1
|
Vestergaard CD, Donia M, Madsen K, Schmidt H, Luczak AA, Bastholt L, Ellebaek E, Svane IM. Impact of assessment-to-treatment interval and metastatic biopsy site on the predictive value of PD-L1 expression at the 1 % cut-off level in melanoma. Eur J Cancer 2025; 221:115402. [PMID: 40222200 DOI: 10.1016/j.ejca.2025.115402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/13/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Intratumoral PD-L1 expression at the 1 % cut-off predicts clinical outcomes and may guide first-line immune checkpoint inhibitor (ICI) selection for metastatic melanoma (MM). However, the impact of the interval between PD-L1 assessment and ICI initiation and the metastatic site used for PD-L1 evaluation, remains unclear. METHODS In this nationwide cohort study we used the Danish Metastatic Melanoma Database (DAMMED) and the Danish Pathology Registry to analyze patients with MM treated with anti-PD-1 or anti-PD-1 plus anti-CTLA-4 from January 2017 to February 2024. Progression-free survival (PFS) and overall survival (OS) were analyzed using Log-rank tests and Cox regression. RESULTS Data from 1137 patients were analyzed. Among patients with PD-L1 assessed within 90 days of treatment (n = 964; 55.2 % PD-L1 <1 %, 44.8 % PD-L1 ≥1 %), combination therapy improved outcomes in PD-L1 < 1 % (PFS adjusted (a)HR 0.62; 95 % CI 0.48-0.80; p < 0.001, OS aHR 0.64; 95 % CI 0.48-0.85; p = 0.002), while outcomes were comparable for PD-L1 ≥ 1 % patients (PFS aHR 0.90; 95 % CI 0.62-1.30; p = 0.57, OS aHR 0.97; 95 % CI 0.60-1.57; p = 0.89). For PD-L1 assessed > 90 days prior (n = 173), this pattern was less pronounced. Among 48 paired PD-L1 assessments from the same organ, discordance occurred in 25 %. Combination therapy improved PFS for patients with PD-L1 < 1 % skin/subcutaneous (aHR 0.51; 95 % CI 0.34-0.76; p < 0.001) and visceral metastases (aHR 0.65; 95 % CI 0.42-1.02; p = 0.060) while this association was not evident for lymph node metastases (aHR 0.79; 95 % CI 0.48-1.29; p = 0.35). CONCLUSIONS PD-L1 seems a reliable predictive biomarker in MM, when assessed on tissue obtained within 90 days prior to ICI initiation. Non-nodal metastatic sites appear preferable.
Collapse
Affiliation(s)
- Cecilie D Vestergaard
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Marco Donia
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Kasper Madsen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus N 8200, Denmark
| | - Adam A Luczak
- Department of Oncology, Aalborg University Hospital, Aalborg 9000, Denmark
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense C 5000, Denmark
| | - Eva Ellebaek
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Inge M Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev 2730, Denmark.
| |
Collapse
|
2
|
Yang CY, Shih JY. Response to the comments on "PD-L1 expression and immune profiling cannot predict osimertinib efficacy in lung cancer with EGFR T790 M mutation: A translational study". J Formos Med Assoc 2025:S0929-6646(25)00199-8. [PMID: 40307152 DOI: 10.1016/j.jfma.2025.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2025] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 05/02/2025] Open
Affiliation(s)
- Ching-Yao Yang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
3
|
Hughes DJ, Chand G, Johnson J, Tegala R, Bailey D, Adamson K, Edmonds S, Meszaros LK, Moore AEB, Manickavasagar T, Ndagire S, Gennatas S, Georgiou A, Ghosh S, Josephs D, Karapanagiotou E, McLean E, Ting HH, Spicer J, Goh V, Cook GJR. PD-L1 imaging with [ 99mTc]NM-01 SPECT/CT is associated with metabolic response to pembrolizumab with/without chemotherapy in advanced lung cancer. Br J Cancer 2025:10.1038/s41416-025-02991-w. [PMID: 40188291 DOI: 10.1038/s41416-025-02991-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/15/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) immunohistochemistry is a predictive biomarker for anti-PD-(L)1 therapy in non-small cell lung cancer (NSCLC). It is not a reliable predictor of clinical benefit with non-invasive imaging providing a potential solution. We present the PECan study, the aim of which to assess the relationship of [99mTc]-labeled anti-PD-L1 single-domain antibody (NM-01) single-photon emission computed tomography (SPECT)/CT with metabolic response to anti-PD-(L)1. METHODS PD-L1 tumour proportion score (TPS) measured using SP263 assay. [99mTc]NM-01 SPECT/CT and [18F]FDG PET/CT performed before and 9-weeks following pembrolizumab with/without chemotherapy in patients with advanced NSCLC. Tumor (T) to blood pool (BP) maximum region of interest (ROImax) measurements performed in primary and metastatic lesions using SPECT/CT images. RESULTS Fifteen patients were included (median age 63 years, 9 male). Intertumoural heterogeneity evident in 10(67%) patients. Mean [99mTc]NM-01 T:BP demonstrated moderate correlation with PD-L1 TPS (r = 0.45, p < 0.05). Depth of [18F]FDG PET/CT metabolic response at 9-weeks (n = 13), correlated strongly with baseline [99mTc]NM-01 T:BP (r = -0.73, p < 0.05), but only moderately with PD-L1 TPS (r = -0.46, p = 0.06). CONCLUSION [99mTc]NM-01 SPECT/CT allows non-invasive quantification of PD-L1 in primary tumour and metastases in NSCLC. [99mTc]NM-01 uptake moderately correlates with PD-L1 immunohistochemistry, determines heterogeneity, and is associated with early metabolic response to anti-PD-1 pembrolizumab. CLINICAL TRIALS REGISTRATION PD-L1 Expression in Cancer (PECan) study (NCT04436406), registered 18 June 2020 https://clinicaltrials.gov/ct2/show/NCT04436406.
Collapse
Affiliation(s)
- Daniel Johnathan Hughes
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- King's College London & Guy's and St. Thomas' PET Centre, St Thomas' Hospital, London, UK
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Gitasha Chand
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Nanomab Technology (UK) Limited, Borehamwood, Hertfordshire, UK
| | - Jessica Johnson
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Ronan Tegala
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Damion Bailey
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Kathryn Adamson
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Scott Edmonds
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | | | - Amelia Elizabeth Broomfield Moore
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
| | - Thubeena Manickavasagar
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Department of Radiology, Guy's and St. Thomas' NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Susan Ndagire
- King's Health Partners Cancer Biobank, Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London, UK
| | - Spyridon Gennatas
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Alexandros Georgiou
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Sharmistha Ghosh
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Debra Josephs
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Eleni Karapanagiotou
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Emma McLean
- Department of Histopathology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Hong Hoi Ting
- Nanomab Technology (UK) Limited, Borehamwood, Hertfordshire, UK
| | - James Spicer
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Vicky Goh
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Department of Radiology, Guy's and St. Thomas' NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Gary J R Cook
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK.
- King's College London & Guy's and St. Thomas' PET Centre, St Thomas' Hospital, London, UK.
| |
Collapse
|
4
|
Saw SPL, Takano A, Zhou S, Hlaing NO, James A, Joseph C, Lai GGY, Lim DWT, Kanesvaran R, Ang MK, Ng QS, Jain A, Tan WL, Teh YL, Tan AC, Ong BH, Lim TKH, Yeong JPS, Tan SH, Tan DSW. EGFR mutation status affects intra-tumoural heterogeneity of PD-L1 expression but not agreement between assays in resectable non-small cell lung cancer. Lung Cancer 2025; 202:108463. [PMID: 40023889 DOI: 10.1016/j.lungcan.2025.108463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND The predictive value of PD-L1 to select patients for immunotherapy in resectable NSCLC remains imprecise, confounded by different assays used across trials and intra-tumoural heterogeneity (ITH). We sought to compare the concordance between 3 PD-L1 antibodies stratified by EGFR mutation status, evaluate ITH and implications on survival outcomes. METHODS Tissue microarrays were constructed from stage IA-IIIA NSCLC with 3 tumour cores per patient. Tumour proportion score (TPS) was evaluated by 3 pathologists for SP263, SP142, 22C3 and analysed in tertiles of < 1 %, 1-49 % and ≥ 50 %. ITH was defined as discordant TPS in ≥ 2/3 tumour cores. Cohen's kappa test was used to assess agreement. Survival outcomes were estimated using Kaplan-Meier. RESULTS A total of 561 patients were included, 59.5% (334/561) were EGFR-mutant. Stage IA comprised 45.5%(255/561), IB 24.1%(135/561), IIA 12.7%(71/561), IIB 4.5%(25/561) and IIIA 13.4%(75/561). Across 1683 tumour cores, SP263 and 22C3 had the highest concordance (Kappa = 0.689), followed by 22C3 and SP142 (Kappa = 0.354), then SP263 and SP142 (Kappa = 0.284), similar between EGFR-mutant and EGFR-wildtype. Agreement between pathologists was almost perfect. ITH by SP263 was observed in 14.1 % of EGFR-mutant versus 24.2 % in EGFR-wildtype(p = 0.002). Discordance was highest among TPS 1-49 % at 92.6 % (88/95) followed by ≥ 50 % at 37.8 % (14/37) and least among < 1 % at 0 % (0/429) (p < 0.001). For tumour cores scored 1-49 %, 63 %/70 % of adjacent cores were scored < 1 % for EGFR-wildtype/mutant respectively. Histological grade was the only independent predictor of PD-L1 ITH on multivariable analysis. PD-L1 ITH was not associated with survival on multivariable analysis. CONCLUSION PD-L1 scoring by SP263 and 22C3 are interchangeable but not SP142 regardless of EGFR status. PD-L1 ITH was more common in EGFR-wildtype versus EGFR-mutant tumours. Extra care should be taken to select the most representative tumour core for tumours with high histological grade or TPS 1-49% as this may influence peri-operative treatment decisions.
Collapse
Affiliation(s)
- Stephanie P L Saw
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore.
| | - Angela Takano
- Department of Pathology, Singapore General Hospital, Singapore
| | - Siqin Zhou
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore
| | - Nwe Oo Hlaing
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Anne James
- Department of Pathology, Singapore General Hospital, Singapore
| | - Craig Joseph
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Gillianne G Y Lai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Darren W T Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Ravindran Kanesvaran
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Mei-Kim Ang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Quan Sing Ng
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Amit Jain
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Wan Ling Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Yi Lin Teh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Aaron C Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Boon-Hean Ong
- Department of Cardiothoracic Surgery, National Heart Centre Singapore, Singapore
| | - Tony K H Lim
- Duke-NUS Medical School, National University of Singapore, Singapore; Department of Pathology, Singapore General Hospital, Singapore
| | - Joe P S Yeong
- Duke-NUS Medical School, National University of Singapore, Singapore; Department of Pathology, Singapore General Hospital, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Sze Huey Tan
- Duke-NUS Medical School, National University of Singapore, Singapore; Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore; Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore
| |
Collapse
|
5
|
Lin X, Liu Z, Zhou K, Li Y, Huang G, Zhang H, Shu T, Huang Z, Wang Y, Zeng W, Liao Y, Bin J, Shi M, Liao W, Zhou W, Huang N. Intratumoral and peritumoral PET/CT-based radiomics for non-invasively and dynamically predicting immunotherapy response in NSCLC. Br J Cancer 2025; 132:558-568. [PMID: 39930148 PMCID: PMC11920075 DOI: 10.1038/s41416-025-02948-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 12/17/2024] [Accepted: 01/23/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND We aimed to develop a machine learning model based on intratumoral and peritumoral 18F-FDG PET/CT radiomics to non-invasively and dynamically predict the response to immunotherapy in non-small cell lung cancer (NSCLC). METHODS This retrospective study included 296 NSCLC patients, including a training cohort (N = 183), a testing cohort (N = 78), and a TCIA radiogenomic cohort (N = 35). The extreme gradient boosting algorithm was employed to develop the radiomic models. RESULTS The COMB-Radscore, which was developed by combining radiomic features from PET, CT, and PET/CT images, had the most satisfactory predictive performance with AUC (ROC) 0.894 and 0.819 in the training and testing cohorts, respectively. Survival analysis has demonstrated that COMB-Radscore is an independent prognostic factor for progression-free survival and overall survival. Moreover, COMB-Radscore demonstrates excellent dynamic predictive performance, with an AUC (ROC) of 0.857, enabling the earlier detection of potential disease progression in patients compared to radiological evaluation solely relying on tumor size. Further radiogenomic analysis showed that the COMB-Radscore was associated with infiltration abundance and functional status of CD8 + T cells. CONCLUSIONS The radiomic model holds promise as a precise, personalized, and dynamic decision support tool for the treatment of NSCLC patients.
Collapse
Affiliation(s)
- Xianwen Lin
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
- Foshan Key Laboratory of Translational Medicine in Oncology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Zhiwei Liu
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kun Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuedan Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Genjie Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Zhang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tingting Shu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenhua Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuanyuan Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Zeng
- Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
- Foshan Key Laboratory of Translational Medicine in Oncology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianping Bin
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China.
- Foshan Key Laboratory of Translational Medicine in Oncology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China.
| | - Wenlan Zhou
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Oh MS, Abascal J, Rennels AK, Salehi-Rad R, Dubinett SM, Liu B. Tumor Heterogeneity and the Immune Response in Non-Small Cell Lung Cancer: Emerging Insights and Implications for Immunotherapy. Cancers (Basel) 2025; 17:1027. [PMID: 40149360 PMCID: PMC11941341 DOI: 10.3390/cancers17061027] [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: 12/31/2024] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Resistance to immune checkpoint inhibitors (ICIs) represents a major challenge for the effective treatment of non-small cell lung cancer (NSCLC). Tumor heterogeneity has been identified as an important mechanism of treatment resistance in cancer and has been increasingly implicated in ICI resistance. The diversity and clonality of tumor neoantigens, which represent the target epitopes for tumor-specific immune cells, have been shown to impact the efficacy of immunotherapy. Advances in genomic techniques have further enhanced our understanding of clonal landscapes within NSCLC and their evolution in response to therapy. In this review, we examine the role of tumor heterogeneity during immune surveillance in NSCLC and highlight its spatial and temporal evolution as revealed by modern technologies. We explore additional sources of heterogeneity, including epigenetic and metabolic factors, that have come under greater scrutiny as potential mediators of the immune response. We finally discuss the implications of tumor heterogeneity on the efficacy of ICIs and highlight potential strategies for overcoming therapeutic resistance.
Collapse
Affiliation(s)
- Michael S. Oh
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.S.O.); (J.A.); (A.K.R.); (R.S.-R.); (S.M.D.)
| | - Jensen Abascal
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.S.O.); (J.A.); (A.K.R.); (R.S.-R.); (S.M.D.)
| | - Austin K. Rennels
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.S.O.); (J.A.); (A.K.R.); (R.S.-R.); (S.M.D.)
| | - Ramin Salehi-Rad
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.S.O.); (J.A.); (A.K.R.); (R.S.-R.); (S.M.D.)
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA
| | - Steven M. Dubinett
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.S.O.); (J.A.); (A.K.R.); (R.S.-R.); (S.M.D.)
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA
| | - Bin Liu
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.S.O.); (J.A.); (A.K.R.); (R.S.-R.); (S.M.D.)
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
7
|
Zhang Y, Xu Y, Jin H, Liu T, Zhong H, Xu J, Lou Y, Zhong R. Spatial Heterogeneity of PD-L1 Expression as a Biomarker for Third-Generation EGFR-TKI Response in Advanced EGFR-Mutant NSCLC. Cancer Sci 2025. [PMID: 40102299 DOI: 10.1111/cas.70060] [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: 01/02/2025] [Revised: 02/07/2025] [Accepted: 03/13/2025] [Indexed: 03/20/2025] Open
Abstract
The association between the spatial heterogeneity of programmed cell death ligand 1 (PD-L1) expression and the efficacy of third-generation epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) in EGFR-mutant non-small cell lung cancer (NSCLC) remains elusive. This retrospective study analyzed data from 4171 NSCLC patients with EGFR-sensitive mutations treated at Shanghai Chest Hospital from August 2019 to September 2023. Among them, 182 patients receiving third-generation EGFR-TKIs monotherapy as a first-line treatment were enrolled. Patients were categorized by biopsy sites into primary lung lesions (n = 112) and metastatic lymph nodes (n = 70). PD-L1 expression was stratified based on tumor cell proportion score (TPS): < 1%, 1%-49%, and ≥ 50%. The median progression-free survival (PFS) for the entire cohort was 18.33 months. In the PD-L1 TPS group, PFS was 18.87 months for TPS < 1%, 17.6 months for TPS 1%-49%, and 13.6 months for TPS ≥ 50%, with significant differences across groups (p = 0.026). Moreover, multivariate analysis identified smoking history [HR = 1.653, 95% CI (1.132-2.414), p = 0.009] and TPS ≥ 50% [HR = 2.069, 95% CI (1.183-3.618), p = 0.011] as independent risk factors. In primary lesions, the median PFS was 21.93 months for TPS < 1%, 18.57 months for TPS 1%-49%, and 10.17 months for TPS ≥ 50%, with significant differences (p < 0.001). However, PD-L1 expression in metastatic lymph nodes was not associated with PFS (p = 0.973). In advanced EGFR-mutant NSCLC, high PD-L1 expression may suggest reduced efficacy of third-generation EGFR-TKIs. The spatial heterogeneity of PD-L1 expression could influence its predictive accuracy for third-generation EGFR-TKI efficacy.
