1
|
Li M, Lin C, Lin J, Chen S, Weng L, He Z. Efficacy analysis of immunotherapy‑based combinations for patients with EGFR‑mutant advanced non‑small cell lung cancer after TKI failure. Oncol Lett 2024; 28:504. [PMID: 39233818 PMCID: PMC11369851 DOI: 10.3892/ol.2024.14637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 07/30/2024] [Indexed: 09/06/2024] Open
Abstract
Treatment options for epidermal growth factor receptor (EGFR)-mutant advanced non-small cell lung cancer (NSCLC) following tyrosine kinase inhibitor (TKI) failure are limited, and platinum-based chemotherapy remains the main treatment. The development of effective immunotherapy for this disease has been challenging. In the present study, 37 patients with EGFR-mutant advanced NSCLC who were treated with programmed cell death-1 (PD-1) inhibitor-based combinations after TKI failure were reviewed. The total cohort had a median progression-free survival (mPFS) of 5.2 months (95% CI, 4.077-6.323 months) and a median overall survival (mOS) of 18.3 months (95% CI, 12.932-23.668 months). Patients with Eastern Cooperative Oncology Group performance-status (ECOG-PS) scores of 0 or 1 had longer mPFS than those with ECOG-PS scores of 2 (5.4 vs. 2.4 months; P=0.006). In addition, a PFS benefit was observed in patients with EGFR T790M-negative compared with EGFR T790M-positive tumors (mPFS 6.2 vs. 4.4 months; P=0.041). Patients treated with immunotherapy-based combinations as a front-line therapy had a longer mPFS than those in which the combinations were used as a late-line therapy (6.2 vs. 2.4 months; P<0.001). PD-1 inhibitor combined with chemotherapy and bevacizumab did not show a clear advantage over PD-1 inhibitor combined with chemotherapy alone (mPFS, 6.2 vs. 4.4 months; P=0.681), although it resulted in an improved overall response rate (ORR) and disease control rate. Notably, the 7 patients with a programmed cell death ligand-1 (PD-L1) tumor proportion score of ≥50% had an ORR of 100% and an mPFS of 8.3 months. Therefore, it is suggested that PD-1 inhibitor-based combinations should be a priority treatment option in selective populations, such as those with low ECOG-PS scores, T790M-negative status or high PD-L1 expression in EGFR-mutant NSCLC after TKI failure. The use of immunotherapy and chemotherapy in combination with antiangiogenic agents appears to be a promising combination therapy for such patients.
Collapse
Affiliation(s)
- Meifang Li
- Department of Thoracic Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Cheng Lin
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Jinghui Lin
- Department of Thoracic Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Shijie Chen
- Department of Thoracic Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Lihong Weng
- Department of Thoracic Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Zhiyong He
- Department of Thoracic Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| |
Collapse
|
2
|
Jin X, Pan Y, Cheng C, Shen H, Zhai C, Yin K, Zhu X, Pan H, You L. Optimizing first-line TKI treatment efficacy in PD-L1-positive EGFR-mutated NSCLC: the impact of antiangiogenic agents. Front Pharmacol 2024; 15:1391972. [PMID: 39161896 PMCID: PMC11330760 DOI: 10.3389/fphar.2024.1391972] [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: 02/26/2024] [Accepted: 07/26/2024] [Indexed: 08/21/2024] Open
Abstract
Background In individuals receiving treatment with epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs), those exhibiting positive PD-L1 expression might experience reduced progression-free survival (PFS). However, the effects on overall survival (OS) and the determination of efficacious treatment approaches are still not well-defined. Methods In our retrospective study, we examined data from 193 NSCLC patients with advanced EGFR mutations who received first-line TKI treatments, treated at two centers of Shaw Hospital in Zhejiang, China. This analysis covered a period from 1 January 2016 to 30 April 2023. Results Patients with PD-L1 positivity exhibited a markedly shorter average PFS (9.5 months versus 17.8 months, P < 0.001) and OS (44.4 months versus 65.7 months, P = 0.016) relative to those without PD-L1 expression. This difference in both PFS and OS remained statistically significant even after adjusting for multiple factors (P < 0.001 for PFS and P = 0.028 for OS). In the PD-L1-positive cohort, introducing combination antiangiogenic significantly extended both PFS (from 9.1 to 25.7 months, P = 0.026) and OS (from 42 to 53.5 months, P = 0.03). Post-first-line TKI therapy, 39.3% of PD-L1-positive patients and 54.5% of PD-L1-negative patients developed the T790M mutation (P = 0.212), with no notable difference in PFS from second-line TKI treatments between the groups. Additionally, subsequent combination therapy with immunotherapy markedly prolonged OS in the PD-L1-positive group. However, for PD-L1-negative patients, neither combination antiangiogenic therapy nor later-line immunotherapy demonstrated significant benefits in PFS or OS. Conclusion For PD-L1-positive patients, combined antiangiogenic treatments and immunotherapy can significantly improve survival outcomes. In contrast, PD-L1-negative patients show less benefit from these therapies, highlighting the greater efficacy of these treatments in PD-L1-positive individuals.
Collapse
Affiliation(s)
- Xuanhong Jin
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Pan
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
| | - Cheng Cheng
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hangchen Shen
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chongya Zhai
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Kailai Yin
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
| | - Xinyu Zhu
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Liangkun You
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
3
|
Fukuda A, Okuma Y. From Rarity to Reality: Osimertinib's Promising Horizon in Treating Uncommon EGFR Mutations in Non-Small Cell Lung Cancer. Clin Cancer Res 2024; 30:3128-3136. [PMID: 38767589 DOI: 10.1158/1078-0432.ccr-23-4035] [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: 12/31/2023] [Revised: 03/01/2024] [Accepted: 04/16/2024] [Indexed: 05/22/2024]
Abstract
In the realm of advanced non-small cell lung cancer (NSCLC) harboring epidermal growth factor receptor (EGFR) therapy with tyrosine kinase inhibitors (TKI), addressing optimal treatment for uncommon EGFR mutations like G719X in exon 18, S768I in exon 20, and L861Q in exon 21 remains a pivotal yet challenging frontier. Contrary to the well-established efficacy of EGFR-TKIs in common EGFR mutations, these uncommon alterations pose unmet medical needs due to a lack of comprehensive evidence. While afatinib, a second-generation EGFR-TKI, has received FDA approval for patients with these uncommon EGFR mutations, the approval was based on a post-hoc analysis of randomized clinical trials. Recent developments include multiple clinical trials investigating the efficacy of both second- and third-generation EGFR-TKIs in patients with uncommon EGFR mutations. A noteworthy example is a prospective phase II trial of osimertinib including the landmark UNICORN study, which has shown promising results in treating uncommon EGFR mutations. Despite various reports on the efficacy of afatinib and osimertinib in treating uncommon EGFR mutations, the appropriate use of these TKIs remains unclear. This review aims to consolidate the findings from the latest clinical trials focused on uncommon EGFR mutations, outlining variations in the therapeutic efficacy of these TKIs based on the specific genetic mutation. By synthesizing these findings, we aim to guide oncologists toward more informed decisions in employing TKIs for NSCLC with uncommon EGFR mutations other than exon 20 insertion. Additionally, we explore potential treatment strategies tailored to these patient populations to address the challenges posed by these mutations.
Collapse
Affiliation(s)
- Akito Fukuda
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
4
|
Lafon M, Cousin S, Alamé M, Nougaret S, Italiano A, Crombé A. Metastatic Lung Adenocarcinomas: Development and Evaluation of Radiomic-Based Methods to Measure Baseline Intra-Patient Inter-Tumor Lesion Heterogeneity. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024:10.1007/s10278-024-01163-1. [PMID: 39020153 DOI: 10.1007/s10278-024-01163-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 07/19/2024]
Abstract
Radiomics has traditionally focused on individual tumors, often neglecting the integration of metastatic disease, particularly in patients with non-small cell lung cancer. This study sought to examine intra-patient inter-tumor lesion heterogeneity indices using radiomics, exploring their relevance in metastatic lung adenocarcinoma. Consecutive adults newly diagnosed with metastatic lung adenocarcinoma underwent contrast-enhanced CT scans for lesion segmentation and radiomic feature extraction. Three methods were devised to measure distances between tumor lesion profiles within the same patient in radiomic space: centroid to lesion, lesion to lesion, and primitive to lesion, with subsequent calculation of mean, range, and standard deviation of these distances. Associations between HIs, disease control rate, objective response rate to first-line treatment, and overall survival were explored. The study included 167 patients (median age 62.3 years) between 2016 and 2019, divided randomly into experimental (N = 117,546 lesions) and validation (N = 50,232 tumor lesions) cohorts. Patients without disease control/objective response and with poorer survival consistently systematically exhibited values of all heterogeneity indices. Multivariable analyses revealed that the range of primitive-to-lesion distances was associated with disease control in both cohorts and with objective response in the validation cohort. This metrics showed univariable associations with overall survival in the experimental. In conclusion, we proposed original methods to estimate the intra-patient inter-tumor lesion heterogeneity using radiomics that demonstrated correlations with patient outcomes, shedding light on the clinical implications of inter-metastases heterogeneity. This underscores the potential of radiomics in understanding and potentially predicting treatment response and prognosis in metastatic lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Mathilde Lafon
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Sophie Cousin
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Mélissa Alamé
- Department of Biopathology, Institut Bergonié, Bordeaux, France
| | - Stéphanie Nougaret
- Medical Imaging Department, Montpellier Cancer Institute, Montpellier Cancer Research Institute (U1194), University of Montpellier, Montpellier, France
| | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
- SARCOTARGET Team, Bordeaux Research Institute in Oncology (BRIC) INSERM U1312 & University of Bordeaux, Bordeaux, France
| | - Amandine Crombé
- SARCOTARGET Team, Bordeaux Research Institute in Oncology (BRIC) INSERM U1312 & University of Bordeaux, Bordeaux, France.
- Department of Radiology, Institut Bergonié, Bordeaux, France.
- Department of Radiology, Pellegrin University Hospital, Bordeaux, France.
| |
Collapse
|
5
|
Masson-Grehaigne C, Lafon M, Palussière J, Leroy L, Bonhomme B, Jambon E, Italiano A, Cousin S, Crombé A. Enhancing Immunotherapy Response Prediction in Metastatic Lung Adenocarcinoma: Leveraging Shallow and Deep Learning with CT-Based Radiomics across Single and Multiple Tumor Sites. Cancers (Basel) 2024; 16:2491. [PMID: 39001553 PMCID: PMC11240700 DOI: 10.3390/cancers16132491] [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: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
This study aimed to evaluate the potential of pre-treatment CT-based radiomics features (RFs) derived from single and multiple tumor sites, and state-of-the-art machine-learning survival algorithms, in predicting progression-free survival (PFS) for patients with metastatic lung adenocarcinoma (MLUAD) receiving first-line treatment including immune checkpoint inhibitors (CPIs). To do so, all adults with newly diagnosed MLUAD, pre-treatment contrast-enhanced CT scan, and performance status ≤ 2 who were treated at our cancer center with first-line CPI between November 2016 and November 2022 were included. RFs were extracted from all measurable lesions with a volume ≥ 1 cm3 on the CT scan. To capture intra- and inter-tumor heterogeneity, RFs from the largest tumor of each patient, as well as lowest, highest, and average RF values over all lesions per patient were collected. Intra-patient inter-tumor heterogeneity metrics were calculated to measure the similarity between each patient lesions. After filtering predictors with univariable Cox p < 0.100 and analyzing their correlations, five survival machine-learning algorithms (stepwise Cox regression [SCR], LASSO Cox regression, random survival forests, gradient boosted machine [GBM], and deep learning [Deepsurv]) were trained in 100-times repeated 5-fold cross-validation (rCV) to predict PFS on three inputs: (i) clinicopathological variables, (ii) all radiomics-based and clinicopathological (full input), and (iii) uncorrelated radiomics-based and clinicopathological variables (uncorrelated input). The Models' performances were evaluated using the concordance index (c-index). Overall, 140 patients were included (median age: 62.5 years, 36.4% women). In rCV, the highest c-index was reached with Deepsurv (c-index = 0.631, 95%CI = 0.625-0.647), followed by GBM (c-index = 0.603, 95%CI = 0.557-0.646), significantly outperforming standard SCR whatever its input (c-index range: 0.560-0.570, all p < 0.0001). Thus, single- and multi-site pre-treatment radiomics data provide valuable prognostic information for predicting PFS in MLUAD patients undergoing first-line CPI treatment when analyzed with advanced machine-learning survival algorithms.
