1
|
Cohen RB, Jimeno A, Hreno J, Sun L, Wallén-Öhman M, Millrud CR, Sanfridson A, Garcia-Ribas I. Safety, tolerability, and preliminary efficacy of nadunolimab, an anti-IL- 1 receptor accessory protein monoclonal antibody, in combination with pembrolizumab in patients with solid tumors. Invest New Drugs 2025:10.1007/s10637-025-01538-3. [PMID: 40310569 DOI: 10.1007/s10637-025-01538-3] [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: 03/08/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
Interleukin (IL)-1 signaling has an essential role in tumor progression and immunosuppression and is linked to acquired resistance to anti-PD-1/PD-L1 treatment. Nadunolimab is an IL1RAP (IL-1 receptor accessory protein)-targeting antibody that blocks IL-1α/IL-1β signaling and has enhanced antibody-dependent cellular cytotoxicity. We investigated the safety and preliminary efficacy of nadunolimab with pembrolizumab in patients with metastatic solid tumors who had progressed on previous checkpoint inhibitor treatment, suggesting acquired checkpoint inhibitor resistance (NCT04452214). This phase 1b trial enrolled patients with metastatic disease who had exhausted or declined standard-of-care alternatives. Patients received nadunolimab (5 mg/kg) and standard-dose pembrolizumab. The primary objective was to assess safety. Secondary objectives were anti-tumor response as per iRECIST, pharmacokinetics, and changes in immune mediators. Fifteen patients with stage IV cancer (head and neck squamous cell carcinoma, non-small cell lung cancer, melanoma) entered the trial. Grade ≥ 3 adverse events were reported for 7 patients (47%). There was one dose-limiting toxicity of febrile neutropenia. The most frequent grade ≥ 3 adverse event was dysphagia (two patients). Seven patients (47%) had reductions in target lesion size. Median iPFS was 3.4 months (95% CI 1.4-8.6). Median OS was 19.7 months (95% CI 4.3-28.7) with 67% 1-year survival. Survival was significantly longer in patients with higher baseline tumor infiltration of CD163 + macrophages and natural killer cells and in patients with reduced on-treatment circulating IL-6 levels or neutrophil-to-lymphocyte ratio. Nadunolimab with pembrolizumab had an acceptable safety profile, and prolonged disease control was observed in a subset of patients. The results support further development of nadunolimab in combination with checkpoint inhibitors.
Collapse
Affiliation(s)
- Roger B Cohen
- Division of Hematology-Oncology, University of Pennsylvania, Philadelphia, PA, USA.
| | - Antonio Jimeno
- Department of Medicine, Division of Medical Oncology, Developmental Therapeutics Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jennifer Hreno
- Department of Medicine, Division of Medical Oncology, Developmental Therapeutics Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lova Sun
- Division of Hematology-Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
2
|
Nair R, Somasundaram V, Kuriakose A, Krishn SR, Raben D, Salazar R, Nair P. Deciphering T-cell exhaustion in the tumor microenvironment: paving the way for innovative solid tumor therapies. Front Immunol 2025; 16:1548234. [PMID: 40236693 PMCID: PMC11996672 DOI: 10.3389/fimmu.2025.1548234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
In solid tumors, the tumor microenvironment (TME) is a complex mix of tumor, immune, stromal cells, fibroblasts, and the extracellular matrix. Cytotoxic T lymphocytes (CTLs) constitute a fraction of immune cells that may infiltrate into the TME. The primary function of these T-cells is to detect and eliminate tumor cells. However, due to the immunosuppressive factors present in the TME primarily mediated by Myeloid-Derived Suppressor Cells (MDSCs), Tumor associated macrophages (TAMs), Cancer Associated Fibroblasts (CAFs) as well as the tumor cells themselves, T-cells fail to differentiate into effector cells or become dysfunctional and are unable to eliminate the tumor. In addition, chronic antigen stimulation within the TME also leads to a phenomenon, first identified in chronic lymphocytic choriomeningitis virus (LCMV) infection in mice, where the T-cells become exhausted and lose their effector functions. Exhausted T-cells (Tex) are characterized by the presence of remarkably conserved inhibitory receptors, transcription and signaling factors and the downregulation of key effector molecules. Tex cells have been identified in various malignancies, including melanoma, colorectal and hepatocellular cancers. Recent studies have indicated novel strategies to reverse T-cell exhaustion. These include checkpoint inhibitor blockade targeting programmed cell death protein 1 (PD-1), T-cell immunoglobulin and mucin-domain containing-3 (Tim-3), cytotoxic T-lymphocyte associated protein 4 (CTLA-4), or combinations of different immune checkpoint therapies (ICTs) or combination of ICTs with cytokine co-stimulation. In this review, we discuss aspects of T-cell dysfunction within the TME with a focus on T-cell exhaustion. We believe that gaining insight into the mechanisms of T-cell exhaustion within the TME of human solid tumors will pave the way for developing therapeutic strategies to target and potentially re-invigorate exhausted T-cells in cancer.
Collapse
Affiliation(s)
- Reshmi Nair
- Syngene International Limited, Bengaluru, India
| | | | | | | | - David Raben
- Bicara Therapeutics, Boston, MA, United States
| | | | - Pradip Nair
- Syngene International Limited, Bengaluru, India
| |
Collapse
|
3
|
Sun S, Zhang F, Zhang J, Yu H, Hu Z, Xu X, Zhao X, Chen S, Zhang Y, Nian B, Lin Y, Li Z, Wu Z, Yu B, Wu X, Wang H, Hui X, Zhang D, Wang J. Small extracellular vesicle miRNAs as biomarkers for predicting antitumor efficacy in lung adenocarcinoma treated with chemotherapy and checkpoint blockade. Front Immunol 2025; 16:1573043. [PMID: 40230863 PMCID: PMC11994727 DOI: 10.3389/fimmu.2025.1573043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
Checkpoint blockade combined with chemotherapy has become an important treatment option for lung cancer patients in clinical settings. However, biomarkers that effectively identify true responders remain lacking. We assessed the potential of plasma small extracellular vesicle (sEV)-derived microRNAs (miRNAs) as biomarkers for predicting and identifying responders to combined immunochemotherapy. A total of 29 patients with lung adenocarcinoma who received pembrolizumab combined with pemetrexed and carboplatin were enrolled. The efficacy evaluation revealed that 24 patients obtained durable clinical benefits from combined immunochemotherapy, and the rest experienced disease progression. Using unsupervised hierarchical clustering, 56 differentially expressed miRNAs (DEMs) were identified between responders and nonresponders. Efficacy prediction models incorporating a combination of sEV miRNAs were established and showed good performance (area under the curve (AUC) > 0.9). In addition, we found that miR-96-5p and miR-6815-5p were notably downregulated in the nonresponder group, while miR-99b-3p, miR-100-5p, miR-193a-5p, and miR-320d were upregulated. These findings were further confirmed by clinical imaging. sEV miRNAs derived from patients with lung cancer showed promise for identifying true responders to combined immunochemotherapy.
Collapse
Affiliation(s)
- Si Sun
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Thoracic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Fuchuang Zhang
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Jiyang Zhang
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Hui Yu
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhihuang Hu
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoya Xu
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Xinmin Zhao
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sheng Chen
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Yao Zhang
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Baoning Nian
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Ying Lin
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhikuan Li
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Zhenhua Wu
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bo Yu
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xianghua Wu
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Huijie Wang
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaohua Hui
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dadong Zhang
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Jialei Wang
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Thoracic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
4
|
Yang Z, Luo Y, Lu R, Liu X, Liu H, Liu S, Huang C, Tian J, Zhang L. Incidence Rates of Cutaneous Immune-Related Adverse Events in Patients with Lung Cancer: A Systematic Review and Meta-Analysis. Curr Oncol 2025; 32:195. [PMID: 40277752 PMCID: PMC12025845 DOI: 10.3390/curroncol32040195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
OBJECTIVE Cutaneous immune-related adverse events (cirAEs) represent a prevalent manifestation of adverse reactions linked to immune checkpoint inhibitors (ICIs) therapy, substantially affecting patients' quality of life. This systematic review and meta-analysis aimed to quantify the pooled incidence of cirAEs in this population and strengthen clinical awareness for early recognition and management. METHODS A comprehensive search of PubMed, Embase, CINAHL, Cochrane Library, CBM, CNKI, and Wanfang databases was conducted from inception to December 2022. Literature that reported the incidence of cirAEs in patients with lung cancer receiving ICIs therapy was included. A meta-analysis was conducted using R software, version 4.4.1 to estimate the pooled incidence of cirAEs, and a random-effects model was used for data synthesis. Begg's rank correlation and funnel plots were used to assess publication bias. RESULTS A total of 99 articles involving 23,814 patients with lung cancer receiving ICIs therapy were included, with publication dates ranging from 2012 to 2022. The meta-analysis results reveal that the incidence of cirAEs in patients with lung cancer was 20.26% (95% confidence interval [CI (17.12-23.81)]. Significant differences were observed between all subgroups, including continent, study type, combination therapy, dual ICIs therapy, and diagnostic criteria for cirAEs for Grade 1-2 and Grade 3-4 incidences. CONCLUSIONS The incidence of cirAEs in patients with lung cancer is relatively high, particularly undergoing combined or dual ICIs therapy. To comprehensively characterize cirAEs in patients with lung cancer, large-scale multicenter studies integrating real-world pharmacovigilance data are warranted to establish precise incidence estimates and identify clinically significant risk factors. IMPLICATIONS FOR CLINICAL PRACTICE This review's insights aroused clinical staff's attention and concern about cirAEs, potentially enhancing the quality of life of patients with cancer.
Collapse
Affiliation(s)
- Zhihui Yang
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
- Evidence Based Nursing and Midwifery Practice PR China: A JBI Centre of Excellence, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
| | - Yuanyuan Luo
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
| | - Ruiqi Lu
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
| | - Xinqi Liu
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
| | - Hanyu Liu
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
| | - Suting Liu
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
| | - Chen Huang
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
| | - Jinhui Tian
- Evidence Based Medicine Centre, School of Basic Medical Sciences, Lanzhou University, No. 199, Donggang West Road, Lanzhou 730000, China
| | - Lili Zhang
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
- Evidence Based Nursing and Midwifery Practice PR China: A JBI Centre of Excellence, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
| |
Collapse
|
5
|
Czajkowski M, Wierzbicki PM, Dolny M, Matuszewski M, Hakenberg OW. Inflammation in Penile Squamous Cell Carcinoma: A Comprehensive Review. Int J Mol Sci 2025; 26:2785. [PMID: 40141426 PMCID: PMC11943298 DOI: 10.3390/ijms26062785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Inflammation appears to play a crucial role in the development and progression of penile cancer (PeCa). Two molecular pathways of PeCa are currently described: HPV-dependent and HPV-independent. The tumor immune microenvironment (TIME) of PeCa is characterized by the presence of tumor-associated macrophages, cancer-associated fibroblasts, and tumor-infiltrating lymphocytes. The components of the TIME produce pro-inflammatory cytokines and chemokines, which have been found to be overexpressed in PeCa tissues and are associated with tumor progression and unfavorable prognoses. Additionally, the nuclear factor kappa B (NF-κB) pathway and secreted phosphoprotein 1 (SPP1) have been implicated in PeCa pathogenesis. Elevated C-reactive protein (CRP) levels and the neutrophil-to-lymphocyte ratio (NLR) have been identified as potential prognostic biomarkers in PeCa. This overview presents the complex contribution of the inflammatory process and collates projects aimed at modulating TIME in PeCa.
Collapse
Affiliation(s)
- Mateusz Czajkowski
- Department of Urology, Medical University of Gdańsk, Mariana Smoluchowskiego 17 Street, 80-214 Gdansk, Poland; (M.D.); (M.M.)
| | - Piotr M. Wierzbicki
- Department of Histology, Medical University of Gdańsk, Dębinki, 80-211 Gdansk, Poland;
| | - Maciej Dolny
- Department of Urology, Medical University of Gdańsk, Mariana Smoluchowskiego 17 Street, 80-214 Gdansk, Poland; (M.D.); (M.M.)
| | - Marcin Matuszewski
- Department of Urology, Medical University of Gdańsk, Mariana Smoluchowskiego 17 Street, 80-214 Gdansk, Poland; (M.D.); (M.M.)
| | - Oliver W. Hakenberg
- Department of Urology, University Medical Center Rostock, 18055 Rostock, Germany;
- Department of Urology, Jena University Hospital, 07747 Jena, Germany
| |
Collapse
|
6
|
Wang F, Wei X, Yang M, Lu C, Yang X, Deng J, Chen Z, Zhou Q. A Novel DNA Repair-Gene Model to Predict Responses to Immunotherapy and Prognosis in Patients With EGFR-Mutant Non-Small Cell Lung Cancer. Thorac Cancer 2025; 16:e70025. [PMID: 39994841 PMCID: PMC11850292 DOI: 10.1111/1759-7714.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND The epidermal growth factor receptor mutant (EGFRm) non-small cell lung cancer (NSCLC) has a unique "cold" immune profile. DNA damage repair (DDR) genes are closely related to tumorigenesis and the effectiveness of immunotherapy in many tumors. However, the role and mechanism of DDR in the genesis and progression of EGFRm NSCLC remain unclear. METHODS This study included 101 EGFRm NSCLC samples from The Cancer Genome Atlas (TCGA) dataset and a GSE31210 dataset (external set) from the GEO database. Cluster analysis was used to identify different subtypes of EGFRm NSCLC based on the expression of DDR genes. Univariate and LASSO regression analysis was used to develop a DDR-based predictive model. The prognostic significance of this model was assessed using Cox regression, Kaplan-Meier, and receiver operating characteristic (ROC) curve analyses. Bioinformatics analysis was performed to investigate the clinicopathological characteristics and immune profiles associated with this model. In vitro experiment was performed to testify the role of DDR genes in EGFRm NSCLC. RESULTS We identified two subtypes of EGFRm NSCLC: DDR-activated and DDR-suppressed. The DDR-activated subtype showed more aggressive clinical behavior and poorer prognosis and was more responsive to immunotherapy. A prognostic model for EGFRm NSCLC was constructed using four DDR genes: CAPS, FAM83A, IGLV8-61, and SLC7A5. The derived risk score could serve as an independent prognostic indicator. High- and low-risk patients exhibited distinct clinicopathological characteristics, immune profiles, and responses to immunotherapy. The T-cell inflammation and Tumor Immune Dysfunction and Exclusion (TIDE) scores differed between the high- and low-risk subgroups, with both showing enhanced effectiveness of immunotherapy in the low-risk subgroup. Targeted therapy such as BI.2536, an inhibitor of polo-like kinase 1, could be effective for patients with high-risk EGFRm NSCLC. Meanwhile, in vitro detection approved the role of DDR genes in EGFRm NSCLC response. CONCLUSION This study demonstrated a diversity of DDR genes in EGFRm NSCLC and developed a predictive model using these genes. This model could assist in identifying potential candidates for immunotherapy and in assessing personalized treatment and prognosis of patients with EGFRm NSCLC.
Collapse
Affiliation(s)
- Fen Wang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Xue‐Wu Wei
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Ming‐Yi Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Chang Lu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Xiao‐Rong Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Jia‐Yi Deng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Zhi‐Hong Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| |
Collapse
|
7
|
Kashyap P, Raj KV, Sharma J, Dutt N, Yadav P. Classification of NSCLC subtypes using lung microbiome from resected tissue based on machine learning methods. NPJ Syst Biol Appl 2025; 11:11. [PMID: 39824879 PMCID: PMC11742043 DOI: 10.1038/s41540-025-00491-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 01/07/2025] [Indexed: 01/20/2025] Open
Abstract
Classification of adenocarcinoma (AC) and squamous cell carcinoma (SCC) poses significant challenges for cytopathologists, often necessitating clinical tests and biopsies that delay treatment initiation. To address this, we developed a machine learning-based approach utilizing resected lung-tissue microbiome of AC and SCC patients for subtype classification. Differentially enriched taxa were identified using LEfSe, revealing ten potential microbial markers. Linear discriminant analysis (LDA) was subsequently applied to enhance inter-class separability. Next, benchmarking was performed across six different supervised-classification algorithms viz. logistic-regression, naïve-bayes, random-forest, extreme-gradient-boost (XGBoost), k-nearest neighbor, and deep neural network. Noteworthy, XGBoost, with an accuracy of 76.25%, and AUROC (area-under-receiver-operating-characteristic) of 0.81 with 69% specificity and 76% sensitivity, outperform the other five classification algorithms using LDA-transformed features. Validation on an independent dataset confirmed its robustness with an AUROC of 0.71, with minimal false positives and negatives. This study is the first to classify AC and SCC subtypes using lung-tissue microbiome.