Collapse
Affiliation(s)
- Yidan Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingqi Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongping Jin
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tengfei Liu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianlin Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuqing Lou
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runbo Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Wang Y, Safi M, Hirsch FR, Lu S, Peters S, Govindan R, Rosell R, Park K, Zhang JJ. Immunotherapy for advanced-stage squamous cell lung cancer: the state of the art and outstanding questions. Nat Rev Clin Oncol 2025; 22:200-214. [PMID: 39762577 DOI: 10.1038/s41571-024-00979-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 02/26/2025]
Abstract
Immune-checkpoint inhibitors (ICIs) have transformed the treatment paradigm for advanced-stage squamous non-small-cell lung cancer (LUSC), a histological subtype associated with inferior outcomes compared with lung adenocarcinoma. However, only a subset of patients derive durable clinical benefit. In the first-line setting, multiple ICI regimens are available, including anti-PD-(L)1 antibodies as monotherapy, in combination with chemotherapy, or with an anti-CTLA4 antibody with or without chemotherapy. Several important questions persist regarding the optimal regimen for individual patients, particularly how to identify patients who might benefit from adding chemotherapy and/or anti-CTLA4 antibodies to anti-PD-(L)1 antibodies. An urgent need exists for predictive biomarkers beyond PD-L1 to better guide precision oncology approaches. Deeper knowledge of the underlying molecular biology of LUSC and its implications for response to ICIs will be important in this regard. Integration of this knowledge into multi-omics methods coupled with artificial intelligence might enable the development of more robust biomarkers. Finally, several novel therapeutic strategies, including novel ICIs, bispecific antibodies and personalized cancer vaccines, are emerging. Addressing these unresolved questions through innovative clinical trials and translational research will be crucial to further improving the outcomes of patients with LUSC. In this Review, we provide a comprehensive overview of current immunotherapeutic approaches, unresolved challenges and emerging strategies for patients with LUSC.
Collapse
Affiliation(s)
- Yibei Wang
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mohammed Safi
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Fred R Hirsch
- Center for Thoracic Oncology, Tisch Cancer Institute and Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Shun Lu
- Department of Medical Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Solange Peters
- Oncology Department, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | | | - Rafael Rosell
- Dr. Rosell Oncology Institute, Dexeus University Hospital, Barcelona, Spain
| | - Keunchil Park
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
- Division of Hematology/Oncology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Jianjun J Zhang
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
9
|
Song P, Pan G, Zhang Y, Ni Y, Wang Q, Shi J, Peng Y, Jing R, Luo D. Prospects and Challenges of Immunotherapy for Thyroid Cancer. Endocr Pract 2025; 31:373-379. [PMID: 39631664 DOI: 10.1016/j.eprac.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/21/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Thyroid cancer generally boasts a favorable prognosis; however, advanced and refractory cases exhibit aggressive characteristics and resistance to conventional therapies, necessitating the investigation of innovative treatment modalities. Immunotherapy, which harnesses the body's immune system to target cancer cells, has shown considerable promise for specific thyroid cancer subtypes. OBJECTIVE This review article aims to encapsulate the latest advancements in immunotherapy for thyroid cancer, examining its mechanisms, therapeutic efficacy, ongoing challenges, and the potential benefits of combination therapy approaches. METHODS An extensive literature review and critical analysis of clinical trial data were conducted to inform this synthesis. RESULTS The review reveals that immunotherapy strategies, encompassing immune checkpoint inhibitors, CAR-T cell therapy, tumor vaccines, and immunomodulators, are demonstrating efficacy in the treatment of thyroid cancer. Notably, checkpoint inhibitors have been particularly effective in anaplastic and poorly differentiated thyroid cancers, albeit with challenges such as treatment resistance and adverse effects. The application of CAR-T cell therapy, successful in hematologic cancers, provides a novel perspective for thyroid cancer treatment, although its efficacy in solid tumors requires further study. Additionally, research into tumor vaccines and immunomodulators is advancing, with preliminary evidence suggesting their therapeutic potential for thyroid cancer patients. CONCLUSION The recognition of the immune microenvironment's role in treatment responsiveness is pivotal for enhancing the care of thyroid cancer patients. This review underscores the significance of combination therapy as a means to optimize treatment outcomes and charts a course for future research endeavors to broaden the spectrum of effective treatment options available to thyroid cancer patients.
Collapse
Affiliation(s)
- Ping Song
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Gang Pan
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Zhang
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Yeqin Ni
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Qianyu Wang
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jingjng Shi
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - You Peng
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Ruirui Jing
- Department of Translational Medicine and Clinical Research, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Dingcun Luo
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China; The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; College of Mathematical Medicine, Zhejiang Normal University, Jinhua, Zhejiang, China.
| |
Collapse
|
10
|
Ma X, Hu B, Zhou X, Wang L, Chen H, Xie F, Zhu H, Jia B, Yang Z. Development and First-in-Human evaluation of a Site-Specific [ 18F]-Labeled PD-L1 nanobody PET radiotracer for noninvasive imaging in NSCLC. Bioorg Chem 2025; 156:108222. [PMID: 39889552 DOI: 10.1016/j.bioorg.2025.108222] [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: 12/24/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Immunohistochemistry (IHC) for PD-L1 detection is limited by its invasiveness and heterogeneity of tumors. To address these challenges, a new PD-L1-targeted nanobody-based immune-PET radiotracer [18F]AlF-APN09 was developed using the site-specific radiolabeling method with the complexing agent (Mal-RESCA) under mild conditions. [18F]AlF-APN09 was prepared at room temperature (pH 4.6-4.8) within 20 min with satisfactory radiochemical yields (45.8 ± 4.48 %, non-decay corrected), high radiochemical purity (>98 %) and moderate apparent molar activity (15-35 GBq/μmol), and remained stable in both PBS and 5 % HSA after 4 h (>90 %). Cell uptake studies indicated variable levels of surface PD-L1 expression in the following order: A549PD-L1 > H1975 > A549. In micro-PET/CT imaging, A549PD-L1 and H1975 tumors were distinctly visualized in a 6.0:1 and 3.2:1 ratios over PD-L1-negative A549 tumors in vivo. Ex vivo biodistribution studies showed tumor uptake values of 6.47 ± 1.06 %ID/g (A549PD-L1) and 2.27 ± 0.19 %ID/g (H1975), significantly higher than 0.90 ± 0.28 %ID/g in A549 tumors. The estimated effective radiation dose in humans was 8.65E-03 mSv/MBq, lower than that of conventional [18F]FDG. First-in-human imaging was conducted on a single resectable non-small cell lung cancer (NSCLC) subject without any adverse reactions. The radiotracer exhibited renal excretion with minimal hepatobiliary clearance. Tumor uptake reached SUVmax 4.20 at 2 h post-injection, demonstrating high contrast and rapid clearance. After PD-1 inhibitor immunotherapy and chemotherapy, the subject showed a therapeutic response and postoperative pathological specimens confirmed a major pathological response (MPR). These results suggest that we have successfully developed a new PD-L1-targeted nanobody PET tracer using the site-specific labeling method with the complexing agent (Mal-RESCA) within 20 min under mild conditions and [18F]AlF-APN09 is a promising noninvasive PET radiotracer for visualizing PD-L1 expression in tumors, offering rapid tumor targeting, excellent signal-to-noise ratios, and favorable clearance properties.
Collapse
Affiliation(s)
- Xiaopan Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142 China; Department of Nuclear Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441138 China
| | - Biao Hu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191 China
| | - Xin Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142 China
| | - Lei Wang
- Department of Laboratory Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441138 China
| | - Hui Chen
- Department of Nuclear Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441138 China
| | - Fei Xie
- Department of Nuclear Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441138 China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142 China.
| | - Bing Jia
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191 China.
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142 China.
| |
Collapse
|
11
|
Zheng M, Hu M, Liu Y, Li X, Wang G, Zhang T, Zhao Y. A Cohort Study on Dual Predictive Markers of Immune Combination Therapy for Advanced Non-Small Cell Lung Cancer. Biomark Insights 2025; 20:11772719251319641. [PMID: 39957864 PMCID: PMC11829304 DOI: 10.1177/11772719251319641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 01/24/2025] [Indexed: 02/18/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) hold a great promise in treatment of non-small cell lung cancer (NSCLC), while only a portion of patients benefited from the treatment, and others could not achieve optimal therapeutic effects from initial immunotherapy, even for those patients with PD-L1 (Programed cell death ligand 1) tested positive. However, the clinical markers for the selection of patients who will benefit from ICIs combination treatment beforehand are largely unknown. Objectives The purpose of this study was to explore the non-invasive biomarkers that can predict the efficacy of immune combination therapy in advanced/metastatic NSCLC patients. Design This study employed a retrospective cohort design to analyze dual predictive biomarkers in advanced non-small cell lung cancer (NSCLC) patients with immune combination therapy. Method An analysis was conducted on baseline information of 144 patients with advanced/metastatic NSCLC who received ICIs treatment from the November of 2018 to the January of 2023 in Beijing Chest Hospital. We established a scoring group chart to make quantitative prediction for overall survival (OS) and progression-free survival (PFS) based on 4 variables, and set up the nomogram model as well as Decision curve analysis (DCA) to assess clinical benefits of ICIs combination in treatment of patients with advanced/metastatic NSCLC. Results We found that serum globulin (GLB) >26.6 (g/L) (HR = 1.865, P = .002), absolute neutrophil counts (ANC) (109/L) > 5 (HR = 2.146, P < .001), and bone metastasis (HR = 2.148, P < .001) were independent factors affecting the PFS of NSCLC patients. GLB > 26.6 (g/L) (HR = 1.741, P = .018), ANC (109/L) >5 (HR = 1.807, P = .008), bone metastasis (HR = 1.651, P = .002), and PD-L1 Negative (HR = 2.432, P = .032) were independent factors affecting the OS of NSCLC patients. Same variables and cut-off value have good predictive efficacy in both PFS and OS. Conclusion In patients with advanced/metastatic NSCLC receiving ICIs combination treatment, the GLB, ANC, bone metastasis, and PD-L1 may serve as useful predictive markers for the prognosis of NSCLC patients with ICIs combination treatment.
Collapse
Affiliation(s)
- Maike Zheng
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Tongzhou District, Beijing, China
| | - Mingming Hu
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Tongzhou District, Beijing, China
| | - Yanxia Liu
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Tongzhou District, Beijing, China
| | - Xiaomi Li
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Guirong Wang
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Tongzhou District, Beijing, China
| | - Tongmei Zhang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Tongzhou District, Beijing, China
| | - Yan Zhao
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Tongzhou District, Beijing, China
| |
Collapse
|
12
|
Trisolini R, Cetoretta V, Sotgiu G, Cancellieri A, Puci M, Viscuso M, Livi V, Cani M, Scambia G, Cappuzzo F, Bria E, Novello S. Supraclavicular Lymph Node Metastases in Advanced Lung Cancer: Prevalence and Analysis of Demographic, Clinical and Molecular Characteristics. Clin Lung Cancer 2025:S1525-7304(25)00033-6. [PMID: 40037966 DOI: 10.1016/j.cllc.2025.02.003] [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: 08/29/2024] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND The prevalence of supraclavicular lymph nodes metastases (SNM) in advanced lung cancer has not been systematically evaluated, nor has then been a comparison of demographic, clinical, or molecular characteristics between patients with and without SNM. METHODS In this prospective cohort study, the presence of SNM was evaluated using imaging studies (CT, PET, neck ultrasonography) in patients with suspected advanced lung cancer referred for biopsy aimed at diagnosis and molecular profiling. Ultrasound-guided biopsy confirmed or excluded metastatic involvement when suspicious supraclavicular nodes were identified. We assessed the prevalence of SNM and compared the demographic, clinicopathologic and molecular characteristics of patients with and without SNM. RESULTS Among the 348 patients with advanced lung cancer, 94 (27%) had SMN. SMN was more common in small cell lung cancer (24/48, 50%) and adenocarcinoma (61/248, 24.6%) than in squamous cell carcinoma (4/35, 11.4%). Compared to patients without SMN, those with SMN were more likely to have small-cell lung cancer, N2/3 disease (97.9 vs. 83.9%, P < .0001), liver metastases (29.8% vs. 16.1% P = .006), and metastases to less common sites (33.7% vs. 14.1%, P < .0001). The prevalence of genomic alterations and PD-L1 expression did not differ between biopsy samples obtained from SNM and those from the primary tumor or other metastatic sites. CONCLUSION SNM is common in patients with advanced small-cell lung cancer and adenocarcinoma. Ultrasound-guided biopsy of SNM is a simple and relatively inexpensive method for obtaining adequate tissue samples for diagnosis and comprehensive molecular profiling.
Collapse
Affiliation(s)
- Rocco Trisolini
- Interventional Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Hearth, Rome, Italy
| | - Valeria Cetoretta
- Interventional Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Hearth, Rome, Italy; Department of Oncology, University of Turin, Azienda Ospedaliero-Universitaria San Luigi, Turin, Italy
| | - Giovanni Sotgiu
- Clinical Epidemiology and Medical Statistics Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | | | - Mariangela Puci
- Clinical Epidemiology and Medical Statistics Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Marta Viscuso
- Interventional Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Hearth, Rome, Italy; Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Vanina Livi
- Interventional Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Hearth, Rome, Italy
| | - Massimiliano Cani
- Department of Oncology, University of Turin, Azienda Ospedaliero-Universitaria San Luigi, Turin, Italy
| | - Giovanni Scambia
- Gynecologic Oncology Unit, Department of Woman, Child and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Federico Cappuzzo
- Medical Oncology Division, IRCCS Regina Elena National Cancer Institute, Rome, Italy Medical
| | - Emilio Bria
- Oncology Division, Gemelli Isola Hospital, Catholic University of the Sacred Hearth, Rome, Italy
| | - Silvia Novello
- Department of Oncology, University of Turin, Azienda Ospedaliero-Universitaria San Luigi, Turin, Italy.
| |
Collapse
|
13
|
Das L, Das S. A comprehensive insights of cancer immunotherapy resistance. Med Oncol 2025; 42:57. [PMID: 39883235 DOI: 10.1007/s12032-025-02605-8] [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: 12/07/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
Cancer is a major global health issue that is usually treated with multiple therapies, such as chemotherapy and targeted therapies like immunotherapy. Immunotherapy is a new and alternative approach to treating various types of cancer that are difficult to treat with other methods. Although immune checkpoint inhibitors have shown promise for long-term efficacy, they have limited effectiveness in common cancer types such as breast, prostate, and lung. Some patients do not respond to immunotherapy, while others develop resistance to the treatment over time, which is classified as primary or acquired resistance. Cancer immunotherapy, specifically immune checkpoint inhibitor-based resistance involves multiple factors such as genes, metabolism, inflammation, and angiogenesis. However, cutting-edge research has identified the mechanisms of immunotherapy resistance and possible solutions. Current research may improve biomarker identification and modify treatment strategies, which will lead to better clinical outcomes. This review provides a comprehensive discussion of the current mechanisms of immunotherapy resistance, related biomarker modulation, and strategies to overcome resistance.
