isabele Pagani Pavan
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - João Pedro Sarcinelli Chagas
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - José Henrique Borges Bourguignon
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Juliana Ribeiro de Oliveira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Karen Ruth Michio Barbosa
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Lorena Souza Castro Altoé
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Luana Santos Louro
- Centro de Ciências da Saúde, Curso de Medicina, Universidade Federal do Espírito Santo (UFES), Vitória 29090-040, Brazil
| | - Luiza Poppe Merigueti
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Lyvia Neves Rebello Alves
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Marlon Ramos Rosado Machado
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Maria Luísa Rodrigues Oliveira Roque
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Pedro Santana Prates
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Sayuri Honorio de Paula Segáua
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Taissa dos Santos Uchiya
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Thomas Erik Santos Louro
- Escola Superior de Ciências da Santa Casa de Misericórdia de Vitória (EMESCAM), Curso de Medicina, Vitória 29027-502, Brazil
| | - Vinicius Eduardo Daleprane
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Yasmin Moreto Guaitolini
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Creuza Rachel Vicente
- Departamento de Medicina Social, Universidade Federal do Espírito Santo, Vitória 29090-040, Brazil
| | - Raquel Silva dos Reis Trabach
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Bruno Cancian de Araújo
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Eldamária de Vargas Wolfgramm dos Santos
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Flávia de Paula
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Tiago José S. Lopes
- Department of Reproductive Biology, National Center for Child Health and Development Research Institute, Tokyo 157-8535, Japan
| | - Elizeu Fagundes de Carvalho
- Instituto de Biologia Roberto Alcântara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20551-030, Brazil
| | - Iúri Drumond Louro
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| |
Collapse
|
17
|
Qiu G, Wang F, Xie X, Liu T, Zeng C, Chen Z, Zhou M, Deng H, Yang Y, Lin X, Xie Z, Sun G, Zhou C, Liu M. A retrospective real-world experience of immunotherapy in patients with extensive stage small-cell lung cancer. Cancer Med 2023; 12:14881-14891. [PMID: 37462138 PMCID: PMC10417203 DOI: 10.1002/cam4.5843] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/28/2023] [Accepted: 03/12/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND The treatment of extensive stage small-cell lung cancer (ES-SCLC) has only made modest progress in the past decade, with two immune checkpoint inhibitors (ICIs), atezolizumab and durvalumab, approved for the treatment of SCLC by January 2022. However, currently, there is limited real-world data on ES-SCLC patients received immunotherapy. METHODS We retrospectively collected and analyzed the demographic and treatment data of ES-SCLC patients at the First Affiliated Hospital of Guangzhou Medical University from January 2017 to January 2022. Survival and prognosis information was obtained through follow-up. RESULTS A total of 353 ES-SCLC patients were included, of which 165 received immunotherapy combined with chemotherapy as the first-line (FL) treatment (chemo-immune group), and 188 received chemotherapy (chemotherapy group). The objective response rate (ORR) and disease control rate (DCR) of patients receiving immunotherapy as the FL treatment were better than the chemotherapy group (76.97% vs. 48.40%, p < 0.001, and 83.03% vs. 68.09%, p < 0.001). Moreover, the progression-free survival (PFS) and overall survival (OS) of ES-SCLC patients receiving immunotherapy as the FL treatment were better than the chemotherapy group (6.7 months vs. 5.1 months, p < 0.001, and 12.5 months vs. 11.2 months, p < 0.001). Furthermore, the OS of ES-SCLC patients who received immunotherapy as second-line treatment was better than that in the chemotherapy group (15.9 months vs. 12.9 months, p = 0.036). CONCLUSION ICIs combined with chemotherapy as the FL treatment could be beneficial to the ORR, DCR, PFS, and OS of ES-SCLC patients. Furthermore, ES-SCLC patients can benefit from ICIs in the second-line treatment, even if they had not received ICIs in the FL treatment.
Collapse
Affiliation(s)
- Guihuan Qiu
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Fei Wang
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Xiaohong Xie
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Ting Liu
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Chen Zeng
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Ziyao Chen
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Maolin Zhou
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Haiyi Deng
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Yilin Yang
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Xinqing Lin
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Zhanhong Xie
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Gengyun Sun
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Chengzhi Zhou
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Ming Liu
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
18
|
Alturki NA. Review of the Immune Checkpoint Inhibitors in the Context of Cancer Treatment. J Clin Med 2023; 12:4301. [PMID: 37445336 DOI: 10.3390/jcm12134301] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Checkpoint proteins are an integral part of the immune system and are used by the tumor cells to evade immune response, which helps them grow uncontrollably. By blocking these proteins, immune checkpoint inhibitors can restore the capability of the immune system to attack cancer cells and stop their growth. These findings are backed by adequate clinical trial data and presently, several FDA-approved immune checkpoint inhibitors exist in the market for treating various types of cancers, including melanoma, hepatocellular, endometrial, lung, kidney and others. Their mode of action is inhibition by targeting the checkpoint proteins CTLA-4, PD-1, PD-L1, etc. They can be used alone as well as in amalgamation with other cancer treatments, like surgery, radiation or chemotherapy. Since these drugs target only specific immune system proteins, their side effects are reduced in comparison with the traditional chemotherapy drugs, but may still cause a few affects like fatigue, skin rashes, and fever. In rare cases, these inhibitors are known to have caused more serious side effects, such as cardiotoxicity, and inflammation in the intestines or lungs. Herein, we provide an overview of these inhibitors and their role as biomarkers, immune-related adverse outcomes and clinical studies in the treatment of various cancers, as well as present some future perspectives.
Collapse
Affiliation(s)
- Norah A Alturki
- Clinical Laboratory Science Department, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| |
Collapse
|
19
|
Mao J, Shi X, Hua L, Yang M, Shen Y, Ruan Z, Li B, Xi X. Arsenic Inhibits Proliferation and Induces Autophagy of Tumor Cells in Pleural Effusion of Patients with Non-Small Cell Lung Cancer Expressing EGFR with or without Mutations via PI3K/AKT/mTOR Pathway. Biomedicines 2023; 11:1721. [PMID: 37371816 PMCID: PMC10295848 DOI: 10.3390/biomedicines11061721] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
To clarify whether arsenic could exert inhibitory effects on tumor cells in pleural effusions of patients with non-small cell lung cancer (NSCLC), 36 NSCLC pleural effusion samples were collected from Changzheng Hospital and Ruijin Hospital, from 2019 to 2022. The genotype of epidermal growth factor receptor (EGFR) was identified. Tumor cells were isolated and treated with arsenic trioxide (ATO) or/and gefitinib. Additionally, six patients were intrapleurally administrated with ATO. Results showed that 25 samples bore EGFR wild type (WT) and 11 harbored EGFR mutations, including 6 with L858R, 3 with ΔE746-A750, and 2 with T790M. ATO diminished the number of tumor cells from patients with WT and mutant EGFR, down-regulated the expression or phosphorylation of EGFR, pmTOR, PI3K, PTEN, and p4E-BP1, and up-regulated the expression of LC3. Immunofluorescent experiments showed that ATO enhanced LC3 and P62. By contrast, gefitinib was only effective in those harboring EGFR sensitizing mutations. Notably, in patients with intrapleural ATO injection, the pleural effusion underwent a bloody to pale yellow color change, the volume of the pleural effusion was reduced, and the number of the tumor cells was significantly reduced. In conclusion, arsenic is effective against NSCLC with various EGFR genotypes in vitro and in vivo, and potentially circumvents gefitinib resistance.
Collapse
Affiliation(s)
- Jianhua Mao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (J.M.); (Z.R.)
| | - Xiaoqian Shi
- Department of Respiratory and Critical Care Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, 1279 Sanmen Road, Shanghai 200434, China;
| | - Li Hua
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Menghang Yang
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai 200433, China;
| | - Yan Shen
- Research Center for Experimental Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Zheng Ruan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (J.M.); (Z.R.)
| | - Bing Li
- Department of Respiratory and Critical Care Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, 1279 Sanmen Road, Shanghai 200434, China;
- Department of Respiratory and Critical Care Medicine, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Xiaodong Xi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (J.M.); (Z.R.)
| |
Collapse
|
20
|
Wang J, Zhang J, Wen W, Wang F, Wu M, Chen D, Yu J. Exploring low-dose radiotherapy to overcome radio-immunotherapy resistance. Biochim Biophys Acta Mol Basis Dis 2023:166789. [PMID: 37302425 DOI: 10.1016/j.bbadis.2023.166789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/13/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the current treatment landscape for cancer, yet the response rates of ICIs remain unmet. Synergistic with immunotherapy, low-dose radiotherapy (LDRT) has been demonstrated to activate anti-tumor immunity - a transition from traditional radiation therapy geared toward local radical treatment to a type of immunological adjuvant. As such, studies utilizing LDRT to enhance the efficacy of immunotherapy have been increasing preclinically and clinically. This paper reviews the recent strategies of using LDRT to overcome the resistance of ICIs, as well as providing potential opportunities in cancer treatment. Despite the potential of LDRT in immunotherapy is recognized, the mechanisms behind this form of treatment remain largely elusive. Thus, we reviewed history, mechanisms and challenges associated with this form of treatment, as well as different modes of its application, to provide relatively accurate practice standards for LDRT as a sensitizing treatment when combined with immunotherapy or radio-immunotherapy.