Collapse
Affiliation(s)
- Pragya Kashyap
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, India
| | - Kalbhavi Vadhi Raj
- Department of Electrical Engineering, Indian Institute of Technology, Jodhpur, Rajasthan, India
| | - Jyoti Sharma
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, India
| | - Naveen Dutt
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Pankaj Yadav
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, India.
- School of Artificial Intelligence and Data Science, Indian Institute of Technology, Jodhpur, Rajasthan, India.
| |
Collapse
|
8
|
Lu C, Zhu W, Han X, Du X, Zhang H, Yao Q, Liu T, Zhang C. Clinicopathological characteristics of invasive stratified mucinous carcinoma of the cervix and the expression and clinical significance of SLC7A11, SLC3A2 and PD-L1. Front Oncol 2024; 14:1492498. [PMID: 39807126 PMCID: PMC11725564 DOI: 10.3389/fonc.2024.1492498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
Introduction Invasive Stratified Mucin-producing Carcinoma (ISMC) of the cervix is a newly named cervical adenocarcinoma associated with Human Papilloma virus (HPV). Due to its relative rarity, clinical data, pathological features, and molecular characteristics of ISMC are still under exploration. This study aims to retrospectively analyze the clinical data and pathological features of ISMC patients, summarizing the clinical and pathological morphological characteristics of ISMC. Immunohistochemistry for SLC7A11, SLC3A2, and PD-L1 will be performed on tumor tissues from ISMC patients to preliminarily explore potential therapeutic targets for ISMC. Methods We retrospectively reviewed the electronic medical records and pathological slides of 22 ISMC patients, and performed immunohistochemical staining for solute carrier family 7 member 11 (SLC7A11), solute carrier family 3 member 2 (SLC3A2), and programmed death-ligand 1 (PD-L1). Results The patients were aged between 31 and 70 years old. The most common symptoms were abnormal vaginal bleeding and unusual vaginal discharge. HPV testing indicated that the infection rate of HPV type 18 was the highest. All patients underwent extensive hysterectomy and pelvic lymph node dissection. The progression-free survival (PFS) ranged from 3 to 112 months, with a postoperative recurrence rate of 22.7% (5/22). ISMC exhibited diverse characteristic microstructures. Immunohistochemistry results showed that the positive rates of SLC7A11 and SLC3A2 were both 91.0% (20/22). The staining intensity of SLC7A11 in frequent ISMC recurrence cases was significantly stronger than in non-recurrent ISMC cases. PD-L1 positivity was observed in 86.4% (19/22) of cases, defined as having a Combined Positive Score(CPS)≥1. Discussion ISMC demonstrates a high rate of lymph node metastasis and a high recurrence rate, indicating strong invasiveness. Additionally, ISMC exhibits a wide morphological spectrum. SLC7A11, SLC3A2, and PD-L1 are all highly expressed in ISMC tissues. The high expression of SLC7A11 may indicate a high recurrence rate for ISMC. Immunotherapy with checkpoint inhibitors and iron death-related treatments show potential in the treatment of ISMC, with SLC7A11, SLC3A2, and PD-L1 serving as potential therapeutic targets for ISMC.
Collapse
Affiliation(s)
- Changyu Lu
- The Affiliated Hospital of Qingdao University, Qingdao, China
- Qilu Hospital, Shandong University, Jinan, China
| | - Wei Zhu
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiahui Han
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiuzhen Du
- The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Hui Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
| | - Qin Yao
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ting Liu
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
| | - Ce Zhang
- Shandong Second Medical University, Weifang, China
| |
Collapse
|
9
|
Chen YC, Chen AYC, Hong R, Huang BE, Pirooznia M. Optimizing T cell inflamed signature through a combination biomarker approach for predicting immunotherapy response in NSCLC. Sci Rep 2024; 14:31382. [PMID: 39733163 PMCID: PMC11682295 DOI: 10.1038/s41598-024-82903-9] [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: 10/09/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
The introduction of anti-PD-1/PD-L1 therapies revolutionized treatment for advanced non-small cell lung cancer (NSCLC), yet response rates remain modest, underscoring the need for predictive biomarkers. While a T cell inflamed gene expression profile (GEP) has predicted anti-PD-1 response in various cancers, it failed in a large NSCLC cohort from the Stand Up To Cancer-Mark (SU2C-MARK) Foundation. Re-analysis revealed that while the T cell inflamed GEP alone was not predictive, its performance improved significantly when combined with gene signatures of myeloid cell markers. These additional signatures, however, showed negative contributions to prediction, hinting at immune alterations affecting therapy. Based on this, we proposed a combination biomarker approach that integrates the T cell inflamed GEP with immune-altered signatures, derived from the SU2C-MARK cohort using a machine-learning approach, as novel biomarkers. These signatures consisted of genes highly expressed in myeloid and stromal cells. We then assessed the predictive ability of these combined biomarkers in six independent cancer cohorts treated with anti-PD-1. The combined biomarkers demonstrated enhanced performance in NSCLC and gastric cancer cohorts, but not in melanoma cohorts. Our study introduces new biomarkers for predicting anti-PD-(L)1 response in NSCLC and offers mechanistic insights into treatment efficacy.
Collapse
Affiliation(s)
- Yun-Ching Chen
- Interventional Oncology, Johnson & Johnson Enterprise Innovation, Inc, 10th Floor 255 Main St, 02142, Cambridge, Boston, MA, USA.
| | - Ariel Yung-Chia Chen
- Interventional Oncology, Johnson & Johnson Enterprise Innovation, Inc, 10th Floor 255 Main St, 02142, Cambridge, Boston, MA, USA
| | - Rui Hong
- Interventional Oncology, Johnson & Johnson Enterprise Innovation, Inc, 10th Floor 255 Main St, 02142, Cambridge, Boston, MA, USA
| | - Bevan Emma Huang
- Interventional Oncology, Johnson & Johnson Enterprise Innovation, Inc, 10th Floor 255 Main St, 02142, Cambridge, Boston, MA, USA
| | - Mehdi Pirooznia
- Interventional Oncology, Johnson & Johnson Enterprise Innovation, Inc, 10th Floor 255 Main St, 02142, Cambridge, Boston, MA, USA.
| |
Collapse
|
10
|
Dowdell AK, Meng RC, Vita A, Bapat B, Hanes D, Chang SC, Harold L, Wong C, Poon H, Schroeder B, Weerasinghe R, Leidner R, Urba WJ, Bifulco CB, Piening BD. Widespread Adoption of Precision Anticancer Therapies After Implementation of Pathologist-Directed Comprehensive Genomic Profiling Across a Large US Health System. JCO Oncol Pract 2024; 20:1523-1532. [PMID: 39531849 PMCID: PMC11623383 DOI: 10.1200/op.24.00226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/25/2024] [Accepted: 08/16/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSE Precision therapies and immunotherapies have revolutionized cancer care, with novel genomic biomarker-associated therapies being introduced into clinical practice rapidly, resulting in notable gains in patient survival. Despite this, there is significant variability in the utilization of tumor molecular profiling that spans the timing of test ordering, comprehensiveness of gene panels, and clinical decision support through therapy and trial recommendations. METHODS To standardize testing, we designed a pathologist-directed test ordering system at the time of diagnosis using a 523-gene DNA/RNA hybrid comprehensive genomic profiling (CGP) panel and extensive clinical decision support tools. To comprehensively characterize the clinical impact of this protocol, we developed a novel natural language processing (NLP)-based approach to extract clinical features from physician chart notes. We assessed test actionability rates, therapy choice, and outcomes across a set of 3,216 patients with advanced cancer. RESULTS We observed 49% of patients had at least one actionable genomic biomarker-driven-approved and/or guideline-recommended targeted or immunotherapy (IO) and 53% of patients would have been eligible for a precision therapy clinical trial from three large basket trials. When assessing CGP versus an in silico 50-gene panel, 67% of tumors compared with 33% harbored actionable alterations including clinical trials. Among patients with 6 months or more of follow-up, over 52% received a targeted therapy (TT) or IO, versus 32% who received conventional chemotherapy alone. Furthermore, patients receiving TT had significantly improved overall survival compared with patients receiving chemotherapy alone (P < .001). CONCLUSION Overall, these data represent a major shift in standard clinical practice toward molecularly guided treatments (targeted and immunotherapies) over conventional systemic chemotherapy. As guidelines continue to evolve and more precision therapeutics gain approval, we expect this gap to continue to widen.
Collapse
Affiliation(s)
- Alexa K. Dowdell
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| | - Ryan C. Meng
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| | | | | | | | | | - Lauren Harold
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| | | | | | | | | | - Rom Leidner
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| | - Walter J. Urba
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| | - Carlo B. Bifulco
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| | - Brian D. Piening
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| |
Collapse
|
11
|
Abulizi A, Yan G, Xu Q, Muhetaer R, Wu S, Abudukelimu K, Chen X, Liu C, Li J. Cardiovascular adverse events and immune-related adverse events associated with PD-1/PD-L1 inhibitors for head and neck squamous cell carcinoma (HNSCC). Sci Rep 2024; 14:25919. [PMID: 39472591 PMCID: PMC11522629 DOI: 10.1038/s41598-024-75099-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 10/01/2024] [Indexed: 11/02/2024] Open
Abstract
While some literature has provided limited information about the potential cardiovascular risk and immune-related adverse events (irAEs) risk associated with PD-1/PD-L1 inhibitors in the treatment of Head and Neck Squamous Cell Carcinoma (HNSCC), the exact relevance is still uncertain. To assess the pharmacovigilance (PV), constituent ratio, severity, and reaction outcomes of major adverse cardiovascular events (MACE) and immune-related adverse events (irAEs) related to PD-1/PD-L1 inhibitors for HNSCC reported to the United States Food and Drug Administration Adverse Event Reporting System (FAERS). We analyzed reports of cardiovascular adverse events and irAEs associated with drug therapy for HNSCC submitted to FAERS from the 1st quarter 2015 to the 3rd quarter of 2023. Three PD-1/PD-L1 inhibitors were identified: nivolumab, pembrolizumab and durvalumab. Our primary composite endpoint was the PV of MACE and irAEs related to PD-1/PD-L1 inhibitors in the treatment of HNSCC, and the secondary endpoint was PV of other cardiovascular events. The software implemented was STATA 17.0 MP. 19,372 suspected drug-adverse event reports related to drug treatment in patients with HNSCC were identified, of which 916 reports were cardiovascular events, including 555 reports of MACE and 361 reports of other cardiovascular events. The PV signal regarding MACE was detected in durvalumab (PRR = 2.12, 95% CI: 1.24-3.61; χ2 = 7.71; ROR = 2.19, 95% CI: 1.24-3.86; IC = 1.01; IC025 = 0.07) but not in nivolumab and pembrolizumab. The constituent ratio of MACE in all adverse events caused by nivolumab (OR = 0.38, 95% CI: 0.19-0.73) and pembrolizumab (OR = 0.48, 95% CI: 0.23-0.99) was significantly decreased, compared with durvalumab. A PV signal about other cardiovascular events was detected in durvalumab (PRR = 3.04, 95% CI: 1.73-5.31; χ2 = 16.13; ROR = 3.15, 95% CI: 1.74-5.70; IC = 1.46; IC025 = 0.48), but it was not detected in nivolumab or pembrolizumab. The constituent ratio of other cardiovascular events in all adverse events caused by nivolumab (OR = 0.25, 95% CI: 0.13-0.48) and pembrolizumab (OR = 0.40, 95% CI: 0.20-0.80) was significantly decreased, compared with durvalumab. The constituent ratio of other cardiovascular events in all adverse events caused by nivolumab (OR = 0.61, 95% CI: 0.38-0.99) was significantly decreased, compared with pembrolizumab. There were 40 cases of hypertension. A PV signal about hypertension was detected in pembrolizumab (PRR = 3.72, 95% CI: 1.87-7.43; χ2 = 15.99; ROR = 3.75, 95% CI: 1.87-7.51; IC = 1.53, IC025 = 0.45), but it was not detected in nivolumab. The constituent ratio of hypertension in all adverse events caused by nivolumab (OR = 0.09, 95% CI: 0.09-0.39) was significantly decreased, compared with pembrolizumab. There were 737 cases of irAEs. A PV signal about irAEs was detected in nivolumab (PPR = 1.27, 95% CI: 1.05-1.53; χ2 = 6.38; ROR = 1.28, 95% CI: 1.06-1.56; IC = 0.29, IC025 = -0.00) and pembrolizumab (PPR = 2.20, 95% CI: 1.79-2.71; χ2 = 56.55; ROR = 2.31, 95% CI: 1.84-2.88; IC = 1.03; IC025 = 0.68), but it was not detected in durvalumab. The constituent ratio of irAEs in all adverse events caused by nivolumab (OR = 0.58, 95% CI: 0.44-0.76) significantly decreased, compared with pembrolizumab. By comparing the PV signals, constituent ratio, severity, and reaction outcome of the three drugs, we suppose that nivolumab can be used as the safest PD-1/PD-L1 inhibitor for HNSCC.
Collapse
Affiliation(s)
- Adila Abulizi
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Guangpeng Yan
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Qian Xu
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Reyihanguli Muhetaer
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Shihan Wu
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Kudelaiti Abudukelimu
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Xi Chen
- School of Health, Brooks College, Sunnyvale, USA
- Department of Epidemiology and Statistics, School of Public Health, Medical College, Zhejiang University, Hangzhou, China
| | - Chengjiang Liu
- Department of General Medicine, Anhui Medical University, Hefei, 230000, China
| | - Jun Li
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China.
| |
Collapse
|
12
|
Wang Z, Zou X, Wang H, Hao Z, Li G, Wang S. Companion diagnostics and predictive biomarkers for PD-1/PD-L1 immune checkpoint inhibitors therapy in malignant melanoma. Front Immunol 2024; 15:1454720. [PMID: 39530091 PMCID: PMC11550933 DOI: 10.3389/fimmu.2024.1454720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024] Open
Abstract
Programmed cell death receptor 1 (PD-1), when bound to the ligand programmed death-ligand 1 (PD-L1), can suppress cellular immunity and play a critical role in the initiation and development of cancer. Immune drugs targeting these two sites have been developed for different cancers, including malignant melanoma. The accompanying diagnostic method has been approved by the FDA to guide patient medication. However, the method of immunohistochemical staining, which varies widely due to the antibody and staining cut-off values, has certain limitations in application and does not benefit all patients. Increasing researches begin to focus on new biomarkers to improve objective response rates and survival in cancer patients. In this article, we enumerated three major groups, including tumour microenvironment, peripheral circulation, and gene mutation, which covered the current main research directions. In the future, we hope those biomarkers may be used to guide the treatment of patients with malignant melanoma.
