1
|
Nishioka N, Naito T, Sugino T, Muramatsu K, Nishihara S, Urashima H, Mamesaya N, Kobayashi H, Omori S, Ko R, Wakuda K, Ono A, Kenmotsu H, Murakami H, Takahashi T. Desensitizing Effect of Intra-Tumoral GDF-15 on Immunotherapy in Patients With Advanced Non-Small Cell Lung Cancer. Thorac Cancer 2025; 16:e70089. [PMID: 40396532 PMCID: PMC12093252 DOI: 10.1111/1759-7714.70089] [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: 04/10/2025] [Revised: 05/02/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Serum growth/differentiation factor 15 (GDF-15) suppresses anti-tumor immunity and predicts prognosis in several malignancies. Elevated GDF-15 levels are linked to cancer cachexia, characterized by weight loss and systemic inflammation, adversely affecting patient outcomes and therapy response. However, serum GDF-15 is not always derived from tumor tissues but also from multiple organs. Therefore, we evaluated whether intra-tumoral GDF-15 could be used as a biomarker for immunotherapy and its potential association with cancer cachexia. METHOD We retrospectively evaluated patients with advanced non-small cell lung cancer (NSCLC) who underwent treatment with programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) inhibitors at the Shizuoka Cancer Center between 2017 and 2021. Patients with histologically confirmed NSCLC (stage III-IV or postoperative recurrence) who had undergone biopsy or surgery within 6 months prior to initiating immunotherapy were included. Expression of tumor-derived GDF-15 was evaluated using immunohistochemical staining of archival biopsy and surgical specimens. We analyzed the correlation between intra-tumoral GDF-15 expression and the incidence of cancer cachexia, as well as its impact on progression-free survival (PFS) and overall survival (OS). RESULT In 6 of 35 cases, tumor cells highly expressed GDF-15. Patients with high intra-tumoral GDF-15 expression had a higher incidence of cancer cachexia (100% vs. 41.4%, p < 0.05), shorter PFS (3.4 vs. 13.4 months, p < 0.05), and shorter OS (9.5 vs. 26.5 months, p < 0.05) than those with low intra-tumoral GDF-15 expression. CONCLUSION Intra-tumoral GDF-15 expression may predict the presence of cancer cachexia and the efficacy of PD-1/PD-L1 inhibitors in patients with advanced non-small cell lung cancer.
Collapse
Affiliation(s)
- Naoya Nishioka
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
- Department of Pulmonary MedicineGraduate School of Medical Science, Kyoto Prefectural University of MedicineKyotoJapan
| | - Tateaki Naito
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
- Division of Cancer Supportive Care CenterShizuoka Cancer CenterShizuokaJapan
| | - Takashi Sugino
- Division of PathologyShizuoka Cancer CenterShizuokaJapan
| | - Koji Muramatsu
- Division of PathologyShizuoka Cancer CenterShizuokaJapan
| | - Shigeki Nishihara
- Department of CNS ResearchOtsuka Pharmaceutical Co., Ltd.TokushimaJapan
| | - Hiroki Urashima
- Osaka Research Center for Drug Discovery, Department of Research Management, Otsuka Pharmaceutical Co., Ltd.OsakaJapan
| | - Nobuaki Mamesaya
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Haruki Kobayashi
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Shota Omori
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Ryo Ko
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Kazushige Wakuda
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | - Akira Ono
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | | | | | | |
Collapse
|
2
|
Messana G, Bortolotto C, Thulasi Seetha S, Marrocco A, Pairazzi C, Sanvito F, Brero F, Robustelli Test A, Cabini RF, Lascialfari A, Zacà D, Stella GM, Agustoni F, Saddi J, Filippi AR, Preda L. Non-invasive PD-L1 stratification in non-small cell lung cancer using dynamic contrast-enhanced MRI. Eur Radiol 2025:10.1007/s00330-025-11524-1. [PMID: 40146425 DOI: 10.1007/s00330-025-11524-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/23/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025]
Abstract