Collapse
Affiliation(s)
- Juan Wang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Jingxin Zhang
- Shandong University Cancer Center, Jinan, Shandong 250117, PR China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Weitao Wen
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Fei Wang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Meng Wu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Dawei Chen
- Shandong University Cancer Center, Jinan, Shandong 250117, PR China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China.
| | - Jinming Yu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China; Shandong University Cancer Center, Jinan, Shandong 250117, PR China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong 250117, PR China.
| |
Collapse
|
21
|
Chen S, Tang J, Liu F, Li W, Yan T, Shangguan D, Yang N, Liao D. Changes of tumor microenvironment in non-small cell lung cancer after TKI treatments. Front Immunol 2023; 14:1094764. [PMID: 36949948 PMCID: PMC10025329 DOI: 10.3389/fimmu.2023.1094764] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common lung cancer diagnosis, among which epidermal growth factor receptor (EGFR), Kirsten rat sarcoma (KRAS), and anaplastic lymphoma kinase (ALK) mutations are the common genetic drivers. Their relative tyrosine kinase inhibitors (TKIs) have shown a better response for oncogene-driven NSCLC than chemotherapy. However, the development of resistance is inevitable following the treatments, which need a new strategy urgently. Although immunotherapy, a hot topic for cancer therapy, has shown an excellent response for other cancers, few responses for oncogene-driven NSCLC have been presented from the existing evidence, including clinical studies. Recently, the tumor microenvironment (TME) is increasingly thought to be a key parameter for the efficacy of cancer treatment such as targeted therapy or immunotherapy, while evidence has also shown that the TME could be affected by multi-factors, such as TKIs. Here, we discuss changes in the TME in NSCLC after TKI treatments, especially for EGFR-TKIs, to offer information for a new therapy of oncogene-driven NSCLC.
Collapse
Affiliation(s)
- Shanshan Chen
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jingyi Tang
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Fen Liu
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Li
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Ting Yan
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Dangang Shangguan
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Nong Yang
- Lung Cancer and Gastrointestinal Unit, Department of Medical Oncology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Dehua Liao
- Department of Pharmacy, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
22
|
Sun N, Luo Y, Zheng B, Zhang Z, Zhang C, Zhang Z, Zhang G, Tan F, Xue Q, Gao S, He J. A novel immune checkpoints-based signature to predict prognosis and response to immunotherapy in lung adenocarcinoma. J Transl Med 2022; 20:332. [PMID: 35879761 PMCID: PMC9310422 DOI: 10.1186/s12967-022-03520-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/07/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Except for B7-CD28 family members, more novel immune checkpoints are being discovered. They are closely associated with tumor immune microenvironment and regulate the function of many immune cells. Various cancer therapeutic studies targeting these novel immune checkpoints are currently in full swing. However, studies concerning novel immune checkpoints phenotypes and clinical significance in lung adenocarcinoma (LUAD) are still limited.
Methods
We enrolled 1883 LUAD cases from nine different cohorts. The samples from The Cancer Genome Atlas (TCGA) were used as a training set, whereas seven microarray data cohorts and an independent cohort with 102 qPCR data were used for validation. The immune profiles and potential mechanism of the system were also explored.
Results
After univariate Cox proportional hazards regression and stepwise multivariable Cox analysis, a novel immune checkpoints-based system (LTA, CD160, and CD40LG) were identified from the training set, which significantly stratified patients into high- and low-risk groups with different survivals. Furthermore, this system has been well validated in different clinical subgroups and multiple validation cohorts. It also acted as an independent prognostic factor for patients with LAUD in different cohorts. Further exploration suggested that high-risk patients exhibited distinctive immune cells infiltration and suffered an immunosuppressive state. Additionally, this system is closely linked to various classical immunotherapy biomarkers.
Conclusion
we constructed a novel immune checkpoints-based system for LUAD, which predicts prognosis and immunotherapeutic implications. We believe that these findings will not only aid in clinical management but will also shed some light on screening appropriate patients for immunotherapy.
Collapse
|
23
|
Yu J, Yin Y, Yu Y, Cheng M, Zhang S, Jiang S, Dong M. Effect of concomitant antibiotics use on patient outcomes and adverse effects in patients treated with ICIs. Immunopharmacol Immunotoxicol 2022; 45:386-394. [PMID: 36382735 DOI: 10.1080/08923973.2022.2145966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Jiuhang Yu
- College of Pharmacy, Jiamusi University, Jiamusi, China
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yichuang Yin
- College of Pharmacy, Jiamusi University, Jiamusi, China
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yang Yu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mengfei Cheng
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuo Zhang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuai Jiang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mei Dong
- College of Pharmacy, Jiamusi University, Jiamusi, China
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
24
|
Peng C, Chen J, Cui W, Li S, Li J, Peng L. Comparative efficacy of various CHIs combined with western medicine for non-small cell lung cancer: A bayesian network meta-analysis of randomized controlled trials. Front Pharmacol 2022; 13:1037620. [PMID: 36438813 PMCID: PMC9686447 DOI: 10.3389/fphar.2022.1037620] [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: 09/06/2022] [Accepted: 10/20/2022] [Indexed: 11/11/2022] Open
Abstract
Background: Given the limitations of Western medicine (WM) for the treatment of non-small cell lung cancer (NSCLC) and the wide exploration of Chinese herbal injections (CHIs), systematically evaluate the efficacy of Various CHIs Combined with WM for Non-small Cell Lung Cancer. In this study, we performed a network meta-analysis to evaluate the comparative efficacy of 16 CHIs combined with WM regimens for the treatment of NSCLC. Methods: Literature databases were searched from their inception to November 2021, and all randomized control trials (RCTs) involving NSCLC patients treated with a combination of Chinese and WM were retrieved. Outcomes, including disease control rate, survival quality score, incidence of gastrointestinal adverse reactions, incidence of leukopenia, and incidence of thrombocytopenia, were analyzed using RevMan (5.3), Stata17, and R software. Surface under the cumulative ranking curve (SUCRA) probability values were calculated to rank the treatments examined, and clustering analysis was used to compare the effects of CHIs on different outcomes. Results: A total of 389 studies involving 31,263 patients and 16 CHIs were included. The 16 CHIs were: Aidi injection (ADI), Huachansu injection (HCSI), oil of Ophiopogon injection (OOMI), disodium cantharidinate and vitamin B6 injection (DCI), Shenfu injection (SFI), Shenmai injection (SMI), Shenqi Fuzheng injection (SQFZI), Chansu injection (CSI), Delisheng injection (DLSI), Fufang Kushen injection (FFKSI), Huangqi injection (HQI), Kangai injection (KAI), Kanglaite injection (KLTI), Shengmai injection (SI), Xiangguduotang injection (XGDTI), and Xiaoaiping injection (XAPI). The results of the network meta-analysis showed that, with WM treatment as a co-intervention, CSI was most likely to improve the disease control rate (SUCRA = 80.90%), HQI had the highest probability of being the best option for improving the survival quality score (SUCRA = 82.60%), DCI had the highest probability of reducing the incidence of gastrointestinal adverse reactions (SUCRA = 85.50%), HCSI + WM had the highest probability of reducing the incidence of thrombocytopenia (SUCRA = 91.30%), while SMI had the highest probability of reducing the incidence of leukopenia (SUCRA = 79.10%). Conclusion: CHIs combined with WM is proved to be more effective than WM alone, which may be beneficial to NSCLC patients. SMI + WM and DCI + WM are most likely the optimal CHI to improve disease control rates, survival quality score, and reduce adverse effects. This study has limitations; therefore, higher quality RCTs and real-world evidence are required to support our conclusions.
Collapse
Affiliation(s)
- Ciyan Peng
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jing Chen
- Department of Pharmacy, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Wei Cui
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Sini Li
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jianhe Li
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Liubao Peng
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Liubao Peng,
| |
Collapse
|
25
|
Cao L, Cao Z, Liu H, Liang N, Bing Z, Tian C, Li S. Detection of Potential Mutated Genes Associated with Common Immunotherapy Biomarkers in Non-Small-Cell Lung Cancer Patients. Curr Oncol 2022; 29:5715-5730. [PMID: 36005189 PMCID: PMC9406727 DOI: 10.3390/curroncol29080451] [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: 07/12/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
Microsatellite instability (MSI), high tumor mutation burden (TMB-H) and programmed cell death 1 ligand 1 (PD-L1) expression are hot biomarkers related to the improvement of immunotherapy response. Two cohorts of non-small-cell lung cancer (NSCLC) were collected and sequenced via targeted next-generation sequencing. Drug analysis was then performed on the shared genes using three different databases: Drugbank, DEPO and DRUGSURV. A total of 27 common genes were mutated in at least two groups of TMB-H-, MSI- and PD-L1-positive groups. AKT1, SMAD4, SCRIB and AXIN2 were severally involved in PI3K-activated, transforming growth factor beta (TGF-β)-activated, Hippo-repressed and Wnt-repressed pathways. This study provides an understanding of the mutated genes related to the immunotherapy biomarkers of NSCLC.
