1
|
Yao J, Lin X, Zhang X, Xie M, Ma X, Bao X, Song J, Liang Y, Wang Q, Xue X. Predictive biomarkers for immune checkpoint inhibitors therapy in lung cancer. Hum Vaccin Immunother 2024; 20:2406063. [PMID: 39415535 PMCID: PMC11487980 DOI: 10.1080/21645515.2024.2406063] [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: 04/23/2024] [Revised: 09/05/2024] [Accepted: 09/15/2024] [Indexed: 10/18/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have changed the treatment mode of lung cancer, extending the survival time of patients unprecedentedly. Once patients respond to ICIs, the median duration of response is usually longer than that achieved with cytotoxic or targeted drugs. Unfortunately, there is still a large proportion of lung cancer patients do not respond to ICI. Effective biomarkers are crucial for identifying lung cancer patients who can benefit from them. The first predictive biomarker is programmed death-ligand 1 (PD-L1), but its predictive value is limited to specific populations. With the development of single-cell sequencing and spatial imaging technologies, as well as the use of deep learning and artificial intelligence, the identification of predictive biomarkers has been greatly expanded. In this review, we will dissect the biomarkers used to predict ICIs efficacy in lung cancer from the tumor-immune microenvironment and host perspectives, and describe cutting-edge technologies to further identify biomarkers.
Collapse
Affiliation(s)
- Jie Yao
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xuwen Lin
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xin Zhang
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Mei Xie
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xidong Ma
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinyu Bao
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Jialin Song
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Yiran Liang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Qiqi Wang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| |
Collapse
|
2
|
Liu X, Kong Y, Qian Y, Guo H, Zhao L, Wang H, Xu K, Ye L, Liu Y, Lu H, He Y. Spatial heterogeneity of infiltrating immune cells in the tumor microenvironment of non-small cell lung cancer. Transl Oncol 2024; 50:102143. [PMID: 39366301 PMCID: PMC11474367 DOI: 10.1016/j.tranon.2024.102143] [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: 05/10/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are essential components of the tumor microenvironment (TME) of non-small cell lung cancer (NSCLC). Still, it is difficult to describe due to their heterogeneity. In this study, five cell markers from NSCLC patients were analyzed. We segmented tumor cells (TCs) and TILs using Efficientnet-B3 and explored their quantitative information and spatial distribution. After that, we simulated multiplex immunohistochemistry (mIHC) by overlapping continuous single chromogenic IHCs slices. As a result, the proportion and the density of programmed cell death-ligand 1 (PD-L1)-positive TCs were the highest in the core. CD8+ T cells were the closest to the tumor (median distance: 41.71 μm), while PD-1+T cells were the most distant (median distance: 62.2μm), and our study found that most lymphocytes clustered together within the peritumoral range of 10-30 μm where cross-talk with TCs could be achieved. We also found that the classification of TME could be achieved using CD8+ T-cell density, which is correlated with the prognosis of patients. In addition, we achieved single chromogenic IHC slices overlap based on CD4-stained IHC slices. We explored the number and spatial distribution of cells in heterogeneous TME of NSCLC patients and achieved TME classification. We also found a way to show the co-expression of multiple molecules economically.
Collapse
Affiliation(s)
- Xinyue Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Yan Kong
- SJTU-Yale Joint Center for Biostatistics and Data Science, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Youwen Qian
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Affiliated to Naval Medical University, Shanghai, China
| | - Haoyue Guo
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Lishu Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Kandi Xu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Li Ye
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Yujin Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 200092, China
| | - Hui Lu
- SJTU-Yale Joint Center for Biostatistics and Data Science, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
3
|
Molero A, Hernandez S, Alonso M, Peressini M, Curto D, Lopez-Rios F, Conde E. Assessment of PD-L1 expression and tumour infiltrating lymphocytes in early-stage non-small cell lung carcinoma with artificial intelligence algorithms. J Clin Pathol 2024:jcp-2024-209766. [PMID: 39419594 DOI: 10.1136/jcp-2024-209766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024]
Abstract
AIMS To study programmed death ligand 1 (PD-L1) expression and tumour infiltrating lymphocytes (TILs) in patients with early-stage non-small cell lung carcinoma (NSCLC) with artificial intelligence (AI) algorithms. METHODS The study included samples from 50 early-stage NSCLCs. PD-L1 immunohistochemistry (IHC) stained slides (clone SP263) were scored manually and with two different AI tools (PathAI and Navify Digital Pathology) by three pathologists. TILs were digitally assessed on H&E and CD8 IHC stained sections with two different algorithms (PathAI and Navify Digital Pathology, respectively). The agreement between observers and methods for each biomarker was analysed. For PD-L1, the turn-around time (TAT) for manual versus AI-assisted scoring was recorded. RESULTS Agreement was higher in tumours with low PD-L1 expression regardless of the approach. Both AI-powered tools identified a significantly higher number of cases equal or above 1% PD-L1 tumour proportion score as compared with manual scoring (p=0.00015), a finding with potential therapeutic implications. Regarding TAT, there were significant differences between manual scoring and AI use (p value <0.0001 for all comparisons). The total TILs density with the PathAI algorithm and the total density of CD8+ cells with the Navify Digital Pathology software were significantly correlated (τ=0.49 (95% CI 0.37, 0.61), p value<0.0001). CONCLUSIONS This preliminary study supports the use of AI algorithms for the scoring of PD-L1 and TILs in patients with NSCLC.
Collapse
Affiliation(s)
- Aida Molero
- Pathology, Complejo Asistencial de Segovia, Segovia, Spain
| | - Susana Hernandez
- Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Marta Alonso
- Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Melina Peressini
- Tumor Microenvironment and Immunotherapy Research Group, Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Daniel Curto
- Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando Lopez-Rios
- Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), CIBERONC, Universidad Complutense de Madrid, Madrid, Spain
| | - Esther Conde
- Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), CIBERONC, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
4
|
Li X, Peng Y, Wu D, Tang J, Wu Y. Efficacy and safety of anlotinib as maintenance therapy in patients with advanced non-small cell lung cancer achieving SD post first-line chemotherapy combined with immunotherapy. J Chemother 2024:1-9. [PMID: 39219263 DOI: 10.1080/1120009x.2024.2397924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/29/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Advanced non-small cell lung cancer (NSCLC) remains a significant clinical challenge, particularly in patients who exhibit stable disease (SD) following first-line chemotherapy combined with immunotherapy. This study aims to evaluate the efficacy and safety of Anlotinib, a novel multitarget tyrosine kinase inhibitor, as maintenance therapy in this patient cohort. This retrospective, single-center study enrolled patients with advanced NSCLC who showed SD after receiving a combination of first-line chemo-immunotherapy for 4 cycles, then add anlotinib to subsequent standard maintenance therapy, continuing treatment until disease progression or the occurrence of intolerable toxic side effects. The primary endpoint was progression-free survival (P FS), overall survival (OS), objective response rate (ORR), disease control rate (DCR) and safety profile. A total of 52 patients were enrolled, the median P FS and OS was 5.0m and 10.0m, respectively. The ORR and DCR was 28.85% and 67.31%. subgroup analysis indicated that its efficacy correlate with certain Adverse Effects (AEs, such as hypertension, proteinuria, and hand-foot syndrome). Further mechanistic analysis suggests that this regimen may likely reduce immune suppression by depleting Tregs, thereby further activating the immune system to exert synergistic anti-tumor effects. Besides promising efficacy, the toxicity can be tolerated. Anlotinib demonstrates promising efficacy as a maintenance therapy in patients with advanced NSCLC who have achieved SD following first-line chemotherapy combined with immunotherapy. The manageable safety profile and the observed extension in P FS and OS suggest that Anlotinib could be a valuable therapeutic option for this challenging patient population. Further large-scale randomized controlled trials are warranted to confirm these findings and to optimize patient selection and management strategies.