Collapse
Affiliation(s)
- Zeping Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaojing Zou
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Haiyan Wang
- Beijing Biomedical Science and Technology Center, Zhaofenghua Biotechnology (Nanjing) Company Limited, Beijing, China
| | - Zhihui Hao
- College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, China
- National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing, China
| | - Gebin Li
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shuaiyu Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, China
- National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing, China
| |
Collapse
|
13
|
Santoro A, Pilar G, Tan DSW, Zugazagoitia J, Shepherd FA, Bearz A, Barlesi F, Kim TM, Overbeck TR, Felip E, Cai C, Simantini E, McCulloch T, Schaefer ES. Spartalizumab in combination with platinum-doublet chemotherapy with or without canakinumab in patients with PD-L1-unselected, metastatic NSCLC. BMC Cancer 2024; 24:1307. [PMID: 39448966 PMCID: PMC11515544 DOI: 10.1186/s12885-024-12841-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 08/21/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Despite promising outcomes of treatment with anti-programmed cell death (PD)-1/PD-ligand (L)1 agents in combination with platinum-doublet chemotherapy (PDC) in the first-line setting, a significant unmet medical need remains in patients with PD-L1-unselected non-small cell lung cancer (NSCLC). METHODS This multicenter, open-label, phase 1b study comprising dose-confirmation and dose-expansion parts investigated the combination of spartalizumab and various PDC regimens, with or without canakinumab, in treatment-naïve patients with PD-L1-unselected, metastatic NSCLC. The primary objectives were to determine maximum tolerated dose (MTD) and/or recommended dose for expansion (RDE) of spartalizumab, with or without canakinumab, in combination with PDC in the dose-confirmation part and antitumor activity of spartalizumab in the dose-expansion part. RESULTS The MTD/RDE of spartalizumab was 300 mg every 3 weeks (Q3W) when administered with either gemcitabine (1250 mg/m2)/cisplatin (75 mg/m2) (group A; no dose-limiting toxicities [DLTs]), pemetrexed (500 mg/m2)/cisplatin (group B; 2 DLTs: grade 2 posterior reversible encephalopathy syndrome and grade 4 hyponatremia), or paclitaxel (200 mg/m2)/carboplatin area under the curve 6 min*mg/mL (group C; 1 DLT: grade 4 neutropenic colitis). The RDE of canakinumab combined with spartalizumab and pemetrexed/cisplatin (group E; no DLTs) was 200 mg Q3W (no dose-expansion part was initiated). No new safety signals were identified. In groups A, B, C, and E, the overall response rates were 57.6%, 55.3%, 51.5%, and 57.1%, respectively. Group B compared with other groups had the longest median progression-free survival (10.4 months vs. 6.2-7.5 months), overall survival (29.7 months vs. 16.1-21.0 months), and duration of response (30.1 months vs. 6.0-8.2 months). CONCLUSIONS The combination of spartalizumab and PDC, with or without canakinumab, was well tolerated across treatment groups. The antitumor activity across treatment groups was comparable with that of pembrolizumab and pemetrexed combination. Canakinumab did not appear to improve the antitumor activity when combined with spartalizumab, pemetrexed and cisplatin. TRIAL REGISTRATION The trial was registered in Clinicaltrials.gov with identifier no. NCT03064854. Date of Registration: 06 February 2017.
Collapse
Affiliation(s)
- Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.
- Department of Oncology and Hematology, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, Rozzano-Milan, 20089, Italy.
| | - Garrido Pilar
- Department of Medical Oncology, Hospital Ramón Y Cajal, Madrid, Spain
| | - Daniel S W Tan
- Department of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Jon Zugazagoitia
- Department of Medical Oncology, University Hospital 12 de Octubre, Madrid, Spain
| | - Frances A Shepherd
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Alessandra Bearz
- Department of Medical Oncology, Centro di Riferimento Oncologico - CRO, Aviano, Italy
| | - Fabrice Barlesi
- Department of Multidisciplinary Oncology and Therapeutic Innovations, Aix Marseille University, CNRS, INSERM, CRCM, APHM, CEPCM, CLIP, Marseille, France
- Faculté de Médecine, Université Paris Saclay, Kremlin Bicêtre, France
- Medical Oncology Department, Gustave Roussy, Villejuif, France
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Tobias R Overbeck
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Enriqueta Felip
- Department of Medical Oncology Service, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Can Cai
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Eddy Simantini
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | - Eric S Schaefer
- Department of Medical Oncology, Highlands Oncology Group, Fayetteville, AZ, USA
| |
Collapse
|
14
|
Liu J, Man Y, Gao J, Wang X, Zhang L, Li M, Yu J. Correlation between PD-L1 expression status and efficacy of immunotherapy as second-line or later-line therapy in advanced non-small cell lung cancer patients. Eur J Cancer Prev 2024; 33:448-460. [PMID: 38386588 DOI: 10.1097/cej.0000000000000880] [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: 02/24/2024]
Abstract
OBJECTIVE The objective of this study is to evaluate the correlation between tumor proportionality scores (TPS) and the effectiveness of immune checkpoint inhibitors (ICIs) as the second or subsequent line therapies for individuals who received diagnoses of advanced non-small cell lung cancer (NSCLC). METHODS The retrospective analysis was conducted on the medical records of a total of 143 patients who received diagnoses of stage IIIB/IV NSCLC and were admitted to our hospital from the beginning of 2019 to the end of September 2022. The follow-up period ended on 01 January 2023. The study used Kaplan-Meier survival curves to assess the progression-free survival (PFS) and overall survival (OS) of patients. Univariate and multivariate Cox proportional risk models were used to analyze the factors associated with the PFS and OS of advanced-stage NSCLC patients who received ICIs as the second or subsequent lines. RESULTS Patients diagnosed with NSCLC who had a TPS ≥1% and got treatment with ICIs exhibit notably elevated rates of partial response, objective response rate, disease control rate and extended PFS in comparison to NSCLC patients with a TPS of <1% ( P < 0.05). NSCLC patients with TPS within 1-49% [hazard ratio (HR) = 0.372; 95% confidence interval (CI), 0.140-0.993; P = 0.048] or ≥50% (HR = 0.276; 95% CI, 0.095-0.796; P = 0.017) were significantly associated with prolonged PFS, which were conducted by multivariate Cox regression analysis. CONCLUSION Programmed death protein-1 expression status may be predictive markers of the effectiveness of ICIs as the second or subsequent lines of therapies in advanced NSCLC are influenced by TPS.
Collapse
Affiliation(s)
- Jingya Liu
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin
| | - Yingchun Man
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin
| | - Jianing Gao
- Department of Urology, Daqing Oilfield General Hospital, Dqing, China
| | - Xinxin Wang
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin
| | - Lijie Zhang
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin
| | - Mingheng Li
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin
| | - Jiahan Yu
- Department of Medical Oncology, Beidahuang Industry Group General Hospital, Harbin
| |
Collapse
|
15
|
Matsuki T, Kawakita D, Takahashi H, Okada T, Sakai A, Ueki Y, Tsuge H, Hanyu K, Momiyama K, Shodo R, Yamauchi M, Asako Y, Hirai H, Nagao T, Tada Y. PD-L1 expression as a predictive biomarker in patients with recurrent or metastatic salivary gland carcinoma treated with pembrolizumab. Sci Rep 2024; 14:19794. [PMID: 39187586 PMCID: PMC11347672 DOI: 10.1038/s41598-024-70779-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024] Open
Abstract
Although immune checkpoint inhibitors (ICIs) are effective in some patients with salivary gland carcinoma (SGC), biomarkers which predict the efficacy and prognosis of SGC patients treated with pembrolizumab have not been identified. We conducted a multi-institutional retrospective cohort study to evaluate the efficacy and safety of pembrolizumab monotherapy in patients with recurrent and/or metastatic SGC and to determine optimal cut-off values of the combined positive score (CPS) and tumor proportion score (TPS) as numerical expression levels of programmed death-ligand 1 (PD-L1), which predict the efficacy of pembrolizumab. Furthermore, we investigated the association of patient characteristics and hematological markers with clinical outcomes, including overall response rate (ORR), progression-free survival (PFS), and overall survival (OS). From 2016 to 2021, 27 patients were included in the analysis. ORR of SGC was 25.9%. Optimal cut-off values of CPS and TPS were 15 and 25%, respectively. ORRs of CPS-high and TPS-high were 55.6 and 75.0%, respectively, and significantly higher than those of CPS-low and TPS-low. Furthermore, patients with a low platelet-lymphocyte ratio (PLR) had a significantly longer PFS. No grade 4 or greater adverse events were observed. This study demonstrated the efficacy and safety of pembrolizumab monotherapy and identified optimal cut-off values of CPS and TPS.
Collapse
Affiliation(s)
- Takashi Matsuki
- Department of Head and Neck Surgery, Kanagawa Cancer Center, Yokohama, Kanagawa, 241-8515, Japan
- Department of Otorhinolaryngology, Head and Neck Surgery, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252‑0375, Japan
| | - Daisuke Kawakita
- Department of Otorhinolaryngology, Head and Neck Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467‑8602, Japan
| | - Hideaki Takahashi
- Department of Otorhinolaryngology, Head and Neck Surgery, School of Medicine, Yokohama City University, Yokohama, Kanagawa, 236‑0004, Japan
| | - Takuro Okada
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University School of Medicine, Tokyo, 160‑0023, Japan
| | - Akihiro Sakai
- Department of Otolaryngology-Head and Neck Surgery, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Yushi Ueki
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951‑8520, Japan
| | - Hiroshi Tsuge
- Department of Otorhinolaryngology, Head and Neck Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467‑8602, Japan
| | - Kenji Hanyu
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University School of Medicine, Tokyo, 160‑0023, Japan
| | - Kaho Momiyama
- Department of Otorhinolaryngology, Head and Neck Surgery, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252‑0375, Japan
| | - Ryusuke Shodo
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951‑8520, Japan
| | - Mayu Yamauchi
- Department of Otolaryngology-Head and Neck Surgery, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
- Department of Head and Neck Oncology and Surgery, International University of Health and Welfare Mita Hospital, 1-4-3 Mita, Minato-Ku, Tokyo, 108-8329, Japan
| | - Yukiko Asako
- Department of Otorhinolaryngology, Head and Neck Surgery, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252‑0375, Japan
- Department of Head and Neck Oncology and Surgery, International University of Health and Welfare Mita Hospital, 1-4-3 Mita, Minato-Ku, Tokyo, 108-8329, Japan
| | - Hideaki Hirai
- Department of Anatomic Pathology, Tokyo Medical University School of Medicine, Tokyo, 160‑0023, Japan
| | - Toshitaka Nagao
- Department of Anatomic Pathology, Tokyo Medical University School of Medicine, Tokyo, 160‑0023, Japan
| | - Yuichiro Tada
- Department of Head and Neck Oncology and Surgery, International University of Health and Welfare Mita Hospital, 1-4-3 Mita, Minato-Ku, Tokyo, 108-8329, Japan.
| |
Collapse
|
16
|
Li C, Zhou Z, Hou L, Hu K, Wu Z, Xie Y, Ouyang J, Cai X. A novel machine learning model for efficacy prediction of immunotherapy-chemotherapy in NSCLC based on CT radiomics. Comput Biol Med 2024; 178:108638. [PMID: 38897152 DOI: 10.1016/j.compbiomed.2024.108638] [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: 01/29/2024] [Revised: 04/16/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024]
Abstract
Lung cancer is categorized into two main types: non-small cell lung cancer (NSCLC) and small cell lung cancer. Of these, NSCLC accounts for approximately 85% of all cases and encompasses varieties such as squamous cell carcinoma and adenocarcinoma. For patients with advanced NSCLC that do not have oncogene addiction, the preferred treatment approach is a combination of immunotherapy and chemotherapy. However, the progression-free survival (PFS) typically ranges only from about 6 to 8 months, accompanied by certain adverse events. In order to carry out individualized treatment more effectively, it is urgent to accurately screen patients with PFS for more than 12 months under this treatment regimen. Therefore, this study undertook a retrospective collection of pulmonary CT images from 60 patients diagnosed with NSCLC treated at the First Affiliated Hospital of Wenzhou Medical University. It developed a machine learning model, designated as bSGSRIME-SVM, which integrates the rime optimization algorithm with self-adaptive Gaussian kernel probability search (SGSRIME) and support vector machine (SVM) classifier. Specifically, the model initiates its process by employing the SGSRIME algorithm to identify pivotal image features. Subsequently, it utilizes an SVM classifier to assess these features, aiming to enhance the model's predictive accuracy. Initially, the superior optimization capability and robustness of SGSRIME in IEEE CEC 2017 benchmark functions were validated. Subsequently, employing color moments and gray-level co-occurrence matrix methods, image features were extracted from images of 60 NSCLC patients undergoing immunotherapy combined with chemotherapy. The developed model was then utilized for analysis. The results indicate a significant advantage of the model in predicting the efficacy of immunotherapy combined with chemotherapy for NSCLC, with an accuracy of 92.381% and a specificity of 96.667%. This lays the foundation for more accurate PFS predictions and personalized treatment plans.
Collapse
Affiliation(s)
- Chengye Li
- Department of Pulmonary and Critical Care Medicine, The First Affliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Zhifeng Zhou
- Wenzhou University Library, Wenzhou, 325035, China.
| | - Lingxian Hou
- Rehabilitation Department, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, 325000, China.
| | - Keli Hu
- Department of Computer Science and Engineering, Shaoxing University, Shaoxing, 312000, China; Information Technology R&D Innovation Center of Peking University, Shaoxing, 312000, China.
| | - Zongda Wu
- Department of Computer Science and Engineering, Shaoxing University, Shaoxing, 312000, China.
| | - Yupeng Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Jinsheng Ouyang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Xueding Cai
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
17
|
Qi C, Li Y, Zeng H, Wei Q, Tan S, Zhang Y, Li W, Tian P. Current status and progress of PD-L1 detection: guiding immunotherapy for non-small cell lung cancer. Clin Exp Med 2024; 24:162. [PMID: 39026109 PMCID: PMC11258158 DOI: 10.1007/s10238-024-01404-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths and represents a substantial disease burden worldwide. Immune checkpoint inhibitors combined with chemotherapy are the standard first-line therapy for advanced NSCLC without driver mutations. Programmed death-ligand 1 (PD-L1) is currently the only approved immunotherapy marker. PD-L1 detection methods are diverse and have developed rapidly in recent years, such as improved immunohistochemical detection methods, the application of liquid biopsy in PD-L1 detection, genetic testing, radionuclide imaging, and the use of machine learning methods to construct PD-L1 prediction models. This review focuses on the detection methods and challenges of PD-L1 from different sources, and discusses the influencing factors of PD-L1 detection and the value of combined biomarkers. Provide support for clinical screening of immunotherapy-advantage groups and formulation of personalized treatment decisions.
Collapse
Affiliation(s)
- Chang Qi
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yalun Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Zeng
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qi Wei
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sihan Tan
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuanyuan Zhang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Panwen Tian
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Respiratory Health and Multimorbidity, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Center/Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
18
|
Yasmeen F, Pirzada RH, Ahmad B, Choi B, Choi S. Understanding Autoimmunity: Mechanisms, Predisposing Factors, and Cytokine Therapies. Int J Mol Sci 2024; 25:7666. [PMID: 39062908 PMCID: PMC11277571 DOI: 10.3390/ijms25147666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Autoimmunity refers to an organism's immune response against its own healthy cells, tissues, or components, potentially leading to irreversible damage to vital organs. Central and peripheral tolerance mechanisms play crucial roles in preventing autoimmunity by eliminating self-reactive T and B cells. The disruption of immunological tolerance, characterized by the failure of these mechanisms, results in the aberrant activation of autoreactive lymphocytes that target self-tissues, culminating in the pathogenesis of autoimmune disorders. Genetic predispositions, environmental exposures, and immunoregulatory disturbances synergistically contribute to the susceptibility and initiation of autoimmune pathologies. Within the realm of immune therapies for autoimmune diseases, cytokine therapies have emerged as a specialized strategy, targeting cytokine-mediated regulatory pathways to rectify immunological imbalances. Proinflammatory cytokines are key players in inducing and propagating autoimmune inflammation, highlighting the potential of cytokine therapies in managing autoimmune conditions. This review discusses the etiology of autoimmune diseases, current therapeutic approaches, and prospects for future drug design.
Collapse
Affiliation(s)
- Farzana Yasmeen
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; (F.Y.); (B.C.)
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Rameez Hassan Pirzada
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; (F.Y.); (B.C.)
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Bilal Ahmad
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, Republic of Korea
| | - Bogeum Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; (F.Y.); (B.C.)