Collapse
Affiliation(s)
- Laavanya Das
- Department of Food and Nutrition, Brainware University, 398, Ramkrishnapur Rd, Barasat, Kolkata, West Bengal, 700125, India
| | - Subhadip Das
- Department of In Vivo Pharmacology, TCG Lifesciences Pvt. Ltd, BN 7, Sector V, Salt Lake City, Kolkata, West Bengal, 700091, India.
| |
Collapse
|
14
|
Zhao Q, Li B, Xu Y, Li X, Yu J, Wang L. IRF4: A potential prognostic biomarker for immunotherapy in NSCLC. Int Immunopharmacol 2024; 143:113411. [PMID: 39437487 DOI: 10.1016/j.intimp.2024.113411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/12/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Immunotherapy is revolutionizing the management of advanced non-small cell lung cancer (NSCLC). However, sustained responses are observed in only a minority of patients. Reliable biomarkers are required to identify potential beneficiaries. Interferon regulatory factor 4 (IRF4) plays a crucial role in immune regulation, suggesting its potential as a prognostic biomarker in NSCLC immunotherapy. This study aimed to investigate the predictive role of IRF4 expression in patients with NSCLC receiving immunotherapy. METHODS Data from three NSCLC cohorts treated with immune checkpoint inhibitors were collected from the Gene Expression Omnibus (GEO) database. The prognostic significance of IRF4 was assessed across these cohorts, and gene set enrichment analysis (GSEA) was performed. IRF4-based nomograms were developed to predict the outcomes of immunotherapy. Correlations among IRF4 expression, immune cell infiltration, and immunotherapy prognosis were evaluated in our cohort. RESULTS Elevated IRF4 expression was associated with improved prognosis in patients with NSCLC undergoing immunotherapy, consistent with both GEO dataset and our cohort. IRF4 emerged as an independent predictor for progression-free survival (PFS) and overall survival (OS) in multivariable Cox regression analysis. GSEA analysis highlighted links between IRF4 expression and immune activation pathways such as Chemokine_Signaling_Pathway, Natural_Killer_Cell_Mediated_Cytotoxicity, B_Cell_Receptor_Signaling_Pathway, and T_Cell_Receptor_Signaling_Pathway. In our cohort, immunohistochemistry demonstrated correlations between IRF4 expression and the infiltration of CD8+ T cells, CD20+ B cells, and PD-L1 expression in the tumor microenvironment. CONCLUSION High IRF4 expression in baseline tumor tissue could serve as a favorable predictor of NSCLC immunotherapy outcomes, aiding in personalized treatment strategies.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430064, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Butuo Li
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Yiyue Xu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Xuanzong Li
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinming Yu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430064, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| | - Linlin Wang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
15
|
Nu er lan STE, Yu B, Yang Y, Shen Y, Xu B, Zhan Y, Liu C. Discover Mutational Differences Between Lung Adenocarcinoma and Lung Squamous Cell Carcinoma and Search for More Effective Biomarkers for Immunotherapy. Cancer Manag Res 2024; 16:1759-1773. [PMID: 39678041 PMCID: PMC11645897 DOI: 10.2147/cmar.s491661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose Lung cancer is a severe malignant tumor. This study aims to more comprehensively characterize lung cancer patients and identify combination markers for immunotherapy. Patients and Methods We gathered data from 166 lung cancer patients at the Cancer Hospital Affiliated with Xinjiang Medical University. The collected samples underwent NGS sequencing using a panel of 616 genes associated with cancer. Subsequently, data analysis was conducted to identify markers that are more suitable for lung cancer immunotherapy. Results In this study, the most common variant genes in LUAD were TP53, EGFR, MST1, KMT2C, RBM10, LRP1B. Meanwhile, the highest mutation frequency genes in LUSC samples were TP53, KMT2D, LRP1B, FAT1, MST1, KMT2C. Mutation frequencies, tumor mutation burden (TMB), PD-L1 expression, and mutant-allele tumor heterogeneity (MATH) values differed between LUAD and LUSC, with LUSC exhibiting higher values than LUAD. Irrespective of LUAD or LUSC, patients with TMB≥10 demonstrated better immunotherapy efficacy compared to patients with TMB<10. Similarly, when PD-L1≥50%, whether in LUAD or LUSC, the immunotherapy effect was superior to that of patients with PD-L1<50%. Combining TMB≥10 and PD-L1≥50% as immunotherapy markers, in both LUAD and LUSC, resulted in a very favorable immunotherapy effect, with the overall response rate (ORR) reaching 100%. Conclusion We observed distinct mutation patterns and clinical factors between LUAD and LUSC, and noted that patients with TMB≥10 and PD-L1≥50% exhibited enhanced immunotherapy effects. Combining TMB≥10 and PD-L1≥50% proved to be a more effective predictor of immunotherapy efficacy.
Collapse
Affiliation(s)
- Sai te er Nu er lan
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| | - Bo Yu
- Beijing USCI Medical Laboratory, Beijing, 100195, People’s Republic of China
| | - Yan Yang
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| | - Yanli Shen
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| | - Bing Xu
- Beijing USCI Medical Laboratory, Beijing, 100195, People’s Republic of China
| | - Yiyi Zhan
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| | - Chunling Liu
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| |
Collapse
|
16
|
He Y, Zhu M, Lai X, Zhang H, Jiang W. The roles of PD-L1 in the various stages of tumor metastasis. Cancer Metastasis Rev 2024; 43:1475-1488. [PMID: 38733457 DOI: 10.1007/s10555-024-10189-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
The interaction between tumor programmed death ligand 1 (PD-L1) and T-cell programmed cell death 1 (PD-1) has long been acknowledged as a mechanism for evading immune surveillance. Recent studies, however, have unveiled a more nuanced role of tumor-intrinsic PD-L1 in reprograming tumoral phenotypes. Preclinical models emphasize the synchronized effects of both intracellular and extracellular PD-L1 in promoting metastasis, with intricate interactions with the immune system. This review aims to summarize recent findings to elucidate the spatiotemporal heterogeneity of PD-L1 expression and the pro-metastatic roles of PD-L1 in the entire process of tumor metastasis. For example, PD-L1 regulates the epithelial-to-mesenchymal transition (EMT) process, facilitates the survival of circulating tumor cells, and induces the formation of immunosuppressive environments at pre-metastatic niches and metastatic sites. And the complexed and dynamic regulation process of PD-L1 for tumor metastasis is related to the spatiotemporal heterogeneity of PD-L1 expression and functions from tumor primary sites to various metastatic sites. This review extends the current understandings for the roles of PD-L1 in mediating tumor metastasis and provides new insights into therapeutic decisions in clinical practice.
Collapse
Affiliation(s)
- Yinjun He
- Department of Colorectal Surgery, First Affiliated Hospital, Zhejiang University Medical School, Hangzhou, 310009, China
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China
| | - Ming Zhu
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China
| | - Xuan Lai
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China
| | - Honghe Zhang
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China.
| | - Weiqin Jiang
- Department of Colorectal Surgery, First Affiliated Hospital, Zhejiang University Medical School, Hangzhou, 310009, China.
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China.
| |
Collapse
|
17
|
Mo DC, Huang JF, Lin P, Huang SX, Wang HL, Luo PH, Liang XJ. The role of PD-L1 in patients with non-small cell lung cancer receiving neoadjuvant immune checkpoint inhibitor plus chemotherapy: a meta-analysis. Sci Rep 2024; 14:26200. [PMID: 39482343 PMCID: PMC11527982 DOI: 10.1038/s41598-024-78159-y] [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: 07/06/2024] [Accepted: 10/29/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND The use of immune checkpoint inhibitors (ICIs) as neoadjuvant therapy is a promising novel approach in resectable non-small-cell lung cancer (NSCLC). This study aimed to investigate the prognostic value of PD-L1 in patients with NSCLC receiving neoadjuvant immune checkpoint inhibitor plus chemotherapy (CT). MATERIALS AND METHODS Several databases (PubMed, Embase, and cochrane central register of controlled trials [CENTRAL]) were systematically searched. Randomized controlled trials (RCTs) investigating pathological and survival outcomes with neoadjuvant ICI + CT versus CT alone in NSCLC were analyzed. RESULTS Overall, eight RCTs (n = 3,404) were included. The analyses showed neoadjuvant ICI + CT significantly improved complete pathological response (pCR) and event-free survival (EFS) in either tumor PD-L1 < 1%, ≥ 1%, 1-49%, or ≥ 50% population (both p < 0.0001) compared with neoadjuvant CT alone. The overall survival (OS) data are not yet mature among all included RCTs, and only three RCTs presented OS data by PD-L1 status of patients. The pooled OS favored neoadjuvant ICI + CT in the PD-L1 ≥ 1% population (hazard ratio [HR], 0.45; 95% CI, 0.31-0.65; p < 0.0001), but not in the PD-L1 < 1% population (HR, 0.89; 95% CI, 0.66-1.19; p = 0.43). CONCLUSIONS Compared with neoadjuvant CT alone, neoadjuvant ICI + CT significantly enhanced pCR and EFS for patients with resectable NSCLC regardless of the expression of PD-L1. It seems that only patients with PD-L1 positive tumors may achieve a better OS, but it's currently inconclusive due to immature data, so future research with long-term follow-up is still needed.
Collapse
Affiliation(s)
- Dun-Chang Mo
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China.
| | - Jian-Feng Huang
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China
| | - Peng Lin
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China
| | - Shang-Xiao Huang
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China
| | - Han-Lei Wang
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China
| | - Peng-Hui Luo
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China
| | - Xiu-Juan Liang
- Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, 530000, Guangxi, China
| |
Collapse
|
18
|
Pan L, Mu L, Lei H, Miao S, Hu X, Tang Z, Chen W, Wang X. Predicting survival benefits of immune checkpoint inhibitor therapy in lung cancer patients: a machine learning approach using real-world data. Int J Clin Pharm 2024:10.1007/s11096-024-01818-7. [PMID: 39470981 DOI: 10.1007/s11096-024-01818-7] [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: 07/26/2024] [Accepted: 10/04/2024] [Indexed: 11/01/2024]
Abstract
BACKGROUND Due to the heterogeneity in the effectiveness of immunotherapy for lung cancer, identifying predictors is crucial. AIM This study aimed to develop a machine learning model to identify predictors of overall survival in lung cancer patients treated with immune checkpoint inhibitors (ICIs). METHOD A retrospective analysis was performed on data from 1314 lung cancer patients at the Chongqing University Cancer Hospital from September 2018 to September 2022. We used the random survival forest (RSF) model to identify survival-influencing factors, using backward elimination for variable selection. A Cox proportional hazards (CPH) model was constructed using the most significant predictors. We assessed model performance and generalizability using time-dependent receiver operating characteristics (ROC) and predictive error curves. RESULTS The RSF model demonstrated better predictive accuracy than the CPH (IBS 0.17 vs. 0.17; C-index 0.91 vs. 0.68), with better discrimination and prediction performance. The influential variables identified included D-dimer, Karnofsky performance status, albumin, surgery, TNM stage, platelet count, and age. The RSF model, which incorporated these variables, achieved area under the curve (AUC) scores of 0.95, 0.94, and 0.98 for 1-, 3-, and 5-year survival predictions, respectively, in the training set. The validation set showed AUCs of 0.94, 0.90, and 0.95, respectively, exceeding the performance of the CPH model. CONCLUSION The study successfully developed a machine learning model that accurately predicted the survival benefits of ICI therapy in lung cancer patients, supporting clinical decision-making in lung cancer treatment.
Collapse
Affiliation(s)
- Lingyun Pan
- Department of Pharmacy, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Li Mu
- Department of Pharmacy, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Haike Lei
- Chongqing Cancer Multi-Omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Siwei Miao
- Centre for Medical Big Data and Artificial Intelligence, First Affiliated Hospital of Third Military Medical University: Southwest Hospital, Chongqing, China
| | - Xiaogang Hu
- Department of Pharmacy, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Zongwei Tang
- Department of Pharmacy, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Wanyi Chen
- Department of Pharmacy, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, China.
- Chongqing University, Chongqing, China.
| | - Xiaoxiao Wang
- Department of Pharmacy, Chongqing University Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, China.
| |
Collapse
|
19
|
El Beaino Z, Dupain C, Marret G, Paoletti X, Fuhrmann L, Martinat C, Allory Y, Halladjian M, Bièche I, Le Tourneau C, Kamal M, Vincent-Salomon A. Pan-cancer evaluation of tumor-infiltrating lymphocytes and programmed cell death protein ligand-1 in metastatic biopsies and matched primary tumors. J Pathol 2024; 264:186-196. [PMID: 39072750 DOI: 10.1002/path.6334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/22/2024] [Accepted: 06/19/2024] [Indexed: 07/30/2024]
Abstract
Tumor immunological characterization includes evaluation of tumor-infiltrating lymphocytes (TILs) and programmed cell death protein ligand-1 (PD-L1) expression. This study investigated TIL distribution, its prognostic value, and PD-L1 expression in metastatic and matched primary tumors (PTs). Specimens from 550 pan-cancer patients of the SHIVA01 trial (NCT01771458) with available metastatic biopsy and 111 matched PTs were evaluated for TILs and PD-L1. Combined positive score (CPS), tumor proportion score (TPS), and immune cell (IC) score were determined. TILs and PD-L1 were assessed according to PT organ of origin, histological subtype, and metastatic biopsy site. We found that TIL distribution in metastases did not vary according to PT organ of origin, histological subtype, or metastatic biopsy site, with a median of 10% (range: 0-70). TILs were decreased in metastases compared to PT (20% [5-60] versus 10% [0-40], p < 0.0001). CPS varied according to histological subtype (p = 0.02) and biopsy site (p < 0.02). TPS varied according to PT organ of origin (p = 0.003), histological subtype (p = 0.0004), and metastatic biopsy site (p = 0.00004). TPS was higher in metastases than in PT (p < 0.0001). TILs in metastases did not correlate with overall survival. In conclusion, metastases harbored fewer TILs than matched PT, regardless of PT organ of origin, histological subtype, and metastatic biopsy site. PD-L1 expression increased with disease progression. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Zakhia El Beaino
- Department of Pathology, Institut Curie, PSL Research University, Paris, France
| | - Célia Dupain
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - Grégoire Marret
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - Xavier Paoletti
- INSERM U900 Research Unit, Institut Curie, Saint-Cloud, France
- Department of Biostatistics, Institut Curie, Paris, France
| | - Laëtitia Fuhrmann
- Department of Pathology, Institut Curie, PSL Research University, Paris, France
| | - Charlotte Martinat
- Department of Pathology, Institut Curie, PSL Research University, Paris, France
| | - Yves Allory
- Department of Pathology, Institut Curie, Saint-Cloud, Versailles Saint-Quentin University, Paris-Saclay, France
| | - Maral Halladjian
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - Ivan Bièche
- Department of Genetics, Institut Curie, Paris, France
- INSERM U1016 Research Unit, Paris, France
- Faculty of Pharmaceutical and Biological Sciences, Paris-Cité University, Paris, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
- INSERM U900 Research Unit, Institut Curie, Saint-Cloud, France
- Paris-Saclay University, Paris, France
| | - Maud Kamal
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | | |
Collapse
|
20
|
Pathak G, Shah R, Castonguay M, Cheng A, Fris J, Murphy R, Darling G, Ednie A, French D, Henteleff H, Mujoomdar A, Plourde M, Wallace A, Xu Z. Temporal Effect on PD-L1 Detection and Novel Insights Into Its Clinical Implications in Non-Small Cell Lung Cancer. Cancer Med 2024; 13:e70262. [PMID: 39382248 PMCID: PMC11462595 DOI: 10.1002/cam4.70262] [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: 07/12/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 10/10/2024] Open
Abstract
OBJECTIVES Several studies rely on archived tissue blocks to assess the PD-L1 scores; however, a detailed analysis of potential variations of scores between fresh and archived tissue blocks still lacks. In addition, the prognostic implications of PD-L1 in lung cancers have not yet been completely understood. Here, we aimed to investigate the temporal variation in PD-L1 scores from clinical samples and the clinical implications of PD-L1 in non-small cell lung cancer (NSCLC). METHODS NSCLC cases from January 2005 to June 2023 were considered for this study, and PD-L1 scores in archived and fresh tissue blocks were analyzed. Association of PD-L1 with various driver mutations was explored, and implications of PD-L1 in progression-free survival (PFS) and overall survival (OS) were analyzed. RESULTS Our study revealed a significant disparity in PD-L1 scores between archived and fresh tissue blocks, and a temporal variation in scores within 6 months of tissue acquisition. Advanced-stage primary tumors, metastatic lymph nodes, and visceral pleural invasion revealed higher PD-L1 expression as presented by tumor proportion score (TPS). Notably, in fully resected stage I/II NSCLC cases, OS was better in the high PD-L1 (≥ 50% TPS) cohort with driver mutations compared to cases without driver mutations (hazard ratio-0.5129, 95% confidence interval 0.2058-1.084, p = 0.0779). In contrast, high PD-L1 was associated with worse OS compared to no PD-L1 (< 1% TPS) (hazard ratio-2.431, 95% confidence interval 1.144-6.656, p = 0.0242) in the cohort without driver mutations. Furthermore, the presence of a KRAS mutation favored the outcome of anti-PD-L1/PD1 immunotherapy in advanced NSCLC. CONCLUSION PD-L1 detection from tissue blocks was found to vary temporally, urging for a prioritized consideration for patients with marginal scores when archived blocks are employed for its detection. Prognostic roles of PD-L1 were associated with driver mutations, and KRAS mutations favored the outcome of anti-PD-L1/PD1 therapy in advanced NSCLC.