Collapse
Affiliation(s)
- Xiaobing Li
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Peng
- Department of Radiotherapy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - De Wu
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Tang
- Department of Lymphoma, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuebing Wu
- Department of Lymphoma, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Xia L, Zhu X, Wang Y, Lu S. The gut microbiota improves the efficacy of immune-checkpoint inhibitor immunotherapy against tumors: From association to cause and effect. Cancer Lett 2024; 598:217123. [PMID: 39033797 DOI: 10.1016/j.canlet.2024.217123] [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: 05/08/2024] [Revised: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Immune-checkpoint inhibitors (ICIs), including anti-PD-1/PD-L1 therapeutic antibodies, have markedly enhanced survival across numerous cancer types. However, the limited number of patients with durable benefits creates an urgent need to identify response biomarkers and to develop novel strategies so as to improve response. It is widely recognized that the gut microbiome is a key mediator in shaping immunity. Additionally, the gut microbiome shows significant potential in predicting the response to and enhancing the efficacy of ICI immunotherapy against cancer. Recent studies encompassing mechanistic analyses and clinical trials of microbiome-based therapy have shown a cause-and-effect relationship between the gut microbiome and the modulation of the ICI immunotherapeutic response, greatly contributing to the establishment of novel strategies that will improve response and overcome resistance to ICI treatment. In this review, we outline the current state of research advances and discuss the future directions of utilizing the gut microbiome to enhance the efficacy of ICI immunotherapy against tumors.
Collapse
Affiliation(s)
- Liliang Xia
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Xiaokuan Zhu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Ying Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, PR China.
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China.
| |
Collapse
|
6
|
Sugawara K, Fukuda T, Murakami C, Oka D, Yoshii T, Amori G, Ishibashi K, Kobayashi Y, Hara H, Kanda H, Motoi N. Impacts of tumor microenvironment during neoadjuvant chemotherapy in patients with esophageal squamous cell carcinoma. Cancer Sci 2024; 115:2819-2830. [PMID: 38693726 PMCID: PMC11309932 DOI: 10.1111/cas.16203] [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: 02/03/2024] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 05/03/2024] Open
Abstract
With the advent of immune checkpoint inhibitors (ICIs), a better understanding of tumor microenvironment (TME) is becoming crucial in managing esophageal squamous cell carcinoma (ESCC) patients. We investigated the survival impact of TME status and changes in patients with ESCC who underwent neoadjuvant chemotherapy (NAC) followed by surgery (n = 264). We examined immunohistochemical status (CD4+, CD8+, CD20+, Foxp3+, HLA class-1+, CD204+, and programmed death ligand-1 [PD-L1+]) on 264 pre-NAC and 204 paired post-NAC specimens. Patients were classified by their pre- and post-NAC immune cell status and their changes following NAC. Our findings showed that pre-NAC TME status was not significantly associated with survival outcomes. In contrast, post-NAC TME status, such as low level of T cells, CD4+ T cells, and high PD-L1 combined positive score (CPS), were significantly associated with poor overall survival (OS). Notably, TME changes through NAC exerted significant survival impacts; patients with consistently low levels of T cells, low levels of CD4+ T cells, or high levels of PD-L1 (CPS) had very poor OS (3-year OS: 35.5%, 40.2%, and 33.3%, respectively). Tumor microenvironment changes of consistently low T cells, low CD4+ T cells, and high PD-L1 were independent predictors of poor OS in multivariate Cox hazards analyses, while factors indicating post-NAC status (T cells, CD4+, and PD-L1 [CPS]) alone were not. Therefore, we suggest that the consistently low T/high PD-L1 group could benefit from additional therapies, such as ICIs, and the importance of stratification by the TME, which has recently been recognized.
Collapse
Affiliation(s)
- Kotaro Sugawara
- Department of Gastroenterological SurgerySaitama Cancer CenterSaitamaJapan
| | - Takashi Fukuda
- Department of Gastroenterological SurgerySaitama Cancer CenterSaitamaJapan
| | - Chiaki Murakami
- Department of PathologySaitama Cancer CenterSaitamaJapan
- Department of PathologySaitama Medical Center, Saitama Medical UniversitySaitamaJapan
| | - Daiji Oka
- Department of Gastroenterological SurgerySaitama Cancer CenterSaitamaJapan
| | - Takako Yoshii
- Department of GastroenterologySaitama Cancer CenterSaitamaJapan
| | - Gulanbar Amori
- Department of PathologySaitama Cancer CenterSaitamaJapan
- Division of PathologyCancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
- Department of PathologyCancer Institute Hospital of JFCR, Japanese Foundation for Cancer ResearchTokyoJapan
| | | | | | - Hiroki Hara
- Department of GastroenterologySaitama Cancer CenterSaitamaJapan
| | - Hiroaki Kanda
- Department of PathologySaitama Cancer CenterSaitamaJapan
| | - Noriko Motoi
- Department of PathologySaitama Cancer CenterSaitamaJapan
- Center for Cancer Genomic MedicineSaitama Cancer CenterSaitamaJapan
| |
Collapse
|
7
|
Inomata M, Minatoyama S, Takata N, Hayashi K, Hirai T, Seto Z, Tokui K, Taka C, Okazawa S, Kambara K, Imanishi S, Miwa T, Hayashi R, Matsui S, Tobe K. Comparison of the efficacy of first‑/second‑generation EGFR‑tyrosine kinase inhibitors and osimertinib for EGFR‑mutant lung cancer with negative or low PD‑L1 expression. Mol Clin Oncol 2024; 20:43. [PMID: 38756869 PMCID: PMC11097130 DOI: 10.3892/mco.2024.2741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 03/28/2024] [Indexed: 05/18/2024] Open
Abstract
In epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC) with negative or low programmed death ligand-1 (PD-L1) expression, the acquisition rate of the T790M mutation is higher after treatment with first-/second-generation EGFR-tyrosine kinase inhibitors (TKIs) and the progression-free survival (PFS) is longer in patients treated with osimertinib. The present study compared the clinical course after the initiation of each EGFR-TKI monotherapy in patients with EGFR-mutant NSCLC with negative or low PD-L1 expression. Data of patients with EGFR-mutant NSCLC with negative or low PD-L1 expression who were treated with EGFR-TKI monotherapy were retrieved and retrospectively analyzed. Between June 2013 and November 2023, 26 and 29 patients were treated with first-/second-generation EGFR-TKIs and osimertinib, respectively. The PFS time was longer in patients treated with osimertinib (median, 22.5 months) than in those treated with first-/second-generation EGFR-TKIs (median, 12.9 months). However, the EGFR-TKI treatment duration, defined as the PFS for osimertinib, or the sum of the PFS for first-/second-generation EGFR-TKIs and sequential osimertinib therapy after the acquisition of the T790M mutation, was similar between patients treated with first-/second-generation EGFR-TKIs (median, 23.0 months) and osimertinib (median, 22.5 months). The Cox proportional hazard model suggested that there was no significant difference in the EGFR-TKI treatment duration between patients treated with first-/second-generation EGFR-TKIs and patients treated with osimertinib (hazard ratio, 1.31, 95% CI, 0.55-3.13). In conclusion, first-/second-generation EGFR-TKIs and osimertinib were associated with a similar EGFR-TKI treatment duration in patients with EGFR-mutant NSCLC with negative or low PD-L1 expression. The findings suggested that both treatments are promising for this population.