Collapse
Affiliation(s)
- Cécile Masson-Grehaigne
- Department of Diagnostic and Interventional Oncologic Imaging, Institut Bergonié, F-33076 Bordeaux, France
- Department of Radiology, Pellegrin University Hospital, F-33000 Bordeaux, France
| | - Mathilde Lafon
- Department of Medical Oncology, Institut Bergonié, F-33076 Bordeaux, France
| | - Jean Palussière
- Department of Diagnostic and Interventional Oncologic Imaging, Institut Bergonié, F-33076 Bordeaux, France
| | - Laura Leroy
- Department of Medical Oncology, Institut Bergonié, F-33076 Bordeaux, France
| | - Benjamin Bonhomme
- Department of Biopathology, Institut Bergonié, F-33076 Bordeaux, France
| | - Eva Jambon
- Department of Radiology, Pellegrin University Hospital, F-33000 Bordeaux, France
| | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, F-33076 Bordeaux, France
- SARCOTARGET Team, Bordeaux Institute of Oncology (BRIC) INSERM U1312, F-33076 Bordeaux, France
| | - Sophie Cousin
- Department of Medical Oncology, Institut Bergonié, F-33076 Bordeaux, France
| | - Amandine Crombé
- Department of Diagnostic and Interventional Oncologic Imaging, Institut Bergonié, F-33076 Bordeaux, France
- Department of Radiology, Pellegrin University Hospital, F-33000 Bordeaux, France
- SARCOTARGET Team, Bordeaux Institute of Oncology (BRIC) INSERM U1312, F-33076 Bordeaux, France
| |
Collapse
|
6
|
Kim HD, Ryu MH, Park YS, Yoo C, Kim SJ, Kang YK. Clinical and Biomarker Analysis of a Phase I/II Study of PDR001 Plus Imatinib for Advanced Treatment-Refractory Gastrointestinal Stromal Tumors. Clin Cancer Res 2024; 30:2743-2750. [PMID: 38662455 DOI: 10.1158/1078-0432.ccr-23-4065] [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: 12/27/2023] [Revised: 02/16/2024] [Accepted: 04/22/2024] [Indexed: 07/02/2024]
Abstract
PURPOSE In this phase Ib/II study, we aimed to evaluate the safety and efficacy of PDR001, an anti-PD1 antibody, in combination with imatinib in patients with treatment-refractory gastrointestinal stromal tumor (GIST). PATIENTS AND METHODS Patients with advanced GIST whose disease had progressed on imatinib, sunitinib, and regorafenib were enrolled. In phase Ib, the standard 3 + 3 dose escalation scheme was applied. Intravenous administration of PDR001 at 400 mg for every 4 weeks plus imatinib (300 and 400 mg daily for dose levels I and II, respectively) was given. The primary outcome for phase II was the disease control rate at 12 weeks. Exploratory biomarker analysis was performed based on PDL1 IHC, next-generation sequencing, and multiplexed IHC. RESULTS No dose-limiting toxicity was observed in the phase Ib part (n = 10), and dose level II was selected as the recommended phase II dose. In the phase II part (n = 29), there was no objective response, and the disease control rate at 12 weeks was 37.9%, not meeting the primary efficacy endpoint. For patients in phase Ib-dose level II and phase II (n = 36), the median progression-free survival (PFS) and overall survival were 2.3 and 9.5 months, respectively. The most common grade 3 to 4 adverse event was anemia. Exploratory biomarker analysis indicated that a higher CD8+ T-cell density was associated with a favorable PFS but to a limited degree. Tumor mutational burden and PDL1 were not associated with better PFS. CONCLUSIONS In patients with treatment-refractory GIST, PDR001 in combination with imatinib was generally tolerable, but it was not effective.
Collapse
Affiliation(s)
- Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young Soo Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Joo Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoon-Koo Kang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
7
|
Favorito V, Ricciotti I, De Giglio A, Fabbri L, Seminerio R, Di Federico A, Gariazzo E, Costabile S, Metro G. Non-small cell lung cancer: an update on emerging EGFR-targeted therapies. Expert Opin Emerg Drugs 2024; 29:139-154. [PMID: 38572595 DOI: 10.1080/14728214.2024.2331139] [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: 10/17/2023] [Accepted: 03/12/2024] [Indexed: 04/05/2024]
Abstract
INTRODUCTION Current research in EGFR-mutated NSCLC focuses on the management of drug resistance and uncommon mutations, as well as on the opportunity to extend targeted therapies' field of action to earlier stages of disease. AREAS COVERED We conducted a review analyzing literature from the PubMed database with the aim to describe the current state of art in the management of EGFR-mutated NSCLC, but also to explore new strategies under investigation. To this purpose, we collected recruiting phase II-III trials registered on Clinicaltrials.govand conducted on EGFR-mutated NSCLC both in early and advanced stage. EXPERT OPINION With this review, we want to provide an exhaustive overview of current and new potential treatments in EGFR-mutated NSCLC, with emphasis on the most promising newly investigated strategies, such as association therapies in the first-line setting involving EGFR-TKIs and chemotherapy (FLAURA2) or drugs targeting different driver pathways (MARIPOSA). We also aimed at unearthing challenges to achieve in this field, specifically the need to fully exploit already available compounds while developing new ones, the management of new emerging toxicities and the necessity to improve our biological understanding of the disease to design trials with a solid scientific rationale and to allow treatment personalization such in case of uncommon mutations.
Collapse
Affiliation(s)
- Valentina Favorito
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Ilaria Ricciotti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea De Giglio
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Laura Fabbri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Renata Seminerio
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alessandro Di Federico
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Eleonora Gariazzo
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Silvia Costabile
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Giulio Metro
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Perugia, Italy
| |
Collapse
|
8
|
Buyukbayram ME, Hannarici Z, Yilmaz A, Turhan A, Caglar AA, Esdur PC, Bilici M, Tekin SB. Inflammatory parameters in NSCLC with driver mutation. Lung Cancer Manag 2024; 13:LMT66. [PMID: 38818368 PMCID: PMC11137794 DOI: 10.2217/lmt-2023-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/26/2024] [Indexed: 06/01/2024] Open
Abstract
Aim: The tumor microenvironment of NSCLC with driver mutations, such as EGFR, ALK and ROS, is less inflammatory. Materials & methods: This retrospective study included 38 patients with NSCLC driver mutations. The relationship between clinical and inflammatory markers concerning progression-free survival and overall survival was analyzed based on Kaplan-Meier curves. Results: The mean age of the patients was 59.8 ± 11.9. Progression-free survival and overall survival were significantly longer in patients under 65 years of age and with low neutrophil-lymphocyte ratio, low systemic immune-inflammation index and high lymphocyte count (p < 0.05). Conclusion: Unlike tumor biology, peripheral inflammatory parameters, such as neutrophil-lymphocyte ratio, systemic immune-inflammation index and lymphocyte count may be associated with survival in NSCLC patients with driver mutations.
Collapse
Affiliation(s)
- Mehmet Emin Buyukbayram
- Department of Medical Oncology, Atatürk University Faculty of Medicine, Erzurum, 25100, Turkey
| | | | - Ali Yilmaz
- Department of medical Oncology, Health Sciences University Yüksek İhtisas Training and Research Hospital, Bursa, 16350, Turkey
| | - Aykut Turhan
- Department of Medical Oncology, Atatürk University Faculty of Medicine, Erzurum, 25100, Turkey
| | - Alperen Akansel Caglar
- Department of Medical Oncology, Atatürk University Faculty of Medicine, Erzurum, 25100, Turkey
| | - Pınar Coban Esdur
- Department of Medical Oncology, Atatürk University Faculty of Medicine, Erzurum, 25100, Turkey
| | - Mehmet Bilici
- Department of Medical Oncology, Atatürk University Faculty of Medicine, Erzurum, 25100, Turkey
| | - Salim Basol Tekin
- Department of Medical Oncology, Atatürk University Faculty of Medicine, Erzurum, 25100, Turkey
| |
Collapse
|
9
|
Passaro A, Al Bakir M, Hamilton EG, Diehn M, André F, Roy-Chowdhuri S, Mountzios G, Wistuba II, Swanton C, Peters S. Cancer biomarkers: Emerging trends and clinical implications for personalized treatment. Cell 2024; 187:1617-1635. [PMID: 38552610 PMCID: PMC7616034 DOI: 10.1016/j.cell.2024.02.041] [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: 12/13/2023] [Revised: 02/21/2024] [Accepted: 02/28/2024] [Indexed: 04/02/2024]
Abstract
The integration of cancer biomarkers into oncology has revolutionized cancer treatment, yielding remarkable advancements in cancer therapeutics and the prognosis of cancer patients. The development of personalized medicine represents a turning point and a new paradigm in cancer management, as biomarkers enable oncologists to tailor treatments based on the unique molecular profile of each patient's tumor. In this review, we discuss the scientific milestones of cancer biomarkers and explore future possibilities to improve the management of patients with solid tumors. This progress is primarily attributed to the biological characterization of cancers, advancements in testing methodologies, elucidation of the immune microenvironment, and the ability to profile circulating tumor fractions. Integrating these insights promises to continually advance the precision oncology field, fostering better patient outcomes.
Collapse
Affiliation(s)
- Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Maise Al Bakir
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Emily G Hamilton
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Fabrice André
- Gustave-Roussy Cancer Center, Paris Saclay University, Villejuif, France
| | - Sinchita Roy-Chowdhuri
- Department of Anatomic Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giannis Mountzios
- Fourth Department of Medical Oncology and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Department of Oncology, University College London Hospitals, London, UK
| | - Solange Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| |
Collapse
|
10
|
Wang X, Meng X, Cai G, Jin P, Bai M, Fu Y, Wang Z, Guo J, Han X. Survival outcomes of targeted and immune consolidation therapies in locally advanced unresectable lung adenocarcinoma. Int Immunopharmacol 2024; 129:111684. [PMID: 38364745 DOI: 10.1016/j.intimp.2024.111684] [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: 12/01/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND Locally advanced non-small cell lung cancer (LA-NSCLC) presents unique challenges due to its progression and tumor heterogeneity. The effectiveness of consolidation therapies, particularly in patients with gene mutations, remains an area of active investigation. METHODS In this retrospective cohort study, we examined data from 3,454 patients with unresectable lung adenocarcinoma (LUAD), narrowing our focus to 242 individuals with stage II/III. We gathered patient data, such as demographics, ECOG status, histology, treatment specifics, and gene expression, from patients in China. The study's primary outcome was overall survival (OS), while progression-free survival (PFS) served as the secondary outcome. RESULTS In this study, 50 % of the 242 patients underwent only radical chemoradiotherapy, with 45.87 % (111/242) exhibiting driver gene mutations, predominantly EGFR (58.57 %), followed by KRAS and ALK. Patients with mutations who received either targeted or immune consolidation therapy demonstrated a significantly longer median PFS (42.97 months vs. 24.87 months, p = 0.014) and improved OS (not reached vs. 24.37 months, p = 0.006), compared to those without consolidation therapy. Targeted therapy in mutant patients resulted in an extended median PFS (42.87 months) compared to immune therapy (27.03 months, p = 0.029), with no significant difference in OS. Median PFS and OS were similar between mutant and wild-type patients receiving immune therapy (p = 0.380 and p = 0.928, respectively). CONCLUSION This study underscores the efficacy of targeted consolidation therapy in enhancing PFS in LUAD patients with genetic mutations. It also shows that immune consolidation therapy provides similar survival benefits to mutant and wild-type patients. Future research should focus on optimizing these therapies for improved patient outcomes.
Collapse
Affiliation(s)
- Xiaohan Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Xue Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Guoxin Cai
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Peng Jin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Menglin Bai
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Ying Fu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Zhehai Wang
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jun Guo
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiao Han
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
11
|
Liu F, Zhang X, Lu M, Liu C, Zhang X, Chu Q, Chen Y, Zhang P. The association of genomic alterations with PD-L1 expression in Chinese patients with EGFR/ALK wild-type lung adenocarcinoma and potential predictive value of Hippo pathway mutations to immunotherapy. Cancer Med 2024; 13:e7038. [PMID: 38396367 PMCID: PMC10891359 DOI: 10.1002/cam4.7038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/23/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The study focuses on PD-L1 expression as an essential biomarker for gauging the response of EGFR/ALK wild-type NSCLC patients to FDA-approved immune checkpoint inhibitors (ICIs). It aims to explore clinical, molecular, and immune microenvironment characteristics associated with PD-L1 expression in EGFR/ALK wild-type lung adenocarcinoma patients eligible for ICI therapy. METHODS In this retrospective study, tumor samples from 359 Chinese EGFR/ALK wild-type lung adenocarcinoma patients underwent comprehensive evaluations for PD-L1 expression and NGS-targeted sequencing. The investigation encompassed the analysis and comparison of clinical traits, gene mutations, pathways, and immune signatures between two groups categorized by PD-L1 status: negative (TPS < 1%) and positive (TPS ≥ 1%). Additionally, the study explored the link between genomic changes and outcomes following immunotherapy. RESULTS High tumor mutational burden correlated significantly with PD-L1 positivity in patients with EGFR/ALK wild-type lung adenocarcinoma. Gene alterations, including TP53, KRAS, and others, were more pronounced in the PD-L1 positive group. Pathway analysis highlighted higher frequencies of alterations in pathways like RTK/RAS, p53, and Hippo in PD-L1-positive patients. The Hippo pathway's relevance was confirmed in separate immunotherapy cohorts, associated with better outcomes. In terms of immune cell infiltration, Hippo mutants exhibited higher levels of CD68+ PD-L1+ macrophages, CD8+ T cells, and CD8+ PD-1- T cells. CONCLUSIONS This study offers insights into genomic features of Chinese EGFR/ALK wild-type lung adenocarcinoma patients based on PD-L1 expression. Notably, Hippo pathway alterations were linked to improved immunotherapy outcomes. These findings suggest connections between the Hippo pathway and PD-L1 expression, warranting further clinical and functional investigations. The research advances our understanding of PD-L1 expression's genomic context and immunotherapy response in EGFR/ALK wild-type lung adenocarcinoma.
Collapse
Affiliation(s)
- Fangfang Liu
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xuemei Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Mengyao Lu
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chun Liu
- Genecast Biotechnology Co., LtdWuxiJiangsuChina
| | | | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuan Chen
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Peng Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
12
|
McMahon DJ, McLaughlin R, Naidoo J. Is Immunotherapy Beneficial in Patients with Oncogene-Addicted Non-Small Cell Lung Cancers? A Narrative Review. Cancers (Basel) 2024; 16:527. [PMID: 38339280 PMCID: PMC10854575 DOI: 10.3390/cancers16030527] [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: 09/23/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024] Open
Abstract
Over the past 20 years, there has been a paradigm shift in the care of patients with non-small cell lung cancer (NSCLC), who now have a range of systemic treatment options including targeted therapy, chemotherapy, immunotherapy (ICI), and antibody-drug conjugates (ADCs). A proportion of these cancers have single identifiable alterations in oncogenes that drive their proliferation and cancer progression, known as "oncogene-addiction". These "driver alterations" are identified in approximately two thirds of patients with lung adenocarcinomas, via next generation sequencing or other orthogonal tests. It was noted in the early clinical development of ICIs that patients with oncogene-addicted NSCLC may have differential responses to ICI. The toxicity signal for patients with oncogene-addicted NSCLC when treated with ICIs also seemed to differ depending on the alteration present and the specific targeted agent used. Developing a greater understanding of the underlying reasons for these clinical observations has become an important area of research in NSCLC. In this review, we analyze the efficacy and safety of ICI according to specific mutations, and consider possible future directions to mitigate safety concerns and improve the outcomes for patients with oncogene-addicted NSCLC.