Collapse
Affiliation(s)
- Lei Cao
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Zhili Cao
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Hongsheng Liu
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Naixin Liang
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Zhongxing Bing
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Caijuan Tian
- Tianjin Marvel Medical Laboratory, Tianjin Marvelbio Technology Co., Ltd., Tianjin 300381, China
| | - Shanqing Li
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
- Correspondence: ; Tel./Fax: +86-010-6915-2630
| |
Collapse
|
26
|
Pan W, Yin L, Guo Y, Pan D, Huang H. Case Report: Good cardiac tolerance to Toripalimab in a CVD patient with oral melanoma. Front Pharmacol 2022; 13:890546. [PMID: 35979233 PMCID: PMC9376878 DOI: 10.3389/fphar.2022.890546] [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: 03/06/2022] [Accepted: 06/28/2022] [Indexed: 11/20/2022] Open
Abstract
Primary oral melanoma is extremely rare, and the prognosis is very poor. With the development of immunotherapy, melanoma’s treatment landscape changed dramatically. Toripalimab, a recombinant programmed death receptor 1 (PD-1) monoclonal antibody, has been approved as second-line therapy for metastatic melanoma. However, the cardiac toxicity of Toripalimab is seldom reported. This article describes the application of Toripalimab on a patient who suffered from primary oral melanoma accompanied with arrhythmic mitral valve prolapse (AMVP). Case Summary: A 55-year-old Chinese female was diagnosed with BRAF wild-type oral malignant melanoma by excisional biopsy and genetic test. The melanoma quickly progressed after complete tumor resection. Combined therapy after surgical resection was applied to control the progression of melanoma. Due to this patient’s basic cardiovascular situation, sacubitril–valsartan, spironolactone, and bisoprolol were used to maintain cardiac function. After five antitumor treatment courses, we re-evaluated the patient systemically from the symptom, physical examination, and auxiliary examination. The result showed that the patient who received Toripalimab combined with chemotherapy and radiotherapy did not present severe side effects on the cardiovascular system. The cardiac function remained well. Conclusions: This case provided evidence of Toripalimab combined with chemotherapy on melanoma patients with complex cardiovascular diseases. Toripalimab demonstrated a manageable safety profile and durable clinical response. In addition, the standard CHF treatment plays a vital role in the protection of cardiac function. In a cancer patient with complex cardiovascular diseases, standard prophylactic CHF treatment should be applied at an early stage.
Collapse
Affiliation(s)
- Wei Pan
- Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Li Yin
- Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yadi Guo
- Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Dachao Pan
- Oncology Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Hui Huang
- Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- *Correspondence: Hui Huang,
| |
Collapse
|
27
|
De Mello RAB, Voscaboinik R, Luciano JVP, Cremonese RV, Amaral GA, Castelo-Branco P, Antoniou G. Immunotherapy in Patients with Advanced Non-Small Cell Lung Cancer Lacking Driver Mutations and Future Perspectives. Cancers (Basel) 2021; 14:122. [PMID: 35008287 PMCID: PMC8749892 DOI: 10.3390/cancers14010122] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/11/2021] [Accepted: 12/22/2021] [Indexed: 12/20/2022] Open
Abstract
From a complete literature review, we were able to present in this paper what is most current in the treatment with immunotherapy for advanced non-small cell lung cancer (NSCLC). Especially the use of immunotherapy, particularly inhibitors of PD-1 (programmed cell death protein 1), PDL-1 (programmed cell death protein ligand 1), and CTLA-4 (cytotoxic T-lymphocyte antigen 4). Since 2015, these drugs have transformed the treatment of advanced NSCLC lacking driver mutations, evolving from second-line therapy to first-line, with excellent results. The arrival of new checkpoint inhibitors such as cemiplimab and the use of checkpoint inhibitors earlier in the therapy of advanced and metastatic cancers has been making the future prospects for treating NSCLC lacking driver mutations more favorable and optimistic. In addition, for those patients who have low PDL-1 positivity tumors, the combination of cytotoxic chemotherapy, VEGF inhibitor, and immunotherapy have shown an important improvement in global survival and progression free survival regardless the PDL-1 status. We also explored the effectiveness of adding radiotherapy to immunotherapy and the most current results about this combination. One concern that cannot be overlooked is the safety profile of immune checkpoint inhibitors (ICI) and the most common toxicities are described throughout this paper as well as tumor resistance to ICI.
Collapse
Affiliation(s)
- Ramon Andrade Bezerra De Mello
- Division of Medical Oncology, Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de São Paulo (UNIFESP), Sao Paulo 04023-900, Brazil; (R.V.); (G.A.A.)
- Faculdade de Medicina, Universidade Nove de Julho, Sao Paulo 04023-900, Brazil; (J.V.P.L.); (R.V.C.)
- Centro Médico Especialidades, Division of Medical Oncology, Hospital 9 de Julho, Sao Paulo 01409-002, Brazil
| | - Rafael Voscaboinik
- Division of Medical Oncology, Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de São Paulo (UNIFESP), Sao Paulo 04023-900, Brazil; (R.V.); (G.A.A.)
| | - João Vittor Pires Luciano
- Faculdade de Medicina, Universidade Nove de Julho, Sao Paulo 04023-900, Brazil; (J.V.P.L.); (R.V.C.)
| | - Rafaela Vilela Cremonese
- Faculdade de Medicina, Universidade Nove de Julho, Sao Paulo 04023-900, Brazil; (J.V.P.L.); (R.V.C.)
| | - Giovanna Araujo Amaral
- Division of Medical Oncology, Escola Paulista de Medicina, Hospital São Paulo, Universidade Federal de São Paulo (UNIFESP), Sao Paulo 04023-900, Brazil; (R.V.); (G.A.A.)
| | - Pedro Castelo-Branco
- Faculty of Medicine and Biomedical Sciences (FMCB), Campus de Gambelas, University of Algarve, 8003-139 Faro, Portugal; (P.C.-B.); (G.A.)
| | - Georgios Antoniou
- Faculty of Medicine and Biomedical Sciences (FMCB), Campus de Gambelas, University of Algarve, 8003-139 Faro, Portugal; (P.C.-B.); (G.A.)
| |
Collapse
|
28
|
Liu C, Wang S, Zheng S, Xu F, Cao Z, Feng X, Wang Y, Xue Q, Sun N, He J. Avoiding Absolute Quantification Trap: A Novel Predictive Signature of Clinical Benefit to Anti-PD-1 Immunotherapy in Non-Small Cell Lung Cancer. Front Immunol 2021; 12:782106. [PMID: 34868057 PMCID: PMC8640493 DOI: 10.3389/fimmu.2021.782106] [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: 09/23/2021] [Accepted: 11/03/2021] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy has been focused on by many oncologists and researchers. While, due to technical biases of absolute quantification, few traditional biomarkers for anti-PD-1 immunotherapy have been applied in regular clinical practice of non-small cell lung cancer (NSCLC). Therefore, there is an urgent and unmet need for a feasible tool—immune to data source bias—for identifying patients who might benefit from ICIs in clinical practice. Using the strategy based on the relative ranking of gene expression levels, we herein proposed the novel BRGP index (BRGPI): four BRGPs significantly related with progression-free survival of NSCLC patients treated with anti-PD-1 immunotherapy in the multicohort analysis. Moreover, stratification and multivariate Cox regression analyses demonstrated that BRGPI was an independent prognostic factor. Notably, compared to PD-L1, BRGPI exerted the best predictive ability. Further analysis showed that the patients in the BRGPI-low and PD-L1-high subgroup derived more clinical benefits from anti-PD-1 immunotherapy. In conclusion, the prospect of applying the BRGPI to real clinical practice is promising owing to its powerful and reliable predictive value.
Collapse
Affiliation(s)
- Chengming Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sihui Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sufei Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Cao
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoli Feng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
29
|
Olszewska P, Pazdrak B, Kruzel ML. A Novel Human Recombinant Lactoferrin Inhibits Lung Adenocarcinoma Cell Growth and Migration with No Cytotoxic Effect on Normal Human Epithelial Cells. Arch Immunol Ther Exp (Warsz) 2021; 69:33. [PMID: 34748082 PMCID: PMC8575758 DOI: 10.1007/s00005-021-00637-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
Lung cancer remains the leading cause of cancer death worldwide. Despite the recent advances in cancer treatment, only a subset of patients responds to targeted and immune therapies, and many patients developing resistance after an initial response. Lactoferrin (Lf) is a natural glycoprotein with immunomodulatory and anticancer activities. We produced a novel recombinant human Lf (rhLf) that exhibits glycosylation profile compatible with the natural hLf for potential parenteral therapeutic applications. The aim of this study was to evaluate the anticancer effects of this novel rhLf in human lung adenocarcinoma cells and its mechanisms of action. The results showed a concentration-dependent inhibition of A549 cancer cell growth in response to rhLf. Treatment with 1 mg/ml of rhLf for 24 h and 72 h resulted in a significant inhibition of cancer cell growth by 32% and 25%, respectively. Moreover, rhLf increased fourfold the percentage of early and late apoptotic cells compared to the control. This effect was accompanied by increased levels of caspase-3 activity and cell cycle arrest at the S phase in rhLf-treated cancer cells. Furthermore, rhLf significantly attenuated A549 cell migration. Importantly, treatment of normal human bronchial epithelial (NHBE) cells with rhLf showed the cell viability and morphology comparable to the control. In contrast, chemotherapeutic etoposide induced cytotoxicity in NHBE cells and reduced the cell viability by 40%. These results demonstrate the selective anticancer effects of rhLf against lung adenocarcinoma cells without cytotoxicity on normal human cells. This study highlights a potential for clinical utility of this novel rhLf in patients with lung cancer.