Collapse
Affiliation(s)
- Minehiko Inomata
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Shuhei Minatoyama
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Naoki Takata
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Kana Hayashi
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Takahiro Hirai
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Zenta Seto
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Kotaro Tokui
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Chihiro Taka
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Seisuke Okazawa
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Kenta Kambara
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Shingo Imanishi
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Toshiro Miwa
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Ryuji Hayashi
- Department of Medical Oncology, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Shoko Matsui
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Toyama University Hospital, Toyama, Toyama 930-0194, Japan
| |
Collapse
|
8
|
Hayashi H, Chamoto K, Hatae R, Kurosaki T, Togashi Y, Fukuoka K, Goto M, Chiba Y, Tomida S, Ota T, Haratani K, Takahama T, Tanizaki J, Yoshida T, Iwasa T, Tanaka K, Takeda M, Hirano T, Yoshida H, Ozasa H, Sakamori Y, Sakai K, Higuchi K, Uga H, Suminaka C, Hirai T, Nishio K, Nakagawa K, Honjo T. Soluble immune checkpoint factors reflect exhaustion of antitumor immunity and response to PD-1 blockade. J Clin Invest 2024; 134:e168318. [PMID: 38557498 PMCID: PMC10977985 DOI: 10.1172/jci168318] [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: 12/22/2022] [Accepted: 01/30/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUNDPrecise stratification of patients with non-small cell lung cancer (NSCLC) is needed for appropriate application of PD-1/PD-L1 blockade therapy.METHODSWe measured soluble forms of the immune-checkpoint molecules PD-L1, PD-1, and CTLA-4 in plasma of patients with advanced NSCLC before PD-1/PD-L1 blockade. A prospective biomarker-finding trial (cohort A) included 50 previously treated patients who received nivolumab. A retrospective observational study was performed for patients treated with any PD-1/PD-L1 blockade therapy (cohorts B and C), cytotoxic chemotherapy (cohort D), or targeted therapy (cohort E). Plasma samples from all patients were assayed for soluble immune-checkpoint molecules with a highly sensitive chemiluminescence-based assay.RESULTSNonresponsiveness to PD-1/PD-L1 blockade therapy was associated with higher concentrations of these soluble immune factors among patients with immune-reactive (hot) tumors. Such an association was not apparent for patients treated with cytotoxic chemotherapy or targeted therapy. Integrative analysis of tumor size, PD-L1 expression in tumor tissue (tPD-L1), and gene expression in tumor tissue and peripheral CD8+ T cells revealed that high concentrations of the 3 soluble immune factors were associated with hyper or terminal exhaustion of antitumor immunity. The combination of soluble PD-L1 (sPD-L1) and sCTLA-4 efficiently discriminated responsiveness to PD-1/PD-L1 blockade among patients with immune-reactive tumors.CONCLUSIONCombinations of soluble immune factors might be able to identify patients unlikely to respond to PD-1/PD-L1 blockade as a result of terminal exhaustion of antitumor immunity. Our data suggest that such a combination better predicts, along with tPD-L1, for the response of patients with NSCLC.TRIAL REGISTRATIONUMIN000019674.FUNDINGThis study was funded by Ono Pharmaceutical Co. Ltd. and Sysmex Corporation.
Collapse
Affiliation(s)
- Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kenji Chamoto
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Immuno-Oncology PDT, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryusuke Hatae
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Kurosaki
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yosuke Togashi
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- Department of Tumor Microenvironment, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazuya Fukuoka
- Clinical Research Center, Kindai University Hospital, Osaka-Sayama, Japan
| | | | - Yasutaka Chiba
- Clinical Research Center, Kindai University Hospital, Osaka-Sayama, Japan
| | - Shuta Tomida
- Department of Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Takayo Ota
- Department of Medical Oncology, Izumi City General Hospital, Izumi, Japan
| | - Koji Haratani
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Takayuki Takahama
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Junko Tanizaki
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Takeshi Yoshida
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tsutomu Iwasa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kaoru Tanaka
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masayuki Takeda
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- Department of Cancer Genomics and Medical Oncology, Nara Medical University School of Medicine, Nara, Japan
| | - Tomoko Hirano
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hironori Yoshida
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Ozasa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuichi Sakamori
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | | | | | | | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tasuku Honjo
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
9
|
Passaro A, Al Bakir M, Hamilton EG, Diehn M, André F, Roy-Chowdhuri S, Mountzios G, Wistuba II, Swanton C, Peters S. Cancer biomarkers: Emerging trends and clinical implications for personalized treatment. Cell 2024; 187:1617-1635. [PMID: 38552610 PMCID: PMC7616034 DOI: 10.1016/j.cell.2024.02.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/21/2024] [Accepted: 02/28/2024] [Indexed: 04/02/2024]
Abstract
The integration of cancer biomarkers into oncology has revolutionized cancer treatment, yielding remarkable advancements in cancer therapeutics and the prognosis of cancer patients. The development of personalized medicine represents a turning point and a new paradigm in cancer management, as biomarkers enable oncologists to tailor treatments based on the unique molecular profile of each patient's tumor. In this review, we discuss the scientific milestones of cancer biomarkers and explore future possibilities to improve the management of patients with solid tumors. This progress is primarily attributed to the biological characterization of cancers, advancements in testing methodologies, elucidation of the immune microenvironment, and the ability to profile circulating tumor fractions. Integrating these insights promises to continually advance the precision oncology field, fostering better patient outcomes.
Collapse
Affiliation(s)
- Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Maise Al Bakir
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Emily G Hamilton
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Fabrice André
- Gustave-Roussy Cancer Center, Paris Saclay University, Villejuif, France
| | - Sinchita Roy-Chowdhuri
- Department of Anatomic Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giannis Mountzios
- Fourth Department of Medical Oncology and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Department of Oncology, University College London Hospitals, London, UK
| | - Solange Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| |
Collapse
|
10
|
Liu L, Zhang Z, Jiang C, Zhu Y, Han R, Wu L, Xu Y. HOXC9 characterizes a suppressive tumor immune microenvironment and integration with multiple immune biomarkers predicts response to PD-1 blockade plus chemotherapy in lung adenocarcinoma. Aging (Albany NY) 2024; 16:4841-4861. [PMID: 38446596 PMCID: PMC10968688 DOI: 10.18632/aging.205637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/13/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND The quest for dependable biomarkers to predict responses to immune checkpoint inhibitors (ICIs) combined with chemotherapy in advanced non-small cell lung cancer remains unfulfilled. HOXC9, known for its role in oncogenesis and creating a suppressive tumor microenvironment (TME), shows promise in enhancing predictive precision when included as a TME biomarker. This study explores the predictive significance of HOXC9 for ICI plus chemotherapy efficacy in lung adenocarcinoma (LUAD). METHODS Following the bioinformatic findings, assays were performed to ascertain the effects of Hoxc9 on oncogenesis and response to programmed death 1 (PD-1) blockade. Furthermore, a cohort of LUAD patients were prospectively enrolled to receive anti-PD-1 plus chemotherapy. Based on the expression levels, baseline characteristics, and clinical outcomes, the predictive potential of HOXC9, PD-L1, CD4, CD8, CD68, and FOXP3 was integrally analyzed. HOXC9 not only mediated oncogenesis, but also corelated with suppressive TME. CMT167 and LLC cell lines unveiled the impacts of Hoxc9 on proliferation, invasion, and migration. Subsequently, tumor-bearing murine models were established to validate the inverse relationship between Hoxc9 expression and effective CD8+ T cells. RESULTS Inhibition of Hoxc9 significantly curtailed tumor growth (P<0.05), independent of PD-1 blockade. In patient studies, while individual markers fell short in prognosticating survival, a notable elevation in CD8-positive expression was observed in responders (P=0.042). Yet, the amalgamation of HOXC9 with other markers provided a more distinct differentiation between responders and non-responders. Notably, patients displaying PD-L1+/HOXC9- and CD8+/HOXC9- phenotypes exhibited significantly prolonged progression-free survival. CONCLUSIONS The expression of HOXC9 may serve as a biomarker to amplifying predictive efficacy for ICIs plus chemotherapy, which is also a viable oncogene and therapeutic target for immunotherapy in LUAD.