Collapse
Affiliation(s)
- David John McMahon
- Trinity St James’s Cancer Institute, St. James’s Hospital, James’s Street, D08 NHY1 Dublin, Ireland
| | | | - Jarushka Naidoo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
- Beaumont RCSI Cancer Centre, D09 V2NO Dublin, Ireland
- RCSI University of Health Sciences, D02 YN77 Dublin, Ireland
- Beaumont Hospital, D09 Y177 Dublin, Ireland
| |
Collapse
|
13
|
Lasvergnas J, Fallet V, Duchemann B, Jouveshomme S, Cadranel J, Chouaïd C. PDL1-status predicts primary resistance of metastatic, EGFR-mutated non small cell lung cancers to EGFR tyrosine-kinase inhibitors. Respir Med Res 2023; 84:101018. [PMID: 37302160 DOI: 10.1016/j.resmer.2023.101018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/03/2023] [Accepted: 04/16/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND EGFR tyrosine-kinase inhibitors (TKIs) are the reference treatment for metastatic, EGFR-mutated, non-small-cell lung cancers (EGFRm NSCLCs). However, 16-20% of those tumors progress early (3-6 months) and factors predicting that resistance are unknown. This study was undertaken to examine PDL1 status as such a factor. METHODS This retrospective analysis included metastatic, EGFRm-NSCLC patients who received first-line 1st-, 2nd- or 3rd-generation EGFR TKIs with PDL1 expression determined in pretreatment biopsies. Kaplan-Meier estimations of probabilities of progression-free survival (PFS) and overall survival (OS) were compared with log-rank test, and logistic-regression analyses. RESULTS PDL1 status of the 145 included patients was ≥1% (47%), 1-49% (33%) or ≥50% (14%). For PDL1-positive vs PDL1-negative patients, respectively, median PFS lasted 8 (95% CI: 6-12) vs 12 (95% CI: 11-17) months (p = 0.008), with 18% vs. 8% (NS) of NSCLCs progressing at 3 months, and 47% vs. 18% (HR 0.25 [95% CI 0.10-0.566], p<0.001) at 6 months. Multivariate analysis retained 1st- or 2nd-generation EGFR TKI, brain metastases and albuminemia <35 g/L at diagnosis as significantly associated with shorter PFS, but not PDL1 status, which was independently associated with progression at 6 months (HR 3.76 [1.23-12.63], p = 0.02). PDL1-negative and PDL1-positive patients' OS lasted 27 (95% CI 24-39) and 22 (95% CI 19-41) months, respectively (NS). Multivariate analysis retained only brain metastases or albuminemia <35 g/L at diagnosis as being independently associated with OS. CONCLUSION PDL1 expression ≥1% seems to be associated with early progression during the first 6 months of first-line EGFR-TKI treatment of metastatic EGFRm NSCLCs, without impacting OS.
Collapse
Affiliation(s)
- Julie Lasvergnas
- Service de Pneumologie, Groupe Hospitalier Paris Saint-Joseph, 195 rue Raymond Losserand, 75014 Paris, France.
| | - Vincent Fallet
- Service de Pneumologie et Oncologie Thoracique, DMU APPROCHES, Hôpital Tenon, APHP, Sorbonne Université, 4 rue de la Chine, 75020 Paris, France
| | - Boris Duchemann
- Service d'Oncologie Médicale et Thoracique, Université Paris Sorbonne Nord, APHP, Hôpital Avicenne, 125 rue de Stalingrad, 93000 Bobigny, France
| | - Stephane Jouveshomme
- Service de Pneumologie, Groupe Hospitalier Paris Saint-Joseph, 195 rue Raymond Losserand, 75014 Paris, France
| | - Jacques Cadranel
- Service de Pneumologie et Oncologie Thoracique, DMU APPROCHES, Hôpital Tenon, APHP, Sorbonne Université, 4 rue de la Chine, 75020 Paris, France
| | - Christos Chouaïd
- Service de Pneumologie, CHI Créteil, 40 av. de Verdun, 94000 Créteil, France; Inserm U955, UPEC, IMRB, Créteil, France
| |
Collapse
|
14
|
Loh J, Low JL, Sachdeva M, Low PQ, Wong RSJ, Huang Y, Chia PL, Soo RA. Management of Oncogene Driven Locally Advanced Unresectable Non-small Cell Lung Cancer. Expert Rev Anticancer Ther 2023; 23:913-926. [PMID: 37551698 DOI: 10.1080/14737140.2023.2245140] [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: 12/11/2022] [Accepted: 08/02/2023] [Indexed: 08/09/2023]
Abstract
INTRODUCTION The current standard of care of locally advanced non-small cell lung cancer (LA-NSCLC) is concurrent chemoradiation, followed by consolidation durvalumab. However, there is evidence that the efficacy of chemoradiation and also immunotherapy in many oncogene-positive LA-NSCLC are attenuated, and dependent on the subgroup. AREAS COVERED We will firstly review the outcomes of standard-of-care therapy in oncogene-driven LA-NSCLC. We looked at various oncogene driven subgroups and the tumor microenvironment that may explain differential response. Finally, we review the role of targeted therapy in the treatment of LA-NSCLC. EXPERT OPINION Each oncogene-positive subgroup should be treated as its own entity, and continued efforts should be undertaken to incorporate targeted therapy, which is likely to yield superior survival outcomes if trial design can be optimized and toxicities can be managed.
Collapse
Affiliation(s)
- Jerold Loh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Jia Li Low
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Manavi Sachdeva
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Peter Qj Low
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Rachel Su Jen Wong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Yiqing Huang
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| | - Puey Ling Chia
- Department of Medical Oncology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Ross A Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, Singapore, Singapore
| |
Collapse
|
15
|
Kwok HH, Yang J, Lam DCL. Breaking the Invisible Barriers: Unleashing the Full Potential of Immune Checkpoint Inhibitors in Oncogene-Driven Lung Adenocarcinoma. Cancers (Basel) 2023; 15:2749. [PMID: 37345086 DOI: 10.3390/cancers15102749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
The rapid development of targeted therapy paved the way toward personalized medicine for advanced non-small cell lung cancer (NSCLC). Lung adenocarcinoma (ADC) harboring actionable genetic alternations including epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), Kirsten rat sarcoma virus (ALK) and c-ros oncogene 1 (ROS1) treated with tyrosine kinase inhibitors (TKIs) incurred lesser treatment toxicity but better therapeutic responses compared with systemic chemotherapy. Angiogenesis inhibitors targeting vascular endothelial growth factor (VEGF) have also shown an increase in overall survival (OS) for NSCLC patients. However, acquired resistance to these targeted therapies remains a major obstacle to long-term maintenance treatment for lung ADC patients. The emergence of immune checkpoint inhibitors (ICIs) against programmed cell death protein 1 (PD-1) or programmed cell death-ligand 1 (PD-L1) has changed the treatment paradigm for NSCLC tumors without actionable genetic alternations. Clinical studies have suggested, however, that there are no survival benefits with the combination of targeted therapy and ICIs. In this review, we will summarize and discuss the current knowledge on the tumor immune microenvironment and the dynamics of immune phenotypes, which could be crucial in extending the applicability of ICIs for this subpopulation of lung ADC patients.
Collapse
Affiliation(s)
- Hoi-Hin Kwok
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jiashuang Yang
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - David Chi-Leung Lam
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
16
|
Mohanty SK, Mishra SK, Amin MB, Agaimy A, Fuchs F. Role of Surgical Pathologist for the Detection of Immuno-oncologic Predictive Factors in Non-small Cell Lung Cancers. Adv Anat Pathol 2023; 30:174-194. [PMID: 37037418 DOI: 10.1097/pap.0000000000000395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Until very recently, surgery, chemotherapy, and radiation therapy have been the mainstay of treatment in non-small cell carcinomas (NSCLCs). However, recent advances in molecular immunology have unveiled some of the complexity of the mechanisms regulating cellular immune responses and led to the successful targeting of immune checkpoints in attempts to enhance antitumor T-cell responses. Immune checkpoint molecules such as cytotoxic T-lymphocyte associated protein-4, programmed cell death protein-1, and programmed death ligand (PD-L) 1 have been shown to play central roles in evading cancer immunity. Thus, these molecules have been targeted by inhibitors for the management of cancers forming the basis of immunotherapy. Advanced NSCLC has been the paradigm for the benefits of immunotherapy in any cancer. Treatment decisions are made based on the expression of PD-L1 on the tumor cells and the presence or absence of driver mutations. Patients with high PD-L1 expression (≥50%) and no driver mutations are treated with single-agent immunotherapy whereas, for all other patients with a lower level of PD-L1 expression, a combination of chemotherapy and immunotherapy is preferred. Thus, PD-L1 blockers are the only immunotherapeutic agents approved in advanced NSCLC without any oncogenic driver mutations. PD-L1 immunohistochemistry, however, may not be the best biomarker in view of its dynamic nature in time and space, and the benefits may be seen regardless of PD -L1 expression. Each immunotherapy molecule is prescribed based on the levels of PD-L1 expression as assessed by a Food and Drug Administration-approved companion diagnostic assay. Other biomarkers that have been studied include tumor mutational burden, the T-effector signature, tumor-infiltrating lymphocytes, radiomic assays, inflammation index, presence or absence of immune-related adverse events and specific driver mutations, and gut as well as local microbiome. At the current time, none of these biomarkers are routinely used in the clinical decision-making process for immunotherapy in NSCLC. However, in individual cases, they can be useful adjuncts to conventional therapy. This review describes our current understanding of the role of biomarkers as predictors of response to immune checkpoint molecules. To begin with a brief on cancer immunology in general and in NSCLC, in particular, is discussed. In the end, recent advancements in laboratory techniques for refining biomarker assays are described.
Collapse
Affiliation(s)
- Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, India and CORE Diagnostics, Gurgaon, HR
| | - Sourav K Mishra
- Department of Medical Oncology, All India Institute of Medical Sciences, DL, India
| | - Mahul B Amin
- Departments of Pathology and Laboratory Medicine and Urology, University of Tennessee Health Science Center, Memphis, TN
| | - Abbas Agaimy
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Fuchs
- Department of Internal Medicine-1, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen University Hospital and Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| |
Collapse
|
17
|
Lee J, Piotrowska Z, Soo R, Cho BC, Lim SM. Combatting acquired resistance to osimertinib in EGFR-mutant lung cancer. Ther Adv Med Oncol 2022; 14:17588359221144099. [PMID: 36544540 PMCID: PMC9761802 DOI: 10.1177/17588359221144099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022] Open
Abstract
The discovery of activating mutations in epidermal growth factor receptor (EGFR) in non-small-cell lung cancer transformed the care and prognosis of patients and heralded the era of 'personalized medicine' in thoracic oncology. Osimertinib, a third-generation EGFR inhibitor, has been established as the preferred EGFR inhibitor for newly diagnosed patients which urged the need to develop treatment options for patients progressing on first-line osimertinib. However, acquired resistance invariably emerges and numerous efforts have been attempted to delay or overcome acquired resistance. In this article, we thoroughly reviewed the current understanding of osimertinib resistance mechanisms and explored the established and emerging treatment options. Newer treatment strategies targeting EGFR-dependent or -independent resistance mechanisms, novel approaches using bispecific antibodies and antibody-drug conjugates will be discussed. Moreover, what to do with brain only progression, and how to incorporate immunotherapy in EGFR-mutant lung cancer will be discussed. Lastly, future perspectives on the ongoing clinical trials and combination of front-line therapy will be introduced.
Collapse
Affiliation(s)
| | | | - Ross Soo
- Department of Haematology-Oncology, National
University Cancer Institute, Singapore, Singapore
| | - Byoung Chul Cho
- Division of Medical Oncology, Yonsei Cancer
Center, Yonsei University College of Medicine, Seoul, Korea
| | | |
Collapse
|
18
|
Wan Y, Qian Y, Wang Y, Fang F, Wu G. Prognostic value of Beclin 1, EGFR and ALK in non-squamous non-small cell lung cancer. Discov Oncol 2022; 13:127. [PMID: 36401689 PMCID: PMC9675885 DOI: 10.1007/s12672-022-00586-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/31/2022] [Indexed: 11/21/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most malignant tumors. The study was carried out to investigate the prognostic value of Beclin 1, EGFR and ALK for this cancer. Patients diagnosed with non-squamous NSCLC and admitted to our hospital from January 2011 to September 2016 were analyzed. Expression of Beclin 1 and mutation of EGFR and ALK were assessed using polymerase chain reaction (PCR) and fluorescent in situ hybridization (FISH) and analyzed for their relationship with demographic and clinical characteristics of the patients. Multivariate Cox regression models were applied to analyze the risk factors associated with survival and receiver response curves (ROC) were plotted to determine the prognostic value of Beclin 1, EGFR and ALK for patients with non-squamous NSCLC. Compared with adjacent normal tissue, Beclin 1 expression was elevated in the cancer tissue significantly; assessments of EGFR and ALK mutations showed that out of the 480 patients, 233 (48.5%) and 75 (12.6%) patients had EGFR and ALK mutations. Univariate analysis revealed that Beclin 1 level, EGFR and ALK mutations were associated with lymph node metastasis, TNM stage, tumor differentiation and prognosis, but not with gender, age and smoking status. The Kaplan-Meier survival analysis indicated that low Beclin 1 expression and positive EGFR and ALK rearrangements were associated with higher survival rate and longer progress-free survival (PFS). Multivariate Cox regression analysis showed that Beclin 1, EGFR, ALK mutations, tumor differentiation grade, TNM stage and lymph node metastasis were independently associated with PFS. ROC analysis showed that Beclin 1, EGFR and ALK were significant predictors for PFS; the areas under curve (AUC) for Beclin 1, EGFR and ALK were 0.812 (P = 0.018, cut-off value: 1.2), 0.781 (P = 0.011, cut-off value: 15%) and 0.722 (P = 0.010, cut-off value: 11%), respectively, suggesting that they have significant prognostic value for lung cancer patients. Our data indicate that Beclin 1, EGFR and ALK genes are associated with the prognosis of patients with non-squamous NSCLC. High Beclin 1 expression and negative EGFR and ALK mutations predict a poor prognosis with PFS.