Collapse
Affiliation(s)
- Paulina Olszewska
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Lodz, Poland.
| | - Barbara Pazdrak
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
30
|
Gupta R, Smalley M, Anusim N, Jindal V, Singh Rahi M, Gupta S, Gupta S, Jaiyesimi I. Paradigm shift in the management of metastatic nonsmall cell lung cancer. Int J Clin Pract 2021; 75:e14533. [PMID: 34129744 DOI: 10.1111/ijcp.14533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 06/06/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Lung cancer is one of the leading causes of cancer mortality in the United States. The use of precision medicine in the past 10 years has significantly changed the therapeutic landscape of lung cancer. Management of advanced nonsmall cell lung cancer (NSCLC) has transitioned from a chemotherapeutic approach to targeted treatments and immunotherapeutic agents. Several tyrosine kinase inhibitors (TKIs) have been approved for patients with targeted mutations and patients who do not have driver mutations; immunotherapy has been recently approved as frontline therapy, which has resulted in marked improvement in overall survival and added a new tool in our armamentarium. AIMS The purpose of this review is to highlight recent advancements in diagnostic approach and management strategies in patients with metastatic NSCLC. MATERIALS AND METHODS A literature search was conducted on Medline (via PubMed) and National Comprehensive Cancer Network Guidelines using the keywords "precision diagnosis," "advanced non-small cell lung cancer," "target therapies," and "immunotherapy." CONCLUSION The use of next-generation sequencing has significantly changed our understanding of molecular oncogenic mechanisms of lung cancer. These advancements have created a paradigm shift in the treatment strategies of metastatic lung cancer from primarily chemotherapeutic approach to increasing use of targeted therapies and immune checkpoint inhibitors (ICI) leading to better survival rates and lesser toxicity.
Collapse
Affiliation(s)
- Ruby Gupta
- Department of Hematology and Medical Oncology, William Beaumont Hospital, Royal Oak, MI, USA
| | - Melanie Smalley
- Department of Internal Medicine, William Beaumont Hospital, Royal Oak, MI, USA
| | - Nwabundo Anusim
- Department of Hematology and Medical Oncology, William Beaumont Hospital, Royal Oak, MI, USA
| | - Vishal Jindal
- Department of Hematology and Medical Oncology, William Beaumont Hospital, Royal Oak, MI, USA
| | - Mandeep Singh Rahi
- Division of Pulmonary Diseases and Critical Care, Yale-New Haven Health Bridgeport Hospital, Bridgeport, CT, USA
| | - Sorab Gupta
- Department of Hematology and Medical Oncology, BronxCare Hospital, Bronx, NY, USA
| | - Sachin Gupta
- Department of Internal Medicine, Tower Health Reading Hospital, Reading, PA, USA
| | - Ishmael Jaiyesimi
- Department of Hematology and Medical Oncology, William Beaumont Hospital, Royal Oak, MI, USA
| |
Collapse
|
31
|
Zhang F, Qiu B, Ji Y, Guo W, Li N, Xue Q, Gao S, He J. Comparison of surgical difficulty in patients with resectable non-small cell lung cancer under different neoadjuvant treatment modes: a retrospective cohort study. J Thorac Dis 2021; 13:5604-5616. [PMID: 34795911 PMCID: PMC8575808 DOI: 10.21037/jtd-21-1007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/10/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Previous studies have reported on the efficacy and safety of neoadjuvant use of a programmed cell death 1 (PD-1) antibody, sintilimab, in patients with non-small cell lung cancer (NSCLC). This study aimed to further evaluate the difficulty of this surgery and the postoperative complication rates in patients with NSCLC receiving neoadjuvant sintilimab. METHODS Patients who received neoadjuvant sintilimab (200 mg) in the Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital from March 2018 to March 2019 were enrolled in the neoadjuvant immunotherapy group (NI group). Another two cohorts who did not receive sintilimab were retrospectively selected by propensity score matching (PSM) at a ratio of 1:1 in the upfront surgery (M-US) and neoadjuvant chemotherapy (M-NC) groups. The postoperative complication rate, postoperative days (PODs), and other detailed objective indicators were compared by t-test or χ2 test. RESULTS Thirty-seven patients were enrolled in each group. Postoperative complications were greater in the NI group (37.8%) than in the M-US (10.8%; P=0.013) or in the M-NC group (16.2%; P=0.036). The number of PODs (7) was greater in the NI group than in the M-US group (P=0.005). The total number of dissected lymph nodes was lower in the NI group than in the M-US group (P<0.001) or in the M-NC group (P<0.001). Lymph node dissection (LND) in the NI group was more difficult than in the M-US group (P=0.015), but intrathoracic adhesion, tumor invasion, and whole procedure difficulty were similar. CONCLUSIONS The administration of neoadjuvant sintilimab increased complications but did not increase the difficulty of surgery. Fewer lymph nodes were dissected in the NI group.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Qiu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Ji
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
32
|
Zhou L, Wei X. Ocular Immune-Related Adverse Events Associated With Immune Checkpoint Inhibitors in Lung Cancer. Front Immunol 2021; 12:701951. [PMID: 34504488 PMCID: PMC8421677 DOI: 10.3389/fimmu.2021.701951] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/05/2021] [Indexed: 02/05/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are novel immunotherapy-based drugs that have become increasingly popular in the treatment of lung cancer. Researchers have recognized ocular immune-related adverse events (irAEs) secondary to ICIs because of their vision-threatening characteristics. However, they are incompletely characterized and no studies have reported the ICI-related ocular irAEs in lung cancer. Therefore, we aimed to comprehensively illustrate the clinical characteristics, contributory factors, diagnosis, and management of ICI-related ocular irAEs in lung cancer, based on previously reported 79 patients. Ophthalmoplegia (40.51%), uveitis (20.25%), and dry eye (17.72%) were the most common ICI-related ocular irAEs in lung cancer. Ptosis was the most common (36.71%) and the highest mortality (23.33%) of ophthalmoplegia. Patients in Asia and patients who underwent combination therapy with programmed cell death-1 and cytotoxic T-lymphocyte-associated antigen 4 inhibitors demonstrated significantly higher frequency of ophthalmoplegia than other ocular irAEs. Most ICI-related ophthalmoplegia and uveitis in lung cancer were observed in the first 10 weeks following the initiation of ICIs. Furthermore, the onset time of dry eye and other ocular irAEs was much longer. In addition, 92.31% of the patients with ocular irAEs other than ophthalmoplegia could be remised. In conclusion, ocular irAEs secondary to ICIs in lung cancer are non-negligible, particularly ophthalmoplegia. Ethnicity and the type of ICIs play important roles in the distribution of ocular irAEs. ICI-related ophthalmoplegia in lung cancer presented with early onset and worse prognosis features, thus necessitating further attention.
Collapse
Affiliation(s)
- Lin Zhou
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Wei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Reischer A, Kruger S, von Bergwelt-Baildon M. [A decade of checkpoint inhibitors: current standard of care and future trends]. Dtsch Med Wochenschr 2021; 146:1108-1118. [PMID: 34448187 DOI: 10.1055/a-1303-8820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Therapy with checkpoint inhibitors still revolutionizes the therapeutical landscape in oncology. Since the first approval of a checkpoint inhibitor for the therapy of malignant melanoma 2011, many other approvals in the field of hematology and oncology followed. Besides monotherapy, a rapidly increasing number of trials is investigating checkpoint inhibitors in different combination therapies for advanced disease. Cumulating evidence suggests checkpoint blockade also as promising option for (neo)-adjuvant treatment. Here we review the different treatment strategies of mono- and combination-therapies. Additionally, important biomarkers for the treatment with checkpoint inhibitors are discussed.