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea; (F.Y.); (B.C.)
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, Republic of Korea
| |
Collapse
|
19
|
Saowapa S, Polpichai N, Siladech P, Wannaphut C, Tanariyakul M, Wattanachayakul P, Lalitnithi P. Pneumonitis Incidence in Patients With Metastatic Non-small Cell Lung Cancer on Immunotherapy: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e63615. [PMID: 39092378 PMCID: PMC11293893 DOI: 10.7759/cureus.63615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2024] [Indexed: 08/04/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, often diagnosed at the advanced stage (metastatic). Treatment options for metastatic NSCLC include radiotherapy, chemotherapy, target drug therapy, and immunotherapy. Immunotherapy (utilization of checkpoint inhibitors) boosts the immune system to recognize and destroy cancer cells. However, it is often associated with immune-related complications such as pneumonitis. This review aims to determine the incidence of pneumonitis in metastatic NSCLC patients treated with different immunotherapy drugs. PubMed, Cochrane Library, and Embase databases were scoured for randomized controlled trials (RCTs) until October 2023. Published RCTs with similar research objectives were included, while non-English articles, reviews, case reports, ongoing trials, non-randomized studies, conference abstracts, and studies on small cell lung cancer (SCLC) were excluded. The Cochrane risk-of-bias tool for randomized trials (RoB 2) was used to assess the risk of bias among the included studies. The statistical analyses were performed with the Comprehensive Meta-Analysis software. The subgroup analysis of the 16 included RCTs showed that metastatic NSCLC patients treated with nivolumab and pembrolizumab had a higher incidence of any grade pneumonitis than those treated with atezolizumab (4.5% and 5.1% vs. 1.6%, respectively). Similarly, the incidence of grade ≥3 pneumonitis was higher among patients receiving nivolumab (1.3%) and pembrolizumab (2.4%) than those receiving atezolizumab (0.7%). Furthermore, the subgroup analysis showed that patients with naive-treated NSCLC on immunotherapy had a higher incidence of any grade pneumonitis than those with previously treated NSCLC (6.5% vs. 3.9%). Treatment-naive patients recorded higher grade ≥3 pneumonitis incidences than those previously treated (3.1% vs. 1.3%). Programmed death 1 (PD-1) inhibitors (i.e., pembrolizumab and nivolumab) have higher incidences of pneumonitis than programmed death-ligand 1 inhibitors (atezolizumab).
Collapse
Affiliation(s)
- Sakditad Saowapa
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, USA
| | | | | | - Chalothorn Wannaphut
- Internal Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA
| | - Manasawee Tanariyakul
- Internal Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA
| | | | | |
Collapse
|
20
|
Sholl LM, Awad M, Basu Roy U, Beasley MB, Cartun RW, Hwang DM, Kalemkerian G, Lopez-Rios F, Mino-Kenudson M, Paintal A, Reid K, Ritterhouse L, Souter LA, Swanson PE, Ventura CB, Furtado LV. Programmed Death Ligand-1 and Tumor Mutation Burden Testing of Patients With Lung Cancer for Selection of Immune Checkpoint Inhibitor Therapies: Guideline From the College of American Pathologists, Association for Molecular Pathology, International Association for the Study of Lung Cancer, Pulmonary Pathology Society, and LUNGevity Foundation. Arch Pathol Lab Med 2024; 148:757-774. [PMID: 38625026 DOI: 10.5858/arpa.2023-0536-cp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/29/2024] [Indexed: 04/17/2024]
Abstract
CONTEXT.— Rapid advancements in the understanding and manipulation of tumor-immune interactions have led to the approval of immune therapies for patients with non-small cell lung cancer. Certain immune checkpoint inhibitor therapies require the use of companion diagnostics, but methodologic variability has led to uncertainty around test selection and implementation in practice. OBJECTIVE.— To develop evidence-based guideline recommendations for the testing of immunotherapy/immunomodulatory biomarkers, including programmed death ligand-1 (PD-L1) and tumor mutation burden (TMB), in patients with lung cancer. DESIGN.— The College of American Pathologists convened a panel of experts in non-small cell lung cancer and biomarker testing to develop evidence-based recommendations in accordance with the standards for trustworthy clinical practice guidelines established by the National Academy of Medicine. A systematic literature review was conducted to address 8 key questions. Using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach, recommendations were created from the available evidence, certainty of that evidence, and key judgments as defined in the GRADE Evidence to Decision framework. RESULTS.— Six recommendation statements were developed. CONCLUSIONS.— This guideline summarizes the current understanding and hurdles associated with the use of PD-L1 expression and TMB testing for immune checkpoint inhibitor therapy selection in patients with advanced non-small cell lung cancer and presents evidence-based recommendations for PD-L1 and TMB testing in the clinical setting.
Collapse
Affiliation(s)
- Lynette M Sholl
- From the Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (Sholl)
| | - Mark Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (Awad)
| | - Upal Basu Roy
- Translational Science Research Program, LUNGevity Foundation, Chicago, Illinois (Basu Roy)
| | - Mary Beth Beasley
- the Department of Anatomic Pathology and Clinical Pathology, Mt. Sinai Medical Center, New York, New York (Beasley)
| | - Richard Walter Cartun
- the Department of Anatomic Pathology, Hartford Hospital, Hartford, Connecticut (Cartun)
| | - David M Hwang
- the Department of Laboratory Medicine & Pathobiology, Sunnybrook Health Science Centre, Toronto, Ontario, Canada (Hwang)
| | - Gregory Kalemkerian
- the Department of Medical Oncology and Internal Medicine, University of Michigan Health, Ann Arbor (Kalemkerian)
| | - Fernando Lopez-Rios
- Pathology Department, Hospital Universitario 12 de Octubre, Madrid, Spain (Lopez-Rios)
| | - Mari Mino-Kenudson
- the Department of Pathology, Massachusetts General Hospital, Boston (Mino-Kenudson)
| | - Ajit Paintal
- the Department of Pathology, NorthShore University Health System, Evanston, Illinois (Paintal)
| | - Kearin Reid
- Governance (Reid) and the Pathology and Laboratory Quality Center for Evidence-based Guidelines, College of American Pathologists, Northfield, Illinois(Ventura)
| | - Lauren Ritterhouse
- the Department of Pathology, Foundation Medicine, Cambridge, Massachusetts (Ritterhouse)
| | | | - Paul E Swanson
- the Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle (Swanson)
| | - Christina B Ventura
- Governance (Reid) and the Pathology and Laboratory Quality Center for Evidence-based Guidelines, College of American Pathologists, Northfield, Illinois(Ventura)
| | - Larissa V Furtado
- the Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee (Furtado)
| |
Collapse
|
21
|
Li T, Ma W, Al-Obeidi E. Evolving Precision First-Line Systemic Treatment for Patients with Unresectable Non-Small Cell Lung Cancer. Cancers (Basel) 2024; 16:2350. [PMID: 39001412 PMCID: PMC11240640 DOI: 10.3390/cancers16132350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
First-line systemic therapy for patients with advanced or metastatic non-small cell lung cancer (NSCLC) has rapidly evolved over the past two decades. First, molecularly targeted therapy for a growing number of gain-of-function molecular targets has been shown to improve progression-free survival (PFS) and overall survival (OS) with favorable toxicity profiles compared to platinum-containing chemotherapy and can be given as first-line systemic therapy in ~25% of patients with NSCLC. Actionable genetic alterations include EGFR, BRAF V600E, and MET exon 14 splicing site-sensitizing mutations, as well as ALK-, ROS1-, RET-, and NTRK-gene fusions. Secondly, inhibitors of programmed cell death protein 1 or its ligand 1 (PD-1/L1) such as pembrolizumab, atezolizumab, or cemiplimab monotherapy have become a standard of care for ~25% of patients with NSCLC whose tumors have high PD-L1 expression (total proportion score (TPS) ≥50%) and no sensitizing EGFR/ALK alterations. Lastly, for the remaining ~50% of patients who are fit and whose tumors have no or low PD-L1 expression (TPS of 0-49%) and no sensitizing EGFR/ALK aberrations, platinum-containing chemotherapy with the addition of a PD-1/L1 inhibitor alone or in combination of a cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitor improves PFS and OS compared to chemotherapy alone. The objectives of this review are to summarize the current data and perspectives on first-line systemic treatment in patients with unresectable NSCLC and propose a practical algorithm for implementing precision biomarker testing at diagnosis.
Collapse
Affiliation(s)
- Tianhong Li
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; (W.M.)
- Medical Service, Hematology/Oncology, Veterans Affairs Northern California Health Care System, 10535 Hospital Way, Mather, CA 95655, USA
| | - Weijie Ma
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; (W.M.)
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Ebaa Al-Obeidi
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; (W.M.)
| |
Collapse
|
22
|
Badenhorst M, Windhorst AD, Beaino W. Navigating the landscape of PD-1/PD-L1 imaging tracers: from challenges to opportunities. Front Med (Lausanne) 2024; 11:1401515. [PMID: 38915766 PMCID: PMC11195831 DOI: 10.3389/fmed.2024.1401515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/20/2024] [Indexed: 06/26/2024] Open
Abstract
Immunotherapy targeted to immune checkpoint inhibitors, such as the program cell death receptor (PD-1) and its ligand (PD-L1), has revolutionized cancer treatment. However, it is now well-known that PD-1/PD-L1 immunotherapy response is inconsistent among patients. The current challenge is to customize treatment regimens per patient, which could be possible if the PD-1/PD-L1 expression and dynamic landscape are known. With positron emission tomography (PET) imaging, it is possible to image these immune targets non-invasively and system-wide during therapy. A successful PET imaging tracer should meet specific criteria concerning target affinity, specificity, clearance rate and target-specific uptake, to name a few. The structural profile of such a tracer will define its properties and can be used to optimize tracers in development and design new ones. Currently, a range of PD-1/PD-L1-targeting PET tracers are available from different molecular categories that have shown impressive preclinical and clinical results, each with its own advantages and disadvantages. This review will provide an overview of current PET tracers targeting the PD-1/PD-L1 axis. Antibody, peptide, and antibody fragment tracers will be discussed with respect to their molecular characteristics and binding properties and ways to optimize them.
Collapse
Affiliation(s)
- Melinda Badenhorst
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| | - Albert D. Windhorst
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| | - Wissam Beaino
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan, Amsterdam, Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands
| |
Collapse
|
23
|
Dong C, Hui K, Gu J, Wang M, Hu C, Jiang X. Plasma sPD-L1 and VEGF levels are associated with the prognosis of NSCLC patients treated with combination immunotherapy. Anticancer Drugs 2024; 35:418-425. [PMID: 38386011 DOI: 10.1097/cad.0000000000001576] [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: 02/23/2024]
Abstract
The clinical significance of plasma soluble programmed cell death ligand 1 (sPD-L1) and vascular endothelial growth factor (VEGF) for non-small cell lung cancer (NSCLC) treated with the combination of anti-angiogenic therapy and anti-PD-L1 antibody (Ab) remain unknown. This study aimed to explore the association between plasma sPD-L1 and VEGF levels and the prognosis of NSCLC patients treated with the combination of Envafolimab and Endostar. Peripheral blood samples were collected from 24 NSCLC patients at baseline and after 6 weeks of treatment and were detected for sPD-L1 and VEGF levels. Both baseline and posttreatment sPD-L1 were significantly higher in progressive disease (PD) group than in controlled disease (CD) group (median: 77.5 pg/ml vs. 64.6 pg/ml, P = 0.036, median: 8451 pg/ml vs. 5563 pg/ml, P = 0.012). In multivariate analysis, lower baseline sPD-L1 levels were significantly associated with longer progression-free survival (PFS) (HR = 6.834, 95% CI: 1.350-34.592, P = 0.020). There were significantly higher posttreatment VEGF levels in PD group compared with CD group (median: 323.7 pg/ml vs. 178.5 pg/ml, P = 0.009). Higher posttreatment VEGF levels were significantly associated with shorter PFS in multivariate analysis (HR = 5.911, 95% CI: 1.391-25.122, P = 0.016). Plasma sPD-L1 and VEGF levels are associated with the clinical response and prognosis of NSCLC patients treated with the combination of PD-L1 inhibitors and anti-angiogenetic therapy.
Collapse
Affiliation(s)
- Changhong Dong
- Department of Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu Province, China
| | | | | | | | | | | |
Collapse
|
24
|
Yang M, Cao H, Wang C, Yu C, Sun P. Incidence of thromboembolic events in non-small cell lung cancer patients treated with immune checkpoint inhibitors: A systematic review and meta-analysis. J Cancer Res Ther 2024; 20:509-521. [PMID: 38687920 DOI: 10.4103/jcrt.jcrt_1031_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 01/08/2024] [Indexed: 05/02/2024]
Abstract
ABSTRACT The incidence of thromboembolic events (TEs) in non-small cell lung cancer (NSCLC) patients treated with immune checkpoint inhibitors (ICIs) has rarely been reported. The MEDLINE, EMBASE, and the Cochrane Library databases were searched. The primary outcome was the incidence of TEs, and the secondary outcome was the relationship between TEs and overall survival (OS) following ICI therapy. A subgroup analysis of TE incidents was performed according to the TE type and combination regimens. The I2 statistic was used to determine the heterogeneity, and funnel plots and Egger's test were used to assess publication bias. A total of 16,602 patients with NSCLC in 63 experimental arms were included in the analysis. The rate of TEs ranged from 0.1% to 13.8%, and the pooled overall incidence of all-grade TEs was 3% (95% confidence interval [CI], 2%-4%). The pooled rate of high-grade TEs was 1% (95% CI, 1%-2%). The venous and arterial TE rates were 3% (95% CI, 2%-4%) and 1% (95% CI, 1%-2%), respectively. Patients who received immunotherapy + chemoradiotherapy had the highest incidence of TEs (7%). The TE pooled rate was higher in patients treated with combined ICIs than in those treated with mono ICIs (4% vs. 2%). The OS was lower in patients with TEs than in those without TEs (hazard ratio, 1.4; 95% CI, 1.02%-1.92%). The incidence of TEs in NSCLC patients treated with ICIs was reasonable. Nonetheless, clinicians must be aware of potential thrombotic complications and treat them promptly.
Collapse
Affiliation(s)
- Miaomiao Yang
- Department of Oncology, Yantai Yuhuangding Hospital, Affiliated with Medical College of Qingdao University, Yantai, Shandong, P.R. China
| | - Hongxin Cao
- Department of Medical Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
| | - Congcong Wang
- Department of Oncology, Yantai Yuhuangding Hospital, Affiliated with Medical College of Qingdao University, Yantai, Shandong, P.R. China
| | - Caiyan Yu
- Department of Oncology, Yantai Yuhuangding Hospital, Affiliated with Medical College of Qingdao University, Yantai, Shandong, P.R. China
| | - Ping Sun
- Department of Oncology, Yantai Yuhuangding Hospital, Affiliated with Medical College of Qingdao University, Yantai, Shandong, P.R. China
| |
Collapse
|
25
|
Privat M, Massot A, Hermetet F, Al Sabea H, Racoeur C, Mabrouk N, Cordonnier M, Moreau M, Collin B, Bettaieb A, Denat F, Bodio E, Bellaye PS, Goze C, Paul C. Development of an Immuno-SPECT/Fluorescent Bimodal Tracer Targeting Human or Murine PD-L1 on Preclinical Models. J Med Chem 2024; 67:2188-2201. [PMID: 38270503 DOI: 10.1021/acs.jmedchem.3c02120] [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: 01/26/2024]
Abstract
Detection of biomarkers to diagnose, treat, and predict the efficacy of cancer therapies is a major clinical challenge. Currently, biomarkers such as PD-L1 are commonly detected from biopsies, but this approach does not take into account the spatiotemporal heterogeneity of their expression in tumors. A solution consists in conjugating monoclonal antibodies (mAbs) targeting these biomarkers with multimodal imaging probes. In this study, a bimodal [111In]-DOTA-aza-BODIPY probe emitting in the near-infrared (NIR) was grafted onto mAbs targeting murine or human PD-L1 either in a site-specific or random manner. In vitro, these bimodal mAbs showed a good stability and affinity for PD-L1. In vivo, they targeted specifically PD-L1 and were detected by both fluorescence and SPECT imaging. A significant benefit of site-specific conjugation on glycans was observed compared to random conjugation on lysine. The potential of this bimodal agent was also highlighted, thanks to a proof of concept of fluorescence-guided surgery in a human PD-L1+ tumor model.