Collapse
Affiliation(s)
- Liang Liu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Zhenshan Zhang
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai 201315, China
| | - Chenxue Jiang
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Yaoyao Zhu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Ruiqin Han
- State Key Laboratory of Common Mechanism Research for Major Disease, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100191, China
| | - Leilei Wu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Yaping Xu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| |
Collapse
|
11
|
Duan XP, Qin BD, Jiao XD, Liu K, Wang Z, Zang YS. New clinical trial design in precision medicine: discovery, development and direction. Signal Transduct Target Ther 2024; 9:57. [PMID: 38438349 PMCID: PMC10912713 DOI: 10.1038/s41392-024-01760-0] [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: 11/30/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
In the era of precision medicine, it has been increasingly recognized that individuals with a certain disease are complex and different from each other. Due to the underestimation of the significant heterogeneity across participants in traditional "one-size-fits-all" trials, patient-centered trials that could provide optimal therapy customization to individuals with specific biomarkers were developed including the basket, umbrella, and platform trial designs under the master protocol framework. In recent years, the successive FDA approval of indications based on biomarker-guided master protocol designs has demonstrated that these new clinical trials are ushering in tremendous opportunities. Despite the rapid increase in the number of basket, umbrella, and platform trials, the current clinical and research understanding of these new trial designs, as compared with traditional trial designs, remains limited. The majority of the research focuses on methodologies, and there is a lack of in-depth insight concerning the underlying biological logic of these new clinical trial designs. Therefore, we provide this comprehensive review of the discovery and development of basket, umbrella, and platform trials and their underlying logic from the perspective of precision medicine. Meanwhile, we discuss future directions on the potential development of these new clinical design in view of the "Precision Pro", "Dynamic Precision", and "Intelligent Precision". This review would assist trial-related researchers to enhance the innovation and feasibility of clinical trial designs by expounding the underlying logic, which be essential to accelerate the progression of precision medicine.
Collapse
Affiliation(s)
- Xiao-Peng Duan
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bao-Dong Qin
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Dong Jiao
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ke Liu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhan Wang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yuan-Sheng Zang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
12
|
Sun W, Qiu F, Zheng J, Fang L, Qu J, Zhang S, Jiang N, Zhou J, Zeng X, Zhou J. CD57-positive CD8 + T cells define the response to anti-programmed cell death protein-1 immunotherapy in patients with advanced non-small cell lung cancer. NPJ Precis Oncol 2024; 8:25. [PMID: 38297019 PMCID: PMC10830454 DOI: 10.1038/s41698-024-00513-0] [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: 07/11/2023] [Accepted: 12/08/2023] [Indexed: 02/02/2024] Open
Abstract
Immune checkpoint inhibitors have transformed the treatment landscape of non-small cell lung cancer (NSCLC). However, accurately identifying patients who will benefit from immunotherapy remains a challenge. This study aimed to discover potential biomarkers for predicting immunotherapy response in NSCLC patients. Single-cell mass cytometry (CyTOF) was utilized to analyze immune cell subsets in peripheral blood mononuclear cells (PBMCs) obtained from NSCLC patients before and 12 weeks after single-agent immunotherapy. The CyTOF findings were subsequently validated using flow cytometry and multiplex immunohistochemistry/immunofluorescence in PBMCs and tumor tissues, respectively. RNA sequencing (RNA-seq) was performed to elucidate the underlying mechanisms. In the CyTOF cohort (n = 20), a high frequency of CD57+CD8+ T cells in PBMCs was associated with durable clinical benefit from immunotherapy in NSCLC patients (p = 0.034). This association was further confirmed in an independent cohort using flow cytometry (n = 27; p < 0.001), with a determined cutoff value of 12.85%. The cutoff value was subsequently validated in another independent cohort (AUC = 0.733). We also confirmed the CyTOF findings in pre-treatment formalin-fixed and paraffin-embedded tissues (n = 90; p < 0.001). RNA-seq analysis revealed 475 differentially expressed genes (DEGs) between CD57+CD8+ T cells and CD57-CD8+ T cells, with functional analysis identifying DEGs significantly enriched in immune-related signaling pathways. This study highlights CD57+CD8+ T cells as a promising biomarker for predicting immunotherapy success in NSCLC patients.
Collapse
Affiliation(s)
- Wenjia Sun
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Fengqi Qiu
- Cancer Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Jing Zheng
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Liangjie Fang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shumeng Zhang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Nan Jiang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xun Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jianya Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
13
|
Shirasawa M, Yoshida T, Ohe Y. Biomarkers of immunotherapy for non-small cell lung cancer. Jpn J Clin Oncol 2024; 54:13-22. [PMID: 37823218 DOI: 10.1093/jjco/hyad134] [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: 06/24/2023] [Accepted: 09/22/2023] [Indexed: 10/13/2023] Open
Abstract
Immunotherapy is revolutionizing the treatment of non-small cell lung cancer by targeting immune checkpoint proteins, including programmed death-1, programmed death ligand 1 and cytotoxic T-lymphocyte-associated antigen 4. Several immune checkpoint inhibitors, including programmed death ligand 1 inhibitors, programmed death-1 inhibitors and cytotoxic T-lymphocyte-associated antigen 4 inhibitors, were approved for the treatment of patients with advanced non-small cell lung cancer. Programmed death ligand 1 expression is currently the only predictive biomarker for immune checkpoint inhibitors to guide the treatment strategy in these patients. However, programmed death ligand 1 expression is not a perfect biomarker for predicting the efficacy of immunotherapy. Therefore, various biomarkers such as tumour mutation burden, tumour microenvironment, gut microbiome and T-cell receptor repertoire have been proposed to predict the efficacy of immunotherapy more accurately. Additionally, combining different biomarkers may provide a more accurate prediction of response to immunotherapy. This article reports the review of the latest evidence of the predictive marker of immunotherapy in patients with advanced non-small cell lung cancer.