Collapse
Affiliation(s)
- Yanhui Wan
- Department of Thoracic Surgery, the First Affiliated Hospital of Shenzhen University/Shenzhen Second People's Hospital, 3002 Futian Road , Shenzhen, 518000, China.
| | - Youhui Qian
- Department of Thoracic Surgery, the First Affiliated Hospital of Shenzhen University/Shenzhen Second People's Hospital, 3002 Futian Road , Shenzhen, 518000, China
| | - Youyu Wang
- Department of Thoracic Surgery, the First Affiliated Hospital of Shenzhen University/Shenzhen Second People's Hospital, 3002 Futian Road , Shenzhen, 518000, China
| | - Fuyuan Fang
- Department of Thoracic Surgery, the First Affiliated Hospital of Shenzhen University/Shenzhen Second People's Hospital, 3002 Futian Road , Shenzhen, 518000, China
| | - Guodong Wu
- Department of Thoracic Surgery, the First Affiliated Hospital of Shenzhen University/Shenzhen Second People's Hospital, 3002 Futian Road , Shenzhen, 518000, China
| |
Collapse
|
19
|
Xie D, Xu F, Zhu W, Pu C, Huang S, Lou K, Wu Y, Huang D, He C, Hu H. Delta radiomics model for the prediction of progression-free survival time in advanced non-small-cell lung cancer patients after immunotherapy. Front Oncol 2022; 12:990608. [PMID: 36276082 PMCID: PMC9583844 DOI: 10.3389/fonc.2022.990608] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022] Open
Abstract
Objective To assess the validity of pre- and posttreatment computed tomography (CT)-based radiomics signatures and delta radiomics signatures for predicting progression-free survival (PFS) in stage III-IV non-small-cell lung cancer (NSCLC) patients after immune checkpoint inhibitor (ICI) therapy. Methods Quantitative image features of the largest primary lung tumours were extracted on CT-enhanced imaging at baseline (time point 0, TP0) and after the 2nd-3rd immunotherapy cycles (time point 1, TP1). The critical features were selected to construct TP0, TP1 and delta radiomics signatures for the risk stratification of patient survival after ICI treatment. In addition, a prediction model integrating the clinicopathologic risk characteristics and phenotypic signature was developed for the prediction of PFS. Results The C-index of TP0, TP1 and delta radiomics models in the training and validation cohort were 0.64, 0.75, 0.80, and 0.61, 0.68, 0.78, respectively. The delta radiomics score exhibited good accuracy for distinguishing patients with slow and rapid progression to ICI treatment. The predictive accuracy of the combined prediction model was higher than that of the clinical prediction model in both training and validation sets (P<0.05), with a C-index of 0.83 and 0.70, respectively. Additionally, the delta radiomics model (C-index of 0.86) had a higher predictive accuracy compared to PD-L1 expression (C-index of 0.50) (P<0.0001). Conclusions The combined prediction model including clinicopathologic characteristics (tumour anatomical classification and brain metastasis) and the delta radiomics signature could achieve the individualized prediction of PFS in ICIs-treated NSCLC patients.
Collapse
Affiliation(s)
- Dong Xie
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Radiology, Shaoxing Second Hospital, Shaoxing, China
| | - Fangyi Xu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenchao Zhu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cailing Pu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shaoyu Huang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Radiology, Ningbo Medical Center LiHuili Hospital, Ningbo, China
| | - Kaihua Lou
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Wu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dingpin Huang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cong He
- Department of Radiology, Shaoxing Second Hospital, Shaoxing, China
| | - Hongjie Hu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Hongjie Hu,
| |
Collapse
|
20
|
Ren X, Li Y, Nishimura C, Zang X. Crosstalk between the B7/CD28 and EGFR pathways: Mechanisms and therapeutic opportunities. Genes Dis 2022; 9:1181-1193. [PMID: 35873032 PMCID: PMC9293717 DOI: 10.1016/j.gendis.2021.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/31/2022] Open
Abstract
Somatic activating mutations in the epidermal growth factor receptor (EGFR) are one of the most common oncogenic drivers in cancers such as non-small-cell lung cancer (NSCLC), metastatic colorectal cancer, glioblastoma, head and neck cancer, pancreatic cancer, and breast cancer. Molecular-targeted agents against EGFR signaling pathways have shown robust clinical efficacy, but patients inevitably experience acquired resistance. Although immune checkpoint inhibitors (ICIs) targeting PD-1/PD-L1 have exhibited durable anti-tumor responses in a subset of patients across multiple cancer types, their efficacy is limited in cancers harboring activating gene alterations of EGFR. Increasing studies have demonstrated that upregulation of new B7/CD28 family members such as B7-H3, B7x and HHLA2, is associated with EGFR signaling and may contribute to resistance to EGFR-targeted therapies by creating an immunosuppressive tumor microenvironment (TME). In this review, we discuss the regulatory effect of EGFR signaling on the PD-1/PD-L1 pathway and new B7/CD28 family member pathways. Understanding these interactions may inform combination therapeutic strategies and potentially overcome the current challenge of resistance to EGFR-targeted therapies. We also summarize clinical data of anti-PD-1/PD-L1 therapies in EGFR-mutated cancers, as well as ongoing clinical trials of combination of EGFR-targeted therapies and anti-PD-1/PD-L1 immunotherapies.
Collapse
Affiliation(s)
- Xiaoxin Ren
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Yixian Li
- Division of Pediatric Hematology/Oncology/Transplant and Cellular Therapy, Children's Hospital at Montefiore, Bronx, NY 10467, USA
| | - Christopher Nishimura
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY 10461, USA.,Department of Medicine, Albert Einstein College of Medicine, New York, NY 10461, USA.,Department of Urology, Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
21
|
[Advances in ICIs Therapy after TKIs Resistance in Patients with EGFR Mutant NSCLC: A Review]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:601-608. [PMID: 36002197 PMCID: PMC9411951 DOI: 10.3779/j.issn.1009-3419.2022.101.37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The follow-up treatment of patients with advanced non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) mutation after drug resistance to EGFR-tyrosine kinase inhibitors (TKIs) have become a hotspot and difficulty at present. Immune checkpoint inhibitors (ICIs) therapy is a new and important choice for these patients, but many studies have shown unsatisfactory efficacy. However, some domestic and foreign studies have shown that ICIs combination therapy is still effective in some patients with positive driver genes and drug resistance after targeted therapy. So, in the era of immunotherapy, what are the differences in the efficacy of different combination immunotherapy strategies for different patients? What are the factors that affect efficacy? What are the interrelationships between these factors and other immunotherapy efficacy prediction biomarkers? All these problems have broad and important research value.
.
Collapse
|
22
|
Liu QX, Wei JG, Chen YY, Wang JF. Efficacy and safety of pembrolizumab monotherapy in EGFR-mutant squamous cell lung cancer with PD-L1 over-expression: A case report. Medicine (Baltimore) 2022; 101:e30099. [PMID: 35984168 PMCID: PMC9387979 DOI: 10.1097/md.0000000000030099] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR)-mutant nonsmall cell lung cancer (NSCLC) patients are less likely to be programmed death-ligand 1 (PD-L1)-positive compared with wild-type EGFR mutant tumors. Given the rarity of actionable driver genes in squamous cell lung cancer (SQCC), the frequency of SQCC patients simultaneously carrying EGFR driver gene mutation and having PD-L1 over-expression is extremely low. Studies on the effectiveness and safety of EGFR-TKIs or immune-checkpoint inhibitors (ICIs) in this subset of patients are lacking. PATIENT CONCERNS The patient suffered from coughing and chest pain for 1 month. A chest CT revealed a mass with a cavity in the right lung, enlarged mediastinal lymph nodes, diffuse pleural thickening in the right pleura, and pleural effusion of the right chest. DIAGNOSIS A pleural biopsy was performed using a video-assisted thoracoscope. The pathological examination revealed a poorly differentiated squamous cell carcinoma of lung. Further genetic testing identified exon 19 deletion mutation in EGFR with abundance of 0.27%. Meanwhile, immunohistochemical PD-L1 analysis showed a TPS of 90%. INTERVENTIONS The patient was initially resistant to EGFR-TKIs but exhibited a rapid and marked response to pembrolizumab. OUTCOMES After 5 cycles of pembrolizumab monotherapy, the patient developed Grade 3 immune-related dermatitis, and ICI therapy was suspended. CONCLUSIONS ICI monotherapy could be an effective therapy in SQCC patients with low-abundance of EGFR mutations and PD-L1 over-expression. However, close attention should be paid to immune-related adverse events.
Collapse
Affiliation(s)
| | - Jian-Guo Wei
- Department of Pathology, Shaoxing People’s Hospital, Shaoxing 312000, Zhejiang Province
| | - Yi-Yi Chen
- Wenzhou Medical University, Department of Clinical Medicine, Wenzhou 325000, Zhejiang Province, China
| | - Jian-Fang Wang
- Department of Medical Oncology
- *Correspondence: Jian-Fang Wang, Department of Medical Oncology, Shaoxing People’s Hospital, No. 568 North Zhongxing Road, Shaoxing 31200, Zhejiang Province, China (e-mail: )
| |
Collapse
|
23
|
Nakagawa N, Kawakami M. Choosing the optimal immunotherapeutic strategies for non-small cell lung cancer based on clinical factors. Front Oncol 2022; 12:952393. [PMID: 36033471 PMCID: PMC9414869 DOI: 10.3389/fonc.2022.952393] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/04/2022] [Indexed: 11/24/2022] Open
Abstract
The treatment landscape of advanced non-small cell lung cancer (NSCLC) has changed dramatically since the emergence of immune checkpoint inhibitors (ICIs). Although some patients achieve long survival with relatively mild toxicities, not all patients experience such benefits from ICI treatment. There are several ways to use ICIs in NSCLC patients, including monotherapy, combination immunotherapy, and combination chemoimmunotherapy. Decision-making in the selection of an ICI treatment regimen for NSCLC is complicated partly because of the absence of head-to-head prospective comparisons. Programmed death-ligand 1 (PD-L1) expression is currently considered a standard biomarker for predicting the efficacy of ICIs, although some limitations exist. In addition to the PD-L1 tumor proportion score, many other clinical factors should also be considered to determine the optimal treatment strategy for each patient, including age, performance status, histological subtypes, comorbidities, status of oncogenic driver mutation, and metastatic sites. Nevertheless, evidence of the efficacy and safety of ICIs with some specific conditions of these factors is insufficient. Indeed, patients with poor performance status, oncogenic driver mutations, or interstitial lung disease have frequently been set as ineligible in randomized clinical trials of NSCLC. ICI use in these patients is controversial and remains to be discussed. It is important to select patients for whom ICIs can benefit the most from these populations. In this article, we review previous reports of clinical trials or experience in using ICIs in NSCLC, focusing on several clinical factors that are associated with treatment outcomes, and then discuss the optimal ICI treatment strategies for NSCLC.
Collapse
|
24
|
Ma T, Jiao J, Huo R, Li X, Fang G, Zhao Q, Liu W, Han X, Xi C, Wang Y, Shang Y. PD-L1 expression, tumor mutational burden, and immune cell infiltration in non-small cell lung cancer patients with epithelial growth factor receptor mutations. Front Oncol 2022; 12:922899. [PMID: 35992815 PMCID: PMC9389166 DOI: 10.3389/fonc.2022.922899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundImmunotherapy using programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) inhibitors seems less effective in non-small cell lung cancer (NSCLC) patients with epithelial growth factor receptor (EGFR) mutations. Varied responses to PD-1/PD-L1 inhibitors have recently been observed in NSCLC patients harboring different types of EGFR mutations. Some EGFR-mutated NSCLC patients may benefit from PD-1/PD-L1 inhibitors. At present, PD-L1 expression, tumor mutational burden (TMB), and tumor immune microenvironment (TIME) are biomarkers for predicting the efficacy of PD-1/PD-L1 inhibitors in NSCLC patients. We retrospectively evaluated PD-L1 expression, TMB, and immune cell infiltration in NSCLC patients with EGFR mutation subtypes.MethodsPD-L1 expression, TMB, and the abundance of immune cell infiltration in NSCLC patients were evaluated in public databases and clinical samples. TMB was detected using the NGS technique, PD-L1 was detected using immunohistochemistry, and the abundance of immune cell infiltration in NSCLC samples was detected using multiple immunohistochemistry.ResultsPD-L1 expression and TMB were lower in EGFR-mutated NSCLCs than in wild-type patients. Differences in the abundance of immune cell infiltration were also observed between EGFR-mutated and wild-type NSCLC. The expression of PD-L1, TMB, and abundance of immune cell infiltration were different in patients harboring different subtypes of EGFR mutations. Patients with uncommon EGFR mutations, especially the G719X mutation, showed higher TMB and expressions of PD-L1 than classical EGFR mutations. M1 macrophages were higher in uncommon EGFR mutations than classical EGFR mutations.ConclusionsThe expression of PD-L1 and TMB in uncommon EGFR-mutated NSCLCs, especially the G719X mutation, were higher than those for classical EGFR-mutated NSCLCs and similar to EGFR wild-type. The abundance of immune cell infiltration in uncommon EGFR-mutated NSCLCs was similar to that in EGFR wild-type. Our findings suggest that uncommon EGFR-mutated NSCLCs may benefit from PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Tiantian Ma
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Jin Jiao
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Ran Huo
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Xiaofang Li
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Guotao Fang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Qi Zhao
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Weiwei Liu
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Xiao Han
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Chenglin Xi
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Yanan Wang
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Yanhong Shang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
- *Correspondence: Yanhong Shang,
| |
Collapse
|
25
|
Punekar SR, Shum E, Grello CM, Lau SC, Velcheti V. Immunotherapy in non-small cell lung cancer: Past, present, and future directions. Front Oncol 2022; 12:877594. [PMID: 35992832 PMCID: PMC9382405 DOI: 10.3389/fonc.2022.877594] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Many decades in the making, immunotherapy has demonstrated its ability to produce durable responses in several cancer types. In the last decade, immunotherapy has shown itself to be a viable therapeutic approach for non-small cell lung cancer (NSCLC). Several clinical trials have established the efficacy of immune checkpoint blockade (ICB), particularly in the form of anti-programmed death 1 (PD-1) antibodies, anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibodies and anti-programmed death 1 ligand (PD-L1) antibodies. Many trials have shown progression free survival (PFS) and overall survival (OS) benefit with either ICB alone or in combination with chemotherapy when compared to chemotherapy alone. The identification of biomarkers to predict response to immunotherapy continues to be evaluated. The future of immunotherapy in lung cancer continues to hold promise with the development of combination therapies, cytokine modulating therapies and cellular therapies. Lastly, we expect that innovative advances in technology, such as artificial intelligence (AI) and machine learning, will begin to play a role in the future care of patients with lung cancer.