Collapse
Affiliation(s)
- Malorie Privat
- LIIC, EA7269, Université de Bourgogne, 21000 Dijon, France
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France
- ICMUB, UMR 6302 CNRS, Université de Bourgogne, 9 av. A. Savary, BP 47870, 21078 Dijon, France
| | - Aurélie Massot
- LIIC, EA7269, Université de Bourgogne, 21000 Dijon, France
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France
| | - François Hermetet
- INSERM, UMR 1231, Label Ligue Nationale contre le Cancer and LipSTIC, 21000 Dijon, France
- CRIGEN, 21000 Dijon, France
| | - Hassan Al Sabea
- ICMUB, UMR 6302 CNRS, Université de Bourgogne, 9 av. A. Savary, BP 47870, 21078 Dijon, France
| | - Cindy Racoeur
- LIIC, EA7269, Université de Bourgogne, 21000 Dijon, France
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France
| | - Nesrine Mabrouk
- LIIC, EA7269, Université de Bourgogne, 21000 Dijon, France
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France
| | - Marine Cordonnier
- INSERM, UMR 1231, Label Ligue Nationale contre le Cancer and LipSTIC, 21000 Dijon, France
| | - Mathieu Moreau
- ICMUB, UMR 6302 CNRS, Université de Bourgogne, 9 av. A. Savary, BP 47870, 21078 Dijon, France
| | - Bertrand Collin
- ICMUB, UMR 6302 CNRS, Université de Bourgogne, 9 av. A. Savary, BP 47870, 21078 Dijon, France
- Centre Régional De Lutte Contre Le Cancer Georges-François Leclerc C.G.F.L, plateforme d'imagerie et de radiothérapie précliniques, 21000, Dijon, France
| | - Ali Bettaieb
- LIIC, EA7269, Université de Bourgogne, 21000 Dijon, France
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France
| | - Franck Denat
- ICMUB, UMR 6302 CNRS, Université de Bourgogne, 9 av. A. Savary, BP 47870, 21078 Dijon, France
| | - Ewen Bodio
- ICMUB, UMR 6302 CNRS, Université de Bourgogne, 9 av. A. Savary, BP 47870, 21078 Dijon, France
| | - Pierre-Simon Bellaye
- Centre Régional De Lutte Contre Le Cancer Georges-François Leclerc C.G.F.L, plateforme d'imagerie et de radiothérapie précliniques, 21000, Dijon, France
| | - Christine Goze
- ICMUB, UMR 6302 CNRS, Université de Bourgogne, 9 av. A. Savary, BP 47870, 21078 Dijon, France
| | - Catherine Paul
- LIIC, EA7269, Université de Bourgogne, 21000 Dijon, France
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France
| |
Collapse
|
26
|
Li J, Hu H, Qin G, Bai F, Wu X, Ke H, Zhang J, Xie Y, Wu Z, Fu Y, Zheng H, Gong L, Xie Z, Deng Y. Biomarkers of Pathologic Complete Response to Neoadjuvant Immunotherapy in Mismatch Repair-Deficient Colorectal Cancer. Clin Cancer Res 2024; 30:368-378. [PMID: 37906636 DOI: 10.1158/1078-0432.ccr-23-2213] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/13/2023] [Accepted: 10/27/2023] [Indexed: 11/02/2023]
Abstract
PURPOSE Immune checkpoint inhibitors (ICI) have become the standard of care for patients with mismatch repair-deficient/microsatellite instability-high (dMMR/MSI-H) colorectal cancer. However, biomarkers of response to ICI are still lacking. EXPERIMENTAL DESIGN Forty-two patients with dMMR colorectal cancer treated with neoadjuvant PD-1 blockade were prospectively enrolled. To identify biomarkers of pathologic complete response (pCR) to neoadjuvant therapy, we analyzed genomic and transcriptomic profiles based on next-generation sequencing, and immune cell density based on multiplex immunofluorescence (mIF) staining. An integrated analysis of single-cell RNA sequencing from our previous study and GSE178341, as well as mIF was performed to further explore the significance of the tumor microenvironment (TME) on pCR response. RESULTS The tumor mutation burden of both tumor tissue and plasma blood samples was comparable between the pCR and non-pCR groups, while HLA-DQA1 and HLA-DQB1 were significantly overexpressed in the pCR group. Gene signature enrichment analysis showed that pathways including T-cell receptor pathway, antigen presentation pathway were significantly enriched in the pCR group. In addition, higher pre-existing CD8+ T-cell density was associated with pCR response (767.47 per.mm2 vs. 326.64 per.mm2, P = 0.013 Wilcoxon test). Further integrated analysis showed that CD8+ T cells with low PD-1 expression (PD-1lo CD8+ T cells) expressing high levels of TRGC2, CD160, and KLRB1 and low levels of proliferated and exhausted genes were significantly associated with pCR response. CONCLUSIONS Immune-associated transcriptomic features, particularly CD8+ T cells were associated with pCR response to ICI in dMMR colorectal cancer. Heterogeneity of TME within dMMR colorectal cancer may help to discriminate patients with complete response to neoadjuvant ICI.
Collapse
Affiliation(s)
- Jianxia Li
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huabin Hu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ge Qin
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fan Bai
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xianrui Wu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haoxian Ke
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianwei Zhang
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuqian Xie
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zehua Wu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yang Fu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | - Zhi Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yanhong Deng
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
27
|
Turner DC, Wada R, Zhou H, Wang X, de Greef R, Valiathan C, Zhang L, Zhang N, Kuchimanchi M, Chen T, Ballas M, Visser SAG. Model-based meta-analysis of non-small cell lung cancer with standard of care PD-1 inhibitors and chemotherapy for early development decision making. CPT Pharmacometrics Syst Pharmacol 2023; 12:1751-1763. [PMID: 36642813 PMCID: PMC10681483 DOI: 10.1002/psp4.12917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/17/2023] Open
Abstract
Single-arm cohorts/trials are often used in early phase oncology programs to support preliminary clinical activity assessments for investigational products, administered alone or in combination with standard of care (SOC) agents. Benchmarking clinical activity of those combinations against other treatments, including SOC, requires indirect comparisons against external trials, which presents challenges including cross-study differences in trial populations/other factors. To facilitate such nonrandomized comparisons, we developed a comprehensive model-based meta-analysis (MBMA) framework to quantitatively adjust for factors related to efficacy in metastatic non-small cell lung cancer (mNSCLC). Data were derived from 15 published studies assessing key programmed cell death protein-1 (PD-1) inhibitors pembrolizumab (n = 8) and nivolumab (n = 7), representing current SOC in mNSCLC. In the first stage, a mixed-effects logistic regression model for overall response rate (ORR) was developed accounting for effects of various population covariates on ORR. The ORR model results indicated an odds ratio (OR) of 1.02 for squamous versus non-squamous histology and OR of 1.20 for PD-ligand 1 tumor proportion score (TPS) per every 10% increase of TPS level. Next, a nonparametric mixed-effects model for overall survival (OS) was developed with ORR/other clinical covariates as input. Subsequently, MBMA simulations of relevant hypothetical scenarios involving single-arm trial design predicted OS hazard ratios as a function of ORR with matched patient characteristics. Findings from this MBMA and derived parameter estimates can be generally applied by the reader as a framework for interpreting efficacy data from early phase trials to support ORR-based go/no-go decisions and futility rules, illustrated through examples in this report.
Collapse
Affiliation(s)
- David C. Turner
- GSKCollegevillePennsylvaniaUSA
- Present address:
GenentechSan FranciscoCaliforniaUSA
| | - Russ Wada
- CertaraMenlo ParkCaliforniaUSA
- Present address:
QuanTx ConsultingMountain ViewCaliforniaUSA
| | | | - Xiaowei Wang
- GSKCollegevillePennsylvaniaUSA
- Present address:
ModernaCambridgeMassachusettsUSA
| | | | - Chandni Valiathan
- GSKCollegevillePennsylvaniaUSA
- Present address:
J&JNew BrunswickNew JerseyUSA
| | | | | | | | | | - Marc Ballas
- GSKCollegevillePennsylvaniaUSA
- Present address:
NovocurePotomacMarylandUSA
| | | |
Collapse
|
28
|
Cameron CM, Richardson B, Golden JB, Phoon YP, Tamilselvan B, Pfannenstiel L, Thapaliya S, Roversi G, Gao XH, Zagore LL, Cameron MJ, Gastman BR. A transcriptional evaluation of the melanoma and squamous cell carcinoma TIL compartment reveals an unexpected spectrum of exhausted and functional T cells. Front Oncol 2023; 13:1200387. [PMID: 38023136 PMCID: PMC10643547 DOI: 10.3389/fonc.2023.1200387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Significant heterogeneity exists within the tumor-infiltrating CD8 T cell population, and exhausted T cells harbor a subpopulation that may be replicating and may retain signatures of activation, with potential functional consequences in tumor progression. Dysfunctional immunity in the tumor microenvironment is associated with poor cancer outcomes, making exploration of these exhausted T cell subpopulations critical to the improvement of therapeutic approaches. Methods To investigate mechanisms associated with terminally exhausted T cells, we sorted and performed transcriptional profiling of CD8+ tumor-infiltrating lymphocytes (TILs) co-expressing the exhaustion markers PD-1 and TIM-3 from large-volume melanoma tumors. We additionally performed immunologic phenotyping and functional validation, including at the single-cell level, to identify potential mechanisms that underlie their dysfunctional phenotype. Results We identified novel dysregulated pathways in CD8+PD-1+TIM-3+ cells that have not been well studied in TILs; these include bile acid and peroxisome pathway-related metabolism and mammalian target of rapamycin (mTOR) signaling pathways, which are highly correlated with immune checkpoint receptor expression. Discussion Based on bioinformatic integration of immunophenotypic data and network analysis, we propose unexpected targets for therapies to rescue the immune response to tumors in melanoma.
Collapse
Affiliation(s)
- Cheryl M. Cameron
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, United States
| | - Brian Richardson
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, United States
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Jackelyn B. Golden
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Yee Peng Phoon
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Banumathi Tamilselvan
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, United States
| | - Lukas Pfannenstiel
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Samjhana Thapaliya
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Gustavo Roversi
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Xing-Huang Gao
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Leah L. Zagore
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Mark J. Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Brian R. Gastman
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
29
|
Xu Z, Liang J, Fu R, Yang L, Xin Chen Y, Ren W, Lu Y, Qiu X, Gu Q. Effect of PD-L1 Expression for the PD-1/L1 Inhibitors on Non-small Cell Lung Cancer: A Meta-analysis Based on Randomised Controlled Trials. Clin Oncol (R Coll Radiol) 2023; 35:640-651. [PMID: 37563075 DOI: 10.1016/j.clon.2023.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/23/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023]
Abstract
AIMS As PD-L1 expression has been proposed as one of the cancer biomarkers for non-small cell lung cancer (NSCLC), the predictive value of tumour proportional score (TPS) in the effect of immunotherapy [programmed death protein-1/ligand 1 (PD-1/L1) inhibitors] for NSCLC is worth exploring further. Here, we aimed to summarise the outcomes of current NSCLC randomised controlled trials (RCTs) and explore the predictive value of TPS in clinical immunotherapy, including immune checkpoint inhibitors (ICIs) with or without chemotherapy. MATERIALS AND METHODS RCTs published by PubMed, Medline, Embase and Scopus before February 2023 comparing immunotherapy (PD-1/L1 with or without other therapy) versus a control group in advanced or metastatic NSCLC were included to assess the prognosis according to the patients' TPS with 1% and 50% as the thresholds. The primary endpoints were overall survival and progression-free survival. RESULTS In total, 28 RCTs containing 17 266 participants with advanced or metastatic NSCLC were included in this meta-analysis. Statistical results showed that compared with TPS <1%, ≥1% or within 1-49%, patients with TPS ≥50% benefited more significantly from the immunotherapy. A subgroup analysis showed that when TPS was <1%, ≥1% or within 1-49%, ICIs + chemotherapy had better efficacy than ICIs alone; PD-1 (such as pembrolizumab) inhibitors had better efficacy than PD-L1 inhibitors (such as atezolizumab). CONCLUSION The efficacy of immunotherapy (PD-1/L1 inhibitors) for advanced or metastatic NSCLC is influenced by TPS.
Collapse
Affiliation(s)
- Z Xu
- Department of Respiratory and Critical Care Medicine, Linhai Second People's Hospital, Taizhou, Zhejiang, China
| | - J Liang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - R Fu
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - L Yang
- Emergency Medical Center, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, China
| | - Y Xin Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - W Ren
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Y Lu
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - X Qiu
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Q Gu
- Department of Respiratory and Critical Care Medicine, Linhai Second People's Hospital, Taizhou, Zhejiang, China.
| |
Collapse
|
30
|
Wang F, Xia T, Li Z, Gao X, Fang X. Current status of clinical trial research and application of immune checkpoint inhibitors for non-small cell lung cancer. Front Oncol 2023; 13:1213297. [PMID: 37727216 PMCID: PMC10505960 DOI: 10.3389/fonc.2023.1213297] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/17/2023] [Indexed: 09/21/2023] Open
Abstract
Immunotherapy has emerged as a hot topic in the treatment of non-small cell lung cancer (NSCLC) with remarkable success. Compared to chemotherapy patients, the 5-year survival rate for immunotherapy patients is 3-fold higher, approximately 4%-5% versus 15%-16%, respectively. Immunotherapies include chimeric antigen receptor T-cell (CAR-T) therapy, tumor vaccines, immune checkpoint inhibitors, and so forth. Among them, immune checkpoint inhibitors are in the spotlight. Common immune checkpoint inhibitors (ICIs) currently in clinical use include programmed death receptor-1(PD-1)/programmed death ligand-1(PD-L1) and cytotoxic T lymphocyte-associated antigen 4(CTLA-4). This article focuses on monotherapy and combination therapy of CTLA-4 and PD-1/PD-L1 immune checkpoint inhibitors. In particular, the combination therapy of ICIs includes the combination of ICIs and chemotherapy, the combination therapy of dual ICIs, the combination of ICIs and anti-angiogenic drugs, the combination of ICIs and radiotherapy, and the combination of ICIs inhibitors and tumor vaccines and so forth. This article focuses on the combination therapy of ICIs with chemotherapy, the combination therapy of dual ICIs, and the combination therapy of ICIs with anti-angiogenic drugs. The efficacy and safety of ICIs as single agents in NSCLC have been demonstrated in many trials. However, ICIs plus chemotherapy regimens offer significant advantages in the treatment of NSCLC with little to no dramatic increase in toxicity, while combined dual ICIs significantly reduce the adverse effects (AEs) of chemotherapy. ICIs plus anti-angiogenic agents regimen improves anti-tumor activity and safety and is expected to be the new paradigm for the treatment of advanced NSCLC. Despite some limitations, these agents have achieved better overall survival rates. In this article, we review the current status and progress of research on ICIs in NSCLC in recent years, aiming to better guide the individualized treatment of NSCLC patients.