Collapse
|
34
|
Liu B, Zhang R, Zhu Y, Hao R. Exosome-derived microRNA-433 inhibits tumorigenesis through incremental infiltration of CD4 and CD8 cells in non-small cell lung cancer. Oncol Lett 2021; 22:607. [PMID: 34188709 PMCID: PMC8227510 DOI: 10.3892/ol.2021.12868] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 04/22/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor-derived exosomal microRNAs (miRNAs/miRs) serve a vital biological role in tumorigenesis and development, but the effects and underlying mechanisms remain unclear. To explore the impact of exosomal miR-433 in non-small cell lung cancer (NSCLC) and understand its mechanism of action in NSCLC progression, the present study isolated the exosomes from the plasma of patients with NSCLC after chemotherapy and found that miR-433 expression was lower in plasma of patients with resistant NSCLC compared with in plasma of patients with sensitive NSCLC and in normal serum. Additionally, miR-433 expression was markedly negatively associated with a large tumor size, distant metastasis, advanced TNM stage and a poor prognosis in patients with NSCLC. miR-433 inhibited tumor growth by blocking the cell cycle in vitro and in vivo, as well as by promoting apoptosis and T-cell infiltration in the tumor microenvironment. Additionally, miR-433 inhibited chemoresistance to cisplatin by regulating DNA damage. Moreover, miR-433 inactivated the WNT/β-catenin signaling pathway by targeting transmembrane p24 trafficking protein 5 in NSCLC. Overall, the current findings may provide a potential prognostic biomarker and therapeutic target for patients with NSCLC.
Collapse
Affiliation(s)
- Boyang Liu
- Department of Radiation, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Ruiping Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Yungang Zhu
- Department of Radiation Oncology, Tianjin Teda Hospital, Tianjin 300457, P.R. China
| | - Ruisheng Hao
- Department of Radiation, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| |
Collapse
|
35
|
Nowosielska EM, Cheda A, Pociegiel M, Cheda L, Szymański P, Wiedlocha A. Effects of a Unique Combination of the Whole-Body Low Dose Radiotherapy with Inactivation of Two Immune Checkpoints and/or a Heat Shock Protein on the Transplantable Lung Cancer in Mice. Int J Mol Sci 2021; 22:6309. [PMID: 34208396 PMCID: PMC8231142 DOI: 10.3390/ijms22126309] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 11/25/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) continues to be the leading cause of cancer death worldwide. Recently, targeting molecules whose functions are associated with tumorigenesis has become a game changing adjunct to standard anti-cancer therapy. As evidenced by the results of preclinical and clinical investigations, whole-body irradiations (WBI) with X-rays at less than 0.1-0.2 Gy per fraction can induce remissions of various neoplasms without inciting adverse side effects of conventional chemo- and radiotherapy. In the present study, a murine model of human NSCLC was employed to evaluate for the first time the anti-neoplastic efficacy of WBI combined with inactivation of CTLA-4, PD-1, and/or HSP90. The results indicate that WBI alone and in conjunction with the inhibition of the function of the cytotoxic T-lymphocyte antigen-4 (CTLA-4) and the programmed death-1 (PD-1) receptor immune checkpoints (ICs) and/or heat shock protein 90 (HSP90) markedly reduced tumorigenesis in mice implanted by three different routes with the syngeneic Lewis lung cancer cells and suppressed clonogenic potential of Lewis lung carcinoma (LLC1) cells in vitro. These results were associated with the relevant changes in the profile of pro- and anti-neoplastic immune cells recruited to the growing tumors and the circulating anti- and pro-inflammatory cytokines. In contrast, inhibition of the tested molecular targets used either separately or in combination with each other did not exert notable anti-neoplastic effects. Moreover, no significant synergistic effects were detected when the inhibitors were applied concurrently with WBI. The obtained results supplemented with further mechanistic explanations provided by future investigations will help design the effective strategies of treatment of lung and other cancers based on inactivation of the immune checkpoint and/or heat shock molecules combined with low-dose radiotherapy.
Collapse
Affiliation(s)
- Ewa M. Nowosielska
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163 Warsaw, Poland; (A.C.); (P.S.); (A.W.)
| | - Aneta Cheda
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163 Warsaw, Poland; (A.C.); (P.S.); (A.W.)
| | - Mateusz Pociegiel
- National Centre for Nuclear Research Radioisotope Centre POLATOM, 7A Soltana St., 05-400 Otwock, Poland;
| | - Lukasz Cheda
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, 101 Żwirki i Wigury St., 02-089 Warsaw, Poland;
| | - Paweł Szymański
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163 Warsaw, Poland; (A.C.); (P.S.); (A.W.)
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, 1 Muszyńskiego St., 90-151 Lodz, Poland
| | - Antoni Wiedlocha
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163 Warsaw, Poland; (A.C.); (P.S.); (A.W.)
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway
- Centre for Cancer Reprograming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway
| |
Collapse
|
36
|
Jain E, Sharma S, Aggarwal A, Bhardwaj N, Dewan A, Kumar A, Jain D, Bhattacharya M, Saurav GK, Kini L, Mohanty SK. PD-L1 expression and its clinicopathologic and genomic correlation in the non-small cell lung carcinoma patients: An Indian perspective. Pathol Res Pract 2021; 228:153497. [PMID: 34053784 DOI: 10.1016/j.prp.2021.153497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Immunotherapy with checkpoint inhibitor programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) antibodies targeting the cellular immune checkpoints is the present area of interest showing promising results in patients with advanced non-small cell lung cancer (NSCLC). As there is paucity of PD-L1 expression data from the Indian perspective, we studied the correlation of clinicopathologic profile and oncogenic driver mutations in these patients. MATERIALS AND METHODS Samples from 252 advanced NSCLCs patients were studied for PD-L1 expression through immunohistochemistry using rabbit anti-human PD-L1 monoclonal antibody (clone SP263) on Ventana BenchMark ULTRA autostainer. Simultaneously, genetic mutations were studied by next generation sequencing (for EGFR, ALK, ROS, MET, and BRAF). PD-L1 expression was analyzed for association with clinicopathologic features and various mutations. RESULTS PD-L1 positivity was seen in 134 patients (53.2 %). It was twice more prevalent in males than females. No significant correlation was observed between PD-L1 expression with age, gender, site of testing (primary vs. metastatic tumors), smoking status, tumor laterality, stage, or histologic type; however, there was significant difference among solid and acinar types of adenocarcinoma combined together vs. other adenocarcinoma subtypes (p = 0.013), and well and moderately differentiated vs. poorly differentiated tumors (p = 0.022). When types/extent of PD-L1 positivity (≥25 %) were compared with demographics, clinical, and pathologic variables, significant differences were observed across the tumor grades (high-grade vs. low-grade) (p = 0.009) and stages (p = 0.039). The PD-L1 expression failed to demonstrate any statistical significance with oncogenic drivers. High PD-L1 expression (TPS ≥ 50) was observed in 27.6 % patients, and it was more prevalent in female patients (32.4 %), aged ≥60 years (33.8 %), smokers (27.3 %), poorly differentiated (36.8 %) and stage IV tumors (28.2 %). Exon 19 deletion was more prevalent in PD-L1 negative tumors whereas exon 21 substitution (L858R) was seen more in PD-L1 positive tumors. CONCLUSIONS This is the largest Indian study demonstrating PD-L1 expression in NSCLC patients comparing with clinicopathologic and genomic parameters. PD-L1 expression was significantly associated with high-grade, solid, and acinar types of adenocarcinoma and advanced tumors. High PD-L1 expression was more prevalent in female patients, aged ≥60 years, smokers, and poorly differentiated and stage IV tumors (28.2 %). Exon 19 deletion was more in PD-L1 negative tumors whereas exon 21 substitution (L858R) was more in PD-L1 positive tumors. PD-L1 is a potential predictive marker stratifying patients who benefit from PD-1 pathway-targeted therapy.
Collapse
Affiliation(s)
- Ekta Jain
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Shivani Sharma
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Aditi Aggarwal
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Nitin Bhardwaj
- Indian Council of Medical Research and National Institute of Malaria Research, New Delhi, 110029, India.
| | - Aditi Dewan
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Abhishek Kumar
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Deepika Jain
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Munmun Bhattacharya
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Gauraw Kumar Saurav
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Lata Kini
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Sambit Kumar Mohanty
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| |
Collapse
|
37
|
Hubertus V, Gempt J, Mariño M, Sommer B, Eicker SO, Stangenberg M, Dreimann M, Janssen I, Wipplinger C, Wagner A, Lange N, Jörger AK, Czabanka M, Rohde V, Schaller K, Thomé C, Vajkoczy P, Onken JS, Meyer B. Surgical management of spinal metastases involving the cervicothoracic junction: results of a multicenter, European observational study. Neurosurg Focus 2021; 50:E7. [PMID: 33932937 DOI: 10.3171/2021.2.focus201067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/24/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Surgical management of spinal metastases at the cervicothoracic junction (CTJ) is highly complex and relies on case-based decision-making. The aim of this multicentric study was to describe surgical procedures for metastases at the CTJ and provide guidance for clinical and surgical management. METHODS Patients eligible for this study were those with metastases at the CTJ (C7-T2) who had been consecutively treated in 2005-2019 at 7 academic institutions across Europe. The Spine Instability Neoplastic Score, neurological function, clinical status, medical history, and surgical data for each patient were retrospectively assessed. Patients were divided into four surgical groups: 1) posterior decompression only, 2) posterior decompression and fusion, 3) anterior corpectomy and fusion, and 4) anterior corpectomy and 360° fusion. Endpoints were complications, surgical revision rate, and survival. RESULTS Among the 238 patients eligible for inclusion this study, 37 were included in group 1 (15%), 127 in group 2 (53%), 18 in group 3 (8%), and 56 in group 4 (24%). Mechanical pain was the predominant symptom (79%, 189 patients). Surgical complications occurred in 16% (group 1), 20% (group 2), 11% (group 3), and 18% (group 4). Of these, hardware failure (HwF) occurred in 18% and led to surgical revision in 7 of 8 cases. The overall complication rate was 34%. In-hospital mortality was 5%. CONCLUSIONS Posterior fusion and decompression was the most frequently used technique. Care should be taken to choose instrumentation techniques that offer the highest possible biomechanical load-bearing capacity to avoid HwF. Since the overall complication rate is high, the prevention of in-hospital complications seems crucial to reduce in-hospital mortality.