Collapse
Affiliation(s)
- Gopal P. Pathak
- Department of PathologyQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Rashmi Shah
- Department of PathologyQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Mathieu Castonguay
- Department of PathologyQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Angela Cheng
- Department of PathologyQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - John Fris
- Department of PathologyQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Rowan Murphy
- Division of Thoracic SurgeryQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Gail Darling
- Division of Thoracic SurgeryQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Alexander Ednie
- Division of Thoracic SurgeryQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Daniel French
- Division of Thoracic SurgeryQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Harry Henteleff
- Division of Thoracic SurgeryQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Aneil Mujoomdar
- Division of Thoracic SurgeryQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Madelaine Plourde
- Division of Thoracic SurgeryQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Alison Wallace
- Division of Thoracic SurgeryQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| | - Zhaolin Xu
- Department of PathologyQEII Health Sciences Centre and Dalhousie UniversityHalifaxNova ScotiaCanada
| |
Collapse
|
21
|
Wang Y, Zhang Y, Chen Y, Wang S, Liu W, Liu Z, Hu M. [ 18F]AlF-NOTA-PCP2: a novel PET/CT tracer for enhanced PD-L1 heterogeneity imaging and comparative analysis with [ 18F]AlF-NOTA-WL12 in glioblastoma xenografts. Eur J Nucl Med Mol Imaging 2024; 51:3161-3175. [PMID: 38713298 DOI: 10.1007/s00259-024-06743-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE The unsatisfactory efficacy of PD-L1 antibodies in glioblastoma (GBM) is largely due to the temporal and spatial heterogeneity of PD-L1 expression. Molecular imaging can enhance understanding of the tumor immune microenvironment and guide immunotherapy. However, highly sensitive imaging agents capable of effectively visualizing PD-L1 heterogeneity are limited. This study introduces a novel PET tracer, offering improved imaging of PD-L1 heterogeneity in GBM xenografts, with a comparative analysis to [18F]AlF-NOTA-WL12. METHODS [18F]AlF-NOTA-PCP2 was synthesized with high purity and its affinity for PD-L1 was characterized using surface plasmon resonance (SPR) and cell binding assays. Its specificity for PD-L1 was evaluated both in vitro using various cell lines and in vivo with GBM xenograft models in NOD/SCID mice. PET/CT imaging was conducted to evaluate the tracer's biodistribution, pharmacokinetics, and ability to quantify tumoral spatial heterogeneity of PD-L1 expression. A focused comparative analysis between [18F]AlF-NOTA-PCP2 and [18F]AlF-NOTA-WL12 was conducted, examining binding affinity, biodistribution, pharmacokinetics, and imaging effectiveness in GBM xenografts. Additionally, human radiation dosimetry estimates compared the safety profiles of both tracers. RESULTS [18F]AlF-NOTA-PCP2 demonstrated high radiochemical purity (> 95%) and a strong affinity for PD-L1, comparable to [18F]AlF-NOTA-WL12. In vitro and in vivo studies confirmed its specificity for PD-L1, with increased uptake in PD-L1 expressing cells and tumors. Toxicological profiles indicated no significant abnormalities in serum biochemical indicators or major organ tissues. MicroPET/CT imaging showed [18F]AlF-NOTA-PCP2's effectiveness in visualizing PD-L1 expression levels and spatial heterogeneity in GBM xenografts. Comparative studies revealed [18F]AlF-NOTA-PCP2's improved pharmacokinetic properties, including higher tumor-to-blood ratios and lower nonspecific liver uptake, as well as reduced radiation exposure compared to [18F]AlF-NOTA-WL12. CONCLUSION [18F]AlF-NOTA-PCP2 distinguishes itself as an exceptionally sensitive PET/CT tracer, adept at non-invasively and accurately quantifying PD-L1 expression and its spatial heterogeneity in tumors, especially in GBM. Its favorable pharmacokinetic properties, safety profile, and high affinity for PD-L1 highlight its potential for enhancing the precision of cancer immunotherapy and guiding individualized treatment strategies. While promising, its clinical translation, especially in brain imaging, necessitates further validation in clinical trials.
Collapse
Affiliation(s)
- Yong Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Yang Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Yunhao Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Shijie Wang
- Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Wei Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Zhiguo Liu
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Man Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
22
|
Alsaab HO, Alzahrani MS, Bahauddin AA, Almutairy B. Circulating tumor DNA (ctDNA) application in investigation of cancer: Bench to bedside. Arch Biochem Biophys 2024; 758:110066. [PMID: 38906310 DOI: 10.1016/j.abb.2024.110066] [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: 04/10/2024] [Revised: 06/02/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Now, genomics forms the core of the precision medicine concept. Comprehensive investigations of tumor genomes have made it possible to characterize tumors at the molecular level and, specifically, to identify the fundamental processes that cause condition. A variety of kinds of tumors have seen better outcomes for patients as a result of the development of novel medicines to tackle these genetic-driving processes. Since therapy may exert selective pressure on cancers, non-invasive methods such as liquid biopsies can provide the opportunity for rich reservoirs of crucial and real-time genetic data. Liquid biopsies depend on the identification of circulating cells from tumors, circulating tumor DNA (ctDNA), RNA, proteins, lipids, and metabolites found in patient biofluids, as well as cell-free DNA (cfDNA), which exists in those with cancer. Although it is theoretically possible to examine biological fluids other than plasma, such as pleural fluid, urine, saliva, stool, cerebrospinal fluid, and ascites, we will limit our discussion to blood and solely cfDNA here for the sake of conciseness. Yet, the pace of wider clinical acceptance has been gradual, partly due to the increased difficulty of choosing the best analysis for the given clinical issue, interpreting the findings, and delaying proof of value from clinical trials. Our goal in this review is to discuss the current clinical value of ctDNA in cancers and how clinical oncology systems might incorporate procedures for ctDNA testing.
Collapse
Affiliation(s)
- Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif, 21944, Saudi Arabia.
| | - Mohammad S Alzahrani
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.
| | - Ammar A Bahauddin
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Medina Al-Munawarah, Saudi Arabia.
| | - Bandar Almutairy
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia.
| |
Collapse
|
23
|
Hummelink K, van der Noort V, Muller M, Schouten RD, van den Heuvel MM, Thommen DS, Smit EF, Meijer GA, Monkhorst K. Head-to-head comparison of composite and individual biomarkers to predict clinical benefit to PD-1 blockade in non-small cell lung cancer. PLoS One 2024; 19:e0293707. [PMID: 39083541 PMCID: PMC11290656 DOI: 10.1371/journal.pone.0293707] [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: 10/17/2023] [Accepted: 04/15/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND The efficacy of PD-1 blocking agents in advanced NSCLC has shown prolonged effectiveness, but only in a minority of patients. Multiple biomarkers have been explored to predict treatment benefit, yet their combined performance remains inadequately examined. In this study, we assessed the combined predictive performance of multiple biomarkers in NSCLC patients treated with nivolumab. METHODS Pretreatment samples from 135 patients receiving nivolumab were used to evaluate the predictive performance of CD8 tumor-infiltrating lymphocytes (TILs), intratumoral (IT) localization of CD8 TILs, PD-1 high expressing TILs (PD1T TILs), CD3 TILs, CD20 B-cells, tertiary lymphoid structures (TLS), PD-L1 tumor proportion score (TPS) and the Tumor Inflammation score (TIS). Patients were randomly assigned to a training (n = 55) and validation cohort (n = 80). The primary outcome measure was Disease Control at 6 months (DC 6m) and the secondary outcome measure was DC at 12 months (DC 12m). RESULTS In the validation cohort, the two best performing composite biomarkers (i.e. CD8+IT-CD8 and CD3+IT-CD8) demonstrated similar or lower sensitivity (64% and 83%) and NPV (76% and 85%) compared to individual biomarkers PD-1T TILs and TIS (sensitivity: 72% and 83%, NPV: 86% and 84%) for DC 6m, respectively. Additionally, at 12 months, both selected composite biomarkers (CD8+IT-CD8 and CD8+TIS) demonstrated inferior predictive performance compared to PD-1T TILs and TIS alone. PD-1T TILs and TIS showed high sensitivity (86% and 100%) and NPV (95% and 100%) for DC 12m. PD-1T TILs could more accurately discriminate patients with no long-term benefit, as specificity was substantially higher compared to TIS (74% versus 39%). CONCLUSION Composite biomarkers did not show improved predictive performance compared to PD-1T TILs and TIS alone for both the 6- and 12-month endpoints. PD-1T TILs and TIS identified patients with DC 12m with high sensitivity. Patients with no long-term benefit to PD-1 blockade were most accurately identified by PD-1T TILs.
Collapse
Affiliation(s)
- Karlijn Hummelink
- Department of Pathology, Division of Diagnostic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Thoracic Oncology, Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Mirte Muller
- Department of Thoracic Oncology, Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Robert D. Schouten
- Department of Thoracic Oncology, Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michel M. van den Heuvel
- Department of Thoracic Oncology, Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daniela S. Thommen
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Egbert F. Smit
- Department of Thoracic Oncology, Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Gerrit A. Meijer
- Department of Pathology, Division of Diagnostic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kim Monkhorst
- Department of Pathology, Division of Diagnostic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Mo DC, Chen L, Wu Y, Huang JF, Liang XJ. PD-L1 Expression and Its Prognostic Value in Different Tumor Specimens in Epidermal Growth Factor Receptor-Mutated Non-Small Cell Lung Cancer. J Clin Oncol 2024; 42:2616-2617. [PMID: 38754073 DOI: 10.1200/jco.24.00230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/01/2024] [Indexed: 05/18/2024] Open
Affiliation(s)
- Dun-Chang Mo
- Dun-Chang Mo, MD, Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; Long Chen, MD, Otorhinolaryngology Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; Yu Wu, MD, Medical Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; and Jian-Feng Huang, MD and Xiu-Juan Liang, MD, Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Long Chen
- Dun-Chang Mo, MD, Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; Long Chen, MD, Otorhinolaryngology Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; Yu Wu, MD, Medical Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; and Jian-Feng Huang, MD and Xiu-Juan Liang, MD, Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yu Wu
- Dun-Chang Mo, MD, Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; Long Chen, MD, Otorhinolaryngology Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; Yu Wu, MD, Medical Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; and Jian-Feng Huang, MD and Xiu-Juan Liang, MD, Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jian-Feng Huang
- Dun-Chang Mo, MD, Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; Long Chen, MD, Otorhinolaryngology Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; Yu Wu, MD, Medical Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; and Jian-Feng Huang, MD and Xiu-Juan Liang, MD, Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiu-Juan Liang
- Dun-Chang Mo, MD, Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; Long Chen, MD, Otorhinolaryngology Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; Yu Wu, MD, Medical Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China; and Jian-Feng Huang, MD and Xiu-Juan Liang, MD, Radiotherapy Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
25
|
Zhang Y, Liu S, Liu D, Zhao Z, Song H, Peng K. Identification and validation of GIMAP family genes as immune-related prognostic biomarkers in lung adenocarcinoma. Heliyon 2024; 10:e33111. [PMID: 38948046 PMCID: PMC11211882 DOI: 10.1016/j.heliyon.2024.e33111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 05/15/2024] [Accepted: 06/14/2024] [Indexed: 07/02/2024] Open
Abstract
Background The GIMAP family genes play a key role in immune function. Increasing evidence suggests that GIMAP genes were implicated in the tumorigenesis of lung adenocarcinoma (LUAD). This study aimed to investigate the clinical significance of GIMAP family genes in LUAD. Methods In this study, we explored the expression, mutation, prognostic value of GIMAP family genes and the correlation with immune microenvironment in LUAD. We further investigated the relationship between GIMAP family genes expression and immunotherapy response in GEO LUAD and melanoma cohorts. Results Among the GIMAP family genes, the expression levels of GIMAP1, GIMAP2, GIMAP4, GIMAP5, GIMAP6, GIMAP7, and GIMAP8 were significantly lower in LUAD tumor tissues than normal tissues. Most GIMAP genes were closely related to age, tumor grade and T stage, but not significantly related to sex, N stage and M stage. In the overall population, patients with high expression of GIMAP family genes had a significant longer overall survival (OS). GO and KEGG enrichment analysis showed that GIMAP family genes were highly enriched in immune-related biological process. The expression of GIMAP family genes was positively correlated with immune cell infiltration and immune checkpoint molecules. Furthermore, high expression of GIMAP family genes were correlated with therapeutic response to immunotherapy in LUAD and melanoma patients. Conclusion In this study, we identified that GIMAP family genes were significantly associated with immune cell infiltration and immune checkpoint molecules. They potentially play a critical role in anti-tumor immunity and serve as immunotherapy biomarkers.
Collapse
Affiliation(s)
- Yanyan Zhang
- Department of Infectious Diseases, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Shan Liu
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease, Guangzhou, Guangdong, China
| | - Deyi Liu
- Department of General Practice, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhuxiang Zhao
- Department of Infectious Diseases, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Haifeng Song
- Department of Oncology, Lianzhou People's Hospital, Lianzhou, Guangdong, China
| | - Kunwei Peng
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy & Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Gao R, Lou N, Li L, Xie T, Xing P, Tang L, Yao J, Han X, Shi Y. Mutational variant allele frequency profile as a biomarker of response to immune checkpoint blockade in non-small cell lung Cancer. J Transl Med 2024; 22:576. [PMID: 38890738 PMCID: PMC11184775 DOI: 10.1186/s12967-024-05400-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
INTRODUCTION Identifying new biomarkers for predicting immune checkpoint inhibitors (ICIs) response in non-small cell lung cancer (NSCLC) is crucial. We aimed to assess the variant allele frequency (VAF)-related profile as a novel biomarker for NSCLC personalized therapy. METHODS We utilized genomic data of 915 NSCLC patients via cBioPortal and a local cohort of 23 patients for model construction and mutational analysis. Genomic, transcriptomic data from 952 TCGA NSCLC patients, and immunofluorescence (IF) assessment with the local cohort supported mechanism analysis. RESULTS Utilizing the random forest algorithm, a 15-gene VAF-related model was established, differentiating patients with durable clinical benefit (DCB) from no durable benefit (NDB). The model demonstrated robust performance, with ROC-AUC values of 0.905, 0.737, and 0.711 across training (n = 313), internal validation (n = 133), and external validation (n = 157) cohorts. Stratification by the model into high- and low-score groups correlated significantly with both progression-free survival (PFS) (training: P < 0.0001, internal validation: P < 0.0001, external validation: P = 0.0066) and overall survival (OS) (n = 341) (P < 0.0001). Notably, the stratification system was independent of PD-L1 (P < 0.0001) and TMB (P < 0.0001). High-score patients exhibited an increased DCB ratio and longer PFS across both PD-L1 and TMB subgroups. Additionally, the high-score group appeared influenced by tobacco exposure, with activated DNA damage response pathways. Whereas, immune/inflammation-related pathways were enriched in the low-score group. Tumor immune microenvironment analyses revealed higher proportions of exhausted/effector memory CD8 + T cells in the high-score group. CONCLUSIONS The mutational VAF profile is a promising biomarker for ICI therapy in NSCLC, with enhanced therapeutic stratification and management as a supplement to PD-L1 or TMB.
Collapse
Affiliation(s)
- Ruyun Gao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Ning Lou
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Lin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Tongji Xie
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Puyuan Xing
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Le Tang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Jiarui Yao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
27
|
Hu Y, Zhang Y, Lu Y, Xu Y, Xu J, Zhong H, Cheng L, Zhong R. Heterogeneity in PD-L1 expression between primary and metastatic lymph nodes: a predictor of EGFR-TKI therapy response in non-small cell lung cancer. Respir Res 2024; 25:233. [PMID: 38840238 PMCID: PMC11151486 DOI: 10.1186/s12931-024-02858-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 05/26/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND There is inconclusive evidence to suggest that the expression of programmed cell death ligand 1 (PD-L1) is a putative predictor of response to EGFR-TKI therapy in advanced EGFR-mutant non-small cell lung cancer (NSCLC). We evaluated the heterogeneity in PD-L1 expression in the primary lung site and metastatic lymph nodes to analyze the association between PD-L1 expression and response for patients treated with EGFR-TKI. METHODS This study reviewed 184 advanced NSCLC patients with EGFR mutations who received first-generation EGFR-TKI as first-line treatment from 2020 to 2021 at Shanghai Chest Hospital. The patients were divided into the primary lung site group (n = 100) and the metastatic lymph nodes group (n = 84) according to the biopsy site. The patients in each group were divided into TPS < 1%, TPS 1-49%, and TPS ≥ 50% groups according to PD-L1 expression. RESULTS The median PFS was 7 (95% CI: 5.7-8.3) months, and the median OS was 26 (95% CI: 23.5-28.5) months for all patients. No correlation existed between PFS or OS and PD-L1 expression. The median PFS in the primary lung site group was 11 months (95% CI: 9.6-12.4) in the TPS < 1% group, 8 months (95% CI: 6.6-9.4) in TPS 1-49% group, and 4 months (95% CI: 3.2-4.8) in TPS ≥ 50% group, with statistically significant differences (p = 0.000). The median OS of the TPS < 1% group and TPS ≥ 50% group showed a statistically significant difference (p = 0.008) in the primary lung site group. In contrast, PD-L1 expression in the lymph nodes of EGFR-mutant patients was unrelated to PFS or OS after EGFR-TKI therapy. CONCLUSION PD-L1 expression from the primary lung site might predict clinical benefit from EGFR-TKI, whereas PD-L1 from metastatic lymph nodes did not. TRIAL REGISTRATION This retrospective study was approved by the Ethics Committee of Shanghai Chest Hospital (ID: IS23060) and performed following the Helsinki Declaration of 1964 (revised 2008).