OBJECTIVES This study aimed to assess whether pharmacokinetic parameters derived from DCE-MRI can stratify Programmed Death-Ligand 1 (PD-L1) expression in NSCLC. The secondary aim was to identify a suitable pharmacokinetic model configuration for anisotropic temporally-spaced DCE-MRI sequences, considering Tofts variants, population-averaged arterial input functions (AIF), and bolus arrival time (BAT) estimation methods. MATERIALS AND METHODS From April 2021 to May 2023, patients with locally advanced non-small cell lung cancer (NSCLC) were prospectively enrolled. Tumors were categorized based on: PD-L1 absence/presence (threshold 1%) and hyperexpression/hypoexpression (threshold 50%). Pharmacokinetic parameters were extracted using several candidate configurations; fit quality was evaluated using coefficient of determination (R²). Mann-Whitney U-test and ROC-AUC were used to assess correlation with PD-L1 for the best-fit configuration. RESULTS Thirty-eight patients (mean age 68 ± 9 years, 28 men) were included. PD-L1 expression was present in 25 patients (66%) and absent in 13 (34%). PD-L1 was hyperexpressed in 13 (34%) patients and hypoexpressed in 25 (66%). Voxel-wise pharmacokinetic parameters were extracted using the best-fit configuration-extended Tofts model (ETM) with Georgiou AIF and Peak-Gradient (PG) BAT estimation (R2 = 0.79). Ktrans median (0.25 vs. 0.12 min-¹, p = 0.02), Ktrans standard deviation (0.32 vs. 0.23 min-¹, p = 0.01) and Kep median (1.09 vs. 0.59 min-¹, p = 0.02) were significantly higher in PD-L1 < 50% group (ROC-AUC 0.71-0.76). CONCLUSION DCE-MRI pharmacokinetic parameters could stratify PD-L1 hypo/hyperexpression in NSCLC. The ETM with PG BAT estimation method and Georgiou AIF was the best-performing pharmacokinetic configuration. KEY POINTS Question Could Dynamic Contrast-Enhanced (DCE) MRI offer a safe and non-invasive way to assess Programmed Death-Ligand 1 (PD-L1) expression? Findings Quantitative DCE-MRI parameters Ktrans (the volume transfer rate) and Kep (the efflux rate constant) show potential for distinguishing PD-L1 hyperexpression from hypoexpression. Clinical relevance Preliminary results suggest that DCE-MRI could be a safe method to stratify PD-L1 hypo/hyperexpression in non-small cell lung cancer, potentially optimizing treatment decisions, given the high cost of immunotherapy.
Collapse
Affiliation(s)
- Gaia Messana
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Chandra Bortolotto
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
- Radiology Institute, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Sithin Thulasi Seetha
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy.
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy.
| | - Alessandra Marrocco
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Carlotta Pairazzi
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Francesco Sanvito
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Francesca Brero
- Department of Physics, University of Pavia, Pavia, Italy
- Istituto Nazionale Di Fisica Nucleare, Sezione Di Pavia, Pavia, Italy
| | - Agnese Robustelli Test
- Department of Physics, University of Pavia, Pavia, Italy
- Istituto Nazionale Di Fisica Nucleare, Sezione Di Pavia, Pavia, Italy
| | | | - Alessandro Lascialfari
- Department of Physics, University of Pavia, Pavia, Italy
- Istituto Nazionale Di Fisica Nucleare, Sezione Di Pavia, Pavia, Italy
| | | | - Giulia Maria Stella
- Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
| | - Francesco Agustoni
- Department of Medical Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Jessica Saddi
- Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Andrea Riccardo Filippi
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
- Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Radiation Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology, University of Milan, Milan, Italy
| | - Lorenzo Preda
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
- Radiology Institute, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
3
|