Collapse
Affiliation(s)
- Masayuki Shirasawa
- Department of Thoracic Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo 104-0045 Japan
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara City, Kanagawa 252-0375, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo 104-0045 Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo 104-0045 Japan
| |
Collapse
|
14
|
Mogavero A, Cantale O, Mollica V, Anpalakhan S, Addeo A, Mountzios G, Friedlaender A, Kanesvaran R, Novello S, Banna GL. First-line immunotherapy in non-small cell lung cancer: how to select and where to go. Expert Rev Respir Med 2023; 17:1191-1206. [PMID: 38294292 DOI: 10.1080/17476348.2024.2302356] [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: 06/11/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024]
Abstract
INTRODUCTION Immunotherapy (IO) has established a new milestone in lung cancer treatment. Several registrational studies have approved immune checkpoint inhibitors (ICIs) in different settings, including the metastatic nonsmall cell lung cancer (NSCLC). As well known, responders are just a certain proportion of patients; therefore, their selection by using predictive factors has stood out as a crucial issue to address in tailoring a patient-centered care. AREAS COVERED In our review we propose a detailed yet handy cross section on ICIs as first-line treatment in metastatic NSCLC, regarding indications, histological, clinical, and blood-based biomarkers, other than their mechanisms of resistance and new immunological actionable targets. We performed a literature search through PubMed entering keywords complying with crucial features of immunotherapy. EXPERT OPINION IO represents the backbone of lung cancer treatment. Trials are currently testing novel immune blockade agents assessing combinatorial approaches with standard ICIs, or antibody drug conjugates (ADC), harboring immunological targets. Perfecting patients' selection is an ongoing challenge and a more and more urgent need in order to best predict responders who will consistently benefit from it.
Collapse
Affiliation(s)
| | | | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Shobana Anpalakhan
- Department of Oncology, Portsmouth Hospitals University NHS Trust, Portsmouth, UK
| | - Alfredo Addeo
- Oncology Department, HUG-Hopitaux Universitaires de Geneve, Geneva, Switzerland
| | - Giannis Mountzios
- Fourth Oncology Department and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece
| | | | - Ravindran Kanesvaran
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Silvia Novello
- Department of Oncology, University of Turin, Turin, Italy
| | - Giuseppe Luigi Banna
- Department of Oncology, Portsmouth Hospitals University NHS Trust, Portsmouth, UK
- Science and Health, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| |
Collapse
|
15
|
Wang L, Geng H, Liu Y, Liu L, Chen Y, Wu F, Liu Z, Ling S, Wang Y, Zhou L. Hot and cold tumors: Immunological features and the therapeutic strategies. MedComm (Beijing) 2023; 4:e343. [PMID: 37638340 PMCID: PMC10458686 DOI: 10.1002/mco2.343] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
The "hotness" or "coldness" of the tumors are determined by the information of the cancer cells themselves, tumor immune characteristics, tumor microenvironment, and signaling mechanisms, which are key factors affecting cancer patients' clinical efficacy. The switch mechanism of "hotness" and "coldness" and its corresponding pathological characteristics and treatment strategies are the frontier and hot spot of tumor treatment. How to distinguish the "hotness" or "coldness" effectively and clarify the causes, microenvironment state, and characteristics are very important for the tumor response and efficacy treatments. Starting from the concept of hot and cold tumor, this review systematically summarized the molecular characteristics, influencing factors, and therapeutic strategies of "hot and cold tumors," and analyzed the immunophenotypes, the tumor microenvironment, the signaling pathways, and the molecular markers that contribute to "hot and cold tumors" in details. Different therapeutic strategies for "cold and hot tumors" based on clinical efficacy were analyzed with drug targets and proteins for "cold and hot tumors." Furthermore, this review combines the therapeutic strategies of different "hot and cold tumors" with traditional medicine and modern medicine, to provide a basis and guidance for clinical decision-making of cancer treatment.
Collapse
Affiliation(s)
- Lianjie Wang
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Hui Geng
- Department of Internal MedicineShanghai International Medical CenterShanghaiChina
| | - Yujie Liu
- Department of NephrologyShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Lei Liu
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yanhua Chen
- Department of the Tumor Research Center, Academy of Integrative MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Fanchen Wu
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zhiyi Liu
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Shiliang Ling
- Department of Medical OncologyNingbo Hospital of Traditional Chinese Medicine, Zhejiang ProvinceNingboChina
| | - Yan Wang
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Lihong Zhou
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
16
|
Haratani K, Nakamura A, Mamesaya N, Mitsuoka S, Yoneshima Y, Saito R, Tanizaki J, Fujisaka Y, Hata A, Tsuruno K, Sakamoto T, Teraoka S, Oki M, Watanabe H, Sato Y, Nakano Y, Otani T, Sakai K, Tomida S, Chiba Y, Ito A, Nishio K, Yamamoto N, Nakagawa K, Hayashi H. Tumor Microenvironment Landscape of NSCLC Reveals Resistance Mechanisms for Programmed Death-Ligand 1 Blockade After Chemoradiotherapy: A Multicenter Prospective Biomarker Study (WJOG11518L:SUBMARINE). J Thorac Oncol 2023; 18:1334-1350. [PMID: 37364849 DOI: 10.1016/j.jtho.2023.06.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023]
Abstract
INTRODUCTION The PACIFIC regimen of consolidation therapy with the programmed cell death-ligand 1 inhibitor durvalumab after definitive concurrent chemoradiation therapy has become a standard of care for individuals with unresectable stage III NSCLC. Nevertheless, approximately half of the treated patients experience disease progression within 1 year, with the mechanisms of treatment resistance being poorly understood. We here performed a nationwide prospective biomarker study to explore the resistance mechanisms (WJOG11518L:SUBMARINE). METHODS A total of 135 patients with unresectable stage III NSCLC who received the PACIFIC regimen were included for comprehensive profiling of the tumor microenvironment by immunohistochemistry, transcriptome analysis, and genomic sequencing of pretreatment tumor tissue and flow cytometric analysis of circulating immune cells. Progression-free survival was compared on the basis of these biomarkers. RESULTS The importance of preexisting effective adaptive immunity in tumors was revealed for treatment benefit regardless of genomic features. We also identified CD73 expression by cancer cells as a mechanism of resistance to the PACIFIC regimen. Multivariable analysis of immunohistochemistry data with key clinical factors as covariables indicated that low CD8+ tumor-infiltrating lymphocyte density and the high CD73+ cancer cells were independently associated with poor durvalumab outcome (hazard ratios = 4.05 [95% confidence interval: 1.17-14.04] for CD8+ tumor-infiltrating lymphocytes; 4.79 [95% confidence interval: 1.12-20.58] for CD73). In addition, whole-exome sequencing of paired tumor samples suggested that cancer cells eventually escaped immune pressure as a result of neoantigen plasticity. CONCLUSIONS Our study emphasizes the importance of functional adaptive immunity in stage III NSCLC and implicates CD73 as a promising treatment target, thus providing insight forming a basis for development of a new treatment approach in NSCLC.