Collapse
Affiliation(s)
- Yaohua Hu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Huaihai West Road No.241, Shanghai, 200030, China
| | - Yidan Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Huaihai West Road No.241, Shanghai, 200030, China
| | - You Lu
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Yanchang Middle Road No.301, Shanghai, 200072, China
| | - Yingqi Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Huaihai West Road No.241, Shanghai, 200030, China
| | - Jianlin Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Huaihai West Road No.241, Shanghai, 200030, China
| | - Hua Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Huaihai West Road No.241, Shanghai, 200030, China
| | - Lei Cheng
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Huaihai West Road No.241, Shanghai, 200030, China.
| | - Runbo Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Huaihai West Road No.241, Shanghai, 200030, China.
| |
Collapse
|
28
|
Chen L, Zhu XZ, Zhao SJ, Yang QW. PD-L1 as a predictive factor for non-small-cell lung cancer prognosis. Lancet Oncol 2024; 25:e233. [PMID: 38821094 DOI: 10.1016/s1470-2045(24)00186-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 06/02/2024]
Affiliation(s)
- Long Chen
- ENT & HN Surgery Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning 530033, China
| | - Xian-Ze Zhu
- Medical Equipment Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning 530033, China.
| | - Shi-Jie Zhao
- ENT & HN Surgery Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning 530033, China
| | - Qiang-Wei Yang
- ENT & HN Surgery Department, The Third Affiliated Hospital of Guangxi Medical University, Nanning 530033, China
| |
Collapse
|
29
|
Wang Z, Li X, Chen J, Hua Y, Zhong X, Tang C, Gong S, Yang L. The significance of inflammatory markers in prognosticating the effectiveness and safety of immunotherapy in conjunction with chemotherapy during the primary intervention of advanced non-small cell lung carcinoma. Lung Cancer 2024; 192:107817. [PMID: 38735177 DOI: 10.1016/j.lungcan.2024.107817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/06/2024] [Accepted: 05/06/2024] [Indexed: 05/14/2024]
Abstract
OBJECTIVE The aim of this study is to scrutinize the prognostic significance of inflammatory biomarkers concerning the effectiveness and safety of combining PD-1 inhibition with chemotherapy in the management of advanced NSCLC. METHODS We conducted a retrospective analysis involving 206 NSCLC patients who received treatment at Qingdao Municipal Hospital. The study encompassed the acquisition of baseline clinical attributes and hematological parameters of these patients. The optimal threshold values for PLT and NLR were ascertained based on pre-treatment evaluations, with a particular focus on their association with PFS. Variables linked to PFS were subject to scrutiny through Kaplan-Meier analysis and logistic regression. The Receiver Operating Characteristic (ROC) curve served as the means to determine the ideal cut-off values for categorizing levels of inflammatory markers into high and low classifications. We employed Chi-square tests to evaluate the relationship between elevated and reduced baseline levels of inflammatory markers and irAE. RESULTS Kaplan-Meier analysis disclosed that patients in the low baseline PLT group and the low NLR group exhibited a substantially more favorable prognosis in contrast to their counterparts in the high baseline PLT and high NLR groups. Multivariate analysis indicated that diminished baseline PLT and NLR levels before treatment independently foretell extended PFS. Chi-square analysis underscored a substantial correlation between baseline WBC, NEUT, LYMPH, MONO, and NLR levels and irAE. CONCLUSION Subdued baseline PLT and NLR levels may serve as indicators of a more auspicious prognosis in patients contending with advanced NSCLC undergoing the combination of PD-1 inhibition and chemotherapy. Elevated baseline levels of inflammatory markers antedating PD-1 therapy in advanced NSCLC may be intimately interrelated with the occurrence of irAE.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Oncology, Nanjing Medical University, Qingdao Municipal Hospital, Nanjing, Jiangsu Province 211166, China
| | - Xiaoxue Li
- Department of Oncology, Nanjing Medical University, Qingdao Municipal Hospital, Nanjing, Jiangsu Province 211166, China
| | - Jiahong Chen
- Department of Otolaryngology, Qingdao Women and Children's Hospital, Qingdao, Shandong Province 266000, China
| | - Yin Hua
- Department of Oncology, Nanjing Medical University, Qingdao Municipal Hospital, Nanjing, Jiangsu Province 211166, China
| | - Xiaoqiang Zhong
- Department of Emergency Medicine, Chuanbei Medical College, Zigong City Third People's Hospital, Zigong, Sichuan Province 643020, China
| | - Chunhui Tang
- Department of Oncology, Nanjing Medical University, Qingdao Municipal Hospital, Nanjing, Jiangsu Province 211166, China
| | - Sumin Gong
- Department of Oncology, Nanjing Medical University, Qingdao Municipal Hospital, Nanjing, Jiangsu Province 211166, China
| | - Ling Yang
- Department of Oncology, Nanjing Medical University, Qingdao Municipal Hospital, Nanjing, Jiangsu Province 211166, China.
| |
Collapse
|
30
|
Qiu J, Cheng Z, Jiang Z, Gan L, Zhang Z, Xie Z. Immunomodulatory Precision: A Narrative Review Exploring the Critical Role of Immune Checkpoint Inhibitors in Cancer Treatment. Int J Mol Sci 2024; 25:5490. [PMID: 38791528 PMCID: PMC11122264 DOI: 10.3390/ijms25105490] [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: 03/24/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
An immune checkpoint is a signaling pathway that regulates the recognition of antigens by T-cell receptors (TCRs) during an immune response. These checkpoints play a pivotal role in suppressing excessive immune responses and maintaining immune homeostasis against viral or microbial infections. There are several FDA-approved immune checkpoint inhibitors (ICIs), including ipilimumab, pembrolizumab, and avelumab. These ICIs target cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and programmed death ligand 1 (PD-L1). Furthermore, ongoing efforts are focused on developing new ICIs with emerging potential. In comparison to conventional treatments, ICIs offer the advantages of reduced side effects and durable responses. There is growing interest in the potential of combining different ICIs with chemotherapy, radiation therapy, or targeted therapies. This article comprehensively reviews the classification, mechanism of action, application, and combination strategies of ICIs in various cancers and discusses their current limitations. Our objective is to contribute to the future development of more effective anticancer drugs targeting immune checkpoints.
Collapse
Affiliation(s)
- Junyu Qiu
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zilin Cheng
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zheng Jiang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Luhan Gan
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Huan Kui School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zixuan Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
| |
Collapse
|
31
|
Chen W, Zhang N, He Z, Li Q, Wang Y, Lou W, Di W. The efficacy of immune checkpoint inhibitors on low PD-L1 cervical cancer: A meta-analysis. Health Sci Rep 2024; 7:e2069. [PMID: 38706804 PMCID: PMC11066176 DOI: 10.1002/hsr2.2069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 03/14/2024] [Accepted: 04/07/2024] [Indexed: 05/07/2024] Open
Abstract
Background and Aims The effectiveness of immune checkpoint inhibitors (ICIs) in low programmed death ligand 1 (PD-L1) expression in cervical cancer (CC) patients remains unknown. We aimed to evaluate the efficacy of ICIs in low PD-L1 expression CC patients. Methods The study is an individual patient data (IPD)-based meta-analysis. IPD were compiled through KMSubtraction and IPDfromKM methodologies from high-quality randomized clinical trials and single-arm studies which reported overall survival (OS) or progression-free survival (PFS) stratified by PD-L1 expression. Kaplan-Meier curves and Cox regression analysis were employed to evaluate the survival benefits of ICIs. Results A total of eight studies and 1110 cases were included in the analysis. Within the low PD-L1 expression subgroup, ICI combination therapy, but not ICI monotherapy, demonstrated significant OS benefits over non-ICI treatment (hazard ratio [HR] = 0.61, 95% confidence interval [CI]: 0.36-1.04, p = 0.06). Concerning PFS, ICI monotherapy was associated with a negative effect compared to non-ICI treatment (HR = 4.59, 95% CI: 2.32-9.07, p < 0.001). Notably, both OS and PFS outcomes were unfavorable for ICI monotherapy compared to both non-ICI and ICI combination therapy in the combined positive score <1 subgroup (OS: HR = 2.60, 95% CI: 1.31-5.16, p = 0.008; PFS: HR = 7.59, 95% CI: 3.53-16.31, p < 0.001). Conclusion In patients with CC and low PD-L1 expression, ICI monotherapy may not be considered as the optimal treatment strategy when compared to non-ICI treatment or ICI combination therapy. Registration CRD42023395103.
Collapse
Affiliation(s)
- Wutao Chen
- Shanghai Key Laboratory of Gynecologic Oncology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
- Department of Obstetrics and Gynecology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Nan Zhang
- Shanghai Key Laboratory of Gynecologic Oncology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
- Department of Obstetrics and Gynecology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Zhihong He
- Shanghai Key Laboratory of Gynecologic Oncology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
- Department of Obstetrics and Gynecology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Qing Li
- Shanghai Key Laboratory of Gynecologic Oncology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
- Department of Obstetrics and Gynecology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
| | - You Wang
- Shanghai Key Laboratory of Gynecologic Oncology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
- Department of Obstetrics and Gynecology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Weihua Lou
- Shanghai Key Laboratory of Gynecologic Oncology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
- Department of Obstetrics and Gynecology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Wen Di
- Shanghai Key Laboratory of Gynecologic Oncology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
- Department of Obstetrics and Gynecology, School of Medicine, Renji HospitalShanghai Jiaotong UniversityShanghaiChina
- State Key Laboratory of Systems Medicine for CancerShanghaiChina
| |
Collapse
|
32
|
Parra ER, Zhang J, Duose DY, Gonzalez-Kozlova E, Redman MW, Chen H, Manyam GC, Kumar G, Zhang J, Song X, Lazcano R, Marques-Piubelli ML, Laberiano-Fernandez C, Rojas F, Zhang B, Taing L, Jhaveri A, Geisberg J, Altreuter J, Michor F, Provencher J, Yu J, Cerami E, Moravec R, Kannan K, Luthra R, Alatrash G, Huang HH, Xie H, Patel M, Nie K, Harris J, Argueta K, Lindsay J, Biswas R, Van Nostrand S, Kim-Schulze S, Gray JE, Herbst RS, Wistuba II, Gettinger S, Kelly K, Bazhenova L, Gnjatic S, Lee JJ, Zhang J, Haymaker C. Multi-omics Analysis Reveals Immune Features Associated with Immunotherapy Benefit in Patients with Squamous Cell Lung Cancer from Phase III Lung-MAP S1400I Trial. Clin Cancer Res 2024; 30:1655-1668. [PMID: 38277235 PMCID: PMC11016892 DOI: 10.1158/1078-0432.ccr-23-0251] [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: 01/31/2023] [Revised: 05/06/2023] [Accepted: 01/24/2024] [Indexed: 01/28/2024]
Abstract
PURPOSE Identifying molecular and immune features to guide immune checkpoint inhibitor (ICI)-based regimens remains an unmet clinical need. EXPERIMENTAL DESIGN Tissue and longitudinal blood specimens from phase III trial S1400I in patients with metastatic squamous non-small cell carcinoma (SqNSCLC) treated with nivolumab monotherapy (nivo) or nivolumab plus ipilimumab (nivo+ipi) were subjected to multi-omics analyses including multiplex immunofluorescence (mIF), nCounter PanCancer Immune Profiling Panel, whole-exome sequencing, and Olink. RESULTS Higher immune scores from immune gene expression profiling or immune cell infiltration by mIF were associated with response to ICIs and improved survival, except regulatory T cells, which were associated with worse overall survival (OS) for patients receiving nivo+ipi. Immune cell density and closer proximity of CD8+GZB+ T cells to malignant cells were associated with superior progression-free survival and OS. The cold immune landscape of NSCLC was associated with a higher level of chromosomal copy-number variation (CNV) burden. Patients with LRP1B-mutant tumors had a shorter survival than patients with LRP1B-wild-type tumors. Olink assays revealed soluble proteins such as LAMP3 increased in responders while IL6 and CXCL13 increased in nonresponders. Upregulation of serum CXCL13, MMP12, CSF-1, and IL8 were associated with worse survival before radiologic progression. CONCLUSIONS The frequency, distribution, and clustering of immune cells relative to malignant ones can impact ICI efficacy in patients with SqNSCLC. High CNV burden may contribute to the cold immune microenvironment. Soluble inflammation/immune-related proteins in the blood have the potential to monitor therapeutic benefit from ICI treatment in patients with SqNSCLC.
Collapse
Affiliation(s)
- Edwin Roger Parra
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiexin Zhang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dzifa Yawa Duose
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Edgar Gonzalez-Kozlova
- Department of Oncological Sciences, Mount Sinai, New York, New York
- Tisch Cancer Institute, Mount Sinai, New York, New York
- Precision Immunology Institute, Mount Sinai, New York, New York
- Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mary W. Redman
- SWOG Statistical Center, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Hong Chen
- Department of Thoracic-Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ganiraju C. Manyam
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gayatri Kumar
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xingzhi Song
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rossana Lazcano
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mario L. Marques-Piubelli
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Caddie Laberiano-Fernandez
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frank Rojas
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Baili Zhang
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Len Taing
- CIMAC-CIDC Network, Pipeline Development and Portal Integration, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Aashna Jhaveri
- CIMAC-CIDC Network, Pipeline Development and Portal Integration, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jacob Geisberg
- CIMAC-CIDC Network, Pipeline Development and Portal Integration, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jennifer Altreuter
- CIMAC-CIDC Network, Pipeline Development and Portal Integration, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Franziska Michor
- CIMAC-CIDC Network, Pipeline Development and Portal Integration, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - James Provencher
- CIMAC-CIDC Network, Pipeline Development and Portal Integration, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joyce Yu
- CIMAC-CIDC Network, Pipeline Development and Portal Integration, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ethan Cerami
- CIMAC-CIDC Network, Pipeline Development and Portal Integration, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Radim Moravec
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland
| | - Kasthuri Kannan
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rajyalakshmi Luthra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gheath Alatrash
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer, Houston, Texas
| | - Hsin-Hui Huang
- Precision Immunology Institute, Mount Sinai, New York, New York
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hui Xie
- Precision Immunology Institute, Mount Sinai, New York, New York
| | | | - Kai Nie
- Precision Immunology Institute, Mount Sinai, New York, New York
| | - Jocelyn Harris
- Precision Immunology Institute, Mount Sinai, New York, New York
| | | | - James Lindsay
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Roshni Biswas
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Stephen Van Nostrand
- CIMAC-CIDC Network, Pipeline Development and Portal Integration, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Seunghee Kim-Schulze
- Department of Oncological Sciences, Mount Sinai, New York, New York
- Tisch Cancer Institute, Mount Sinai, New York, New York
- Precision Immunology Institute, Mount Sinai, New York, New York
- Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Roy S. Herbst
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut
| | - Ignacio I. Wistuba
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Karen Kelly
- International Association for the Study of Lung Cancer, Denver, Colorado
| | - Lyudmila Bazhenova
- University of California San Diego Moores Cancer Center, La Jolla, California
| | - Sacha Gnjatic
- Department of Oncological Sciences, Mount Sinai, New York, New York
- Tisch Cancer Institute, Mount Sinai, New York, New York
- Precision Immunology Institute, Mount Sinai, New York, New York
- Icahn School of Medicine at Mount Sinai, New York, New York
| | - J. Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianjun Zhang
- Department of Thoracic-Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cara Haymaker
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
33
|
Stratmann JA, Althoff FC, Doebel P, Rauh J, Trummer A, Hünerlitürkoglu AN, Frost N, Yildirim H, Christopoulos P, Burkhard O, Büschenfelde CMZ, Becker von Rose A, Alt J, Aries SP, Webendörfer M, Kaldune S, Uhlenbruch M, Tritchkova G, Waller CF, Rittmeyer A, Hoffknecht P, Braess J, Kopp HG, Grohé C, Schäfer M, Schumann C, Griesinger F, Kuon J, Sebastian M, Reinmuth N. Sotorasib in KRAS G12C-mutated non-small cell lung cancer: A multicenter real-world experience from the compassionate use program in Germany. Eur J Cancer 2024; 201:113911. [PMID: 38377774 DOI: 10.1016/j.ejca.2024.113911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 01/29/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Sotorasib is a first-in-class KRAS p.G12C-inhibitor that has entered clinical trials in pretreated patients with non-small cell lung cancer (NSCLC) in 2018. First response rates were promising in the CodeBreaK trials. It remains unclear whether response to sotorasib and outcomes differ in a real-world setting when including patients underrepresented in clinical trials. METHODS Patients with KRAS p.G12C-mutated advanced or metastatic NSCLC received sotorasib within the German multicenter sotorasib compassionate use program between 2020 to 2022. Data on efficacy, tolerability, and survival were analyzed in the full cohort and in subgroups of special interest such as co-occurring mutations and across PD-L1 expression levels. RESULTS We analyzed 163 patients who received sotorasib after a median of two treatment lines (range, 0 to 7). Every fourth patient had a poor performance status and 38% had brain metastases (BM). The objective response rate was 38.7%. The median overall survival was 9.8 months (95% CI, 6.5 to not reached). Median real-world (rw) progression-free survival was 4.8 months (9% CI, 3.9 to 5.9). Dose reductions and permanent discontinuation were necessary in 35 (21.5%) and 7 (4.3%) patients, respectively. Efficacy seems to be influenced by PD-L1 expression and a co-occurring KEAP1 mutation. KEAP1 was associated with an inferior survival. Other factors such as BM, STK11, and TP53 mutations had no impact on response and survival. CONCLUSION First results from a real-world population confirm promising efficacy of sotorasib for the treatment of advanced KRAS p.G12C-mutated NSCLC. Patients with co-occurring KEAP1 mutations seem to derive less benefit.