Collapse
Affiliation(s)
- Fuli Wang
- Department of Oncology, Lianyungang Clinical College Affiliated to Bengbu Medical College, Lianyungang, China
- Department of Oncology, Gaochun Hospital Afliated to Jiangsu University, Nanjing, China
| | - Teng Xia
- Department of Oncology, Gaochun Hospital Afliated to Jiangsu University, Nanjing, China
| | - Zhiqiang Li
- Department of Oncology, Lianyungang Clinical College Affiliated to Bengbu Medical College, Lianyungang, China
| | - Xuzhu Gao
- Department of Oncology, Lianyungang Clinical College Affiliated to Bengbu Medical College, Lianyungang, China
- Department of Oncology, Gaochun Hospital Afliated to Jiangsu University, Nanjing, China
| | - Xinjian Fang
- Department of Oncology, Lianyungang Clinical College Affiliated to Bengbu Medical College, Lianyungang, China
- Department of Oncology, Gaochun Hospital Afliated to Jiangsu University, Nanjing, China
| |
Collapse
|
31
|
Alrehaili AA, Gharib AF, Almalki A, Alghamdi A, Hawsawi NM, Bakhuraysah MM, Alhuthali HM, Etewa RL, Elsawy WH. Soluble Programmed Death-Ligand 1 (sPD-L1) as a Promising Marker for Head and Neck Squamous Cell Carcinoma: Correlations With Clinical and Demographic Characteristics. Cureus 2023; 15:e44338. [PMID: 37779773 PMCID: PMC10539100 DOI: 10.7759/cureus.44338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2023] [Indexed: 10/03/2023] Open
Abstract
Background and objective Head and neck squamous cell carcinoma (HNSCC) is a prevalent cancer type that affects the mucosal lining of the upper aerodigestive tract. Soluble programmed death-ligand 1 (sPD-L1) is a significant factor in hindering T cells' function, which prevents cancer cells from being detected by the immune system. This means that sPD-L1 is an essential component in the immune evasion of cancer. This study aimed to explore the potential of sPD-L1 as a prognostic biomarker for patients with HNSCC undergoing concurrent chemotherapy and radiation therapy. Methodology The study included 106 patients with locally advanced HNSCC who received three courses of induction chemotherapy followed by concurrent chemoradiation and 60 healthy subjects as controls. sPD-L1 levels were measured using an enzyme-linked immunosorbent assay (ELISA) kit, and the cutoff value was determined based on receiver operating characteristic (ROC) curve analysis. Results The results showed that sPD-L1 levels were significantly higher in HNSCC patients compared to healthy controls, with a cutoff value of 31.51 pg/mL. Higher sPD-L1 levels were associated with poorer overall survival rates. Conclusions These findings suggest that sPD-L1 may serve as a valuable prognostic biomarker for HNSCC patients undergoing concurrent chemotherapy and radiation therapy. The study highlights the importance of exploring new biomarkers and therapeutic strategies for HNSCC to improve patient outcomes and reduce morbidity and mortality rates associated with this disease.
Collapse
Affiliation(s)
- Amani A Alrehaili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, SAU
| | - Amal F Gharib
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, SAU
| | - Abdulraheem Almalki
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, SAU
| | - Ahmed Alghamdi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, SAU
| | - Nahed M Hawsawi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, SAU
| | - Maha M Bakhuraysah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, SAU
| | - Hayaa M Alhuthali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, SAU
| | - Rasha L Etewa
- Department of Pathology, College of Medicine, Jouf University, Sakaka, SAU
| | - Wael H Elsawy
- Department of Clinical Oncology, Faculty of Medicine, Zagazig University, Zagazig, EGY
| |
Collapse
|
32
|
Liu YG, Jiang ST, Zhang L, Zheng H, Zhang T, Zhang JW, Zhao HT, Sang XT, Xu YY, Lu X. Worldwide productivity and research trend of publications concerning tumor immune microenvironment (TIME): a bibliometric study. Eur J Med Res 2023; 28:229. [PMID: 37430294 DOI: 10.1186/s40001-023-01195-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 06/25/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND As the complexity and diversity of the tumor immune microenvironment (TIME) are becoming better understood, burgeoning research has progressed in this field. However, there is a scarcity of literature specifically focused on the bibliometric analysis of this topic. This study sought to investigate the development pattern of TIME-related research from 2006 to September 14, 2022, from a bibliometric perspective. METHODS We acquired both articles and reviews related to TIME from the Web of Science Core Collection (WoSCC) (retrieved on September 14, 2022). R package "Bibliometrix" was used to calculate the basic bibliometric features, present the collaborative conditions of countries and authors, and generate a three-field plot to show the relationships among authors, affiliations, and keywords. VOSviewer was utilized for co-authorship analysis of country and institution and keyword co-occurrence analysis. CiteSpace was used for citation burst analysis of keywords and cited references. In addition, Microsoft Office Excel 2019 was used to develop an exponential model to fit the cumulative publication numbers. RESULTS A total of 2545 publications on TIME were included, and the annual publication trend exhibited a significant increase over time. China and Fudan University were the most productive country and institution, with the highest number of publications of 1495 and 396, respectively. Frontiers in Oncology held the highest number of publications. A number of authors were recognized as the main contributors in this field. The clustering analysis revealed six clusters of keywords that highlighted the research hot spots in the fields of basic medical research, immunotherapy, and various cancer types separately. CONCLUSIONS This research analyzed 16 years of TIME-related research and sketched out a basic knowledge framework that includes publications, countries, journals, authors, institutions, and keywords. The finding revealed that the current research hot spots of the TIME domain lie in "TIME and cancer prognosis", "cancer immunotherapy", and "immune checkpoint". Our researchers identified the following areas: "immune checkpoint-based immunotherapy", "precise immunotherapy" and "immunocyte pattern", which may emerge as frontiers and focal points in the upcoming years, offering valuable avenues for further exploration.
Collapse
Affiliation(s)
- Yao-Ge Liu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Shi-Tao Jiang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Lei Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Han Zheng
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Ting Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jun-Wei Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Hai-Tao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xin-Ting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Yi-Yao Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China.
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China.
| |
Collapse
|
33
|
Tuerxun H, Zhao Y, Li Y, Liu X, Wen S, Cao J, Cui J, Zhao Y. Immune Checkpoint Inhibitors as A Threat to Reproductive Function: A Systematic Review. Crit Rev Oncol Hematol 2023:104064. [PMID: 37379960 DOI: 10.1016/j.critrevonc.2023.104064] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/08/2023] [Accepted: 06/25/2023] [Indexed: 06/30/2023] Open
Abstract
In recent years, the indications for immunotherapy in cancer treatment have been expanding. The increased risk of cancer in young people, coupled with the fact that many women or men choose to delay childbearing, has made an increasing number of patients of childbearing age eligible for immunotherapy. Furthermore, with the improvements of various treatments, more young people and children are able to survive cancer. As a result, long-term sequelae of cancer treatments, such as reproductive dysfunction, are increasingly important for survivors. While many anti-cancer drugs are known to cause reproduction dysfunction, the effects of immune checkpoint inhibitors (ICIs) on reproduction function remain largely unknown. Through a retrospective analysis of previous reports and literature, this article aims to elucidate the causes of reproductive dysfunction induced by ICIs and focus on their specific mechanisms, in order to providing some guidance to clinicians and patients.
Collapse
Affiliation(s)
- Halahati Tuerxun
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Yixin Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Yawen Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Xingyu Liu
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Shuhui Wen
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Jingjing Cao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
34
|
Mangone L, Marinelli F, Bisceglia I, Zambelli A, Zanelli F, Pagano M, Alberti G, Morabito F, Pinto C. Changes in the Histology of Lung Cancer in Northern Italy: Impact on Incidence and Mortality. Cancers (Basel) 2023; 15:3187. [PMID: 37370797 DOI: 10.3390/cancers15123187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/06/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
This study assessed the incidence, mortality, and survival of lung cancer subtypes of NSCSLC (non-small-cell lung cancer), SCLC (small-cell lung cancer), and other morphologies. It is an observational epidemiological study using 7197 cases from the Reggio Emilia Cancer Registry recorded between 2001 and 2020 in males and females. The incidence of NSCLC in 5104 males indicates a significant 3% annual increase until 2013 and then a decline of -3.2% that is not statistically significant; until 2014, mortality increased significantly (3.2%), but it then decreased non-significantly (-12.1%), especially squamous cell cancer. In 2093 females, the incidence and mortality trends continued to rise significantly through 2012, and then they began to slightly decline (not statistically significant). The two-year relative survival of NSCLC increased from 32% to 38% in males and from 42% to 56% in females. SCLC in males decreased significantly both in incidence and mortality, while in women, it showed a slight increase (significantly for incidence but not for mortality). This study is important because it analyzes the decrease in lung cancer in males and the increase in females in relation to the different histotypes. Our study's findings confirmed a decline in male incidence and death beginning in 2013. We were unable to determine if the drop in cigarette smoking and the introduction of new drugs such as EGFR in first-line therapy were responsible for the lower incidence.
Collapse
Affiliation(s)
- Lucia Mangone
- Epidemiology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, 42122 Reggio Emilia, RE, Italy
| | - Francesco Marinelli
- Epidemiology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, 42122 Reggio Emilia, RE, Italy
| | - Isabella Bisceglia
- Epidemiology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, 42122 Reggio Emilia, RE, Italy
| | - Alessandro Zambelli
- Epidemiology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, 42122 Reggio Emilia, RE, Italy
| | - Francesca Zanelli
- Medical Oncology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, 42122 Reggio Emilia, RE, Italy
| | - Maria Pagano
- Medical Oncology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, 42122 Reggio Emilia, RE, Italy
| | - Giulia Alberti
- Medical Oncology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, 42122 Reggio Emilia, RE, Italy
| | - Fortunato Morabito
- Biotechnology Research Unit, Azienda Sanitaria Provinciale di Cosenza, 87051 Aprigliano, CS, Italy
| | - Carmine Pinto
- Medical Oncology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, 42122 Reggio Emilia, RE, Italy
| |
Collapse
|
35
|
Stanley R, Flanagan S, Reilly DO, Kearney E, Naidoo J, Dowling CM. Immunotherapy through the Lens of Non-Small Cell Lung Cancer. Cancers (Basel) 2023; 15:cancers15112996. [PMID: 37296957 DOI: 10.3390/cancers15112996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/24/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Immunotherapy has revolutionised anti-cancer treatment in solid organ malignancies. Specifically, the discovery of CTLA-4 followed by PD-1 in the early 2000s led to the practice-changing clinical development of immune checkpoint inhibitors (ICI). Patients with lung cancer, including both small cell (SCLC) and non-small cell lung cancer (NSCLC), benefit from the most commonly used form of immunotherapy in immune checkpoint inhibitors (ICI), resulting in increased survival and quality of life. In NSCLC, the benefit of ICIs has now extended from advanced NSCLC to earlier stages of disease, resulting in durable benefits and the even the emergence of the word 'cure' in long term responders. However, not all patients respond to immunotherapy, and few patients achieve long-term survival. Patients may also develop immune-related toxicity, a small percentage of which is associated with significant mortality and morbidity. This review article highlights the various types of immunotherapeutic strategies, their modes of action, and the practice-changing clinical trials that have led to the widespread use of immunotherapy, with a focus on ICIs in NSCLC and the current challenges associated with advancing the field of immunotherapy.
Collapse
Affiliation(s)
- Robyn Stanley
- School of Medicine, University of Limerick, V94 T9PX Limerick, Ireland
| | - Saoirse Flanagan
- School of Medicine, University of Limerick, V94 T9PX Limerick, Ireland
| | | | - Ella Kearney
- School of Medicine, University of Limerick, V94 T9PX Limerick, Ireland
| | - Jarushka Naidoo
- Beaumont Hospital, D09 V2N0 Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- Sidney Kimmel Comprehensive Cancer Centre, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Catríona M Dowling
- School of Medicine, University of Limerick, V94 T9PX Limerick, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| |
Collapse
|
36
|
Faoro L, Brusegan A, Russi A, Calderone V, Martelli A, Marranconi E, Carpanese D, Berti E, Coppola M. Analysis of the relation between adverse events and overall survival in patients treated with pembrolizumab as a first-line treatment for metastatic NSCLC. BMC Pharmacol Toxicol 2023; 24:32. [PMID: 37189193 PMCID: PMC10184366 DOI: 10.1186/s40360-023-00663-0] [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: 09/09/2021] [Accepted: 03/14/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Many trials supported pembrolizumab as a first-line monotherapy to significantly improve overall survival (OS) in selected patients with previously untreated metastatic Non-Small Cell Lung Cancer (mNSCLC) and a PD-L1 TPS of ≥50% without EGFR/ALK mutations. The aim of this study was to reveal the correlation between OS and adverse events in real-world settings after 42 months. METHODS This retrospective observational study involved 98 patients with mNSCLC, TPS ≥ 50%, and no EGFR/ALK aberrations. Patients were treated with pembrolizumab (200 mg q3w) as a first-line treatment. Clinical data, including PD-L1 expression, Performance Status (ECOG-PS), treatment duration, toxicity, and outcomes were retrieved from local electronic medical records and from the Italian Regulatory Agency Registry. RESULTS The cohort's main characteristics were as follows: median age 73 [44-89] years, 64.3% were male and 35.7% were female, an ECOG-PS score of 0 (n = 73) and 1 or 2 (n = 25), and a PD-L1 > 90% in 29.6% of patients. The entire cohort had stage IV NSCLC at diagnosis. The median number of cycles was 8.5 at a median follow-up of 13 months. The median OS of 13.6 months (95% CI: 11.7-NA) was not influenced by sex and PD-L1, but was significantly associated with ECOG-PS (p = 0.02). Immune-Related Adverse Events (irAEs) occurred in 77.5% of patients (30.1% cutaneous, 27.5% gastrointestinal, and 20.4% endocrinological), but no grade 4 or 5 irAEs were identified. Patients experiencing any type of toxicity had a significantly longer median OS (20.39 months, 95% CI: 13.08-NA) than those with no toxicities (6.46 months, 95% CI: 1.41-NA, p = 0.006). CONCLUSION The percentage of irAEs detected was comparable to that reported in KEYNOTE-024 and KEYNOTE-042. These real-world findings demonstrated the significant correlation between OS and cutaneous toxicities.
Collapse
Affiliation(s)
- Lisa Faoro
- Department of Pharmacy, Specialization School in Hospital Pharmacy, University of Pisa, Pisa, Italy.
| | - Adriana Brusegan
- Pharmacy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.
| | - Alberto Russi
- Pharmacy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, Specialization School in Hospital Pharmacy, University of Pisa, Pisa, Italy
| | - Alma Martelli
- Department of Pharmacy, Specialization School in Hospital Pharmacy, University of Pisa, Pisa, Italy
| | - Ettore Marranconi
- Department of Pharmacy, Specialization School in Hospital Pharmacy, University of Pisa, Pisa, Italy
| | - Debora Carpanese
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Elena Berti
- Pharmacy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Marina Coppola
- Pharmacy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| |
Collapse
|
37
|
Bhagwat B, de Waal Malefyt R, Willingham A. Investigating combination benefit of PD1 and LAG3 co-blockade using an engineered cellular bioassay. Int Immunopharmacol 2023; 119:109566. [PMID: 37044037 DOI: 10.1016/j.intimp.2022.109566] [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: 08/26/2022] [Revised: 11/05/2022] [Accepted: 12/06/2022] [Indexed: 04/14/2023]
Abstract
LAG3 and PD1 are both immunomodulatory receptor that act by inhibiting activation of T cells, producing a more immunosuppressive environment. Even with the recent clinical success of PD1 and LAG3 co-blockade, signal transduction downstream of LAG3 remains largely unknown. We have leveraged an engineered Jurkat (T cell) and Raji (APC) co-culture system to assess simultaneous blockade of PD1 and LAG3 pathways using antibodies. RNA-Seq analysis of cell pellets individually treated with LAG3 or PD1 antibodies revealed modest immune activation however, 5-fold more genes were upregulated upon combination treatment. There were increases in costimulatory genes like CD28, CD5, CD6 as well as intracellular signaling molecules like LCP2 and ITK. Given the role of ERK in immune activation of T cells, pERK levels of Jurkat cells in the assay were evaluated. A very modest activation of pERK was observed with anti-LAG3 compared to anti-PD1 but a combination treatment resulted in prolonged ERK phosphorylation. Treatment of Jurkat cells with a commercial phosphatase inhibitor NSC87877 which can impact many phosphatases resulted in immune activation, measured by increased IL2 levels, only in the presence of LAG3. When NSC87877 was combined with the PD1 antibody, it could phenocopy combination benefit of PD1 and LAG3 blocking antibodies. CD28 has a recognized role in PD1 signaling but the impact on LAG3 signaling remains unknown. CD28 knockout in Jurkat cells affected overall IL-2 response of both LAG3 and PD1 antibody treatment but still retained combination benefit. Taken together this reductionist system highlights differences in downstream effects of LAG3 and PD1 blockade and we believe that the assay may have further utility to dissect convergence of both signaling pathways and augment studies in primary cells.