Collapse
Affiliation(s)
- Koji Haratani
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan.
| | - Atsushi Nakamura
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Miyagi, Japan
| | - Nobuaki Mamesaya
- Division of Thoracic Oncology, Shizuoka Cancer Center, Sunto-gun, Shizuoka, Japan
| | - Shigeki Mitsuoka
- Department of Clinical Oncology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Osaka, Japan
| | - Yasuto Yoneshima
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Ryota Saito
- Department of Respiratory Medicine, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Junko Tanizaki
- Division of Medical Oncology, Kishiwada City Hospital, Kishiwada, Osaka, Japan
| | - Yasuhito Fujisaka
- Medical Oncology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Akito Hata
- Division of Thoracic Oncology, Kobe Minimally Invasive Cancer Center, Kobe, Hyogo, Japan
| | - Kosuke Tsuruno
- Department of Respiratory Medicine, Iizuka Hospital, Iizuka, Fukuoka, Japan
| | - Tomohiro Sakamoto
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Shunsuke Teraoka
- Internal Medicine III, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Masahide Oki
- Department of Respiratory Medicine, National Hospital Organization Nagoya Medical Center, Nagoya, Aichi, Japan
| | - Hiroshi Watanabe
- Department of Respiratory Medicine, Saka General Hospital, Shiogama, Miyagi, Japan
| | - Yuki Sato
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Yusuke Nakano
- Department of Medical Oncology, Izumi City General Hospital, Izumi, Osaka, Japan
| | - Tomoyuki Otani
- Department of Pathology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Shuta Tomida
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Okayama, Japan
| | - Yasutaka Chiba
- Clinical Research Center, Kindai University Hospital, Osaka-Sayama, Osaka, Japan
| | - Akihiko Ito
- Department of Pathology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Nobuyuki Yamamoto
- Internal Medicine III, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| |
Collapse
|
17
|
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
|
18
|
Cooper WA, John T. Rethinking Biomarkers for Combination Chemoimmunotherapy. J Thorac Oncol 2023; 18:841-843. [PMID: 37348991 DOI: 10.1016/j.jtho.2023.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 06/24/2023]
Affiliation(s)
- Wendy A Cooper
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia; School of Medicine, University of Western Sydney, Sydney, Australia.
| | - Thomas John
- Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia
| |
Collapse
|
19
|
Sun D, Liu J, Zhou H, Shi M, Sun J, Zhao S, Chen G, Zhang Y, Zhou T, Ma Y, Zhao Y, Fang W, Zhao H, Huang Y, Yang Y, Zhang L. Classification of Tumor Immune Microenvironment According to Programmed Death-Ligand 1 Expression and Immune Infiltration Predicts Response to Immunotherapy Plus Chemotherapy in Advanced Patients With NSCLC. J Thorac Oncol 2023; 18:869-881. [PMID: 36948245 DOI: 10.1016/j.jtho.2023.03.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/14/2023] [Accepted: 03/10/2023] [Indexed: 03/24/2023]
Abstract
INTRODUCTION According to mechanisms of adaptive immune resistance, tumor immune microenvironment (TIME) is classified into four types: (1) programmed death-ligand 1 (PD-L1)-negative and tumor-infiltrating lymphocyte (TIL)-negative (type I); (2) PD-L1-positive and TIL-positive (type II); (3) PD-L1-negative and TIL-positive (type III); and (4) PD-L1-positive and TIL-negative (type IV). However, the relationship between the TIME classification model and immunotherapy efficacy has not been validated by any large-scale randomized controlled clinical trial among patients with advanced NSCLC. METHODS On the basis of RNA-sequencing and immunohistochemistry data from the ORIENT-11 study, we optimized the TIME classification model and evaluated its predictive value for the efficacy of immunotherapy plus chemotherapy. RESULTS PD-L1 mRNA expression and immune score calculated by the ESTIMATE method were the strongest predictors for the efficacy of immunotherapy plus chemotherapy. Therefore, they were determined as the optimized definition of the TIME classification system. When compared between combination therapy and chemotherapy alone, only the type II subpopulation with high immune score and high PD-L1 mRNA expression was significantly associated with improved progression-free survival (PFS) (hazard ratio = 0.12, 95% confidence interval: 0.06-0.25, p < 0.001) and overall survival (hazard ratio = 0.27, 95% confidence interval: 0.13-0.55, p < 0.001). In the combination group, the type II subpopulation had a much longer survival time, not even reaching the median PFS or overall survival, but the other three subpopulations were susceptible to having similar PFS. In the chemotherapy group, there was no marked association between survival outcomes and TIME subtypes. CONCLUSIONS Only patients with both high PD-L1 expression and high immune infiltration could benefit from chemotherapy plus immunotherapy in first-line treatment of advanced NSCLC. For patients lacking either PD-L1 expression or immune infiltration, chemotherapy alone might be a better treatment option to avoid unnecessary toxicities and financial burdens.
Collapse
Affiliation(s)
- Dongchen Sun
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China; Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiaqing Liu
- State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China; Department of Intensive Care Unit, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Huaqiang Zhou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Mengting Shi
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China; Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiya Sun
- New Drug Biology and Translational Medicine, Innovent Biologics, Inc., Suzhou, People's Republic of China
| | - Shen Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Gang Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Yaxiong Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Ting Zhou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Yuxiang Ma
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Yuanyuan Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Wenfeng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Hongyun Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Yan Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Yunpeng Yang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.
| |
Collapse
|
20
|
Chi A, Nguyen NP. Mechanistic rationales for combining immunotherapy with radiotherapy. Front Immunol 2023; 14:1125905. [PMID: 37377970 PMCID: PMC10291094 DOI: 10.3389/fimmu.2023.1125905] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Immunotherapy consisted mainly of immune checkpoint inhibitors (ICIs) has led to significantly improved antitumor response. However, such response has been observed only in tumors possessing an overall responsive tumor immune micro-environment (TIME), in which the presence of functional tumor-infiltrating lymphocytes (TILs) is critical. Various mechanisms of immune escape from immunosurveillance exist, leading to different TIME phenotypes in correlation with primary or acquired resistance to ICIs. Radiotherapy has been shown to induce antitumor immunity not only in the irradiated primary tumor, but also at unirradiated distant sites of metastases. Such antitumor immunity is mainly elicited by radiation's stimulatory effects on antigenicity and adjuvanticity. Furthermore, it may be significantly augmented when irradiation is combined with immunotherapy, such as ICIs. Therefore, radiotherapy represents one potential therapeutic strategy to restore anti-tumor immunity in tumors presenting with an unresponsive TIME. In this review, the generation of anti-tumor immunity, its impairment, radiation's immunogenic properties, and the antitumor effects of combining radiation with immunotherapy will be comprehensively discussed.
Collapse
Affiliation(s)
- Alexander Chi
- Department of Radiation Oncology, Capital Medical University Xuanwu Hospital, Beijing, China
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Nam Phong Nguyen
- Department of Radiation Oncology, Howard University, Washington, DC, United States
| |
Collapse
|
21
|
Shirasawa M, Yoshida T, Shiraishi K, Takigami A, Takayanagi D, Imabayashi T, Matsumoto Y, Masuda K, Shinno Y, Okuma Y, Goto Y, Horinouchi H, Yotsukura M, Yoshida Y, Nakagawa K, Tsuchida T, Hamamoto R, Yamamoto N, Motoi N, Kohno T, Watanabe SI, Ohe Y. Identification of inflamed-phenotype of small cell lung cancer leading to the efficacy of anti-PD-L1 antibody and chemotherapy. Lung Cancer 2023; 179:107183. [PMID: 37037178 DOI: 10.1016/j.lungcan.2023.107183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/17/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND Platinum etoposide plus anti-programmed cell death ligand-1 (PD-L1) antibody therapy is the standard of care for extensive-stage small cell lung cancer (ES-SCLC). However, patient characteristics associated with the efficacy of the combination therapy in SCLC are unclear. METHODS We retrospectively reviewed post-surgical limited-stage (LS)-SCLC and ES-SCLC patients treated with atezolizumab plus carboplatin and etoposide (ACE). The association between SCLC subtypes based on transcriptomic data and pathological findings, including CD8-positive tumor-infiltrating lymphocyte (TIL) status, was investigated in the LS-SCLC cohort. The association between the efficacy of ACE therapy, pathological subtypes, and TIL status was evaluated in the ES-SCLC cohort. RESULTS The LS-SCLC cohort (N = 48) was classified into four SCLC subtypes (ASCL1 + NEUROD1 [SCLC-A + N, N = 17], POU2F3 [SCLC-P, N = 15], YAP1 [SCLC-Y, N = 10], and inflamed [SCLC-I, N = 6]) based on transcriptomic data. SCLC-I showed enriched immune-related pathways, the highest immune score (CD8A expression and T-cell-inflamed gene expression profiles), and epithelial-mesenchymal transition (EMT), in transcriptional subtypes. Immunohistochemical staining (IHC) showed that SCLC-I had the highest density of CD8-positive TILs in transcriptional subtypes. In the ES-SCLC cohort, the efficacy of ACE therapy did not differ according to pathological subtypes. The progression-free survival (PFS) of TILHigh patients was significantly longer than that of TILLow patients (PFS: 7.3 months vs. 4.0 months, p < 0.001). CONCLUSION Tumors with a high density of TILs, which represent the most immunogenic SCLC subtype (SCLC-I), based on transcriptomic data could benefit from ACE therapy.