Collapse
|
26
|
Shi C, Wang Y, Xue J, Zhou X. Immunotherapy for EGFR-mutant advanced non-small-cell lung cancer: Current status, possible mechanisms and application prospects. Front Immunol 2022; 13:940288. [PMID: 35935943 PMCID: PMC9353115 DOI: 10.3389/fimmu.2022.940288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are effective against advanced and even perioperative non-small-cell lung cancer (NSCLC) and result in durable clinical benefit, regardless of programmed death ligand-1 (PD-L1) expression status in cancer. Existing clinical evidence shows that the effect of immunotherapy in patients with EGFR-mutant NSCLC after the development of tyrosine kinase inhibitor (TKI) resistance is not satisfactory. However, compared with monotherapy, ICIs combined with chemotherapy can improve the efficacy. Encouragingly, compared with that of patients with sensitive mutations, the progression-free survival of patients with rare mutations who were treated with ICIs was increased. Adequately maximizing the efficacy of ICIs in EGFR-mutant NSCLC patients is worth exploring. In this review, we described preclinical and clinical studies of ICIs or combined therapy for EGFR-mutant NSCLC. We further focused on EGFR mutations and the cancer immune response, with particular attention given to the role of EGFR activation in the cancer-immunity cycle. The mechanisms for the natural resistance to ICIs were explored to identify corresponding countermeasures that made more EGFR-mutant NSCLC patients benefit from ICIs.
Collapse
Affiliation(s)
- Chunyan Shi
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- The Department of Oncology, Jiujiang No.1 People’s Hospital, Jiujiang, China
| | - Yan Wang
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Xue
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojuan Zhou
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Xiaojuan Zhou,
| |
Collapse
|
27
|
PD-L1 strong expressions affect the clinical outcomes of osimertinib in treatment naïve advanced EGFR-mutant non-small cell lung cancer patients. Sci Rep 2022; 12:9753. [PMID: 35697720 PMCID: PMC9192769 DOI: 10.1038/s41598-022-13102-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 05/20/2022] [Indexed: 12/14/2022] Open
Abstract
The impact of strong Programmed Death-ligand 1 (PD-L1) expression on the clinical outcomes of osimertinib in treatment naïve advanced Epidermal Growth Factor Receptor (EGFR)-mutant Non-small Cell Lung Cancer (NSCLC) patients remains uncertain. We enrolled advanced NSCLC patients who harbored sensitizing EGFR mutation and were treated first-line with osimertinib between 2017 and 2021. The PD-L1 expression level was also tested. A total of 85 patients were included. The objective response rate to osimertinib was 78.9%, with the disease control rate being 90.8%. Median Progression-free Survival (PFS) was 22.1 months, while median Overall Survival (OS) was not reached (NR). Patients with the exon 19 deletion experienced better PFS than those with the exon 21 L858R mutation (NR vs 12.4 months, aHR 0.24 (95% CI, 0.10 to 0.57); p = 0.001). Seventy-one of these 85 patients had reported on their PD-L1 expression. Patients with a PD-L1 < 50% experienced longer PFS than patients with a PD-L1 ≧50% (26.5 vs 9.7 months, aHR 0.19 (95% CI, 0.06 to 0.67); p = 0.009). Additionally, patients with a PD-L1 < 50% experienced better OS than those with a PD-L1 ≧50% (NR vs 25.4 months, aHR 0.09 (95% CI, 0.01 to 0.70); p = 0.021). Strong expressions of PD-L1 in treatment naïve advanced EGFR-mutant NSCLC patients were associated with poor prognoses in those undergoing treatment with osimertinib as first-line therapy.
Collapse
|
28
|
Madeddu C, Donisi C, Liscia N, Lai E, Scartozzi M, Macciò A. EGFR-Mutated Non-Small Cell Lung Cancer and Resistance to Immunotherapy: Role of the Tumor Microenvironment. Int J Mol Sci 2022; 23:6489. [PMID: 35742933 PMCID: PMC9224267 DOI: 10.3390/ijms23126489] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is a leading cause of cancer-related deaths worldwide. About 10-30% of patients with non-small cell lung cancer (NSCLC) harbor mutations of the EGFR gene. The Tumor Microenvironment (TME) of patients with NSCLC harboring EGFR mutations displays peculiar characteristics and may modulate the antitumor immune response. EGFR activation increases PD-L1 expression in tumor cells, inducing T cell apoptosis and immune escape. EGFR-Tyrosine Kinase Inhibitors (TKIs) strengthen MHC class I and II antigen presentation in response to IFN-γ, boost CD8+ T-cells levels and DCs, eliminate FOXP3+ Tregs, inhibit macrophage polarization into the M2 phenotype, and decrease PD-L1 expression in cancer cells. Thus, targeted therapy blocks specific signaling pathways, whereas immunotherapy stimulates the immune system to attack tumor cells evading immune surveillance. A combination of TKIs and immunotherapy may have suboptimal synergistic effects. However, data are controversial because activated EGFR signaling allows NSCLC cells to use multiple strategies to create an immunosuppressive TME, including recruitment of Tumor-Associated Macrophages and Tregs and the production of inhibitory cytokines and metabolites. Therefore, these mechanisms should be characterized and targeted by a combined pharmacological approach that also concerns disease stage, cancer-related inflammation with related systemic symptoms, and the general status of the patients to overcome the single-drug resistance development.
Collapse
Affiliation(s)
- Clelia Madeddu
- Department of Medical Sciences and Public Health, Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, University of Cagliari, 09100 Cagliari, Italy; (C.M.); (N.L.); (E.L.); (M.S.)
| | - Clelia Donisi
- Department of Medical Sciences and Public Health, Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, University of Cagliari, 09100 Cagliari, Italy; (C.M.); (N.L.); (E.L.); (M.S.)
| | - Nicole Liscia
- Department of Medical Sciences and Public Health, Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, University of Cagliari, 09100 Cagliari, Italy; (C.M.); (N.L.); (E.L.); (M.S.)
| | - Eleonora Lai
- Department of Medical Sciences and Public Health, Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, University of Cagliari, 09100 Cagliari, Italy; (C.M.); (N.L.); (E.L.); (M.S.)
| | - Mario Scartozzi
- Department of Medical Sciences and Public Health, Medical Oncology Unit, “Azienda Ospedaliero Universitaria” of Cagliari, University of Cagliari, 09100 Cagliari, Italy; (C.M.); (N.L.); (E.L.); (M.S.)
| | - Antonio Macciò
- Gynecologic Oncology Unit, ARNAS G. Brotzu, Department of Surgical Sciences, University of Cagliari, 09100 Cagliari, Italy;
| |
Collapse
|
29
|
Chan DWK, Choi HCW, Lee VHF. Treatment-Related Adverse Events of Combination EGFR Tyrosine Kinase Inhibitor and Immune Checkpoint Inhibitor in EGFR-Mutant Advanced Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2022; 14:2157. [PMID: 35565285 PMCID: PMC9102470 DOI: 10.3390/cancers14092157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/25/2022] [Indexed: 12/13/2022] Open
Abstract
(1) Background: We performed a meta-analysis to examine whether combined epidermal growth factor tyrosine kinase inhibitor (EGFR-TKI) and immune checkpoint inhibitor (ICI) increases treatment-related adverse events (trAEs) in advanced non-small cell lung cancer (NSCLC). (2) Methods: Articles from MEDLINE, EMBASE, and Cochrane databases were searched. Proportions and odds ratios (ORs) of the pooled incidence of overall and organ-specific trAEs in combination EGFR-TKI and ICI were compared to TKI monotherapy. (3) Results: Eight studies fulfilled our selection criteria. Any-grade organ-specific trAEs were more common in combination EGFR-TKI and ICI than TKI monotherapy (skin: OR = 1.19, p = 0.012; gastrointestinal tract: OR = 1.04, p = 0.790; ILD: OR = 1.28, p = 0.001). Grade ≥ 3 trAEs were also more frequent in combination treatment (skin: OR = 1.13, p = 0.082; gastrointestinal tract: OR = 1.13, p = 0.076; ILD: OR = 1.16, p = 0.003). (4) Conclusions: A higher proportion of grade ≥3 skin and gastrointestinal trAEs and ILDs was observed in combination TKI and ICI compared to TKI alone. Caution has to be taken when interpreting the results owing to the small number of studies included in this meta-analysis.
Collapse
Affiliation(s)
- Daisy Wai-Ka Chan
- LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| | - Horace Cheuk-Wai Choi
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| | - Victor Ho-Fun Lee
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
- Department of Clinical Oncology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
30
|
Serum Level of CEACAM1 in Patients with Nonsmall Cell Lung Cancer and Its Clinical Significance in Cancer Tissue. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7948010. [PMID: 35449849 PMCID: PMC9018175 DOI: 10.1155/2022/7948010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/23/2022]
Abstract
Objective To analyze the expression of CEACAM1 in serum of patients with nonsmall cell lung cancer (NSCLC) and to explore the correlation and clinical significance between the expression of CEACAM1 and pathological parameters of NSCLC tissue. Methods A total of 100 patients with NSCLC who underwent tumor resection were screened. Another 100 healthy patients in physical examination department were selected as control group. Venous blood and cancer tissue samples were collected. The expression of CEACAM1, TGF-β, VEGF-A, and IL-8 was detected. Results The results of various indicators in the lung cancer group were much higher than those in the healthy group; CEACAM1 was significantly positively correlated with TGF-β expression. The later the clinical stage and the higher the degree of differentiation of cancer tissue specimens, the more the expression of CEACAM1 in serum samples. Conclusions The expression level of CEACAM1 in the serum of NSCLC patients is strongly correlated with TGF-β, VEGF-A, and IL-8, indicating that serum CEACAM1 and TGF-β levels can predict the occurrence, progression, and prognosis of lung cancer and provide a target for future targeted therapy of lung cancer.
Collapse
|
31
|
Risk of Hepatitis B Virus Reactivation in Patients Treated With Immunotherapy for Anti-cancer Treatment. Clin Gastroenterol Hepatol 2022; 20:898-907. [PMID: 34182151 DOI: 10.1016/j.cgh.2021.06.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Hepatitis B virus (HBV) reactivation is a well-known complication in patients with chronic hepatitis B treated with cytotoxic chemotherapy. However, the risk of HBV reactivation through use of immune checkpoint inhibitors (ICIs) is not well understood. Therefore, we aimed to evaluate the risk of HBV reactivation and hepatic adverse events in patients with cancer receiving ICIs according to cancer type and virologic serology. METHODS This historical cohort study included 3465 patients with cancer treated with ICIs between January 2015 and September 2020. The primary outcome was the occurrence of HBV reactivation, and the secondary outcome was presence of hepatic adverse events during ICI treatment. RESULTS The mean patient age was 62.2 years, and 68.8% of patients were men. Of the 3465 eligible patients, 511 (14.7%) showed hepatitis B surface antigen (HBsAg) positivity. The incidence rates of HBV reactivation of the total patients, HBsAg-positive patients, and HBsAg-negative patients were 0.14% (5/3465), 1.0% (5/511), and 0.0% (0/2954), respectively. Among HBsAg-positive patients, HBV reactivation occurred at a rate of 0.5% (2/409) and 2.9% (3/102) in patients with and without hepatocellular carcinoma, respectively. The HBV reactivation rates were 0.4% (2/464) and 6.4% (3/47) in patients with and without antiviral prophylaxis, respectively. Grade 3-4 hepatitis occurred in 23 (4.5%) HBsAg-positive, and 218 (7.4%) HBsAg-negative patients. No HBV-related fatality occurred. Only 2 patients (0.4%) experienced HBsAg seroclearance after ICI treatment among HBsAg-positive patients. CONCLUSIONS In general, HBV reactivation was rarely observed in patients with antiviral prophylaxis while undergoing ICI treatment. However, HBV reactivation may occur in HBsAg-positive patients without antiviral prophylaxis or noncompliant with antiviral prophylaxis.
Collapse
|
32
|
Immunological effect of tyrosine kinase inhibitors on the tumor immune environment in non‑small cell lung cancer (Review). Oncol Lett 2022; 23:165. [PMID: 35414830 PMCID: PMC8988264 DOI: 10.3892/ol.2022.13285] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/28/2022] [Indexed: 02/05/2023] Open
Abstract
Acquired resistance to tyrosine kinase inhibitors (TKIs) limits the duration of antitumor effects and impairs the survival of patients with oncogene-driven non-small cell lung cancer (NSCLC). At present, little is known about the immunomodulatory ability of TKIs during the entire treatment period, including the drug-sensitive and drug-resistant periods. The present review aimed to comprehensively explore the dynamic changes in the tumor microenvironment (TME) during TKI treatment in NSCLC. Previous clinical and preclinical studies from medical and health databases related to NSCLC are reviewed. During the response period, cytotoxic immune cells accumulate in the TME and contribute to the formation of an inflammatory microenvironment. During the resistance period, the number of immunosuppressive cells increases, as does the expression of immune checkpoint proteins, which are critical mechanisms for tumor progression. The combination of targeted therapy and immunotherapy has been explored in multiple studies, and preliminary data showed controversial results. Extensive studies are needed to confirm the criteria of the selected patient subgroups and the toxicity profiles of EGFR TKIs and immune checkpoint inhibitors (ICIs). At present, the reagents targeting other immune cells, cytokines and related pathways remain underexplored compared with the revolutionary effect of ICIs in lung cancer. In the future, the precisely selected regimens for combination treatment should be further investigated in carefully designed xenograft models and clinical trials.