Collapse
Affiliation(s)
| | - Rene de Waal Malefyt
- Discovery Biologics, Merck & Co., Inc, South San Francisco, CA, USA; Synthekine, Inc., Menlo Park, CA, USA
| | | |
Collapse
|
38
|
Mortezaee K, Majidpoor J. Anti-PD-(L)1 therapy of non-small cell lung cancer-A summary of clinical trials and current progresses. Heliyon 2023; 9:e14566. [PMID: 36950599 PMCID: PMC10025922 DOI: 10.1016/j.heliyon.2023.e14566] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Background This review discusses the impact of mono or combination therapy of immune checkpoint inhibitor (ICI) therapy in non-small cell lung cancer (NSCLC) patients, comparing clinical outcomes and safety. Cancer subtype, tumor mutational burden (TMB), programmed death-ligand 1 (PD-L1) expression state and T cell infiltration (TIL) density are considered for interpretations. Besides, current progresses in the field of immunotherapy are discussed. Results Anti-PD-(L)1 is a safe and an effective strategy in patients with advanced/metastatic NSCLC. Clinical responses to nivolumab and pembrolizumab, in particular, are promising. The most desired clinical responses are for patients receiving combination of anti-PD-(L)1 or anti-PD-(L)1/anti-cytotoxic T lymphocyte associated antigen-4 (CTLA-4) with chemotherapy (taxane and platinum). PD-L1 expression state (PD-L1 ≥ 50%), patient performance state (PS: 0-1 ECOG scale) and effector T cell (Teff) immune signature considerably affect ICI responses. Higher ICI responses are also expected in TMB high but EGFR-/ALK- cancer patients. In regard with safety profile, adverse events (AEs) related to anti-PD-(L)1 are lower compared with that for platinum-based and docetaxel therapy. Toripalimab is the safest among various immunotherapy drugs. Bispecific antibodies against anti-PD-(L)1 with dominant signaling or alternative checkpoints in tumor microenvironment (TME) is the current focus in immunotherapy of cancers like NSCLC. Besides, the contribution of extracellular vesicles (EVs) to immune escape and their implication in cancer diagnosis and therapy is on the eye of current investigations. Conclusion Appropriate biomarker selection will improve therapy outcomes in ICI treated NSCLC patients, particularly in cases under combinatory ICI therapy. Application of bispecific antibodies and EV-based targeted therapy are effective novel strategies to improve therapeutic outcomes in cancer patients.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Corresponding author.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
39
|
Li LJ, Xuan JZ, Zheng HN. Correlation of 18F-FDG PET/CT metabolic parameters with the expression of immune biomarkers in the tumour microenvironment in lung adenocarcinoma. Clin Radiol 2023:S0009-9260(23)00075-2. [PMID: 36934052 DOI: 10.1016/j.crad.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 02/02/2023] [Accepted: 02/08/2023] [Indexed: 03/06/2023]
Abstract
AIM To explore the association between metabolic parameters evaluated by integrated 2-[18F]-fluoro-2-deoxy-d-glucose (FDG) positron-emission tomography (PET)/computed tomography (CT) and the expression of immune biomarkers in the tumour microenvironment in lung adenocarcinoma. MATERIALS AND METHODS This study included 134 patients. Metabolic parameters were obtained by PET/CT. Immunohistochemistry analysis was used for FOXP3-TILs (transcription factor forkhead box protein 3 tumour-infiltrating lymphocytes), CD8-TILs, CD4-TILs, CD68-TAMs (tumour-associated macrophages) and galectin-1 (Gal-1) tumour expression. RESULTS There were significant positive associations between FDG PET metabolic parameters and the median percentage of immune reactive areas (IRA%) covered by FOXP3-TILs and CD68-TAMs. Negative associations with the median IRA% covered by CD4-TILs and CD8-TILs were observed: maximal standardised uptake value (SUVmax), metabolic tumour volume (MTV), total lesion glycolysis (TLG), and IRA% for FOXP3-TILs (rho = 0.437, 0.400, 0.414; p<0.0001 for all parameters); SUVmax, MTV, TLG, and IRA% for CD68-TAMs (rho = 0.356, 0.355, 0.354; p<0.0001 for all parameters); SUVmax, MTV, TLG, and IRA% for CD4-TILs (rho = -0.164, -0.190, -0.191; p=0.059, 0.028, 0.027, respectively); SUVmax, MTV, TLG, and IRA% for CD8-TILs (rho = -0.305, -0.316, -0.322; p<0.0001 for all parameters). There were significant positive associations between tumour Gal-1 expression and the median IRA% covered by FOXP3-TILs and CD68-TAMs (rho = 0.379; p<0.0001; rho = 0.370; p<0.0001, respectively), and a significant negative association with the median IRA% covered by CD8-TILs (rho = -0.347; p<0.0001) was observed. Tumour stage (p=0.008), Gal-1 expression (p=0.008), and median IRA% covered by CD8-TILs (p=0.054) were independent risk factors for overall survival. CONCLUSION FDG PET may facilitate a comprehensive evaluation of the tumour microenvironment and predict response to immunotherapy.
Collapse
Affiliation(s)
- L-J Li
- Department of Radiation Oncology, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, Liaoning 116011, People's Republic of China
| | - J-Z Xuan
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, Liaoning 116011, People's Republic of China
| | - H-N Zheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, Liaoning 116011, People's Republic of China.
| |
Collapse
|
40
|
Abu Qubo A, Numan J, Snijder J, Padilla M, Austin JH, Capaccione KM, Pernia M, Bustamante J, O'Connor T, Salvatore MM. Idiopathic pulmonary fibrosis and lung cancer: future directions and challenges. Breathe (Sheff) 2022; 18:220147. [PMID: 36865932 PMCID: PMC9973524 DOI: 10.1183/20734735.0147-2022] [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: 06/06/2022] [Accepted: 09/14/2022] [Indexed: 01/11/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease of pulmonary scarring. New treatments slow disease progression and allow pulmonary fibrosis patients to live longer. Persistent pulmonary fibrosis increases a patient's risk of developing lung cancer. Lung cancer in patients with IPF differs from cancers that develop in the non-fibrotic lung. Peripherally located adenocarcinoma is the most frequent cell type in smokers who develop lung cancer, while squamous cell carcinoma is the most frequent in pulmonary fibrosis. Increased fibroblast foci in IPF are associated with more aggressive cancer behaviour and shorter doubling times. Treatment of lung cancer in fibrosis is challenging because of the risk of inducing an exacerbation of fibrosis. In order to improve patient outcomes, modifications of current lung cancer screening guidelines in patients with pulmonary fibrosis will be necessary to avoid delays in treatment. 2-fluoro-2-deoxy-d-glucose (FDG) positron emission tomography (PET) computed tomography (CT) imaging can help identify cancer earlier and more reliably than CT alone. Increased use of wedge resections, proton therapy and immunotherapy may increase survival by decreasing the risk of exacerbation, but further research will be necessary.
Collapse
Affiliation(s)
- Ahmad Abu Qubo
- Department of Pathology, Faculty of Medicine, Hashemite University, Zarqa, Jordan
| | - Jamil Numan
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Juan Snijder
- Department of Pediatrics, Einstein Medical Center, Philadelphia, PA, USA
| | - Maria Padilla
- Department of Pulmonary Medicine, Mount Sinai, New York, NY, USA
| | - John H.M. Austin
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | | | - Monica Pernia
- Department of Medicine, Metropolitan Hospital, New York, NY, USA
| | - Jean Bustamante
- Department of Oncology, West Virginia University, Morgantown, WV, USA
| | - Timothy O'Connor
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Mary M. Salvatore
- Department of Radiology, Columbia University Medical Center, New York, NY, USA,Corresponding author: Mary M. Salvatore ()
| |
Collapse
|
41
|
Zhang XT, Ge N, Xiang ZJ, Liu T. Immune checkpoint inhibitor-related adverse cardiac events in patients with lung cancer: a systematic review and meta-analysis. Cancer Cell Int 2022; 22:363. [PMCID: PMC9675058 DOI: 10.1186/s12935-022-02760-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/25/2022] [Indexed: 11/21/2022] Open
Abstract
Background Although people are more and more aware of the cardiotoxicity caused by immune checkpoint inhibitors (ICIs) in the treatment of lung cancer, its incidence rate has not been systematically analyzed. This study aims to evaluate the incidence of cardiotoxicity related to the ICI therapies for lung cancer, so as to enhance clinicians' attention to cardiotoxicity, implement proper prevention and intervention for high-risk patients, and minimize the risk of cardiac dysfunction during and after completion of therapy. Methods We conducted a systematic literature search for relevant publications in PubMed and Scopus from inception to 19 April 2022. Pooled incidence and risk ratios with 95% confidence intervals (95% CIs) for cardiotoxicity events were calculated. Results A total of 37 studies covering 38 trials, including 14,342 patients, were identified. The pooled risk ratios of incidence of any cardiac AEs were 1.944 [95% CI 0.8–4.725] (Single ICI versus chemotherapy), 1.677 [95% CI 1.065–2.64] (Single ICI plus chemotherapy versus chemotherapy), and 0.478 [95% CI 0.127–1.798] (Single ICI versus Dual ICI). The incidence of myocarditis and arrhythmia were 0.003[95%CI 0.002–0.006] and 0.014[95%CI 0–0.037], respectively. Conclusion Single ICI did not increase the risk of cardiotoxicity compared with chemotherapy, and single ICI plus chemotherapy increased the risk of cardiotoxicity by 67% compared with chemotherapy alone. Combination immunotherapy did not increase the risk of cardiotoxicity compared with single ICI. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02760-2.
Collapse
Affiliation(s)
- Xiao-Tong Zhang
- grid.506261.60000 0001 0706 7839Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730 China
| | - Nan Ge
- grid.506261.60000 0001 0706 7839Department of Geriatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zi-Jian Xiang
- Beijing Zhiyun Data Technology Co. LTD, Beijing, China
| | - Tao Liu
- grid.506261.60000 0001 0706 7839Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730 China
| |
Collapse
|
42
|
He H, Jin Z, Dai J, Wang H, Sun J, Xu D. Computed tomography‐based radiomics prediction of
CTLA4
expression and prognosis in clear cell renal cell carcinoma. Cancer Med 2022; 12:7627-7638. [PMID: 36397666 PMCID: PMC10067074 DOI: 10.1002/cam4.5449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/31/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES To predict CTLA4 expression levels and prognosis of clear cell renal cell carcinoma (ccRCC) by constructing a computed tomography-based radiomics model and establishing a nomogram using clinicopathologic factors. METHODS The clinicopathologic parameters and genomic data were extracted from 493 ccRCC cases of the Cancer Genome Atlas (TCGA)-KIRC database. Univariate and multivariate Cox regression and Kaplan-Meier analysis were performed for prognosis analysis. Cibersortx was applied to evaluate the immune cell composition. Radiomic features were extracted from the TCGA/the Cancer Imaging Archive (TCIA) (n = 102) datasets. The support vector machine (SVM) was employed to establish the radiomics signature for predicting CTLA4 expression. Receiver operating characteristic curve (ROC), decision curve analysis (DCA), and precision-recall curve were utilized to assess the predictive performance of the radiomics signature. Correlations between radiomics score (RS) and selected features were also evaluated. An RS-based nomogram was constructed to predict prognosis. RESULTS CTLA4 was significantly overexpressed in ccRCC tissues and was related to lower overall survival. A higher CTLA4 expression was independently linked to the poor prognosis (HR = 1.458, 95% CI 1.13-1.881, p = 0.004). The radiomics model for the prediction of CTLA4 expression levels (AUC = 0.769 in the training set, AUC = 0.724 in the validation set) was established using seven radiomic features. A significant elevation in infiltrating M2 macrophages was observed in the RS high group (p < 0.001). The predictive efficiencies of the RS-based nomogram measured by AUC were 0.826 at 12 months, 0.805 at 36 months, and 0.76 at 60 months. CONCLUSIONS CTLA4 mRNA expression status in ccRCC could be predicted noninvasively using a radiomics model based on nephrographic phase contrast-enhanced CT images. The nomogram established by combining RS and clinicopathologic factors could predict overall survival for ccRCC patients. Our findings may help stratify prognosis of ccRCC patients and identify those who may respond best to ICI-based treatments.
Collapse
Affiliation(s)
- Hongchao He
- Department of Urology Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Zhijia Jin
- Department of Radiology Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Jun Dai
- Department of Urology Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Haofei Wang
- Department of Urology Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| | - Jianqi Sun
- School of Biomedical Engineering Shanghai Jiaotong University Shanghai China
| | - Danfeng Xu
- Department of Urology Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine Shanghai China
| |
Collapse
|
43
|
Ling SP, Ming LC, Dhaliwal JS, Gupta M, Ardianto C, Goh KW, Hussain Z, Shafqat N. Role of Immunotherapy in the Treatment of Cancer: A Systematic Review. Cancers (Basel) 2022; 14:5205. [PMID: 36358624 PMCID: PMC9655090 DOI: 10.3390/cancers14215205] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 01/18/2024] Open
Abstract
Tremendous progress has been made in cancer research over the years, and, as a result, immunotherapy has emerged as an important therapy for the treatment of cancer, either as a stand-alone treatment or in conjunction with other cancer therapies. Immunotherapy has demonstrated encouraging outcomes and offers a viable strategy for not only enhancing the quality of life but also dramatically boosting the overall survival rate of cancer patients. The objective of this systematic review was to assess the efficacy of immunotherapy in the treatment of cancer. Databases such as PubMed and Science Direct were searched from their inception until September 2021, using the following keywords: cancer immunotherapy, cancer recurrence, cancer treatment options, and cancer therapies. The systematic review was conducted in accordance with the PRISMA protocol. There were a total of 599 articles; however, after applying the inclusion and exclusion criteria, the final review ended up with 34 publications. In conclusion, the studies have demonstrated that immunotherapy is a viable alternative treatment option for patients with recurrent or metastatic cancer, since the overall survival rate and progression-free survival rate were shown to be successful.
Collapse
Affiliation(s)
- Sia Pei Ling
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Long Chiau Ming
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Jagjit Singh Dhaliwal
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Madhu Gupta
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai 71800, Malaysia
| | - Zahid Hussain
- Faculty of Health, University of Canberra, Bruce, ACT 2617, Australia
| | - Naeem Shafqat
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| |
Collapse
|
44
|
Pang A, Jiali L, Ng A, Cheng J, Wang M, Ng YS, Yao Y, Chun M, Ho F, Tey J. Use of the Cancer and Aging Research Group Predictive Model for Chemotherapy-Related Toxic Effects in a Multiethnic, Older Adult Asian Population. JAMA Netw Open 2022; 5:e2237196. [PMID: 36255721 PMCID: PMC9579905 DOI: 10.1001/jamanetworkopen.2022.37196] [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] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE The Cancer and Aging Research Group (CARG) prediction model for chemotherapy-related toxic effects has been developed but not yet validated in older Asian adults. In view of differences in drug metabolism and toxic effect reporting in the Asian population, the ability of this tool to guide the cancer treatment decision-making process in older Asian adults needs to be assessed. OBJECTIVE To examine the validity of the CARG predictive model in a multiethnic Asian cohort of older adults. DESIGN, SETTING, AND PARTICIPANTS In this prognostic study, patients of various Asian ethnicities 70 years or older with a solid tumor diagnosis receiving chemotherapy at the National University Cancer Institute, Singapore, were accrued from June 1, 2017, to January 1, 2019. Their risks of chemotherapy-related toxic effects were calculated using the CARG tool. A geriatric assessment was performed, and the treating oncologist (blinded to the CARG scores) was asked to give an estimated likelihood of toxic effects (low, medium, or high). Chemotherapy-related toxic effects were recorded during each clinic visit. Validation of the prediction model was performed by calculating the area under the receiver operating characteristic curve. Multivariate analyses were performed to identify variables in other domains in the geriatric assessment predicting for severe toxic effects. MAIN OUTCOMES AND MEASURES Grade 3 to 5 toxic effects and hospitalization. RESULTS The study included 200 patients (median age, 74 years [range, 70-89 years]; 110 [55.0%] male; 177 [88.5%] Chinese, 17 [8.5%] Malay, 4 [2.0%] Indian, and 2 [1.0%] other ethnicities [according to Singapore's national system of race classification]). A total of 137 patients (68.5%) experienced grade 3 to 5 toxic effects, and 131 (65.5%) required hospitalization. The area under the receiver operating characteristic curve for the CARG chemotoxicity prediction model was 0.74 (95% CI, 0.67-0.82), retaining good discrimination in the study population. CONCLUSIONS AND RELEVANCE This prognostic study conducted in a multiethnic Asian cohort of older adults supports the validity of the CARG predictive model in this population, predicting which older adults are at risk of chemotherapy-related toxic effects.