Collapse
Affiliation(s)
- Jan A Stratmann
- Goethe University Frankfurt, University Hospital, Department of Internal Medicine II, Hematology/Oncology, Frankfurt am Main, Germany
| | - Friederike C Althoff
- Goethe University Frankfurt, University Hospital, Department of Internal Medicine II, Hematology/Oncology, Frankfurt am Main, Germany.
| | - Paula Doebel
- Goethe University Frankfurt, University Hospital, Department of Internal Medicine II, Hematology/Oncology, Frankfurt am Main, Germany
| | - Jacqueline Rauh
- Hospital Witten, Medical Specialist Center of Internal Medicine, Witten, Germany
| | - Arne Trummer
- Municipal Clinic Braunschweig, Medical Specialist Center of Hematology/Oncology, Braunschweig, Germany
| | | | - Nikolaj Frost
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Department of Infectious Diseases and Pulmonary Medicine, Berlin, Germany
| | | | - Petros Christopoulos
- University Hospital Heidelberg, Thoraxklinik Heidelberg, Heidelberg, Germany; Translational Lung Research Center Heidelberg, member of the German Center for Lung Research (DZL), Germany
| | - Oswald Burkhard
- Medical Specialist Center of Internal Medicine, Hematology Oncology, Palliative Medicine in Worms, Worms, Germany
| | | | - Aaron Becker von Rose
- Technical University Munich, Klinikum rechts der Isar, Medical Department for Haematology and Oncology, Munich, Germany
| | - Jürgen Alt
- University Medical Center Mainz, Department of Internal Medicine III, Mainz, Germany
| | | | - Maximilian Webendörfer
- University Hospital Essen, West German Cancer Center, Department of Medical Oncology, Essen, Germany
| | - Stefan Kaldune
- RoMed Clinic Rosenheim, Department of Hematology/Oncology, Rosenheim, Germany
| | - Mark Uhlenbruch
- Kaiserswerther Diakonie Florence-Nightingale-Hospital Düsseldorf, Department of Hematology/Oncology, Düsseldorf, Germany
| | - Guergana Tritchkova
- University Hospital Dresden, TU Dresden, Clinic for Internal Medicine I, Dresden, Germany
| | - Cornelius F Waller
- University of Freiburg, University Medical Centre Freiburg, Department of Haematology, Oncology and Stem Cell Transplantation, Freiburg, Germany
| | | | - Petra Hoffknecht
- Niels-Stensen-Kliniken Franziskus Hospital Harderberg, Department of Hematology/Oncology, Hardenberg, Germany
| | - Jan Braess
- Hospital Barmherzige Brüder Regensburg, Regensburg, Germany
| | - Hans-Georg Kopp
- Robert Bosch Center for Tumor Diseases, Robert-Bosch-Hospital, Stuttgart, Germany
| | | | - Monica Schäfer
- Helios Klinikum Emil von Behring GmbH, Lungenklinik Heckeshorn, Berlin, Germany
| | - Christian Schumann
- Klinikverbund Allgäu gGmbH, Clinic for Pneumology, Thoracic Oncology, Sleep and Respiratory Medicine, Kempten and Immenstadt, Germany
| | - Frank Griesinger
- Pius-Hospital, University Medicine Oldenburg, Department of Hematology and Oncology, University Department Internal Medicine-Oncology, Oldenburg, Germany
| | - Jonas Kuon
- Lungenklinik Löwenstein, Department of Oncology, Löwenstein, Germany
| | - Martin Sebastian
- Goethe University Frankfurt, University Hospital, Department of Internal Medicine II, Hematology/Oncology, Frankfurt am Main, Germany
| | | |
Collapse
|
34
|
Guo Z, Wei X, Tang C, Liang J. Non-tumor-related prognostic factors for immunotherapy-chemotherapy or immunotherapy alone as first-line in advanced non-small cell lung cancer (NSCLC). Clin Exp Med 2024; 24:52. [PMID: 38489142 PMCID: PMC10942875 DOI: 10.1007/s10238-024-01298-z] [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: 12/04/2023] [Accepted: 01/16/2024] [Indexed: 03/17/2024]
Abstract
Besides programmed death ligand 1 (PD-L1) expression, rapid, cost-effective and validated scores or models are critical for the prognosis and prediction of patients received immune checkpoint inhibitors (ICIs). In this retrospective study, 182 patients with NSCLC receiving ICIs from 2015 to 2022 were divided 1:1 into a training cohort and a validation cohort. We identified a score established by three factors and analyzed the prognostic implications by Kaplan-Meier approach (Log rank test) and time-dependent receiver operating characteristic (ROC) analyses. A non-tumor-related score (NTRS) was established that could be used as a prognostic factor (HR 2.260, 95% CI 1.559-3.276, P < 0.001 in training cohort; HR 2.114, 95% CI 1.493-2.994, P < 0.001 in validation cohort) and had a high time-dependent ROC for overall survival (OS) (AUC 0.670-0.782 in training cohort; AUC 0.682-0.841 in validation cohort). PD-L1 (1-49%) and NTRS (score = 0, 1, 2, 3) combination significantly improved the assessment of patients' OS and progress-free survival (PFS), which was statistically different in training cohorts (P < 0.001 for OS, 0.012 for PFS) and validation cohorts (P = 0.01 for OS, < 0.001 for PFS). The NTRS provided a better assessment of durable clinical benefit (DCB) compared to PD-L1 expression (P = 0.009 vs. 0.232 in training cohort; P = 0.004 vs. 0.434 in validation cohort). NTRS may help improve prognosis stratification of patients receiving ICIs in first-line NSCLC and may be combined with tumor-related parameters.
Collapse
Affiliation(s)
- Ziwei Guo
- Department of Oncology, Peking University International Hospital, Beijing, 102206, China
| | - Xing Wei
- Department of Oncology, Peking University International Hospital, Beijing, 102206, China
| | - Chuanhao Tang
- Department of Oncology, Peking University International Hospital, Beijing, 102206, China.
| | - Jun Liang
- Department of Oncology, Peking University International Hospital, Beijing, 102206, China.
| |
Collapse
|
35
|
Wu J, Mao L, Lei W, Sun W, Yang X, Zhang Y, Huang X, Lin D. Genomic discordances and heterogeneous mutational burden, PD-L1 expression and immune infiltrates of non-small cell lung cancer metastasis. J Clin Pathol 2024:jcp-2023-209328. [PMID: 38307721 DOI: 10.1136/jcp-2023-209328] [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: 12/02/2023] [Accepted: 01/21/2024] [Indexed: 02/04/2024]
Abstract
AIMS To investigate the genomic discordances and heterogeneous mutational burden, PD-L1 expression and immune cell (IC) infiltrates of non-small cell lung cancer (NSCLC) metastasis. METHODS Surgical samples from 41 cases of NSCLC with metastatic tumours (MTs) and paired primary tumours (PTs) were collected. PD-L1 expression and ICs were quantified using image-based immunohistochemistry profiling. Whole exome sequencing was employed to explore discrepancies in genomic characteristics, tumour mutational burden (TMB) and tumour neoantigen burden (TNB) in 28 cases. RESULTS Non-synonymous mutations in MTs were slightly more than in PTs, with only 42.34% of mutations shared between paired PTs and MTs. The heterogeneity of TMB showed no significant difference (p=0.785) between MTs and PTs, while TNB significantly increased in MTs (p=0.013). MTs generally exhibited a higher density of PD-L1+ cells and a higher tumour proportion score with a lower density of IC infiltrates. Subgroup analysis considering clinicopathological factors revealed that the heterogeneity of immune biomarkers was closely associated with the histology of lung adenocarcinoma, metastatic sites of extrapulmonary, time intervals and treatment history. Prognosis analysis indicated that a high density of CD8+ T cells was a low-risk factor, whereas a high density of PD-L1+ cells in MTs was a high-risk factor for cancer-related death in metastatic NSCLC. CONCLUSIONS The mutational burden, PD-L1 expression and IC infiltrates undergo changes during NSCLC metastasis, which may impact the immunotherapeutic benefits in patients with NSCLC with metastatic progression and should be monitored according to clinical scenarios.
Collapse
Affiliation(s)
- Jianghua Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Luning Mao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wanjun Lei
- Tianjin Medical Laboratory, BGI-Tianjin, BGI-Shenzhen, Tianjin, China
| | - Wei Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanhui Zhang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center of Cancer; Key Laboratory of Cancer Prevention and Therapy; Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Xiaozheng Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Dongmei Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
36
|
Qu FJ, Zhou Y, Wu S. Progress of immune checkpoint inhibitors therapy for non-small cell lung cancer with liver metastases. Br J Cancer 2024; 130:165-175. [PMID: 37945751 PMCID: PMC10803805 DOI: 10.1038/s41416-023-02482-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Nearly one-fifth of patients with non-small cell Lung Cancer (NSCLC) will develop liver metastases (LMs), and the overall treatment strategy of LMs will directly affect the survival of patients. However, some retrospective studies have found that patients receiving chemotherapy or targeted therapy have a poorer prognosis once LMs develop. In recent years, multiple randomised controlled trials (RCTS) have shown significant improvements in outcomes for patients with advanced lung cancer following the introduction of immune checkpoint inhibitors (ICIs) compared to conventional chemotherapy. ICIs is safe and effective in patients with LMs, although patients with LMs are mostly underrepresented in randomised clinical trials. However, NSCLC patients with LMs have a significantly worse prognosis than those without LMs when treated with ICIs, and the mechanism by which LMs induce systemic anti-tumour immunity reduction is unknown, so the management of LMs in patients with NSCLC is a clinical challenge that requires more optimised therapies to achieve effective disease control. In this review, we summarised the mechanism of ICIs in the treatment of LMs, the clinical research and treatment progress of ICIs and their combination with other therapies in patients with LMs from NSCLC.
Collapse
Affiliation(s)
- Fan-Jie Qu
- Department of Oncology, Affiliated Dalian Third People's Hospital of Dalian Medical University, 116033, Dalian, China.
| | - Yi Zhou
- Department of Oncology, Affiliated Dalian Third People's Hospital of Dalian Medical University, 116033, Dalian, China
| | - Shuang Wu
- Department of Oncology, Affiliated Dalian Third People's Hospital of Dalian Medical University, 116033, Dalian, China
| |
Collapse
|
37
|
Zhou D, Wang Y, Ning W, Chen C. Comparison of primary and metastatic site-related PD-L1 expression in predicting ORR in patients with advanced NSCLC who received ICB-based therapy. Thorac Cancer 2024; 15:379-385. [PMID: 38155421 PMCID: PMC10864116 DOI: 10.1111/1759-7714.15201] [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: 09/24/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 12/30/2023] Open
Abstract
BACKGROUND Whether the value of PD-L1 expression from metastatic sites to predict the efficacy of immune checkpoint blockade (ICB)-based treatment is equivalent to that from a primary tumor is uncertain. This study aimed to compare the utility of PD-L1 TPS from a primary lung tumor and metastatic sites to predict the overall response rate (ORR) of first-line ICB-based treatment. METHODS This study included 249 patients with advanced non-small cell lung cancer (NSCLC) who received first-line ICB-based treatment. All subjects underwent PD-L1 testing prior to ICB-based treatment and were divided into two cohorts corresponding to the different biopsy sites: lung primary site-sampled cohort (PT cohort, n = 167) and metastatic site-sampled cohort (MT cohort, n = 82). RESULTS There was no statistical significance in PD-L1 TPS distribution between the two cohorts (p = 0.742). PD-L1 TPS ≥50% was also related to high ORR compared with PD-L1 < 50% in the PT cohort (34.3% vs. 14.1%, p = 0.004). In contrast, ICB-based therapy could bring comparable ORR (35.1% vs. 33.3%, p = 0.871) in the MT cohort regardless of PD-L1 TPS status (≥50%, or <50%). As supported, when the cutoff value of TPS was selected as 50%, it was suggested that PT-related PD-L1 was the independent predictor of ORR (OR 2.870, 95% CI: 1.231-6.694, p = 0.015) rather than MT-related PD-L1 (OR 0.689, 95% CI: 0.236-2.013, p = 0.495). Furthermore, ROC proved that PT-related PD-L1 expression manifested a better AUC of 0.621 (p = 0.026) than that of MT-related PD-L1 (AUC = 0.565, p = 0.362). CONCLUSION Compared with PT-related PD-L1 expression, MT-related PD-L1 expression showed limited value in predicting ORR in patients with advanced NSCLC receiving ICB-based therapy. It was concluded that even patients with low MT-related PD-L1 expression could benefit from ICB-based therapy.
Collapse
Affiliation(s)
- Danhong Zhou
- Department of Respiratory and Critical Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Wang
- Department of Respiratory and Critical Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weiwei Ning
- Department of Respiratory and Critical Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng Chen
- Department of Respiratory and Critical Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
38
|
Wang X, Bai H, Zhang J, Wang Z, Duan J, Cai H, Cao Z, Lin Q, Ding X, Sun Y, Zhang W, Xu X, Chen H, Zhang D, Feng X, Wan J, Zhang J, He J, Wang J. Genetic Intratumor Heterogeneity Remodels the Immune Microenvironment and Induces Immune Evasion in Brain Metastasis of Lung Cancer. J Thorac Oncol 2024; 19:252-272. [PMID: 37717855 DOI: 10.1016/j.jtho.2023.09.276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/18/2023] [Accepted: 09/07/2023] [Indexed: 09/19/2023]
Abstract
INTRODUCTION Brain metastasis, with the highest incidence in patients with lung cancer, significantly worsens prognosis and poses challenges to clinical management. To date, how brain metastasis evades immune elimination remains unknown. METHODS Whole-exome sequencing and RNA sequencing were performed on 30 matched brain metastasis, primary lung adenocarcinoma, and normal tissues. Data from The Cancer Genome Atlas primary lung adenocarcinoma cohort, including multiplex immunofluorescence, were used to support the findings of bioinformatics analysis. RESULTS Our study highlights the key role of intratumor heterogeneity of genomic alterations in the metastasis process, mainly caused by homologous recombination deficiency or other somatic copy number alteration-associated mutation mechanisms, leading to increased genomic instability and genomic complexity. We further proposed a selection model of brain metastatic evolution in which intratumor heterogeneity drives immune remodeling, leading to immune escape through different mechanisms under local immune pressure. CONCLUSIONS Our findings provide novel insights into the metastatic process and immune escape mechanisms of brain metastasis and pave the way for precise immunotherapeutic strategies for patients with lung cancer with brain metastasis.
Collapse
Affiliation(s)
- Xin Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua Bai
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiyang Zhang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianchun Duan
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongqing Cai
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Cao
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qingtang Lin
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaosheng Ding
- Department of Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yiting Sun
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Zhang
- Department of Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Xiaoya Xu
- Department of Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Hao Chen
- Department of Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Dadong Zhang
- Department of Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Xiaoli Feng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinghai Wan
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianjun Zhang
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
39
|
Knetki-Wróblewska M, Dziadziuszko R, Jankowski T, Krawczyk P, Bryl M, Stencel K, Wrona A, Bandura A, Smok-Kalwat J, Rok-Knapińska J, Szydziak-Zwierzyńska K, Rogoziewicz K, Czyżewicz G, Wójtowicz M, Wojtukiewicz M, Kalinka E, Wysocki PJ, Łobacz M, Milanowski J, Pawlik H, Kowalski DM, Krzakowski M. Pembrolizumab-combination therapy for NSCLC- effectiveness and predictive factors in real-world practice. Front Oncol 2024; 14:1341084. [PMID: 38322415 PMCID: PMC10844541 DOI: 10.3389/fonc.2024.1341084] [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: 11/19/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
Introduction Pembrolizumab combined with chemotherapy has become the standard of care for patients with non-small-cell lung cancer (NSCLC) and the expression of programmed death ligand 1 (PD-L1) in <50% of tumour cells (TC). Methods We evaluated the efficacy of the treatment in real-world practice, paying attention to the predictive factors, with a special focus on low level of PD-L1 expression. This study is a multicenter retrospective analysis of patients with stage IV NSCLC. Results A group of 339 consecutive patients was analysed, among them 51% patients with low PD-L1 expression. In the overall population, the ORR was 40.6%, median PFS and OS were 13 months (95% CI 11.4-15) and 16.8 months (95% CI 13.3-20.3), respectively. In multivariate analysis for the entire study population, performance status - ECOG 1 vs. 0 (HR 2.2, 95%CI 1.1-4.6; p=0.02), neutrophil to lymphocyte ratio (NLR)>3 (HR 2.3, 95%CI 1.3-4.2; p=0.04), presence of liver (HR 2.0, 95%CI 1-3.7; p=0. 03) and bone metastases (HR 1.3, 95%CI 1-3; p=0.04), weight loss (HR 1.8, 95%CI 1.1-2.8; p=0.01) and sum of measurable lesions diameters >110 mm (HR 1.7, 95%CI 1-2.9, p=0.049) had a negative impact on OS. Conclusions In the real world, patients can clinically benefit from immunochemotherapy, regardless of the expression of PD-L1 and the histological type. Other clinicopathological factors such as performance status, extent, and location of secondary lesions have prognostic significance.