Collapse
|
33
|
Chen Y, Chen Z, Chen R, Fang C, Zhang C, Ji M, Yang X. Immunotherapy-based combination strategies for treatment of EGFR-TKI-resistant NSCLC. Future Oncol 2022; 18:1757-1775. [PMID: 35232247 DOI: 10.2217/fon-2021-0862] [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] [Indexed: 12/11/2022] Open
Abstract
The rapid development of molecular targeted therapy brings hope to patients with advanced non-small-cell lung cancer (NSCLC). However, drug resistance inevitably occurs during treatment with EGFR-tyrosine kinase inhibitors (TKIs). Osimertinib, a third-generation EGFR-TKI, shows a favorable prognosis in T790M-positive NSCLC. Unfortunately, acquired resistance is still a challenge for both patients and clinicians. There is still no consensus on the optimal treatment. PD-1 and its ligand receptor 1 (PD-L1) inhibitors have yielded great progress, especially in patients with no actionable mutations. In this review, the authors take stock of the relationship between EGFR mutations and PD-L1 expression and summarize the important clinical studies on immunotherapy-inhibitor-based treatment in patients with EGFR-TKI-resistant NSCLC.
Collapse
Affiliation(s)
- Yan Chen
- Department of Oncology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China
| | - Zijun Chen
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China
| | - Rui Chen
- Department of Oncology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China
| | - Cheng Fang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China
| | - Chu Zhang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China
| | - Mei Ji
- Department of Oncology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China
| | - Xin Yang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, no 185 Juqian Road, Tianning District, Changzhou, 213003, China
| |
Collapse
|
34
|
Takahara Y, Tanaka T, Ishige Y, Shionoya I, Yamamura K, Sakuma T, Nishiki K, Nakase K, Nojiri M, Kato R, Shinomiya S, Fujimoto Y, Oikawa T, Mizuno S. Efficacy and predictors of rechallenge with immune checkpoint inhibitors in non-small cell lung cancer. Thorac Cancer 2022; 13:624-630. [PMID: 34989146 PMCID: PMC8841726 DOI: 10.1111/1759-7714.14309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The efficacy of rechallenge with immune checkpoint inhibitors (ICIs) in non-small cell lung cancer (NSCLC) patients has not yet been fully clarified. This study aimed to identify the clinical characteristics of patients with NSCLC who benefited from rechallenge with ICIs. METHODS We retrospectively reviewed the clinical records of 24 patients who were diagnosed with NSCLC and rechallenged with ICIs between August 2016 and July 2021. RESULTS Of the 24 patients included in the study, 11 were in the responder group (45.8%) and 13 in the nonresponder group (54.2%). The number of patients who used a different ICI from that used in the initial therapy was significantly higher in the responder group than in the nonresponder group (p = 0.006). Multivariate analysis identified lung metastasis and female sex as significant independent risk factors for nonresponse to rechallenge with ICIs. Compared to the nonresponder group, the duration of treatment after rechallenge with ICIs was significantly longer in the responder group (p = 0.016), and there was a trend toward longer overall survival (p = 0.059). CONCLUSIONS Patients with lung cancer who were rechallenged with ICIs and without progressive disease after initial ICI therapy were able to continue ICI therapy for a longer period of time. This may be associated with longer survival. Patients with lung metastases and female patients are more likely to be nonresponsive to rechallenge with ICIs. Administration of a different type of ICI from that used in the initial ICI therapy may result in disease control.
Collapse
Affiliation(s)
- Yutaka Takahara
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Takuya Tanaka
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Yoko Ishige
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Ikuyo Shionoya
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Kouichi Yamamura
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Takashi Sakuma
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Kazuaki Nishiki
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Keisuke Nakase
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Masafumi Nojiri
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Ryo Kato
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Shohei Shinomiya
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Yuki Fujimoto
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Taku Oikawa
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Shiro Mizuno
- Department of Respiratory Medicine, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| |
Collapse
|
35
|
What is the current role of immunotherapy in EGFR mutant advanced NSCLC? Lung Cancer 2021; 166:253-254. [PMID: 35031176 DOI: 10.1016/j.lungcan.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 11/30/2021] [Accepted: 12/03/2021] [Indexed: 11/23/2022]
|
36
|
White MN, Piper-Vallillo AJ, Gardner RM, Cunanan K, Neal JW, Das M, Padda SK, Ramchandran K, Chen TT, Sequist LV, Piotrowska Z, Wakelee HA. Chemotherapy Plus Immunotherapy Versus Chemotherapy Plus Bevacizumab Versus Chemotherapy Alone in EGFR-Mutant NSCLC After Progression on Osimertinib. Clin Lung Cancer 2021; 23:e210-e221. [PMID: 34887193 DOI: 10.1016/j.cllc.2021.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/04/2021] [Accepted: 11/04/2021] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Patients with EGFR-mutant lung cancer who have had disease progression on osimertinib commonly receive platinum doublet chemotherapy, but whether adding immunotherapy or bevacizumab provides additional benefit is unknown. MATERIALS AND METHODS This was a retrospective analysis at 2 university-affiliated institutions. Patients with EGFR-mutant lung cancer who had progression on osimertinib and received next-line therapy with platinum doublet chemotherapy (chemo), platinum doublet chemotherapy plus immunotherapy (chemo-IO), or platinum doublet chemotherapy plus bevacizumab (chemo-bev), were identified; patients who continued osimertinib with these regimens were included. Efficacy outcomes including duration on treatment (DOT) and overall survival (OS) from the start of chemotherapy were assessed. Associations of treatment regimen with outcomes were evaluated using adjusted Cox regression models, using pairwise comparisons between groups. RESULTS 104 patients were included: 57 received chemo, 12 received chemo-IO, and 35 received chemo-bev. In adjusted models, patients who received chemo-IO had worse OS than did those who received chemo (hazard ratio (HR) 2.66, 95% CI 1.25-5.65; P= .011) or those who received chemo-bev (HR 2.37, 95% CI 1.09-5.65; P= .030). A statistically significant difference in OS could not be detected in patients who received chemo-bev versus those who received chemo (HR 1.50, 95% CI 0.84-2.69; P= .17). CONCLUSION In this retrospective study, giving immunotherapy with platinum doublet chemotherapy after progression on osimertinib was associated with a worse OS compared with platinum doublet chemotherapy alone. Platinum doublet chemotherapy without immunotherapy (with consideration of continuation of osimertinib, in selected cases) is a reasonable choice in this setting, while we await results of clinical trials examining optimal next-line chemotherapy-based regimens in EGFR-mutant lung cancer.
Collapse
Affiliation(s)
- Maya N White
- Department of Medicine, Division of Oncology, Stanford University, Stanford CA
| | - Andrew J Piper-Vallillo
- Department of Medicine, Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA; Department of Medicine, Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA
| | - Rebecca M Gardner
- Quantitative Sciences Unit, Stanford University School of Medicine, Stanford, CA
| | - Kristen Cunanan
- Quantitative Sciences Unit, Stanford University School of Medicine, Stanford, CA
| | - Joel W Neal
- Department of Medicine, Division of Oncology, Stanford University, Stanford CA
| | - Millie Das
- Department of Medicine, Division of Oncology, Stanford University, Stanford CA; Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA
| | - Sukhmani K Padda
- Department of Medicine, Division of Oncology, Stanford University, Stanford CA
| | - Kavitha Ramchandran
- Department of Medicine, Division of Oncology, Stanford University, Stanford CA
| | | | - Lecia V Sequist
- Department of Medicine, Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA
| | - Zofia Piotrowska
- Department of Medicine, Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA
| | - Heather A Wakelee
- Department of Medicine, Division of Oncology, Stanford University, Stanford CA.
| |
Collapse
|
37
|
Peng J, Zhao X, Zhao K, Meng X. Case Report: Long Progression-Free Survival of Immunotherapy for Lung Adenocarcinoma With Epidermal Growth Factor Receptor Mutation. Front Oncol 2021; 11:731429. [PMID: 34631564 PMCID: PMC8495127 DOI: 10.3389/fonc.2021.731429] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 09/02/2021] [Indexed: 12/26/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have been clinically proven to be efficient in non-small cell lung cancer (NSCLC). However, it has also been found that immunotherapy is not effective for all patients. For instance, some patients with epidermal growth factor receptor (EGFR) mutation tumors have a low overall response rate to ICIs. As a result, we retrospectively analyzed the efficacy of anti-programmed death-ligand 1 (anti-PD-L1) blockade (atezolizumab) treatment for a patient with EGFR mutation, and we explored the interaction between immunotherapy and EGFR mutations in NSCLC. Case Presentation A patient, 62-year-old non-smoking female, with lung adenocarcinoma was initially misdiagnosed as EGFR wild type and received a third-line treatment with atezolizumab, experiencing partial response (PR) and progression-free survival (PFS) for 23 months. She had later been confirmed with EGFR L858R mutation prior to taking atezolizumab. On top of that, the patient developed T790M mutation after being administered with atezolizumab instead of EGFR tyrosine kinase inhibitors (TKIs). She started with osimertinib, although the lesion continued to progress. Tumor mutational burden (TMB), PD-L1 expression, and tumor-infiltrating lymphocytes (TILs) had been tested for further analysis. Conclusion The case report and literature review indicate that ICIs might be more effective for L858R mutation than for other EGFR mutant subtypes, which correlates with certain potential predictors such as TMB and concurrent PD-L1 plus CD8+ TIL expression. However, there is no report on progression from non-primary EGFR T790M mutation to T790M mutation of patients who neither previously suffered from EGFR-TKIs nor responded to osimertinib. This case report will offer some information to guide the investigation on how to identify those who can benefit from immunotherapy and those who do not respond to EGFR-TKIs among the patients with EGFR mutations.
Collapse
Affiliation(s)
- Jianfeng Peng
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xianguang Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital, Jinan, China
| | - Kaikai Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China.,Department of Radiation Oncology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital, Jinan, China
| |
Collapse
|
38
|
Sun S, Du W, Sun Q, Zhao X, Qin B, Shi D, Wan C, Wu Z. Driver gene alterations profiling of Chinese non-small cell lung cancer and the effects of co-occurring alterations on immunotherapy. Cancer Med 2021; 10:7360-7372. [PMID: 34599863 PMCID: PMC8525092 DOI: 10.1002/cam4.4178] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/05/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Molecular testing for alterations in oncogenic driver genes and targeted therapies have become standard procedures for non-small cell lung cancer (NSCLC) patients. However, little evidence has shed light on the pattern of co-existence of driver genes in NSCLC, and whether they may have different tumor features affecting immunotherapy is still unclarified. METHODS Genomic alterations in 14 lung cancer-related genes were conducted in 3440 Chinese NSCLC patients using next-generation sequencing. Meanwhile, tumor mutational burden and immunotherapy dataset from the Memorial sloan kettering cancer center (MSKCC) and lung adenocarcinoma dataset from The Cancer Genome Atlas (TCGA) were utilized for analyzing the impact of the co-occurring alterations on patients' survival following immunotherapy. RESULTS In this cohort, 90.17% of patients had at least one somatic alteration in the 14 genes, including 51% of co-occurring alterations. TP53 and epidermal growth factor receptor (EGFR) were the most prevalent genes (54.74% and 53.55%, respectively), followed by KRAS, ERBB2, ALK, PIK3CA, ROS1, RET, MET, BRAF, KIT, FGFR1, PDGFRA, and NRAS. The prevalence of TP53, EGFR, and ERBB2 in our cohort were significantly higher than that from the TCGA database, whereas KRAS, BRAF, and PDGFRA were significantly lower than the latter. Furthermore, the patients who harbored multiple alterations (8.86%, 31/350) in eight driver genes survived longer and have a higher tumor mutation burden compared to the patients with a single alteration. Similar result was found between the patients with co-occurring alteration of EGFR and other driver genes and the patients with single EGFR alteration. Meanwhile, we found a distinct immune cell infiltration feature between patients with single and multiple driver gene alterations, as well as between patients with only EGFR alteration and co-occurring groups. CONCLUSION This study identified a unique driver gene feature and found patients harboring co-occurring alterations of EGFR and other driver genes may benefit from immunotherapy, which may provide more therapeutic selections for EGFR-mutated NSCLC patients and merit additional investigation.
Collapse
Affiliation(s)
- Shengjie Sun
- Medical Oncology DepartmentThe Fifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Wenjuan Du
- Training Department of Medical Service DepartmentGeneral Hospital of People’s Liberation ArmyBeijingChina
| | - Qiong Sun
- Medical Oncology DepartmentThe Fifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Xiao Zhao
- Medical Oncology DepartmentThe Fifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Boyu Qin
- Medical Oncology DepartmentThe Fifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Duozhi Shi
- Department of MedicineLifehealthcare Clinical LaboratoriesHangzhouChina
| | - Chong Wan
- Department of MedicineLifehealthcare Clinical LaboratoriesHangzhouChina
| | - Zhiyong Wu
- Medical Oncology DepartmentThe Fifth Medical Center of Chinese PLA General HospitalBeijingChina
| |
Collapse
|
39
|
Wang J, Shi W, Miao Y, Gan J, Guan Q, Ran J. Evaluation of tumor microenvironmental immune regulation and prognostic in lung adenocarcinoma from the perspective of purinergic receptor P2Y13. Bioengineered 2021; 12:6286-6304. [PMID: 34494914 PMCID: PMC8806861 DOI: 10.1080/21655979.2021.1971029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tumor-infiltrating immune cells (TICs) can serve as an important indicator to evaluate the prognosis and therapeutic response in lung adenocarcinoma (LUAD). The identification of mutated genes that can affect the abundance of TICs and prognosis has practical implications. In the presented study, tumor microenvironment (TME) scoring was performed by the ESTIMATE scoring system on 598 RNA transcripts selected from the TCGA database to determine the proportions of immune cells and stromal cells. The infiltration difference of TICs in LUAD samples was obtained by CIBERSORT. The 'immuneeconv' R software package, which integrates six latest algorithms, including TIMER, xCell, MCP-counter, CIBERSORT, EPIC and quanTIseq were used to verify the correlation between purinergic receptor P2Y13 (P2RY13) and immune cells. Based on RNA sequencing analysis of the Lewis lung cancer-bearing model in C57BL/6 mice and immunohistochemistry (IHC) of human LUAD tissues, the expression of P2RY13 and associated pathways were verified. It was shown that differentially expressed genes (DEGs) obtained by interactive analysis based on Immunescore and Stromalscore were significantly enriched in immune-related pathways. The expression of P2RY13 was significantly associated with prognosis and clinicopathological characteristics of LUAD patients. More importantly, this gene played an important role in maintaining the immune dominant environment and changing the regulation of TICs. P2RY13 expression was positively correlated with the infiltration of dendritic cells (DCs) in various of tumor tissues as validated by the PanglaoDB scRNA-seq database. Therefore, P2RY13 is expected to be a potential biomarker for predicting TME and the prognosis of LUAD after verification.