Lachkar S, Gervereau D, Loïc P, De Marchi M, Morisse H, Dantoing E, Piton N, Thiberville L, Salaün M, Guisier F. Correlation of programmed death-ligand 1 expression in tumour cells between diagnostic small biopsies performed by radial EBUS and surgical specimens of peripheral lung cancer. BMJ Open Respir Res 2024; 11:e002312. [PMID: 39414327 PMCID: PMC11481116 DOI: 10.1136/bmjresp-2024-002312] [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: 01/13/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Expression of programmed death-ligand 1 (PD-L1) in tumour cells (TCs) is predictive of immunotherapy efficacy in non-small cell lung cancer (NSCLC). Small biopsy samples collected by bronchoscopy are often used to diagnose peripheral lung cancer. It is questionable whether these small samples from radial endobronchial ultrasonography (r-EBUS) procedures are representative of PD-L1 expression in TCs. METHODS We retrieved data of consecutive patients who had surgery for NSCLC and previous r-EBUS biopsy sampling, from 2017 to 2019 in our centre. PD-L1 expression in tumour cells was categorised as <1%, 1%-49% and ≥50%. PD-L1 expression was compared between r-EBUS samples and surgical specimens. RESULTS Among 1026 patients who had r-EBUS, 521 had a diagnosis of lung cancer on r-EBUS sample. PD-L1 testing was indicated in 356 cases and results were considered contributive in 325 cases (91%). 82 patients with PD-L1 expression in r-EBUS samples had subsequent surgical resection of the nodule and were included in the study. PD-L1 expression was identical between r-EBUS samples and surgical specimens in 67% of cases, with kappa 0.44 (p<0.001). 82% of patients with PD-L1≥50% in surgical specimens were identified in r-EBUS samples. Nonetheless, 31% of patients with no PD-L1 expression in r-EBUS samples had some expression in surgical specimens. CONCLUSION Small samples obtained by r-EBUS are adequate for assessment of PD-L1 expression in tumour cells, with moderate concordance compared to surgical specimens. Reassessment of PD-L1 expression in larger samples may be useful to guide therapy in patients with no PD-L1 expression in r-EBUS samples.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Mathieu Salaün
- Pulmonology, CHU de Rouen, Rouen, France
- Department of Pneumology and Inserm CIC-CRB 1404, Normandie Univ, UNIROUEN, LITIS Lab QuantIF team EA4108, CHU Rouen, Rouen, France
| | - Florian Guisier
- Department of Pneumology and Inserm CIC-CRB 1404, Normandie Univ, UNIROUEN, LITIS Lab QuantIF team EA4108, CHU Rouen, Rouen, France
- CHU de Rouen, Rouen, France
| |
Collapse
|
4
|
Sarova P, Mosleh B, Zehetmayer S, Oberndorfer F, Widder J, Prosch H, Aigner C, Idzko M, Hoda MA, Gompelmann D. PD-L1 expression in patients with non-small-cell lung cancer is associated with sex and genetic alterations: A retrospective study within the Caucasian population. Thorac Cancer 2024; 15:1598-1606. [PMID: 38860475 PMCID: PMC11246784 DOI: 10.1111/1759-7714.15336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/21/2024] [Accepted: 05/01/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Programmed cell death-ligand 1 (PD-L1) expression is a well-established biomarker for predicting responses to immune checkpoint inhibitors and certain targeted therapies. As a result, treatment strategies for patients vary based on their PD-L1 expression status. Understanding the clinical features of patients with distinct PD-L1 levels is crucial for personalized treatment approaches. METHODS Demographic and clinicopathological characteristics of 227 patients (54% male, mean age 67 ± 9.9 years) newly diagnosed with non-small-cell lung cancer (NSCLC) between April 2020 and December 2022 were retrospectively compared among three groups based on the PD-L1 expression: PD-L1 Tumor Proportion Score (TPS) negative, 1-50%, and ≥50%. Logistic regression analysis was performed to evaluate predictors for high PD-L1 expression ≥50%. RESULTS PD-L1 expression levels were distributed as follows: negative in 29% of patients, between 1% and 50% in 41%, and greater than 50% (high) in 29%. In comparison to negative PD-L1 expression, low and high PD-L1 expression was associated with female sex (32.9% vs. 52.7% vs. 50.7%, p = 0.031), with the absence of epidermal growth factor receptor (EGFR) mutations (83.6% vs. 91.1% vs. 98.1% p = 0.029), and with the absence of ERBB2 (HER2) tyrosine kinase mutations (90.9% vs. 100% vs. 98.1% p = 0.007), respectively. Age, smoking status, histological subtype, and disease stage showed no significant differences among the three patient groups. In the univariate logistic regression, EGFR mutation appeared to be the only predictor for PD-L1 expression, although it did not reach statistical significance (p = 0.06). CONCLUSION Although sex and genomic alterations are associated with PD-L1 expression in patients with NSCLC, no clinical characteristics seem to predict PD-L1 expression significantly.