Collapse
Affiliation(s)
- Angela Pang
- Department of Haematology Oncology, National University Cancer Institute, Singapore
| | - Low Jiali
- Department of Haematology Oncology, National University Cancer Institute, Singapore
| | - Alex Ng
- Yong Loo Lin School of Medicine, National University of Singapore
| | - Joseph Cheng
- Yong Loo Lin School of Medicine, National University of Singapore
| | - Meng Wang
- Department of Medicine, National University Hospital, Singapore
| | - Yean Shin Ng
- Department of Haematology Oncology, National University Cancer Institute, Singapore
| | - Yao Yao
- Department of Pharmacy, National University Hospital, Singapore
| | - Meiling Chun
- Department of Surgery, Ng Teng Fong General Hospital, Singapore
| | - Francis Ho
- Department of Radiation Oncology, National University Cancer Institute, Singapore
| | - Jeremy Tey
- Department of Radiation Oncology, National University Cancer Institute, Singapore
| |
Collapse
|
45
|
Lian J, Zhang G, Zhang Y, Liu H, Zhang J, Nan P, Tian W. PD-L1 and HER2 expression in gastric adenocarcinoma and their prognostic significance. Dig Liver Dis 2022; 54:1419-1427. [PMID: 35123909 DOI: 10.1016/j.dld.2022.01.128] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 01/10/2022] [Accepted: 01/16/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND The upregulation of programmed death-ligand 1 (PD-L1) and epidermal growth factor receptor 2 (HER2) may play a role in gastric adenocarcinoma (GAC). AIM To study PD-L1 and HER-2 expression and prognosis in GAC. METHODS PD-L1 and HER2 expression was determined in tumor tissues of 75 patients with GAC. The correlations between PD-L1, HER2 expression, and clinicopathological factors were analyzed. RESULTS The positive expression rate for PD-L1 was 57.3% (43/75) and the HER2 over-expression rate was 17.3% (13/75). PD-L1 expression negatively correlated with the grade of GAC differentiation (r =-0.26, P<0.05). Approximately 85% of HER2-positive GACs were found to be PD-L1-positive and PD-L1 expression positively correlated with HER2 overexpression. The TNM stage and combined HER2 and PD-L1 expression were independent prognostic factors affecting the survival of patients with GAC. The median overall survival and recurrence-free survival of groups I (HER2 overexpression and PD-L1 positive), II (HER2 overexpression and PD-L1 negative), III (No HER2 overexpression and PD-L1 positive) and IV (No HER2 overexpression and PD-L1 negative) were (47 (17-77), 15 (0-44), 81 (62-101), and 78 (60-98) months, respectively. CONCLUSION PD-L1 expression is upregulated in more than half of patients with GAC. Anti-PD-L1 treatment combined with anti-HER2 therapy may benefit patients with locally advanced GAC with HER2 overexpression.
Collapse
Affiliation(s)
- Jie Lian
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an 710061, China
| | - Guanjun Zhang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an 710061, China.
| | - Yun Zhang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an 710061, China
| | - Heng Liu
- Department of Radiology, Affiliated Hospital of Zunyi Medical University, Medical Imaging Center of Guizhou Province, Zunyi, Guizhou 563003, P.R. China
| | - Jiaojiao Zhang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an 710061, China
| | - Pengfei Nan
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an 710061, China
| | - Wei Tian
- Department of Pathology, No. 215 Hospital of Shanxi Nuclear Industry, Xianyang 712000, China
| |
Collapse
|
46
|
Kim EJ, Cho YH, Kim DH, Ko DH, Do EJ, Kim SY, Kim YM, Jung JS, Kang Y, Ji W, Choi MG, Lee JC, Rho JK, Choi CM. A Phase I/IIa Randomized Trial Evaluating the Safety and Efficacy of SNK01 Plus Pembrolizumab in Patients with Stage IV Non-Small Cell Lung Cancer. Cancer Res Treat 2022; 54:1005-1016. [PMID: 34856706 PMCID: PMC9582480 DOI: 10.4143/crt.2021.986] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/29/2021] [Indexed: 12/03/2022] Open
Abstract
PURPOSE The aim of this study is to evaluate the safety and efficacy of ex vivo activated and expanded natural killer (NK) cell therapy (SNK01) plus pembrolizumab in a randomized phase I/IIa clinical trial. MATERIALS AND METHODS Overall, 18 patients with advanced non-small cell lung cancer (NSCLC) and a programmed death ligand 1 tumor proportion score of 1% or greater who had a history of failed frontline platinum-based therapy were randomized (2:1) to receive pembrolizumab every 3 weeks +/- 6 weekly infusions of SNK01 at either 2×109 or 4×109 cells per infusion (pembrolizumab monotherapy vs. SNK01 combination). The primary endpoint was safety, whereas the secondary endpoints were the objective response rate (ORR), progression-free survival (PFS), overall survival, and quality of life. RESULTS Since no dose-limiting toxicity was observed, the maximum tolerated dose was determined as SNK01 4×109 cells/dose. The safety data did not show any new safety signals when SNK01 was combined with pembrolizumab. The ORR and the 1-year survival rate in the NK combination group were higher than those in patients who underwent pembrolizumab monotherapy (ORR, 41.7% vs. 0%; 1-year survival rate, 66.7% vs. 50.0%). Furthermore, the median PFS was higher in the SNK01 combination group (6.2 months vs. 1.6 months, p=0.001). CONCLUSION Based on the findings of this study, the NK cell combination therapy may consider as a safe treatment method for stage IV NSCLC patients who had a history of failed platinum-based therapy without an increase in adverse events.
Collapse
Affiliation(s)
- Eo Jin Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
- Division of Hematology/Oncology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Yong-Hee Cho
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
- Data Convergence Drug Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon,
Korea
| | - Dong Ha Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Dae-Hyun Ko
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Eun-Ju Do
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Sang-Yeob Kim
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | | | | | | | - Wonjun Ji
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Myeong Geun Choi
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Jae Cheol Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Jin Kyung Rho
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Chang-Min Choi
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| |
Collapse
|
47
|
Mortezaee K, Majidpoor J. Extracellular vesicle-based checkpoint regulation and immune state in cancer. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:225. [PMID: 36175741 DOI: 10.1007/s12032-022-01837-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/28/2022] [Indexed: 10/14/2022]
Abstract
Tumor cells exploit several mechanisms for hijacking an immunosuppressive tumor ecosystem in order to evade immune surveillance and to progress toward metastasis. Equipment of extracellular vesicles (EVs) with checkpoints is an example of cancer control over anti-tumor responses from immune system. Programmed death-ligand 1 (PD-L1) is a checkpoint highly expressed in a tumor at progressive stage. Interactions between PD-L1 with its receptor programmed death-1 receptor (PD-1) expressed on T cells will block the effector function of CD8+ T cells, known as one of the most important defensive cells against cancer. Evaluation of circulatory exosomal PD-L1 can be a prognostic biomarker in tumor diagnosis and responses to the immune checkpoint inhibitor (ICI) therapy, and can be considered as a tool in clinical practice for exploiting personalized therapy. Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) is also a checkpoint that its engagement with CD80/CD86 expressed on antigen-presenting cells (APCs), such as dendritic cells (DCs) hamper the priming phase of CD4+ and CD8+ T cells. Harvesting EVs from tumor and their modification with desired anti-checkpoint antibodies can be a promising strategy in cancer immunotherapy. The aim of this review is to discuss about EV roles in checkpoint regulation, cancer diagnosis and ICI responses, and to survey possible application of such vesicles in cancer immunotherapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
48
|
Lu L, Zheng D, Qu J, Zhuang Y, Peng J, Lan S, Zhang S, Huang F. METTL16 predicts a favorable outcome and primes antitumor immunity in pancreatic ductal adenocarcinoma. Front Cell Dev Biol 2022; 10:759020. [PMID: 36158188 PMCID: PMC9500295 DOI: 10.3389/fcell.2022.759020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic carcinogenesis is a complicated and multi-step process. It is substantially assisted by N6-methyladenosine (m6A) RNA modification, especially when mutations of driver genes (KRAS, TP53, CDKN2A, and SMAD4) occur. However, the underlying mechanism remains obscure. In this research, we identified m6A regulators as potential biomarkers when mutations of driver genes occur, and investigated the role of these m6A candidates in pancreatic ductal adenocarcinoma (PDA). We first estimated the abnormal expression patterns of potential m6A regulators when all the driver genes are mutated, using The Cancer Genome Atlas and Gene Expression Omnibus databases. METTL16, an m6A“writer,” was chosen as a unique candidate of PDA, owing to its markedly differential expression under mutations of all driver genes (KRAS, TP53, CDKN2A, and SMAD4) and its favorable prognostic value. Moreover, METTL16 was under-expressed in PDA tissues and cell lines. Consistently, gain- and loss-of-function experiments indicated that it had a tumor suppressor role in vitro and in vivo. Further, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed that METTL16 may have an effect on the tumor microenvironment. Notably, a markedly positive association between METTL16 expression and infiltration of B cells and CD8+ T cells was observed according to the CIBERSORT and TIMER databases. Enhanced expression of immune checkpoints and cytokines was elicited in patients with over-expression of METTL16. Notably, decreased expression of PD-L1 was observed when upregulation of METTL16 expression occurred in MIA PaCa-2 cells, while increased expression of PD-L1 existed when downregulation of METTL16 happened in HPAF-II cells. Collectively, these findings highlight the prognostic value of METTL16, and indicate that it is a potential immunotherapy target that could be used to regulate the tumor microenvironment and promote antitumor immunity in PDA.
Collapse
Affiliation(s)
- Liting Lu
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dandan Zheng
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Junchi Qu
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanyan Zhuang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Juanfei Peng
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Sihua Lan
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shineng Zhang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Shineng Zhang, ; Fengting Huang,
| | - Fengting Huang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Shineng Zhang, ; Fengting Huang,
| |
Collapse
|
49
|
Khanniche A, Yang Y, Zhang J, Liu S, Xia L, Duan H, Yao Y, Zhao B, Zhao GP, Hu C, Wang Y, Lu S. Early-like differentiation status of systemic PD-1 +CD8 + T cells predicts PD-1 blockade outcome in non-small cell lung cancer. Clin Transl Immunology 2022; 11:e1406. [PMID: 35910005 PMCID: PMC9327560 DOI: 10.1002/cti2.1406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 05/24/2022] [Accepted: 07/08/2022] [Indexed: 12/02/2022] Open
Abstract
Objectives Despite remarkable advances in the treatment of non‐small cell lung cancer (NSCLC) with anti‐programmed death (PD)‐1 therapy; only a fraction of patients derives durable clinical benefit. In this study, we investigated whether the differentiation status of systemic CD8+ T cells predicts the outcome of PD‐1 blockade in NSCLC. Methods We carried out a prospective study on a total of 77 NSCLC patients receiving anti‐PD‐1 blockers, among which 47 patients were assigned as a discovery cohort and 30 patients as a validation cohort. Peripheral blood samples were obtained at baseline and upon multiple therapy cycles and analyzed by multi‐parameter flow cytometry. Results We found that a higher baseline ratio of PD‐1+ early effector memory CD8+ T cells (CD28+CD27−CD45RO+, TEEM) to PD‐1+ effector CD8+ T cells (CD28−CD27−CD45RO−, TE) delineated responders to PD‐1 blockade from progressors and was associated with prolonged progression‐free survival (PFS) and durable clinical benefit. Moreover, PD‐1+CD8 TEEM cells exhibited early responses after anti‐PD‐1 therapy and was the major fraction of cycling PD‐1+Ki67+CD8+ T cells to expand specifically with positive impact on PFS. Conclusion These findings provide insights into how the baseline differentiation status of the peripheral immune system determines responses to PD‐1‐targeted therapies.
Collapse
Affiliation(s)
- Asma Khanniche
- Department of Immunology and Microbiology, Shanghai Institute of Immunology Shanghai Jiao Tong University School of Medicine Shanghai China.,Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen China
| | - Yi Yang
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital Shanghai Jiao Tong University Shanghai China
| | - Jie Zhang
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital Shanghai Jiao Tong University Shanghai China
| | - Shiqing Liu
- Department of Respiratory Medicine, Xiangya Lung Cancer Center, Xiangya Hospital Central South University Changsha China
| | - Liliang Xia
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital Shanghai Jiao Tong University Shanghai China
| | - Huangqi Duan
- Department of Immunology and Microbiology, Shanghai Institute of Immunology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yaxian Yao
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital Shanghai Jiao Tong University Shanghai China
| | - Bingrong Zhao
- Department of Respiratory Medicine, Xiangya Lung Cancer Center, Xiangya Hospital Central South University Changsha China
| | - Guo-Ping Zhao
- Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen China
| | - Chengping Hu
- Department of Respiratory Medicine, Xiangya Lung Cancer Center, Xiangya Hospital Central South University Changsha China
| | - Ying Wang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Shun Lu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital Shanghai Jiao Tong University Shanghai China
| |
Collapse
|
50
|
Clinical Efficacy and Safety Analysis of PD-1/PD-L1 Inhibitor vs. Chemotherapy in the Treatment of Advanced Non-Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9500319. [PMID: 36033563 PMCID: PMC9402310 DOI: 10.1155/2022/9500319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 01/05/2023]
Abstract
Objective To systematically evaluate the efficacy and safety of pembrolizumab (PD-1/PD-L inhibitor) and adjuvant chemotherapy to treat NSCLC and provide evidence-based reference for clinical use. Methods By searching the Cochrane Library, EMBASE, PubMed, and Web of Science, according to the inclusion criteria, literature selection, data extraction, and quality evaluation were carried out for the included literature. The I 2 test was used to evaluate heterogeneity between studies, and the meta-analysis was performed using RevMan 5.3 software provided by Cochrane. Results Finally, 14 relevant documents meeting the standards were included. It is a statistical difference in one-year survival rate [OR = 1.50, 95% CI (1.28, 1.76), P < 0.00001, I 2 = 0%, Z = 4.99]; overall response rate[OR =1.57, 95% CI (1.29, 1.90), P < 0.00001, I 2 = 0%, Z = 4.58]; progression-free survival [OR = 2.99, 95% CI (2.29, 3.91), P < 0.00001, I 2 = 26%, Z = 8.00]; and overall survival [OR = 1.38, 95% CI (1.07, 1.78), P = 0.01, I 2 = 46%, Z = 2.50] and reduces the incidence of adverse drug reactions [OR = 2.54, 95% CI (1.99, 3.25), P < 0.00001, I 2 = 69%, Z = 7.43]. Conclusion Pembrolizumab adjuvant chemotherapy is effective in the treatment of advanced NSCLC, but attention should be paid to the occurrence of adverse reactions in clinical. Due to the limitations of the methodology included in the study, this conclusion required more validation of large-sample RCT.
Collapse
|