Collapse
Affiliation(s)
- Vanessa Hubertus
- 1Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin
| | - Jens Gempt
- 2Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich
| | - Michelle Mariño
- 1Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin
| | - Björn Sommer
- 3Department of Neurosurgery, Universitätsmedizin Göttingen
| | - Sven O Eicker
- 4Department of Neurosurgery and Interdisciplinary University Spine Center, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| | - Martin Stangenberg
- 5Department of Trauma and Orthopedic Surgery and Interdisciplinary University Spine Center, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Dreimann
- 5Department of Trauma and Orthopedic Surgery and Interdisciplinary University Spine Center, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Insa Janssen
- 6Department of Neurosurgery, Hôpitaux Universitaires de Genève, Switzerland; and
| | - Christoph Wipplinger
- 7Department of Neurosurgery, Medizinische Universität Innsbruck, Innsbruck, Austria
| | - Arthur Wagner
- 2Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich
| | - Nicole Lange
- 2Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich
| | - Ann-Kathrin Jörger
- 2Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich
| | - Marcus Czabanka
- 1Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin
| | - Veit Rohde
- 3Department of Neurosurgery, Universitätsmedizin Göttingen
| | - Karl Schaller
- 6Department of Neurosurgery, Hôpitaux Universitaires de Genève, Switzerland; and
| | - Claudius Thomé
- 7Department of Neurosurgery, Medizinische Universität Innsbruck, Innsbruck, Austria
| | - Peter Vajkoczy
- 1Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin
| | - Julia S Onken
- 1Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin
| | - Bernhard Meyer
- 2Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich
| |
Collapse
|
38
|
Alves M, Borges DDP, Kimberly A, Martins Neto F, Oliveira AC, de Sousa JC, Nogueira CD, Carneiro BA, Tavora F. Glycogen Synthase Kinase-3 Beta Expression Correlates With Worse Overall Survival in Non-Small Cell Lung Cancer-A Clinicopathological Series. Front Oncol 2021; 11:621050. [PMID: 33767989 PMCID: PMC7985549 DOI: 10.3389/fonc.2021.621050] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/27/2021] [Indexed: 12/16/2022] Open
Abstract
Background Glycogen Synthase Kinase-3 beta (GSK-3β) regulates diverse cell functions including metabolic activity, signaling and structural proteins. GSK-3β phosphorylates target pro-oncogenes and regulates programmed cell death-ligand 1 (PD-L1). This study investigated the correlation between GSK-3β expression and clinically relevant molecular features of lung adenocarcinoma (PDL1 score, PTEN expression and driver mutations). Methods We evaluated 95 lung cancer specimens from biopsies and surgical resections. Immunohistochemistry was performed to analyze the expression of GSK-3β, PTEN, and PDL1. Epidemiological data, molecular characteristics and staging were evaluated from medical records. The histologic classification was performed by an experienced pulmonary pathologist. Results Most patients were female (52.6%) and the majority had a positive smoking history. The median age was 68.3 years, with individuals over 60 years accounting for 82.1%. The predominant histological subtype was adenocarcinoma (69.5%), followed by squamous cell carcinoma (20.0%). GSK-3β expression in tumors was cytoplasmic with a dotted pattern and perinuclear concentration, with associated membranous staining. Seven (7.3%) tumors had associated nuclear expression localization. Seventy-seven patients (81.1%) had advanced clinical-stage tumors. GSK-3β was positive in 75 tumors (78%) and GSK3-positive tumors tended to be diagnosed at advanced stages. Among stage III/IV tumors, 84% showed GSK3 positivity (p= 0.007). We identified a statistically significant association between GSK-3β and PTEN in the qualitative analysis (p 0.021); and when comparing PTEN to GSK-3β intensity 2+ (p 0.001) or 3+ expression (> 50%) – p 0.013. GSK-3β positive tumors with a high histological score had a worse overall survival. Conclusion We identified the histological patterns of GSK-3β expression and evaluated its potential as marker for overall survival, establishing a simple histological score to measure the evaluated status in resected tissues. The use of GSK-3β expression as an immune response biomarker remains a challenge. Future studies will seek to explain the role of its interaction with PTEN.
Collapse
Affiliation(s)
- Marclesson Alves
- Department of Pathology, Federal University of Ceará, Fortaleza, Brazil
| | | | - Aline Kimberly
- Department of Pathology, Federal University of Ceará, Fortaleza, Brazil.,Argos Pathology Laboratory, Department of Investigative Pathology, Fortaleza, Brazil
| | - Francisco Martins Neto
- Departments of Patholoy, Oncology and Thoracic Surgery, Messejana Heart and Lung Hospital, Fortaleza, Brazil
| | - Ana Claudia Oliveira
- Departments of Patholoy, Oncology and Thoracic Surgery, Messejana Heart and Lung Hospital, Fortaleza, Brazil
| | - Juliana Cordeiro de Sousa
- Department of Pathology, Federal University of Ceará, Fortaleza, Brazil.,Argos Pathology Laboratory, Department of Investigative Pathology, Fortaleza, Brazil
| | - Cleto D Nogueira
- Department of Pathology, Federal University of Ceará, Fortaleza, Brazil.,Argos Pathology Laboratory, Department of Investigative Pathology, Fortaleza, Brazil
| | - Benedito A Carneiro
- Division of Hematology/Oncology, Lifespan Cancer Institute, The Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Fabio Tavora
- Department of Pathology, Federal University of Ceará, Fortaleza, Brazil.,Argos Pathology Laboratory, Department of Investigative Pathology, Fortaleza, Brazil.,Departments of Patholoy, Oncology and Thoracic Surgery, Messejana Heart and Lung Hospital, Fortaleza, Brazil
| |
Collapse
|
39
|
Is There a Place for Immune Checkpoint Inhibitors in Vulvar Neoplasms? A State of the Art Review. Int J Mol Sci 2020; 22:ijms22010190. [PMID: 33375467 PMCID: PMC7796178 DOI: 10.3390/ijms22010190] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/15/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022] Open
Abstract
Vulvar cancer (VC) is a rare neoplasm, usually arising in postmenopausal women, although human papilloma virus (HPV)-associated VC usually develop in younger women. Incidences of VCs are rising in many countries. Surgery is the cornerstone of early-stage VC management, whereas therapies for advanced VC are multimodal and not standardized, combining chemotherapy and radiotherapy to avoid exenterative surgery. Randomized controlled trials (RCTs) are scarce due to the rarity of the disease and prognosis has not improved. Hence, new therapies are needed to improve the outcomes of these patients. In recent years, improved knowledge regarding the crosstalk between neoplastic and tumor cells has allowed researchers to develop a novel therapeutic approach exploiting these molecular interactions. Both the innate and adaptive immune systems play a key role in anti-tumor immunesurveillance. Immune checkpoint inhibitors (ICIs) have demonstrated efficacy in multiple tumor types, improving survival rates and disease outcomes. In some gynecologic cancers (e.g., cervical cancer), many studies are showing promising results and a growing interest is emerging about the potential use of ICIs in VC. The aim of this manuscript is to summarize the latest developments in the field of VC immunoncology, to present the role of state-of-the-art ICIs in VC management and to discuss new potential immunotherapeutic approaches.
Collapse
|
40
|
Yao Q, Gu L, Su R, Chen B, Cao H. Efficacy and safety of combination PD-1/PD-L1 checkpoint inhibitors for malignant solid tumours: A systematic review. J Cell Mol Med 2020; 24:13494-13506. [PMID: 33078904 PMCID: PMC7701512 DOI: 10.1111/jcmm.15991] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/18/2020] [Accepted: 09/29/2020] [Indexed: 12/20/2022] Open
Abstract
Treatment of multiple malignant solid tumours with programmed death (PD)-1/PD ligand (PD-L) 1 inhibitors has been reported. However, the efficacy and immune adverse effects of combination therapies are controversial. This meta-analysis was performed with PubMed, Web of Science, Medline, EMBASE and Cochrane Library from their inception until January 2020. Random-effect model was adopted because of relatively high heterogeneity. We also calculated hazard ratio (HR) of progression-free survival (PFS), overall survival (OS) and risk ratio (RR) of adverse events (AEs), the incidence of grade 3-5 AEs by tumour subgroup, therapeutic schedules and therapy lines. Nineteen articles were selected using the search strategy for meta-analysis. Combined PD-1/PD-L1 inhibitors prolonged OS and PFS (HR 0.72, P < 0.001) and (HR 0.66, P < 0.001). In addition, incidence of all-grade and grade 3-5 AEs was not significant in the two subgroup analyses (HR 1.01, P = 0.31) and (HR 1.10, P = 0.07), respectively. Our meta-analysis indicated that combination therapy with PD-1/PD-L1 inhibitors had greater clinical benefits and adverse events were not increased significantly.