Collapse
Affiliation(s)
- P Sarova
- Division of Pulmonology, Department of Internal Medicine II, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - B Mosleh
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - S Zehetmayer
- Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - F Oberndorfer
- Department of Pathology, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - J Widder
- Department of Radiation Oncology, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - H Prosch
- Department of Biomedical Imaging and Image-guided Therapy, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - C Aigner
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - M Idzko
- Division of Pulmonology, Department of Internal Medicine II, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - M A Hoda
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - D Gompelmann
- Division of Pulmonology, Department of Internal Medicine II, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Yan F, Da Q, Yi H, Deng S, Zhu L, Zhou M, Liu Y, Feng M, Wang J, Wang X, Zhang Y, Zhang W, Zhang X, Lin J, Zhang S, Wang C. Artificial intelligence-based assessment of PD-L1 expression in diffuse large B cell lymphoma. NPJ Precis Oncol 2024; 8:76. [PMID: 38538739 PMCID: PMC10973523 DOI: 10.1038/s41698-024-00577-y] [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: 11/07/2023] [Accepted: 03/13/2024] [Indexed: 11/12/2024] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is an aggressive blood cancer known for its rapid progression and high incidence. The growing use of immunohistochemistry (IHC) has significantly contributed to the detailed cell characterization, thereby playing a crucial role in guiding treatment strategies for DLBCL. In this study, we developed an AI-based image analysis approach for assessing PD-L1 expression in DLBCL patients. PD-L1 expression represents as a major biomarker for screening patients who can benefit from targeted immunotherapy interventions. In particular, we performed large-scale cell annotations in IHC slides, encompassing over 5101 tissue regions and 146,439 live cells. Extensive experiments in primary and validation cohorts demonstrated the defined quantitative rule helped overcome the difficulty of identifying specific cell types. In assessing data obtained from fine needle biopsies, experiments revealed that there was a higher level of agreement in the quantitative results between Artificial Intelligence (AI) algorithms and pathologists, as well as among pathologists themselves, in comparison to the data obtained from surgical specimens. We highlight that the AI-enabled analytics enhance the objectivity and interpretability of PD-L1 quantification to improve the targeted immunotherapy development in DLBCL patients.
Collapse
Affiliation(s)
- Fang Yan
- Shanghai Artificial Intelligence Laboratory, Shanghai, China
| | - Qian Da
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongmei Yi
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shijie Deng
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lifeng Zhu
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mu Zhou
- Department of Computer Science, Rutgers University, New Brunswick, NJ, USA
| | - Yingting Liu
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Feng
- College of Electronic and Information Engineering, Tongji University, Shanghai, China
| | - Jing Wang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuan Wang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuxiu Zhang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjing Zhang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofan Zhang
- Shanghai Artificial Intelligence Laboratory, Shanghai, China.
| | - Jingsheng Lin
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Shaoting Zhang
- Shanghai Artificial Intelligence Laboratory, Shanghai, China.
- Centre for Perceptual and Interactive Intelligence (CPII) Ltd. under InnoHK, HongKong, China.