Collapse
Affiliation(s)
- Jiangtao Wang
- Department of Radiation Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, PR China.,The First Clinical Medical College of Lanzhou University, Lanzhou, PR China
| | - Weiwei Shi
- Clinical Skills Center of Yantai Affiliated Hospital of Binzhou Medical College, Yantai, PR China
| | - Yandong Miao
- The First Clinical Medical College of Lanzhou University, Lanzhou, PR China
| | - Jian Gan
- The First Clinical Medical College of Lanzhou University, Lanzhou, PR China
| | - Quanlin Guan
- The First Clinical Medical College of Lanzhou University, Lanzhou, PR China.,Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, PR China
| | - Juntao Ran
- Department of Radiation Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, PR China.,The First Clinical Medical College of Lanzhou University, Lanzhou, PR China
| |
Collapse
|
40
|
Gu S, Wu Q, Li Y, Jia W, Zhang D, Jiao L, Zhang Y. Investigation of differentially expressed gene profile for cisplatin-treated lung cancer patients. Anticancer Drugs 2021; 32:875-881. [PMID: 33967201 DOI: 10.1097/cad.0000000000001075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The purpose of the study was to establish a comprehensive differential gene profile for lung cancer patients treated with cisplatin compared with control patients without any chemotherapy drug treatment. The RNA sequencing data and miRNA sequencing data of 108 lung cancer patients treated with cisplatin only and 232 lung cancer patients treated without any chemotherapeutic drugs, were analyzed using differential expression, protein-protein interaction, and immune cell infiltration ratio analysis. Compared with control patients, the cisplatin-treated patients demonstrated 336 differentially expressed genes, which included 48 upregulated genes and 288 downregulated genes. Meanwhile, 12 differentially expressed miRNAs (DEMs), including 7 upregulated miRNAs and 5 downregulated miRNAs showed a differentially expressed pattern. With further instigation, five miRNAs (hsa-miR-548ah, hsa-miR-466, hsa-miR-552, hsa-miR-371a, and hsa-miR-4445) were suggested to be the key targets in the cisplatin-treated patients. At the same time, we also found a significant correlation between the cisplatin treatment and six immune checkpoints including programmed cell death ligand. This study helped us better understand the potential targets and underline molecular mechanisms for cisplatin treatment and provided references to eliminate existing side effects in the future.
Collapse
Affiliation(s)
- Songtao Gu
- Tianjin Medical University
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Qi Wu
- Tianjin Medical University General Hospital
| | - Yuechuan Li
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Wei Jia
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Dongrui Zhang
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Lina Jiao
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Yating Zhang
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| |
Collapse
|
41
|
Li D, Gui Q, Xu C, Shen M, Chen K. Dramatic response to osimertinib combined with crizotinib in EGFR T790 M mutation only in blood and Met amplification only in tumor tissue expressive non-small cell lung cancer: A case report. Medicine (Baltimore) 2021; 100:e26375. [PMID: 34397683 PMCID: PMC8322536 DOI: 10.1097/md.0000000000026375] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/02/2021] [Indexed: 01/04/2023] Open
Abstract
RATIONALE Besides the T790 M mutation, it may coexist with bypass pathway activation in real clinical cases for patients with EGFR mutations who resisted to the first- and second-generation tyrosine kinase inhibitors (TKIs) in non-small cell lung cancer (NSCLC). There are limited clinical trial data describing the efficacy of osimertinib combined with MET inhibition in EGFR T790M-mutant NSCLC patients with Met amplification. PATIENT CONCERNS A non-smoking 53-year-old male patient with lung adenocarcinoma underwent gefitinib, afatinib, and osimertinib combined with crizotinib treatment and developed different EGFR resistance mutations. DIAGNOSES The patient was diagnosed with lung adenocarcinoma (stage cT4N2M0, IIIB). After resistance to the therapy targeting EGFR exon 21 L858R point mutation, T790 M mutation was detected in liquid biopsy and Met amplification was detected via tissue biopsy by next-generation sequencing (NGS). INTERVENTIONS The patient received systemic treatments, including chemotherapy, gefitinib, afatinib, and osimertinib combined with crizotinib. OUTCOMES The patient died of multisystem organ failure and had an overall survival of 24 months. LESSONS Although osimertinib combined with crizotinib therapy showed dramatic tumor shrinkage in both the primary tumor and bone metastasis to an EGFR T790M-mutant NSCLC patient with MET amplification, the progression-free survival (PFS) was only two months.
Collapse
|
42
|
Shah PP, Franke JL, Medikonda R, Jackson CM, Srivastava S, Choi J, Forde PM, Brahmer JR, Ettinger DS, Feliciano JL, Levy BP, Marrone KA, Naidoo J, Redmond KJ, Kleinberg LR, Lim M. Mutation status and postresection survival of patients with non-small cell lung cancer brain metastasis: implications of biomarker-driven therapy. J Neurosurg 2021; 136:56-66. [PMID: 34087798 DOI: 10.3171/2020.10.jns201787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/02/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Non-small cell lung cancer (NSCLC) is the most common primary tumor to develop brain metastasis. Prognostic markers are needed to better determine survival after neurosurgical resection of intracranial disease. Given the importance of mutation subtyping in determining systemic therapy and overall prognosis of NSCLC, the authors examined the prognostic value of mutation status for postresection survival of patients with NSCLC brain metastasis. METHODS The authors retrospectively analyzed all cases of NSCLC brain metastasis with available molecular testing data that were resected by a single surgeon at a single academic center from January 2009 to February 2019. Mutation status, demographic characteristics, clinical factors, and treatments were analyzed. Association between predictive variables and overall survival after neurosurgery was determined with Cox regression. RESULTS Of the included patients (n = 84), 40% were male, 76% were smokers, the mean ± SD Karnofsky Performance Status was 85 ± 14, and the mean ± SD age at surgery was 63 ± 11 years. In total, 23%, 26%, and 4% of patients had EGFR, KRAS, and ALK/ROS1 alterations, respectively. On multivariate analysis, survival of patients with EGFR (HR 0.495, p = 0.0672) and KRAS (HR 1.380, p = 0.3617) mutations were not significantly different from survival of patients with wild-type (WT) tumor. However, the subgroup of patients with EGFR mutation who also received tyrosine kinase inhibitor (TKI) therapy had significantly prolonged survival (HR 0.421, p = 0.0471). In addition, postoperative stereotactic radiosurgery (HR 0.409, p = 0.0177) and resected tumor diameter < 3 cm (HR 0.431, p = 0.0146) were also significantly associated with prolonged survival, but Graded Prognostic Assessment score ≤ 1.0 (HR 2.269, p = 0.0364) was significantly associated with shortened survival. CONCLUSIONS Patients with EGFR mutation who receive TKI therapy may have better survival after resection of brain metastasis than patients with WT tumor. These results may inform counseling and decision-making regarding the appropriateness of resection of NSCLC brain metastasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Patrick M Forde
- 2Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Julie R Brahmer
- 2Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - David S Ettinger
- 2Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Josephine L Feliciano
- 2Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Benjamin P Levy
- 2Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Kristen A Marrone
- 2Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Jarushka Naidoo
- 2Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Kristin J Redmond
- 3Radiation Oncology, Johns Hopkins University School of Medicine; and
| | | | | |
Collapse
|
43
|
McLean L, Leal JL, Solomon BJ, John T. Immunotherapy in oncogene addicted non-small cell lung cancer. Transl Lung Cancer Res 2021; 10:2736-2751. [PMID: 34295674 PMCID: PMC8264320 DOI: 10.21037/tlcr-20-772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/10/2020] [Indexed: 12/25/2022]
Abstract
The use of immune checkpoint inhibitors (ICIs) targeting the programmed cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1) has led to notable changes in treatment strategies for patients with advanced non-small cell lung cancer (NSCLC) and now forms a part of standard of care treatment in patients with advanced disease. However, most patients do not respond to ICI monotherapy, which may be explained by significant variations in efficacy according to different immune and molecular profiles in tumours. Improved response rates have been observed in smokers and are associated with tumors that have high mutation loads, with a higher tendency to form neoantigens. This premise itself defies the eventual significance of ICIs for oncogene-driven NSCLC, which in general are more common in never smokers and potentially have reduced capacity for neoantigen formation. Furthermore, pivotal trials investigating ICIs in advanced NSCLC have usually excluded patients with oncogenic drivers, hence the outcome of these agents in this population is poorly characterized. In this article, we aim to review the most current evidence, encompassing clinical and preclinical data focused on a wide range of oncogene-addicted NSCLCs.
Collapse
Affiliation(s)
- Luke McLean
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jose Luis Leal
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Benjamin J. Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Thomas John
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
44
|
Ghosh S, Nataraj NB, Noronha A, Patkar S, Sekar A, Mukherjee S, Winograd-Katz S, Kramarski L, Verma A, Lindzen M, Garcia DD, Green J, Eisenberg G, Gil-Henn H, Basu A, Lender Y, Weiss S, Oren M, Lotem M, Geiger B, Ruppin E, Yarden Y. PD-L1 recruits phospholipase C and enhances tumorigenicity of lung tumors harboring mutant forms of EGFR. Cell Rep 2021; 35:109181. [PMID: 34038737 PMCID: PMC8170369 DOI: 10.1016/j.celrep.2021.109181] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 10/20/2020] [Accepted: 05/04/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer immunotherapy focuses on inhibitors of checkpoint proteins, such as programmed death ligand 1 (PD-L1). Unlike RAS-mutated lung cancers, EGFR mutant tumors have a generally low response to immunotherapy. Because treatment outcomes vary by EGFR allele, intrinsic and microenvironmental factors may be involved. Among all non-immunological signaling pathways surveyed in patients’ datasets, EGFR signaling is best associated with high PD-L1. Correspondingly, active EGFRs stabilize PD-L1 transcripts and depletion of PD-L1 severely inhibits EGFR-driven tumorigenicity and metastasis in mice. The underlying mechanisms involve the recruitment of phospholipase C-γ1 (PLC-γ1) to a cytoplasmic motif of PD-L1, which enhances PLC-γ1 activation by EGFR. Once stimulated, PLC-γ1 activates calcium flux, Rho GTPases, and protein kinase C, collectively promoting an aggressive phenotype. Anti-PD-L1 antibodies can inhibit these intrinsic functions of PD-L1. Our results portray PD-L1 as a molecular amplifier of EGFR signaling and improve the understanding of the resistance of EGFR+ tumors to immunotherapy. Unlike promoter-mediated PD-L1 induction by IFN-γ, EGFR rapidly stabilizes PD-L1 mRNA Once induced, PD-L1 enhances metastasis in vivo and chemotaxis toward EGF PD-L1 physically binds with and enhances activation of phospholipase C-γ1 by EGFR PLC-γ1 binds a PD-L1’s cytoplasmic segment implicated in protection from cytotoxicity
Collapse
Affiliation(s)
- Soma Ghosh
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | - Ashish Noronha
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Sushant Patkar
- Department of Computer Science, University of Maryland College Park, College Park, MD, USA; Cancer Data Science Laboratory, National Cancer Institute, NIH, Rockville, MD, USA
| | - Arunachalam Sekar
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Saptaparna Mukherjee
- Department of Molecular Cell Biology Weizmann Institute of Science, Rehovot, Israel
| | - Sabina Winograd-Katz
- Department of Molecular Cell Biology Weizmann Institute of Science, Rehovot, Israel
| | - Lior Kramarski
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Aakanksha Verma
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Moshit Lindzen
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Diana Drago Garcia
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Joseph Green
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Galit Eisenberg
- Sharett Institute of Oncology, Hadassah Medical School, Jerusalem, Israel
| | - Hava Gil-Henn
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Arkaprabha Basu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yan Lender
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Shimon Weiss
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Moshe Oren
- Department of Molecular Cell Biology Weizmann Institute of Science, Rehovot, Israel
| | - Michal Lotem
- Sharett Institute of Oncology, Hadassah Medical School, Jerusalem, Israel
| | - Benjamin Geiger
- Department of Molecular Cell Biology Weizmann Institute of Science, Rehovot, Israel
| | - Eytan Ruppin
- Cancer Data Science Laboratory, National Cancer Institute, NIH, Rockville, MD, USA
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
45
|
Qiao M, Jiang T, Liu X, Mao S, Zhou F, Li X, Zhao C, Chen X, Su C, Ren S, Zhou C. Immune Checkpoint Inhibitors in EGFR-Mutated NSCLC: Dusk or Dawn? J Thorac Oncol 2021; 16:1267-1288. [PMID: 33915248 DOI: 10.1016/j.jtho.2021.04.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023]
Abstract
Although immune checkpoint inhibitors (ICIs) that target programmed cell death protein-1/programmed cell death ligand-1 axis have significantly shifted the treatment paradigm in advanced NSCLC, clinical benefits of these agents are limited in patients with EGFR-mutated NSCLC. Several predictive biomarkers (e.g., programmed cell death ligand-1 expression, tumor mutation burden), which have been validated in EGFR-wild type NSCLC, however, are not efficacious in EGFR-mutated tumors, suggesting the unique characteristics of tumor microenvironment of EGFR-mutated NSCLC. Here, we first summarized the clinical evidence on the efficacy of ICIs in patients with EGFR-mutated NSCLC. Then, the cancer immunogram features of EGFR-mutated NSCLC was depicted to visualize the state of cancer-immune system interactions, including tumor foreignness, tumor sensitivity to immune effectors, metabolism, general immune status, immune cell infiltration, cytokines, and soluble molecules. We further discussed the potential subpopulations with EGFR mutations that could benefit from ICI treatment. Lastly, we put forward future strategies to adequately maximize the efficacy of ICI treatment in patients with EGFR-mutated NSCLC in the upcoming era of combination immunotherapies.