Collapse
Affiliation(s)
- Qigu Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou CityChina
| | - Lihu Gu
- Department of General SurgeryHwaMei HospitalUniversity of Chinese Academy of SciencesNingbo CityChina
| | - Rong Su
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou CityChina
| | - Bangsheng Chen
- Emergency Medical CenterThe Second Hospital of YinzhouNingbo CityChina
| | - Hongcui Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhou CityChina
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic‐chemical Injury DiseasesHangzhou CityChina
| |
Collapse
|
41
|
Chae YK, Kim WB, Davis AA, Park LC, Anker JF, Simon NI, Rhee K, Song J, Cho A, Chang S, Ko T, Oh M, Bhave M, Viveiros P. Mass spectrometry-based serum proteomic signature as a potential biomarker for survival in patients with non-small cell lung cancer receiving immunotherapy. Transl Lung Cancer Res 2020; 9:1015-1028. [PMID: 32953481 PMCID: PMC7481587 DOI: 10.21037/tlcr-20-148] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background VeriStrat test is a serum assay which uses a mass spectrometry (MS)-based proteomic signature derived from machine learning. It is currently used as a prognostic marker for patients with non-small cell lung cancer (NSCLC) receiving chemotherapy. However, little is known about its role for NSCLC patients receiving immune checkpoint inhibitors (ICIs). Methods This is a retrospective study that includes 47 patients with advanced stage NSCLC without an activating EGFR mutation, who underwent the VeriStrat test from 2016 to 2018. Spectra from blood samples were evaluated to assign patients into the VeriStrat ‘Good’ (VS-G) or VeriStrat ‘Poor’ (VS-P) risk group. The clinical outcomes of 32 patients who received programmed cell death 1 (PD-1) inhibitors nivolumab or pembrolizumab were analyzed by VeriStrat status. Results The VS-G group demonstrated significantly higher progression-free survival (PFS) and overall survival (OS) compared to the VS-P group among overall NSCLC patients regardless of treatment (median PFS of 7.1 vs. 4.2 months, P=0.013, and median OS, not reached vs. 17.2 months, P=0.012). Among NSCLC patients treated with ICIs, VS-G classification was associated with significantly increased PFS in comparison to VS-P classification (median PFS of 6.2 vs. 3.0 months, P=0.012), while the differences in OS trended towards significance (median OS, not reached vs. 16.5 months P=0.076). Multivariate analysis showed that the VeriStrat status was significantly correlated with PFS and OS in NSCLC patients treated with ICIs (P=0.017, P=0.034, respectively). Conclusions MS-based serum proteomic signature has potential as a biomarker for survival outcome in NSCLC patients receiving immunotherapy.
Collapse
Affiliation(s)
- Young Kwang Chae
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Won Bin Kim
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andrew A Davis
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lee Chun Park
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Division of Hematology/Oncology, Internal Medicine, Kosin University, Busan, Republic of Korea
| | - Jonathan F Anker
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nicholas I Simon
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kyunghoon Rhee
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Junho Song
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Anderson Cho
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sangmin Chang
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Taeyeong Ko
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Michael Oh
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Manali Bhave
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Pedro Viveiros
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
42
|
Prognostic impact of PD-L1 expression in correlation with neutrophil-to-lymphocyte ratio in squamous cell carcinoma of the lung. Sci Rep 2020; 10:1243. [PMID: 31988315 PMCID: PMC6985257 DOI: 10.1038/s41598-019-57321-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 12/21/2019] [Indexed: 12/25/2022] Open
Abstract
The prognostic impact of tumoral programmed death-ligand 1 (PD-L1) expression in correlation with neutrophil-to-lymphocyte ratio (NLR) was retrospectively assessed in 83 patients with completely resected stage I squamous cell carcinoma of the lung, as PD-L1 is a potent regulator of cancer immunity and NLR is a potential surrogate of immune status. Forty-three patients (51.8%) had tumor with positive PD-L1 expression. There was no significant correlation between PD-L1 expression and NLR. PD-L1-positivity failed to provide a significant prognostic impact (overall survival [OS] rate at 5 years, 53.0% in PD-L1-positive patients versus 70.1% in PD-L1-negative patients; P = 0.117). Among NLR-low (<2.2) patients, however, PD-L1-positivity was significantly correlated with a poor prognosis (OS rate at 5 years, 46.1% versus 86.0%; P = 0.020). In contrast, among NLR-high (≥2.2) patients, PD-L1-positivity provided no prognostic impact (P = 0.680). When NLR status and tumoral PD-L1 status were combined, “NLR-low and PD-L1-negative” was a significant and independent factor to predict a favorable recurrence-free survival (hazard ratio, 0.237 [95% confidence interval, 0.083 to 0.674]; P = 0.007) and OS (hazard ratio, 0.260 [0.091 to 0.745]; P = 0.012). These results suggest the prognostic impact of tumoral PD-L1 expression might be influenced by the status of NLR.
Collapse
|
43
|
Zhang B, Liu Y, Zhou S, Jiang H, Zhu K, Wang R. Predictive effect of PD-L1 expression for immune checkpoint inhibitor (PD-1/PD-L1 inhibitors) treatment for non-small cell lung cancer: A meta-analysis. Int Immunopharmacol 2020; 80:106214. [PMID: 31982822 DOI: 10.1016/j.intimp.2020.106214] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/06/2020] [Accepted: 01/13/2020] [Indexed: 10/25/2022]
Abstract
BACKGROUND Programmed death-ligand-1 (PD-L1) is a well-known predictive biomarker in non-small cell lung cancer (NSCLC) patients, however, its accuracy remains controversial. Here, we investigated the correlation between PD-L1 expression level and efficacy of its inhibitors, and hence assessed the predictive effect of PD-L1 expression. METHODS Studies that evaluated the efficacy of programmed death-1 (PD-1)/ PD-L1 inhibitors in advanced NSCLC patients according to tumor PD-L1 expression levels were searched for on Medline, Cochrane Library, and Embase. The pooled risk ratio (RR) and 95% confidence intervals (95% CIs) were calculated for the objective response rate (ORR) with overall survival (OS) and progression-free survival (PFS) were measured in terms of hazard ratio (HR) and the corresponding 95% CIs. RESULTS 1432 NSCLC patients from six randomized controlled trials (RCTs) were included and three PD-1/PD-L1 inhibitors (atezolizumab, nivolumab, and pembrolizumab) were used to treat the patients. A significantly higher ORR was observed in the high PD-L1 expression group compared to the low expression group (0.35 [95% CI, 0.30-0.40] vs 0.11 [95% CI, 0.09-0.14]). The results of the subgroup analysis, grouped by the type of drugs and antibodies which assess immune checkpoint inhibitors were identical with the pooled result. However, our study showed that PD-L1 expression was neither prognostic nor predictive of overall survival (OS) or progression-free survival (PFS) in patients treated with PD-1/PD-L1 inhibitors compared to chemotherapy. CONCLUSIONS PD-L1 can be a predictive biomarker for ORR. Nevertheless, PD-L1 expression is not a good predictive tool for OS and PFS.
Collapse
Affiliation(s)
- Binbin Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yi Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Sijing Zhou
- Hefei Prevention and Treatment Center for Occupational Diseases, Hefei 230022, China.
| | - Huihui Jiang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Ke Zhu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Ran Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
44
|
Liu Y, Zhou S, Du Y, Sun L, Jiang H, Zhang B, Sun G, Wang R. Efficacy and safety of programmed death 1 inhibitors in patients with advanced non-small cell lung cancer: a meta-analysis. Cancer Manag Res 2019; 11:4619-4630. [PMID: 31191023 PMCID: PMC6535411 DOI: 10.2147/cmar.s193394] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 04/11/2019] [Indexed: 12/26/2022] Open
Abstract
Objective: This study aims to perform systematic review and meta-analysis of all randomized controlled trials that compare the efficacy and safety of programmed death 1 (PD-1) inhibitors versus chemotherapy alone in previously untreated advanced non-small cell lung cancer (NSCLC). Materials and methods: Several databases, including Medline, Cochrane Library, Embase, and Web of Science, were searched. The main outcome measures included overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and adverse events (AEs). Results: The results of meta-analysis are expressed as the hazard ratio (HR) or risk ratio (RR) with their corresponding 95% confidence intervals (CIs). The final analysis included six trials for 3,930 patients. PD-1 inhibitors led to a statistically superior survival benefit over chemotherapy in patients with advanced NSCLC. OS was longer in patients who received PD-1 inhibitors (HR =0.71, 95% CI =0.62–0.74, P=0.000). Furthermore, PD-1 inhibitors had significantly higher objective response rate than chemotherapy (RR =0.20,95% CI =0.17–0.23, P=0.000). Meta-analysis showed that the AEs of any grade with PD-1 inhibitors were lower than those with chemotherapy (RR =0.78; 95% CI =0.75–0.81, P=0.000). Conclusion: PD-1 inhibitors showed a clinically meaningful survival benefit and an improved safety profile in patients with previously treated NSCLC.