Collapse
|
22
|
Characterization of Infiltrating Immune Cells and Secretory or Membrane-Associated Proteins in KRAS Lung Adenocarcinoma. J Immunol Res 2023; 2023:4987832. [PMID: 36793588 PMCID: PMC9925262 DOI: 10.1155/2023/4987832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/31/2022] [Accepted: 01/16/2023] [Indexed: 02/09/2023] Open
Abstract
Background This study identified the expression and prognosis significance of secretory or membrane-associated proteins in KRAS lung adenocarcinoma (LUAD) and depicted the characteristics between the immune cell infiltration and the expression of these genes. Methods Gene expression data of LUAD samples (n = 563) were accessed from The Cancer Genome Atlas (TCGA). The expression of secretory or membrane-associated proteins was compared among the KRAS-mutant, wild-type, and normal groups, as well as the subgroup of the KRAS-mutant group. We identified the survival-related differentially expressed secretory or membrane-associated proteins and conducted the functional enrichment analysis. Then, the characterization and association between their expression and the 24 immune cell subsets were investigated. We also constructed a scoring model to predict KRAS mutation by LASSO and logistic regression analysis. Results Secretory or membrane-associated genes with differential expression (n = 74) across three groups (137 KRAS LUAD, 368 wild-type LUAD, and 58 normal groups) were identified, and the results of GO and KEGG indicated that they were strongly associated with immune cell infiltrations. Among them, ten genes were significantly related to the survival of patients with KRAS LUAD. The expression of IL37, KIF2, INSR, and AQP3 had the most significant correlations with immune cell infiltration. In addition, eight DEGs from the KRAS subgroups were highly correlated with immune infiltrations, especially TNFSF13B. Using LASSO-logistic regression, a KRAS mutation prediction model based on the 74 differentially expressed secretory or membrane-associated genes was built, and the accuracy was 0.79. Conclusion The research investigated the relationship between the expression of KRAS-related secretory or membrane-associated proteins in LUAD patients with prognostic prediction and immune infiltration characterization. Our study demonstrated that secretory or membrane-associated genes were closely associated with the survival of KRAS LUAD patients and were strongly correlated to immune cell infiltration.
Collapse
|
23
|
Jia W, Guo H, Wang M, Li J, Yu J, Zhu H, Wu G. High post-chemotherapy TIL and increased CD4+TIL are independent prognostic factors of surgically resected NSCLC following neoadjuvant chemotherapy. MedComm (Beijing) 2023; 4:e213. [PMID: 36789099 PMCID: PMC9911612 DOI: 10.1002/mco2.213] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/28/2022] [Accepted: 01/11/2023] [Indexed: 02/12/2023] Open
Abstract
Neoadjuvant chemotherapy (NCT) has significantly improved the overall survival of patients with operable non-small cell lung cancer (NSCLC). Chemotherapy can remodel the tumor immune microenvironment (TIME) and has an important influence on antitumor immunity. For patients who underwent surgery for resected NSCLC following NCT (NCT-NSCLC), a prognostic value comparison between naïve and post-chemotherapy TIME is absent. We enrolled 89 patients with NCT-NSCLC in this study; the tumor-infiltrating lymphocyte (TIL), CD4+TIL, and CD8+TIL levels in naïve and post-chemotherapy tumor tissues were detected using immunohistochemistry staining and divided into high and low groups. Kaplan-Meier analysis revealed that major pathology response, pathological tumor, node, and metastasis stage post-NCT (ypTNM), high post-chemotherapy TIL, high post-chemotherapy CD8+TIL, low naïve CD4+TIL, low naïve CD4+/CD8+TIL ratio, and increased CD4+TIL levels post-chemotherapy were favorable prognostic factors in patients with NCT-NSCLC. Multivariate Cox analysis found that ypTNM, high post-chemotherapy TIL, and increased CD4+TIL levels post-chemotherapy were independent prognostic factors in patients with NCT-NSCLC. These results indicate that a TIME remodeled by chemotherapy plays an important role in antitumor immunity and has a better prognostic value than the naïve TIME.