Collapse
Affiliation(s)
- Magdalena Knetki-Wróblewska
- Department of Lung Cancer and Chest Tumours, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Rafał Dziadziuszko
- Department of Oncology and Radiotherapy, Medical University of Gdansk, Gdansk, Poland
| | - Tomasz Jankowski
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| | - Maciej Bryl
- Department of Clinical Oncology with the Subdepartment of Diurnal Chemotherapy E. J. Zeyland Wielkopolska Center of Pulmonology and Thoracic Surgery, Poznan, Poland
| | - Katarzyna Stencel
- Department of Clinical Oncology with the Subdepartment of Diurnal Chemotherapy E. J. Zeyland Wielkopolska Center of Pulmonology and Thoracic Surgery, Poznan, Poland
| | - Anna Wrona
- Department of Oncology and Radiotherapy, Medical University of Gdansk, Gdansk, Poland
| | - Artur Bandura
- Department of Oncology and Radiotherapy, Medical University of Gdansk, Gdansk, Poland
| | | | | | | | - Krzysztof Rogoziewicz
- Department of Clinical Oncology, Lower Silesian Oncology, Pulmonology and Hematology Center, Wroclaw, Poland
| | - Grzegorz Czyżewicz
- Department of Clinical Oncology, The John Paul II Specialist Hospital, Cracow, Poland
| | - Monika Wójtowicz
- Department of Clinical Oncology, Medical University of Bialystok, Bialystok, Poland
| | - Marek Wojtukiewicz
- Department of Clinical Oncology, Medical University of Bialystok, Bialystok, Poland
| | - Ewa Kalinka
- Department of Clinical Oncology, Polish Mother’s Memorial Hospital Research Institute, Łódź, Poland
| | - Piotr J. Wysocki
- Department of Clinical Oncology, Jagiellonian University-Collegium Medicum Hospital, Cracow, Poland
| | - Mateusz Łobacz
- Department of Clinical Oncology, Jagiellonian University-Collegium Medicum Hospital, Cracow, Poland
| | - Janusz Milanowski
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| | - Hubert Pawlik
- Computational Oncology Department, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Dariusz M. Kowalski
- Department of Lung Cancer and Chest Tumours, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maciej Krzakowski
- Department of Lung Cancer and Chest Tumours, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
40
|
Li Y, Liang X, Li H, Chen X. Reconstruction of unreported subgroup survival data with PD-L1-low expression in advanced/metastatic triple-negative breast cancer using innovative KMSubtraction workflow. J Immunother Cancer 2024; 12:e007931. [PMID: 38212119 PMCID: PMC10806559 DOI: 10.1136/jitc-2023-007931] [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] [Accepted: 12/13/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Among patients with advanced/metastatic triple-negative breast cancer (TNBC) with high/positive programmed death-ligand 1 (PD-L1) expression, a superior survival outcome has been demonstrated with immune checkpoint inhibitors (ICIs). However, it remains unclear whether ICIs are beneficial for patients with low PD-L1 levels. Here, we derived survival data for subgroups with low PD-L1-expressing and conducted a pooled analysis. METHODS After a systematic search of Embase, PubMed, MEDLINE, and CENTRAL from inception until May 18, 2023, randomized controlled trials (RCTs) reporting progression-free survival (PFS), overall survival (OS), or duration of response (DOR) for metastatic TNBC treated with ICI-based regimens were included. Kaplan-Meier curves were extracted for the intention-to-treat population and high PD-L1 subgroups. KMSubtraction was used when survival curves were not provided for subgroups with low PD-L1 expression. A pooled analysis of survival data was then conducted. RESULTS A total of 3022 patients were included in four RCTs: Impassion130, Impassion131, KEYNOTE-119, and KEYNOTE-355. Unreported low PD-L1-expressing subgroups were identified, including PD-L1 immune cell (IC)<1%, combined positive score (CPS)<1, and 1≤CPS<10. Compared with chemotherapy, ICI-chemotherapy combinations did not significantly differ in OS, PFS, or DOR in the Impassion PD-L1<1%, KEYNOTE-355 PD-L1 CPS<1, and KEYNOTE-355 1≤CPS<10 subgroups. In the KEYNOTE-119 CPS<1 subgroup, the risk of tumor progression was increased with pembrolizumab (HR, 2.23; 95% CI, 1.62 to 3.08; p<0.001), as well as in the 1≤CPS<10 subgroup (HR, 1.64; 95% CI, 1.22 to 2.20; p<0.001). A pooled analysis using a scoring system found no significant difference in OS and PFS among the subgroups with an IC of <1% between immunochemotherapy and chemotherapy. OS (HR, 1.07; 95% CI, 0.91 to 1.26), PFS (HR, 0.96; 95% CI, 0.84 to 1.10), and DOR were also not significantly different in pooled analysis of first-line trials for those with low PD-L1 expression. CONCLUSION ICI-based regimens are not associated with a survival benefit versus chemotherapy in subgroups of advanced/metastatic TNBC that express low PD-L1 levels.
Collapse
Affiliation(s)
- Yan Li
- Department of Clinical Pharmacy, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xueyan Liang
- Phase 1 Clinical Trial Laboratory, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Huijuan Li
- Phase 1 Clinical Trial Laboratory, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xiaoyu Chen
- Department of Clinical Pharmacy, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Phase 1 Clinical Trial Laboratory, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
41
|
Ndembe G, Intini I, Moro M, Grasselli C, Panfili A, Panini N, Bleve A, Occhipinti M, Borzi C, Garassino MC, Marabese M, Canesi S, Scanziani E, Sozzi G, Broggini M, Ganzinelli M. Caloric restriction and metformin selectively improved LKB1-mutated NSCLC tumor response to chemo- and chemo-immunotherapy. J Exp Clin Cancer Res 2024; 43:6. [PMID: 38163906 PMCID: PMC10759660 DOI: 10.1186/s13046-023-02933-5] [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: 09/07/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND About 10% of NSCLCs are mutated in KRAS and impaired in STK11/LKB1, a genetic background associated with poor prognosis, caused by an increase in metastatic burden and resistance to standard therapy. LKB1 is a protein involved in a number of biological processes and is particularly important for its role in the regulation of cell metabolism. LKB1 alterations lead to protein loss that causes mitochondria and metabolic dysfunction that makes cells unable to respond to metabolic stress. Different studies have shown how it is possible to interfere with cancer metabolism using metformin and caloric restriction (CR) and both modify the tumor microenvironment (TME), stimulating the switch from "cold" to "hot". Given the poor therapeutic response of KRASmut/LKB1mut patients, and the role of LKB1 in cell metabolism, we examined whether the addition of metformin and CR enhanced the response to chemo or chemo-immunotherapy in LKB1 impaired tumors. METHODS Mouse cell lines were derived from lung nodules of transgenic mice carrying KRASG12D with either functional LKB1 (KRASG12D/LKB1wt) or mutated LKB1 (KRASG12D/LKB1mut). Once stabilized in vitro, these cell lines were inoculated subcutaneously and intramuscularly into immunocompetent mice. Additionally, a patient-derived xenograft (PDX) model was established by directly implanting tumor fragments from patient into immunocompromised mice. The mice bearing these tumor models were subjected to treatment with chemotherapy or chemo-immunotherapy, both as standalone regimens and in combination with metformin and CR. RESULTS Our preclinical results indicate that in NSCLC KRASmut/LKB1mut tumors, metformin and CR do enhance the response to chemo and chemo-immunotherapy, inducing a metabolic stress condition that these tumors are not able to overcome. Analysis of immune infiltrating cells did not bring to light any strong correlation between the TME immune-modulation and the tumor response to metformin and CR. CONCLUSION Our in vitro and in vivo preliminary studies confirm our hypothesis that the addition of metformin and CR is able to improve the antitumor activity of chemo and chemoimmunotherapy in LKB1 impaired tumors, exploiting their inability to overcome metabolic stress.
Collapse
Affiliation(s)
- Gloriana Ndembe
- Laboratory of Molecular Pharmacology, Department of Experimental Oncology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ilenia Intini
- Laboratory of Molecular Pharmacology, Department of Experimental Oncology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Massimo Moro
- Tumor Genomics Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Chiara Grasselli
- Immunopharmacology Unit, Department of Experimental Oncology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Andrea Panfili
- Immunopharmacology Unit, Department of Experimental Oncology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Nicolò Panini
- Immunopharmacology Unit, Department of Experimental Oncology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Augusto Bleve
- Immunopharmacology Unit, Department of Experimental Oncology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Mario Occhipinti
- Thoracic Unit, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Cristina Borzi
- Tumor Genomics Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Marina Chiara Garassino
- Thoracic Unit, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Mirko Marabese
- Laboratory of Molecular Pharmacology, Department of Experimental Oncology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Simone Canesi
- Mouse & Animal Pathology Lab, Fondazione Filarete, Milan, Italy
- Department of Veterinary Medicine, University of Milan, Milan, Italy
| | - Eugenio Scanziani
- Mouse & Animal Pathology Lab, Fondazione Filarete, Milan, Italy
- Department of Veterinary Medicine, University of Milan, Milan, Italy
| | - Gabriella Sozzi
- Tumor Genomics Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Massimo Broggini
- Laboratory of Molecular Pharmacology, Department of Experimental Oncology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - Monica Ganzinelli
- Thoracic Unit, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| |
Collapse
|
42
|
He J, Wang X, Cai L, Jia Z, Liu C, Sun X, Wu S, Ding C, Zhang Z, Liu Y. Effect of storage time of paraffin sections on the expression of PD-L1 (SP142) in invasive breast cancer. Diagn Pathol 2023; 18:131. [PMID: 38053121 DOI: 10.1186/s13000-023-01423-8] [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: 07/19/2023] [Accepted: 11/25/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND PD-L1 staining using long-stored paraffin sections may not be consistent with the true PD-L1 expression of patients. Therefore, it is necessary to explore the expression of PD-L1(SP142) in paraffin sections of invasive breast cancer with different storage times and the optimal storage temperature for unstained paraffin sections. METHODS The study included 71 cases of PD-L1(SP142) positive breast cancer. The unstained paraffin sections were stored at room temperature conditions (20-25 °C), 4 °C, -20 °C and - 80 °C, respectively. PD-L1 staining was performed at 1, 2, 3, 4, 8, 12 and 24 weeks of storage. PD-L1 expression was assessed with a continuity score. RESULTS The PD-L1 antigen was gradually lost as the storage time of paraffin sections increased. The PD-L1 positivity rate was 97.18% at 1 week for the sections stored at room temperature, and decreased from 83.10 to 71.83% for the sections stored for 2 weeks to 4 weeks, and 61.97%, 54.93%, and 32.93% for 8, 12, and 24 weeks, respectively. When stored at low temperatures of 4 °C, -20 °C and - 80 °C, the positivity rate decreases with the same trend but more slowly compared to room temperature. The mean IC score of PD-L1 also showed a gradual decrease in all cases. In the consistency analysis, PD-L1 expression in slices stored at room temperature for 2 weeks was consistent with PD-L1 expression in fresh slices (ICC ≥ 0.9 for all slices), and PD-L1 expression in slices stored at 4 °C or -20 °C for 4 weeks was consistent with PD-L1 expression in fresh slices (ICC ≥ 0.9 for all slices). When stored under refrigeration at -80 °C, PD-L1 expression in slices stored for 3 weeks was consistent with that in fresh slices (ICC ≥ 0.9). CONCLUSIONS To our knowledge, this is the first article on the effect of preservation time and preservation temperature of paraffin sections on PD-L1 expression in breast cancer. Long-term storage of paraffin sections of unstained invasive breast cancer can lead to antigen loss of PD-L1 (SP142). Refrigerated storage of paraffin sections can delay antigen loss, with best results at 4 °C or -20 °C, and a storage time of no more than 4 weeks is recommended.
Collapse
Affiliation(s)
- Jiankun He
- Department of Pathology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, Hebei, 050011, China
| | - Xinran Wang
- Department of Pathology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, Hebei, 050011, China
| | - Lijing Cai
- Department of Pathology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, Hebei, 050011, China
| | - Zhanli Jia
- Department of Pathology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, Hebei, 050011, China
| | - Chang Liu
- Department of Pathology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, Hebei, 050011, China
| | - Xuemei Sun
- Department of Pathology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, Hebei, 050011, China
| | - Si Wu
- Department of Pathology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, Hebei, 050011, China
| | - Chunyan Ding
- Department of Pathology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, Hebei, 050011, China
| | - Zi Zhang
- Department of Pathology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, Hebei, 050011, China
| | - Yueping Liu
- Department of Pathology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, Hebei, 050011, China.
| |
Collapse
|
43
|
Placke JM, Kimmig M, Griewank K, Herbst R, Terheyden P, Utikal J, Pföhler C, Ulrich J, Kreuter A, Mohr P, Gutzmer R, Meier F, Dippel E, Welzel J, Engel DR, Kreft S, Sucker A, Lodde G, Krefting F, Stoffels I, Klode J, Roesch A, Zimmer L, Livingstone E, Hadaschik E, Becker JC, Weichenthal M, Tasdogan A, Schadendorf D, Ugurel S. Correlation of tumor PD-L1 expression in different tissue types and outcome of PD-1-based immunotherapy in metastatic melanoma - analysis of the DeCOG prospective multicenter cohort study ADOREG/TRIM. EBioMedicine 2023; 96:104774. [PMID: 37660535 PMCID: PMC10483509 DOI: 10.1016/j.ebiom.2023.104774] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 09/05/2023] Open
Abstract
BACKGROUND PD-1-based immune checkpoint inhibition (ICI) is the major backbone of current melanoma therapy. Tumor PD-L1 expression represents one of few biomarkers predicting ICI therapy outcome. The objective of the present study was to systematically investigate whether the type of tumor tissue examined for PD-L1 expression has an impact on the correlation with ICI therapy outcome. METHODS Pre-treatment tumor tissue was collected within the prospective DeCOG cohort study ADOREG/TRIM (CA209-578; NCT05750511) between February 2014 and May 2020 from 448 consecutive patients who received PD-1-based ICI for non-resectable metastatic melanoma. The primary study endpoint was best overall response (BOR), secondary endpoints were progression-free (PFS) and overall survival (OS). All endpoints were correlated with tumor PD-L1 expression (quantified with clone 28-8; cutoff ≥5%) and stratified by tissue type. FINDINGS Tumor PD-L1 was determined in 95 primary tumors (PT; 36.8% positivity), 153 skin/subcutaneous (34.0% positivity), 115 lymph node (LN; 50.4% positivity), and 85 organ (40.8% positivity) metastases. Tumor PD-L1 correlated with BOR if determined in LN (OR = 0.319; 95% CI = 0.138-0.762; P = 0.010), but not in skin/subcutaneous metastases (OR = 0.656; 95% CI = 0.311-1.341; P = 0.26). PD-L1 positivity determined on LN metastases was associated with favorable survival (PFS, HR = 0.490; 95% CI = 0.310-0.775; P = 0.002; OS, HR = 0.519; 95% CI = 0.307-0.880; P = 0.014). PD-L1 positivity determined in PT (PFS, HR = 0.757; 95% CI = 0.467-1.226; P = 0.27; OS; HR = 0.528; 95% CI = 0.305-0.913; P = 0.032) was correlated with survival to a lesser extent. No relevant survival differences were detected by PD-L1 determined in skin/subcutaneous metastases (PFS, HR = 0.825; 95% CI = 0.555-1.226; P = 0.35; OS, HR = 1.083; 95% CI = 0.698-1.681; P = 0.72). INTERPRETATION For PD-1-based immunotherapy in melanoma, tumor PD-L1 determined in LN metastases was stronger correlated with therapy outcome than that assessed in PT or organ metastases. PD-L1 determined in skin/subcutaneous metastases showed no outcome correlation and therefore should be used with caution for clinical decision making. FUNDING Bristol-Myers Squibb (ADOREG/TRIM, NCT05750511); German Research Foundation (DFG; Clinician Scientist Program UMEA); Else Kröner-Fresenius-Stiftung (EKFS; Medical Scientist Academy UMESciA).