- SenseTime Research, Shanghai, China.
| | - Chaofu Wang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Gompelmann D, Sarova P, Mosleh B, Papaporfyriou A, Oberndorfer F, Idzko M, Hoda MA. PD-L1 assessment in lung cancer biopsies-pitfalls and limitations. Int J Biol Markers 2024; 39:3-8. [PMID: 38111297 DOI: 10.1177/03936155231214273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
The programmed cell death-ligand 1 (PD-L1) protein expression on tumor cells predicts the efficacy of immunotherapy in patients with non-small cell lung cancer. However, the assessment of PD-L1 expression on tumor cells has limited power for selecting patients for immunotherapy due to intra-tumoral heterogeneity and inter-tumoral heterogeneity of PD-L1 expression, the inter-observer variability in scoring PD-L1 staining, and reproducibility. These difficulties and pitfalls in interpreting the PD-L1 assessment are discussed in detail in this review.
Collapse
Affiliation(s)
- Daniela Gompelmann
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Pavla Sarova
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Berta Mosleh
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Anastasia Papaporfyriou
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | | | - Marco Idzko
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Shima Y, Sato Y, Morimoto T, Hara S, Hirabayashi R, Nagata K, Nakagawa A, Tachikawa R, Hamakawa H, Takahashi Y, Tomii K. Predictive performance of PD-L1 tumor proportion score for nivolumab response evaluated using archived specimens in patients with non-small cell lung cancer experiencing a postoperative recurrence. Invest New Drugs 2023; 41:35-43. [PMID: 36334214 DOI: 10.1007/s10637-022-01309-4] [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: 04/10/2022] [Accepted: 09/29/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVES Postoperative recurrence in patients with non-small-cell lung carcinoma (NSCLC) is a major issue for life expectancy. Programmed cell death ligand 1 (PD-L1) expression on tumor cells is important in the prognosis of NSCLC. However, the predictive ability of PD-L1 evaluated with archived surgical specimens for nivolumab treatment have remained unknown. This study was aimed to analyze the predictive ability of the PD-L1 tumor proportion score (TPS) for nivolumab response in patients with NSCLC experiencing a postoperative recurrence using archived surgical specimens. MATERIALS AND METHODS This retrospective cohort study involved patients with advanced NSCLC (N = 78) treated with nivolumab between April 2016 and September 2018. They were categorized into postoperative recurrence (N = 24) and non-postoperative recurrence (N = 54) groups. The predictive ability of PD-L1 TPS for response to nivolumab treatment in these two groups was determined using receiver operating characteristic (ROC) analysis. Additionally, we evaluated the predictive ability of PD-L1 TPS using rebiopsy specimens collected from the recurrent lesions in six patients of the postoperative recurrence group. RESULTS PD-L1 TPS exhibited lower predictive performance in the postoperative recurrent group (area under the curve [AUC] = 0.58) compared with that in the non-post operative recurrent group (AUC = 0.81). Furthermore, PD-L1 TPS was significantly increased in rebiopsy specimens. The predictive performance of PD-L1 TPS in these specimens was higher (AUC = 0.90) than that in the archived surgical specimens. CONCLUSION The study revealed that archived surgical specimens are inadequate for assessing the predictive ability of PD-L1 for nivolumab response, while rebiopsy specimens are adequate.
Collapse
Affiliation(s)
- Yusuke Shima
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, 650-0047, Kobe, Japan
| | - Yuki Sato
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, 650-0047, Kobe, Japan.
| | - Takeshi Morimoto
- Department of Clinical Research Center, Kobe City Medical Center General Hospital, Kobe, Japan
- Department of Clinical Epidemiology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Shigeo Hara
- Department of Clinical Pathology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Ryosuke Hirabayashi
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, 650-0047, Kobe, Japan
| | - Kazuma Nagata
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, 650-0047, Kobe, Japan
| | - Atsushi Nakagawa
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, 650-0047, Kobe, Japan
| | - Ryo Tachikawa
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, 650-0047, Kobe, Japan
| | - Hiroshi Hamakawa
- Department of Thoracic Surgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Yutaka Takahashi
- Department of Thoracic Surgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Keisuke Tomii
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, 650-0047, Kobe, Japan
| |
Collapse
|