Collapse
Affiliation(s)
- Yi Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Sijing Zhou
- Hefei Prevention and Treatment Center for Occupational Diseases, Hefei 230022, People's Republic of China
| | - Yongsheng Du
- Department of General Medicine, Hefei Second People's Hospital, Hefei 230022, People's Republic of China
| | - Li Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Huihui Jiang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Binbin Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Ran Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| |
Collapse
|
45
|
Abstract
BACKGROUND Melanoma-associated antigen-A (MAGE-A) was recognized as high-expressed in many solid tumors including esophageal carcinoma (EC), nevertheless, was reported to be low/not-expressed in normal tissues. Thus, it was considered as an extraordinary appropriate target for treatment especially in immunotherapy. Therefore, it demanded more detail knowledge on the precise function of MAGE-A. METHODS In this study, we used the data from the Cancer Genome Atlas dataset (TCGA-ESCA) to analyze the expression and survival for MAGE A3/4/11 (the subtype of MAGE-A) using the online tool of UALCAN. Furthermore, the high-throughput sequencing data of the patients with esophageal squamous-cell carcinoma (ESCC) from TCGA dataset were performed to analyze the correlation test, gene ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment of MAGE A3/4/9/11 using LinkeDomics (online tool) and ClueGO (inner software of Cytoscape). Finally, relative gene expressions of MAGE A3/4/9/11 were verified by quantitative real-time PCR (q-PCR) in the patients with EC. RESULTS MAGE A3/4/11 was high-expressed in tissues of patients with ESCC, and there was no difference in survival time for patients between the high-expressed with the low/medium-expressed. The Go enrichment analysis showed that the 4 MAGE-A subtypes (MAGE-A3/4/9/11) were enriched in the regulation of the adaptive immune response, translational initiation, interleukin-4 production, response to type I interferon, and skin development, respectively. The KEGG results showed that they were enriched in T cell receptor signaling pathway (MAGE-A3), Th1 and Th2 differentiation, antigen processing and presentation (MAGE-A4), cytokine-cytokine receptor interaction (MAGE-A9), and chemokine signaling pathway (MAGE-A11). CONCLUSION MAGE A3/4/9/11 was high-expressed in EC, and were enrolled in the regulation of immune response. They may consider as candidate immune target for EC treatment and provided the messages for further research in the function of MAGE-A.
Collapse
Affiliation(s)
- Xiaohua Chen
- Oncology of Panyu Central Hospital, Panyu Cancer Institute
| | - Sina Cai
- Oncology of The Hospital of Third Affiliated Southern Medical University, Guangzhou, Guangdong
| | - Liping Wang
- The First People's Hospital of Chenzhou, Chenzhou, Hunan
| | - Xiaona Zhang
- Graceland Medical Center, The Sixth Affiliated Hospital of Sun Yat–Sen University, Guangzhou, Guangdong, China
| | - Wenhui Li
- Oncology of Panyu Central Hospital, Panyu Cancer Institute
| | - Xiaolong Cao
- Oncology of Panyu Central Hospital, Panyu Cancer Institute
| |
Collapse
|
46
|
Martin-Fernandez ML, Clarke DT, Roberts SK, Zanetti-Domingues LC, Gervasio FL. Structure and Dynamics of the EGF Receptor as Revealed by Experiments and Simulations and Its Relevance to Non-Small Cell Lung Cancer. Cells 2019; 8:E316. [PMID: 30959819 PMCID: PMC6523254 DOI: 10.3390/cells8040316] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/29/2019] [Accepted: 03/30/2019] [Indexed: 12/25/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is historically the prototypical receptor tyrosine kinase, being the first cloned and the first where the importance of ligand-induced dimer activation was ascertained. However, many years of structure determination has shown that EGFR is not completely understood. One challenge is that the many structure fragments stored at the PDB only provide a partial view because full-length proteins are flexible entities and dynamics play a key role in their functionality. Another challenge is the shortage of high-resolution data on functionally important higher-order complexes. Still, the interest in the structure/function relationships of EGFR remains unabated because of the crucial role played by oncogenic EGFR mutants in driving non-small cell lung cancer (NSCLC). Despite targeted therapies against EGFR setting a milestone in the treatment of this disease, ubiquitous drug resistance inevitably emerges after one year or so of treatment. The magnitude of the challenge has inspired novel strategies. Among these, the combination of multi-disciplinary experiments and molecular dynamic (MD) simulations have been pivotal in revealing the basic nature of EGFR monomers, dimers and multimers, and the structure-function relationships that underpin the mechanisms by which EGFR dysregulation contributes to the onset of NSCLC and resistance to treatment.
Collapse
Affiliation(s)
- Marisa L Martin-Fernandez
- Central Laser Facility, Research Complex at Harwell, STFC Rutherford Appleton Laboratory, Harwell Oxford, Didcot, Oxford OX11 0QX, UK.
| | - David T Clarke
- Central Laser Facility, Research Complex at Harwell, STFC Rutherford Appleton Laboratory, Harwell Oxford, Didcot, Oxford OX11 0QX, UK.
| | - Selene K Roberts
- Central Laser Facility, Research Complex at Harwell, STFC Rutherford Appleton Laboratory, Harwell Oxford, Didcot, Oxford OX11 0QX, UK.
| | - Laura C Zanetti-Domingues
- Central Laser Facility, Research Complex at Harwell, STFC Rutherford Appleton Laboratory, Harwell Oxford, Didcot, Oxford OX11 0QX, UK.
| | | |
Collapse
|
47
|
Vesci L, Carollo V, Rosi A, De Santis R. Therapeutic efficacy of intra-tumor AvidinOX and low systemic dose biotinylated cetuximab, with and without cisplatin, in an orthotopic model of head and neck cancer. Oncol Lett 2019; 17:3529-3536. [PMID: 30867794 DOI: 10.3892/ol.2019.10003] [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: 07/19/2018] [Accepted: 01/17/2019] [Indexed: 11/05/2022] Open
Abstract
In a previous study, the efficacy of low intraperitoneal doses of biotinylated cetuximab (bCet) in mice with subcutaneous tumor xenografts of human head and neck cancer (HNC) treated intra-tumors with AvidinOX was reported. Taking into account that the current standard treatment for HNC is the combination of cetuximab and cisplatin, the present study investigated the activity of AvidinOX-targeted bCet with and without cisplatin in an orthotopic model. The results confirmed that administration of intra-tumor AvidinOX makes an otherwise inactive dose of bCet effective in reducing tumor growth, and the addition of a low dose of cisplatin further improved tumor growth inhibition. Supporting the in vivo data, immunohistochemical staining of tumor masses from mice treated with AvidinOX, bCet and cisplatin exhibited the highest tumor cell damage and the lowest angiogenic activity among all treatment groups, measured as the number of γ-H2A.X and cleaved caspase-3-positive cells, and vascular endothelial growth factor-C and platelet and endothelial cell adhesion molecule 1-positive cells, respectively. AvidinOX is currently under clinical investigation to assess its use in delivering radioactive biotin to inoperable tumor lesions (ClinicalTrials.gov: NCT02053324 and NCT03188328). The present study further supported the potential clinical use of AvidinOX to target low bCet doses to inoperable tumor lesions, with or without an additional low dose of cisplatin. Since low doses of highly expensive monoclonal antibodies become effective with AvidinOX and low dose cisplatin, such therapies promise to be cheaper and less toxic than current treatments.
Collapse
Affiliation(s)
- Loredana Vesci
- Biotechnology R&D, Alfasigma S.p.A., Pomezia, I-00071 Rome, Italy
| | | | - Antonio Rosi
- Biotechnology R&D, Alfasigma S.p.A., Pomezia, I-00071 Rome, Italy
| | - Rita De Santis
- Biotechnology R&D, Alfasigma S.p.A., Pomezia, I-00071 Rome, Italy
| |
Collapse
|
48
|
Chikaishi Y, Hirai A, Imanishi N, Ichiki Y, Tanaka F. We should be done in such a way that patients with stage IV non-small cell lung cancer who would benefit from surgery are not overlooked. J Thorac Dis 2018; 10:S3257-S3259. [PMID: 30370131 DOI: 10.21037/jtd.2018.08.63] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Yasuhiro Chikaishi
- Second Department of Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Ayako Hirai
- Second Department of Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Naoko Imanishi
- Second Department of Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshinobu Ichiki
- Second Department of Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Fumihiro Tanaka
- Second Department of Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|