Collapse
Affiliation(s)
- Jan-Malte Placke
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| | - Mona Kimmig
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Klaus Griewank
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | | | | | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical School, Homburg/Saar, Germany.
| | - Jens Ulrich
- Skin Cancer Center, Department of Dermatology, Harz Clinics, Quedlinburg, Germany.
| | - Alexander Kreuter
- Department of Dermatology, Venereology and Allergology, Helios St. Elisabeth Klinik Oberhausen, University Witten-Herdecke, Oberhausen, Germany.
| | - Peter Mohr
- Department of Dermatology, Elbe Kliniken Buxtehude, Buxtehude, Germany.
| | - Ralf Gutzmer
- Department of Dermatology, Skin Cancer Center Minden, Minden, Germany.
| | - Friedegund Meier
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.
| | - Edgar Dippel
- Department of Dermatology, Ludwigshafen Medical Center, Ludwigshafen, Germany.
| | - Julia Welzel
- Department of Dermatology, Augsburg Medical Center, Augsburg, Germany.
| | - Daniel Robert Engel
- Department of Immunodynamics, Institute for Experimental Immunology and Imaging, Medical Research Centre, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| | - Sophia Kreft
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany.
| | - Antje Sucker
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Georg Lodde
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Frederik Krefting
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Ingo Stoffels
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Joachim Klode
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Eva Hadaschik
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany.
| | - Jürgen C Becker
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany; Translational Skin Cancer Research, West German Cancer Center, University Medicine Essen, Essen, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| | - Michael Weichenthal
- Department of Dermatology, University Hospital of Schleswig-Holstein, Kiel, Germany.
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany.
| |
Collapse
|
44
|
Yu Z, Qin L, Yu G. The progresses of relevant factors on the efficacy of immune checkpoint inhibitors in the non-small cell lung cancer patients. Cancer Treat Res Commun 2023; 37:100758. [PMID: 37776694 DOI: 10.1016/j.ctarc.2023.100758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/29/2023] [Accepted: 09/02/2023] [Indexed: 10/02/2023]
Abstract
Lung cancer has the highest mortality rate of all cancers worldwide. Although immune checkpoint inhibitor (ICI)-based therapy can improve the survival of patients with lung cancer, its efficacy is affected by many factors. Therefore, it is necessary to identify factors that affect the efficacy of ICI-based treatment and establish a model for predicting drug response and resistance before and during treatment for individualized and accurate treatment of patients. This review summarizes the clinical and biological factors related to ICI-based treatment of non-small cell lung cancer (NSCLC) and the recent research progress of predictive models for assessing ICI efficacy.
Collapse
Affiliation(s)
- Zhaoqing Yu
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Li Qin
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Guifang Yu
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
45
|
Li A, Luo L, Du W, Yu Z, He L, Fu S, Wang Y, Zhou Y, Yang C, Yang Y, Fang W, Zhang L, Hong S. Deciphering transcriptomic determinants of the divergent link between PD-L1 and immunotherapy efficacy. NPJ Precis Oncol 2023; 7:87. [PMID: 37696887 PMCID: PMC10495439 DOI: 10.1038/s41698-023-00443-3] [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: 02/28/2023] [Accepted: 08/31/2023] [Indexed: 09/13/2023] Open
Abstract
Programmed cell death ligand 1 (PD-L1) expression remains the most widely used biomarker for predicting response to immune checkpoint inhibitors (ICI), but its predictiveness varies considerably. Identification of factors accounting for the varying PD-L1 performance is urgently needed. Here, using data from three independent trials comprising 1239 patients, we have identified subsets of cancer with distinct PD-L1 predictiveness based on tumor transcriptome. In the Predictiveness-High (PH) group, PD-L1+ tumors show better overall survival, progression-free survival, and objective response rate with ICI than PD-L1- tumors across three trials. However, the Predictiveness-Low (PL) group demonstrates an opposite trend towards better outcomes for PD-L1- tumors. PD-L1+ tumors from the PH group demonstrate the superiority of ICI over chemotherapy, whereas PD-L1+ tumors from the PL group show comparable efficacy between two treatments or exhibit an opposite trend favoring chemotherapy. This observation of context-dependent predictiveness remains strong regardless of immune subtype (Immune-Enriched or Non-Immune), PD-L1 regulation mechanism (adaptative or constitutive), tumor mutation burden, or neoantigen load. This work illuminates avenues for optimizing the use of PD-L1 expression in clinical decision-making and trial design, although this exploratory concept should be further confirmed in large trials.
Collapse
Affiliation(s)
- Anlin Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Linfeng Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei Du
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhixin Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lina He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sha Fu
- Department of Cellular & Molecular Diagnostics Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-Sen University, Guangzhou, China
| | - Yuanyuan Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yixin Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chunlong Yang
- Department of Oncology, The People's Hospital of Fengqing, Lincang, China
| | - Yunpeng Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenfeng Fang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Shaodong Hong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Department of Oncology, The People's Hospital of Fengqing, Lincang, China.
| |
Collapse
|
46
|
Takagi H, Muto S, Enta A, Fukuhara M, Asano S, Shio Y, Suzuki H. A case of discordant histology and expression of programmed death ligand 1 between primary tumor and brain metastases in adenosquamous carcinoma of the lung. Thorac Cancer 2023; 14:2707-2711. [PMID: 37545057 PMCID: PMC10493479 DOI: 10.1111/1759-7714.15061] [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: 04/25/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023] Open
Abstract
A patient presented with vomiting and gait disturbance. Investigation revealed a single cerebellar tumor and another tumor in the upper lobe of the left lung. Based on the severe vomiting and gait disturbance, we removed the cerebellar tumor first, achieving resolution of symptoms. The cerebellar tumor was pathologically diagnosed as metastatic lung adenocarcinoma. No other metastases were identified, including in the mediastinal lymph nodes. We therefore resected the primary lung tumor. On final pathological analysis, the tumor in the upper lobe of the left lung was diagnosed as adenosquamous carcinoma with no lymph node metastasis. PD-L1 expression was low in the primary lung adenosquamous carcinoma and high in the cerebellar metastasis. Furthermore, both tumors were KRASG12C -positive. Tumor PD-L1 expression is considered important for immune escape. In this case, adenocarcinoma cells in the primary adenosquamous carcinoma may have migrated to form a cerebellar metastasis. In advanced lung cancer, tumor growth may be observed in some lesions even when many other lesions are controlled by chemo- or immunotherapy. Biopsy to confirm histology and PD-L1 expression is worth considering, depending on the location of the metastases and the invasiveness of the biopsy procedure.
Collapse
Affiliation(s)
- Hironori Takagi
- Department of Chest SurgeryIwaki City Medical CenterIwakiJapan
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| | - Satoshi Muto
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| | - Akio Enta
- Department of Chest SurgeryIwaki City Medical CenterIwakiJapan
| | | | | | - Yutaka Shio
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| | - Hiroyuki Suzuki
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| |
Collapse
|
47
|
Wang D, Qiu B, Liu Q, Xia L, Liu S, Zheng C, Liu H, Mo Y, Zhang X, Hu Y, Zheng S, Zhou Y, Fu J, Chen N, Liu F, Zhou R, Guo J, Fan W, Liu H. Patlak-Ki derived from ultra-high sensitivity dynamic total body [ 18F]FDG PET/CT correlates with the response to induction immuno-chemotherapy in locally advanced non-small cell lung cancer patients. Eur J Nucl Med Mol Imaging 2023; 50:3400-3413. [PMID: 37310427 DOI: 10.1007/s00259-023-06298-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/01/2023] [Indexed: 06/14/2023]
Abstract
PURPOSE This study aimed to investigate the predictive value of metabolic features in response to induction immuno-chemotherapy in patients with locally advanced non-small cell cancer (LA-NSCLC), using ultra-high sensitivity dynamic total body [18F]FDG PET/CT. METHODS The study analyzed LA-NSCLC patients who received two cycles of induction immuno-chemotherapy and underwent a 60-min dynamic total body [18F]FDG PET/CT scan before treatment. The primary tumors (PTs) were manually delineated, and their metabolic features, including the Patlak-Ki, Patlak-Intercept, maximum SUV (SUVmax), metabolic tumor volume (MTV) and total lesion glycolysis (TLG) were evaluated. The overall response rate (ORR) to induction immuno-chemotherapy was evaluated according to RECIST 1.1 criteria. The Patlak-Ki of PTs was calculated from the 20-60 min frames using the Patlak graphical analysis. The best feature was selected using Laplacian feature importance scores, and an unsupervised K-Means method was applied to cluster patients. ROC curve was used to examine the effect of selected metabolic feature in predicting tumor response to treatment. The targeted next generation sequencing on 1021 genes was conducted. The expressions of CD68, CD86, CD163, CD206, CD33, CD34, Ki67 and VEGFA were assayed through immunohistochemistry. The independent samples t test and the Mann-Whitney U test were applied in the intergroup comparison. Statistical significance was considered at P < 0.05. RESULTS Thirty-seven LA-NSCLC patients were analyzed between September 2020 and November 2021. All patients received two cycles of induction chemotherapy combined with Nivolumab/ Camrelizumab. The Laplacian scores showed that the Patlak-Ki of PTs had the highest importance for patient clustering, and the unsupervised K-Means derived decision boundary of Patlak-Ki was 2.779 ml/min/100 g. Patients were categorized into two groups based on their Patlak-Ki values: high FDG Patlak-Ki (H-FDG-Ki, Patlak-Ki > 2.779 ml/min/100 g) group (n = 23) and low FDG Patlak-Ki (L-FDG-Ki, Patlak-Ki ≤ 2.779 ml/min/100 g) group (n = 14). The ORR to induction immuno-chemotherapy was 67.6% (25/37) in the whole cohort, with 87% (20/23) in H-FDG-Ki group and 35.7% (5/14) in L-FDG-Ki group (P = 0.001). The sensitivity and specificity of Patlak-Ki in predicting the treatment response were 80% and 75%, respectively [AUC = 0.775 (95%CI 0.605-0.945)]. The expression of CD3+/CD8+ T cells and CD86+/CD163+/CD206+ macrophages were higher in the H-FDG-Ki group, while Ki67, CD33+ myeloid cells, CD34+ micro-vessel density (MVD) and tumor mutation burden (TMB) were comparable between the two groups. CONCLUSIONS The total body [18F]FDG PET/CT scanner performed a dynamic acquisition of the entire body and clustered LA-NSCLC patients into H-FDG-Ki and L-FDG-Ki groups based on the Patlak-Ki. Patients with H-FDG-Ki demonstrated better response to induction immuno-chemotherapy and higher levels of immune cell infiltration in the PTs compared to those with L-FDG-Ki. Further studies with a larger patient cohort are required to validate these findings.
Collapse
Affiliation(s)
- DaQuan Wang
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Bo Qiu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - QianWen Liu
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - LiangPing Xia
- Department of VIP, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - SongRan Liu
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | | | - Hui Liu
- United Imaging Healthcare, Shanghai, China
| | - YiWen Mo
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Xu Zhang
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - YingYing Hu
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - ShiYang Zheng
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Yin Zhou
- SuZhou TongDiao Company, Suzhou, China
| | - Jia Fu
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - NaiBin Chen
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - FangJie Liu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Rui Zhou
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - JinYu Guo
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Wei Fan
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China.
| | - Hui Liu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People's Republic of China.
| |
Collapse
|
48
|
Wei F, Azuma K, Nakahara Y, Saito H, Matsuo N, Tagami T, Kouro T, Igarashi Y, Tokito T, Kato T, Kondo T, Murakami S, Usui R, Himuro H, Horaguchi S, Tsuji K, Murotani K, Ban T, Tamura T, Miyagi Y, Sasada T. Machine learning for prediction of immunotherapeutic outcome in non-small-cell lung cancer based on circulating cytokine signatures. J Immunother Cancer 2023; 11:e006788. [PMID: 37433717 DOI: 10.1136/jitc-2023-006788] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI) therapy has substantially improved the overall survival (OS) in patients with non-small-cell lung cancer (NSCLC); however, its response rate is still modest. In this study, we developed a machine learning-based platform, namely the Cytokine-based ICI Response Index (CIRI), to predict the ICI response of patients with NSCLC based on the peripheral blood cytokine profiles. METHODS We enrolled 123 and 99 patients with NSCLC who received anti-PD-1/PD-L1 monotherapy or combined chemotherapy in the training and validation cohorts, respectively. The plasma concentrations of 93 cytokines were examined in the peripheral blood obtained from patients at baseline (pre) and 6 weeks after treatment (early during treatment: edt). Ensemble learning random survival forest classifiers were developed to select feature cytokines and predict the OS of patients undergoing ICI therapy. RESULTS Fourteen and 19 cytokines at baseline and on treatment, respectively, were selected to generate CIRI models (namely preCIRI14 and edtCIRI19), both of which successfully identified patients with worse OS in two completely independent cohorts. At the population level, the prediction accuracies of preCIRI14 and edtCIRI19, as indicated by the concordance indices (C-indices), were 0.700 and 0.751 in the validation cohort, respectively. At the individual level, patients with higher CIRI scores demonstrated worse OS [hazard ratio (HR): 0.274 and 0.163, and p<0.0001 and p=0.0044 in preCIRI14 and edtCIRI19, respectively]. By including other circulating and clinical features, improved prediction efficacy was observed in advanced models (preCIRI21 and edtCIRI27). The C-indices in the validation cohort were 0.764 and 0.757, respectively, whereas the HRs of preCIRI21 and edtCIRI27 were 0.141 (p<0.0001) and 0.158 (p=0.038), respectively. CONCLUSIONS The CIRI model is highly accurate and reproducible in determining the patients with NSCLC who would benefit from anti-PD-1/PD-L1 therapy with prolonged OS and may aid in clinical decision-making before and/or at the early stage of treatment.
Collapse
Affiliation(s)
- Feifei Wei
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Koichi Azuma
- Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Yoshiro Nakahara
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| | - Haruhiro Saito
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Norikazu Matsuo
- Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tomoyuki Tagami
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co Inc, Kawasaki, Japan
| | - Taku Kouro
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Yuka Igarashi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Takaaki Tokito
- Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Terufumi Kato
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Tetsuro Kondo
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Shuji Murakami
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Ryo Usui
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Hidetomo Himuro
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Shun Horaguchi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Kayoko Tsuji
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Kenta Murotani
- Biostatistics Center, Kurume University School of Medicine, Kurume, Japan
| | - Tatsuma Ban
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yohei Miyagi
- Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
49
|
Hong TH, Bang YH, Joe CY, Hwang S, Lee B, Lee N, Park S, Jung HA, Sun JM, Ahn JS, Ahn MJ, Choi YL, Lee SH. Programmed Death-Ligand 1 Copy Number Alteration as an Adjunct Biomarker of Response to Immunotherapy in Advanced NSCLC. J Thorac Oncol 2023; 18:896-906. [PMID: 37028596 DOI: 10.1016/j.jtho.2023.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/08/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
INTRODUCTION This study aimed to evaluate the value of programmed death-ligand 1 (PD-L1) copy number (CN) alteration as an additional biomarker to standard immunohistochemistry (IHC) in predicting response to immune checkpoint inhibitor (ICI) therapy in advanced NSCLC. METHODS Before ICI monotherapy, tumor PD-L1 CN alteration (gain, neutral, or loss) was called using whole-exome sequencing data and compared with IHC results (tumor proportion score ≥50, 1-49, or 0). Progression-free survival (PFS) and overall survival were correlated with both biomarkers. In addition, the impact of CN alteration was further evaluated in two independent cohorts using next-generation sequencing panel. RESULTS A total of 291 patients with advanced-stage NSCLC met the study inclusion criteria. Although the IHC classification distinguished the best responsive group (tumor proportion score ≥ 50), the CN-based classification distinguished the worst responsive group (CN loss) from the others (PFS, p = 0.020; overall survival, p = 0.004). After adjusting for IHC results, CN loss was an independent risk factor for progression (adjusted hazard ratio = 1.32, 95% confidence interval: 1.00-1.73, p = 0.049) and death (adjusted hazard ratio = 1.39, 95% confidence interval: 1.05-1.85, p = 0.022). A risk classification system was developed on the basis of IHC and CN profiles, which outperformed the conventional IHC system. In the validation cohorts, CN loss determined by next-generation sequencing panel was independently associated with worse PFS after ICI treatment, revealing its practical value. CONCLUSIONS This is the first study to directly compare CN alterations with IHC results and survival outcomes after anti-PD-(L)1 therapy. Tumor PD-L1 CN loss can serve as an adjunct biomarker to predict the lack of response. Prospective studies are required to further validate this biomarker.
Collapse
Affiliation(s)
- Tae Hee Hong
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Digital Health, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Yeong Hak Bang
- Department of Digital Health, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea; Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Cheol Yong Joe
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute of Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Soohyun Hwang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Boram Lee
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Naeun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute of Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun-Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute of Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
50
|
Le X, Elamin YY, Zhang J. New Actions on Actionable Mutations in Lung Cancers. Cancers (Basel) 2023; 15:cancers15112917. [PMID: 37296880 DOI: 10.3390/cancers15112917] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Actionable mutations refer to DNA alterations that, if detected, would be expected to affect patients' response to treatments [...].
Collapse
Affiliation(s)
- Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yasir Y Elamin
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|