Collapse
Affiliation(s)
- Meng Qiao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xinyu Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Shiqi Mao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xuefei Li
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Chao Zhao
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xiaoxia Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
46
|
Chen Y, Yang Z, Wang Y, Hu M, Zhang B, Zhang Y, Qian F, Zhang W, Han B. Pembrolizumab Plus Chemotherapy or Anlotinib vs. Pembrolizumab Alone in Patients With Previously Treated EGFR-Mutant NSCLC. Front Oncol 2021; 11:671228. [PMID: 33937084 PMCID: PMC8085497 DOI: 10.3389/fonc.2021.671228] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 03/22/2021] [Indexed: 12/22/2022] Open
Abstract
Objectives More and more encouraging evidence revealed that immunotherapy could improve clinical outcomes in patients with previously treated non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) variations. However, immunotherapy is still a controversy for NSCLC patients with EGFR mutation. Method In this retrospective analysis, we compared the clinical efficacy of pembrolizumab monotherapy (PM), pembrolizumab combined with chemotherapy (P+C) and pembrolizumab combined with anlotinib (P+A) in NSCLC patients with EGFR mutation who had failed on EGFR-TKI and platinum-based chemotherapy. Result Eighty-six patients were included in this study. The overall median progression free survival (PFS) was 3.24 months. Multivariate analysis suggested that EGFR L858R and combined therapy were positive prognostic factors of PFS. The overall median OS was 12.28 months. Multivariate analysis found that high PD-L1 expression (≥50%) and combined therapy seemed to be positive prognostic factors of OS. Among the population, 32 patients received PM, 26 patients received P+C and 28 patients received P+A. Up to Jan 30, 2021, the median progression-free survival was 1.5 months in the PM group, 4.30 months in the P+C group and 3.24 months in the P+A group. The median OS were 7.41, 14.92 and 15.97 months, respectively. The ORR were 3.1%, 23.1% and 21.4%. Conclusion The addition of chemotherapy or antiangiogenic therapy to pembrolizumab resulted in significantly longer PFS, OS and ORR than pembrolizumab alone in our study. EGFR L858R might be a positive prognostic factor of PFS and high PD-L1 expression might be a positive prognostic factor of OS.
Collapse
Affiliation(s)
- Ya Chen
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengyu Yang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanan Wang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Minjuan Hu
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanwei Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fangfei Qian
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Baohui Han
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
47
|
Zhou J, Zhao J, Jia Q, Chu Q, Zhou F, Chu X, Zhao W, Ren S, Zhou C, Su C. Peripheral Blood Autoantibodies Against to Tumor-Associated Antigen Predict Clinical Outcome to Immune Checkpoint Inhibitor-Based Treatment in Advanced Non-Small Cell Lung Cancer. Front Oncol 2021; 11:625578. [PMID: 33816260 PMCID: PMC8010683 DOI: 10.3389/fonc.2021.625578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/02/2021] [Indexed: 01/11/2023] Open
Abstract
Background Peripheral blood biomarkers to immunotherapy have attracted more and more attentions owing to noninvasive nature. This study was designed to identify a panel of tumor associated autoantibodies (TAAbs) in plasma to predict the clinical outcome of ICIs-based treatment in advanced NSCLC patients and correlation between TAAbs and checkpoint inhibitor pneumonitis (CIP) would also be investigated. Materials and Methods Baseline plasma was collected from patients with advanced NSCLC before receiving ICIs-based treatment. ELISA was used to detect concentration of autoantibodies. Clinical efficacy was evaluated according to RECIST v1.1. Results We have identified a panel of five-TAAbs to predict responses of ICIs-based treatment in a discovery cohort (n = 37), and confirmed its predictive value in a validation cohort (n = 129). In the validation cohort, the positivity of this 5-TAAbs panel was significantly associated with better response (ORR: 44.4% vs. 13.6%, P < 0.001) and longer PFS (7.6 vs. 3.3m, P < 0.001). This significant association was remained in subgroup of patients treated with combination therapy (ORR: 43.8% vs. 13.7%, P = 0.004,PFS: 6.7 vs. 3.7m, P = 0 .017). Furthermore, this 5-TAAs panel worked better in patients who received subsequent-line treatment (ORR: 42.4% vs. 7.7%, P = 0.001, PFS: 6.2 vs. 3.0m, P = 0.004) than those received first-line treatment (ORR: 46.7% vs. 35.7%, P = 0.345, PFS: NR vs. 10.48m, P = 0.146). In addition, the CIP incidence in patients with 5-TAAbs positive was significantly higher comparing to negative patients (20.4% vs. 5.9%, P = 0.015). Conclusion Our 5-TAAbs panel is a potential predictive biomarker for responses and toxicities to ICIs-based treatment in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Jing Zhao
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Qingzhu Jia
- Department of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Zhou
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Xiangling Chu
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Wencheng Zhao
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Shengxiang Ren
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Caicun Zhou
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Chunxia Su
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
48
|
Li Y, Chen Z, Tao W, Sun N, He J. Tumor mutation score is more powerful than tumor mutation burden in predicting response to immunotherapy in non-small cell lung cancer. Cancer Immunol Immunother 2021; 70:2367-2378. [PMID: 33533944 DOI: 10.1007/s00262-021-02868-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022]
Abstract
Tumor mutation burden (TMB) predicts response to immunotherapy in non-small cell lung cancer (NSCLC). The current TMB evaluation is expensive and not satisfactory. Here, novel tumor mutation score (TMS) was defined as the number of genes with mutations in candidate genes and compared with TMB and PD-L1 in 240 NSCLC patients and validated in 34 NSCLC patients. Eighteen genes were significantly associated with longer progression-free survival (PFS) or better response. The number of mutated genes within 18 favorable genes were defined as TMS18. TMS18 (HR = 0.307, P < 0.001) had smaller hazard ratio and P value than TMB (HR = 0.455, P = 0.004) and PD-L1 expression (HR = 0.403, P = 0.005) in survival analysis. Moreover, TMS18 had significantly higher AUC than TMB and TMS18 combined with PD-L1 improved the accuracy. Universal cutoff of TMS18 enriched more patients with benefits. These findings were largely consistent in the validation cohort. Taken together, TMS18 was more powerful than TMB in predicting response of ICIs in NSCLC. Selective TMS was more feasible and cost-effective than unselective TMB. TMS18 combined with PD-L1 might yield better efficiency in predicting response of ICIs in NSCLC with future validation in larger cohorts.
Collapse
Affiliation(s)
- Yuan Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zuhua Chen
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Weiping Tao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.18 Panjiayuannanli, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.18 Panjiayuannanli, Beijing, 100021, China.
| |
Collapse
|
49
|
Leng X, Wei S, Mei J, Deng S, Yang Z, Liu Z, Guo C, Deng Y, Xia L, Cheng J, Zhao K, Gan F, Li C, Merrell KW, Molina JR, Metro G, Liu L. Identifying the prognostic significance of B3GNT3 with PD-L1 expression in lung adenocarcinoma. Transl Lung Cancer Res 2021; 10:965-980. [PMID: 33718036 PMCID: PMC7947420 DOI: 10.21037/tlcr-21-146] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background As a novel treatment, programmed cell death protein 1 (PD-1) inhibitor appears to be less effective in tumors of lung adenocarcinoma patients with epidermal growth factor receptor (EGFR) mutation. Beta-1,3-N-acetylglucosaminyltransferase 3 (B3GNT3) has reported to be associated with programmed death ligand 1 (PD-L1)/PD-1 interaction. However, the relationship between B3GNT3 and PD-L1 and its prognostic significance in EGFR mutant status are still unknown. Methods B3GNT3 was identified through transcriptome sequencing and The Cancer Genome Atlas Lung Adenocarcinoma (TCGA-LUAD) database. Flow cytometry and real-time polymerase chain reaction were performed to investigate the association between B3GNT3, PD-L1, and EGFR. Then, B3GNT3 and PD-L1 expression were evaluated by immunohistochemical analysis in 145 surgically resected primary lung adenocarcinomas. The relationships between survival and B3GNT3, PD-L1, and EGFR status were assessed, and the potential prognostic factors in patients with B3GNT3 expression were identified. Results We found that EGFR activation induced PD-L1 expression, and EGFR tyrosine kinase inhibitor (TKI) could reduce PD-L1 protein in EGFR-TKI-sensitive HCC827 and PC9 cell lines. Subsequent analysis showed that EGFR inhibitor could also lead to both decreased PD-L1 and B3GNT3 mRNA expression. A total of 145 lung adenocarcinoma patients were included. PD-L1 >1% and B3GNT3-positive expression in patients might contribute to worse prognosis in both overall survival (OS) [hazard ratio (HR), 2.63; 95% confidence interval (CI), 0.98–7.06; P=0.048] and disease-free survival (DFS) (HR, 3.04; 95% CI, 1.13–8.14; P=0.019), especially in the PD-L1 ≥50% group. However, when patients were negative for B3GNT3, PD-L1, and EGFR (or “triple negative”), there were significant decreases in OS (HR, 5.44; 95% CI, 0.99–29.83; P=0.029) and DFS (HR, 7.24; 95% CI, 1.32–39.73; P=0.008). Positive B3GNT3 expression was a significant risk factor associated with lower DFS (HR, 3.30; P=0.043). Conclusions Our results indicate that the B3GNT3 expression is tightly correlated with PD-L1 expression and EGFR mutation status. B3GNT3 is associated with poor prognosis in lung adenocarcinoma patients. Collectively, these findings may offer new insight into enhancing immune therapy efficacy for lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Xuefeng Leng
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China.,Division of Thoracic Surgery, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China (UESTC), Chengdu, China
| | - Shiyou Wei
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| | - Jiandong Mei
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| | - Senyi Deng
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| | - Zhenyu Yang
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| | - Zheng Liu
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| | - Chenglin Guo
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| | - Yulan Deng
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| | - Liang Xia
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| | - Jiahan Cheng
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| | - Kejia Zhao
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| | - Fanyi Gan
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| | - Chuan Li
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| | | | - Julian R Molina
- Division of Medical Oncology and Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Giulio Metro
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, via Dottori, 1, 06156, Perugia, Italy
| | - Lunxu Liu
- Department of Thoracic Surgery, Institute of Thoracic Oncology/West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
50
|
Zhang H, Yang X, Li K, Wang J, Lv J, Li X, Zhang X, Qin N, Zhang Q, Wu Y, Ma L, Gai F, Hu Y, Zhang S. [A Real-world Study on the Assessment of Pathological Characteristics and Targeted Therapeutic Effect of Non-small Cell Lung Cancer Patients with Positive Driving Genes and High PD-L1 Expression]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2021; 24:78-87. [PMID: 33478196 PMCID: PMC7936084 DOI: 10.3779/j.issn.1009-3419.2021.104.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
背景与目的 驱动基因突变阳性患者行靶向治疗,驱动基因阴性但程序性死亡配体1(programmed death-ligand 1, PD-L1)高表达患者行免疫抑制剂治疗,是晚期非小细胞肺癌(non-small cell lung cancer, NSCLC)患者一线治疗的首选,但对于驱动基因阳性且PD-L1高表达患者的治疗选择仍值得探究。 方法 以315例NSCLC患者为研究对象,分析驱动基因阳性且PD-L1高表达患者的临床病理特征及靶向治疗疗效。 结果 本研究纳入的315例NSCLC患者中,驱动基因突变总阳性率为62.2%,PD-L1高表达率(≥50.0%)为11.2%,驱动基因阳性且PD-L1高表达的患者比例为10.7%。其中表皮生长因子受体(epidermal growth factor receptor, EGFR)突变、KRAS 突变、ALK融合、BRAF突变和MET 14外显子跳跃突变患者中均有PD-L1高表达,比例分别为7.8%(11/141)、18.2%(4/22)、23.1%(3/13)、50.0%(2/4)和100.0%(1/1)。EGFR突变且PD-L1高表达患者主要为Ⅳ期肺腺癌患者,KRAS突变且PD-L1高表达患者主要为有吸烟史的患者。其中详细跟踪了两例分别为ALK融合阳性且PD-L1高表达(90.0%)和EGFR L858R突变且PD-L1高表达(70.0%)患者的靶向治疗全过程,两例患者总生存期分别仅为5个月和2个月。 结论 NSCLC患者各驱动基因突变与PD-L1高表达共存的比例和临床病理特征有较大差异。发生敏感突变且PD-L1高表达的患者靶向治疗疗效和预后可能更差。
Collapse
Affiliation(s)
- Hui Zhang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor
Research Institute, Beijing 101149, China
| | - Xinjie Yang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor
Research Institute, Beijing 101149, China
| | - Kun Li
- Department of Pathology, Beijing Chest Hospital, Capital Medical University/Beijing
Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Jinghui Wang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor
Research Institute, Beijing 101149, China
| | - Jialin Lv
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor
Research Institute, Beijing 101149, China
| | - Xi Li
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor
Research Institute, Beijing 101149, China
| | - Xinyong Zhang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor
Research Institute, Beijing 101149, China
| | - Na Qin
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor
Research Institute, Beijing 101149, China
| | - Quan Zhang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor
Research Institute, Beijing 101149, China
| | - Yuhua Wu
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor
Research Institute, Beijing 101149, China
| | - Li Ma
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor
Research Institute, Beijing 101149, China
| | - Fei Gai
- Amoy Diagnostics Co.,Ltd, Xiamen 361000, China
| | - Ying Hu
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor
Research Institute, Beijing 101149, China
| | - Shucai Zhang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor
Research Institute, Beijing 101149, China
| |
Collapse
|