Collapse
Affiliation(s)
- Wenxiao Jia
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical UniversityShandong Academy of Medical SciencesJinanShandongChina
| | - Hongbo Guo
- Department of Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical UniversityShandong Academy of Medical SciencesJinanShandongChina
| | - Min Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical UniversityShandong Academy of Medical SciencesJinanShandongChina
| | - Ji Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical UniversityShandong Academy of Medical SciencesJinanShandongChina
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical UniversityShandong Academy of Medical SciencesJinanShandongChina
| | - Hui Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical UniversityShandong Academy of Medical SciencesJinanShandongChina
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
24
|
Zhang X, Wang X, Hou L, Xu Z, Liu Y, Wang X. Nanoparticles overcome adaptive immune resistance and enhance immunotherapy via targeting tumor microenvironment in lung cancer. Front Pharmacol 2023; 14:1130937. [PMID: 37033636 PMCID: PMC10080031 DOI: 10.3389/fphar.2023.1130937] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/16/2023] [Indexed: 04/11/2023] Open
Abstract
Lung cancer is one of the common malignant cancers worldwide. Immune checkpoint inhibitor (ICI) therapy has improved survival of lung cancer patients. However, ICI therapy leads to adaptive immune resistance and displays resistance to PD-1/PD-L1 blockade in lung cancer, leading to less immune response of lung cancer patients. Tumor microenvironment (TME) is an integral tumor microenvironment, which is involved in immunotherapy resistance. Nanomedicine has been used to enhance the immunotherapy in lung cancer. In this review article, we described the association between TME and immunotherapy in lung cancer. We also highlighted the importance of TME in immunotherapy in lung cancer. Moreover, we discussed how nanoparticles are involved in regulation of TME to improve the efficacy of immunotherapy, including Nanomedicine SGT-53, AZD1080, Nanomodulator NRF2, Cisplatin nanoparticles, Au@PG, DPAICP@ME, SPIO NP@M-P, NBTXR3 nanoparticles, ARAC nanoparticles, Nano-DOX, MS NPs, Nab-paclitaxel, GNPs-hPD-L1 siRNA. Furthermore, we concluded that targeting TME by nanoparticles could be helpful to overcome resistance to PD-1/PD-L1 blockade in lung cancer.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xuemei Wang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Lijian Hou
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Zheng Xu
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yu’e Liu
- School of Medicine, Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, Tongji University, Shanghai, China
| | - Xueju Wang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
- *Correspondence: Xueju Wang,
| |
Collapse
|
25
|
Mino-Kenudson M, Schalper K, Cooper W, Dacic S, Hirsch FR, Jain D, Lopez-Rios F, Tsao MS, Yatabe Y, Beasley MB, Yu H, Sholl LM, Brambilla E, Chou TY, Connolly C, Wistuba I, Kerr KM, Lantuejoul S. Predictive Biomarkers for Immunotherapy in Lung Cancer: Perspective From the International Association for the Study of Lung Cancer Pathology Committee. J Thorac Oncol 2022; 17:1335-1354. [PMID: 36184066 DOI: 10.1016/j.jtho.2022.09.109] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 10/14/2022]
Abstract
Immunotherapy including immune checkpoint inhibitors (ICIs) has become the backbone of treatment for most lung cancers with advanced or metastatic disease. In addition, they have increasingly been used for early stage tumors in neoadjuvant and adjuvant settings. Unfortunately, however, only a subset of patients experiences meaningful response to ICIs. Although programmed death-ligand 1 (PD-L1) protein expression by immunohistochemistry (IHC) has played a role as the principal predictive biomarker for immunotherapy, its performance may not be optimal, and it suffers multiple practical issues with different companion diagnostic assays approved. Similarly, tumor mutational burden (TMB) has multiple technical issues as a predictive biomarker for ICIs. Now, ongoing research on tumor- and host immune-specific factors has identified immunotherapy biomarkers that may provide better response and prognosis prediction, in particular in a multimodal approach. This review by the International Association for the Study of Lung Cancer Pathology Committee provides an overview of various immunotherapy biomarkers, including updated data on PD-L1 IHC and TMB, and assessments of neoantigens, genetic and epigenetic signatures, immune microenvironment by IHC and transcriptomics, and microbiome and pathologic response to neoadjuvant immunotherapies. The aim of this review is to underline the efficacy of new individual or combined predictive biomarkers beyond PD-L1 IHC and TMB.
Collapse
Affiliation(s)
- Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts
| | - Kurt Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Wendy Cooper
- Royal Prince Alfred Hospital, NSW Health Pathology and University of Sydney, Camperdown, Australia
| | - Sanja Dacic
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Fred R Hirsch
- Center for Thoracic Oncology, The Tisch Cancer Institute, New York, New York; Icahn School of Medicine, Mount Sinai Health System, New York, New York
| | - Deepali Jain
- All India Institute of Medical Sciences, New Delhi, India
| | - Fernando Lopez-Rios
- Department of Pathology, "Doce de Octubre" University Hospital, Madrid, Spain
| | - Ming Sound Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | - Mary Beth Beasley
- Icahn School of Medicine, Mount Sinai Health System, New York, New York
| | - Hui Yu
- Center for Thoracic Oncology, The Tisch Cancer Institute, New York, New York; Icahn School of Medicine, Mount Sinai Health System, New York, New York
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, Massachusetts
| | | | | | - Casey Connolly
- International Association for the Study of Lung Cancer, Denver, Colorado
| | - Ignacio Wistuba
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Keith M Kerr
- Department of Pathology, Aberdeen Royal Infirmary, Aberdeen, United Kingdom
| | - Sylvie Lantuejoul
- Université Grenoble Alpes, Grenoble, France; Centre Léon Bérard Unicancer, Lyon, France.
| |
Collapse
|
26
|
Wu F, Jiang T, Chen G, Huang Y, Zhou J, Lin L, Feng J, Wang Z, Shu Y, Shi J, Hu Y, Wang Q, Cheng Y, Chen J, Lin X, Wang Y, Huang J, Cui J, Cao L, Liu Y, Zhang Y, Pan Y, Zhao J, Wang L, Chang J, Chen Q, Ren X, Zhang W, Fan Y, He Z, Fang J, Gu K, Dong X, Zhang T, Shi W, Zou J, Bai X, Ren S, Zhou C. Multiplexed imaging of tumor immune microenvironmental markers in locally advanced or metastatic non-small-cell lung cancer characterizes the features of response to PD-1 blockade plus chemotherapy. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 42:1331-1346. [PMID: 36331328 PMCID: PMC9759770 DOI: 10.1002/cac2.12383] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 09/16/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Although programmed cell death 1 (PD-1) blockade plus chemotherapy can significantly prolong the progression-free survival (PFS) and overall survival (OS) in first-line settings in patients with driver-negative advanced non-small-cell lung cancer (NSCLC), the predictive biomarkers remain undetermined. Here, we investigated the predictive value of tumor immune microenvironmental marker expression to characterize the response features to PD-1 blockade plus chemotherapy. METHODS Tumor tissue samples at baseline were prospectively collected from 144 locally advanced or metastatic NSCLC patients without driver gene alterations who received camrelizumab plus chemotherapy or chemotherapy alone. Tumor immune microenvironmental markers, including PD-1 ligand (PD-L1), CD8, CD68, CD4 and forkhead box P3, were assessed using multiplex immunofluorescence (mIF) assays. Kaplan-Meier curves were used to determine treatment outcome differences according to their expression status. Mutational profiles were compared between tumors with distinct expression levels of these markers and their combinations. RESULTS Responders had significantly higher CD8/PD-L1 (P = 0.015) or CD68/PD-L1 co-expression levels (P = 0.021) than non-responders in the camrelizumab plus chemotherapy group, while no difference was observed in the chemotherapy group. Patients with high CD8/PD-L1 or CD68/PD-L1 co-expression level was associated with significantly longer PFS (P = 0.002, P = 0.024; respectively) and OS (P = 0.006, P = 0.026; respectively) than those with low co-expression in camrelizumab plus chemotherapy group. When comparing survival in the camrelizumab plus chemotherapy with chemotherapy by CD8/PD-L1 co-expression stratification, significantly better PFS (P = 0.003) and OS (P = 0.032) were observed in high co-expression subgroups. The predictive value of CD8/PD-L1 and CD68/PD-L1 co-expression remained statistically significant for PFS and OS when adjusting clinicopathological features. Although the prevalence of TP53 or KRAS mutations was similar between patients with and without CD8/PD-L1 or CD68/PD-L1 co-expression, the positive groups had a significantly higher proportion of TP53/KRAS co-mutations than the negative groups (both 13.0% vs. 0.0%, P = 0.023). Notably, enriched PI3K (P = 0.012) and cell cycle pathway (P = 0.021) were found in the CD8/PD-L1 co-expression group. CONCLUSION Tumor immune microenvironmental marker expression, especially CD8/PD-L1 or CD68/PD-L1 co-expression, was associated with the efficacy of PD-1 blockade plus chemotherapy as first-line treatment in patients with advanced NSCLC.
Collapse
|
27
|
Zeng Z, Qian X, Liu F, Wang Y, Yuan Y, Fang C, Zhang X, Yuan S, Chen R, Yu B, Wang T, Yin Y, Li Y, Liu A. The efficacy and safety analysis of first-line immune checkpoint inhibitors in pulmonary sarcomatoid carcinoma. Front Immunol 2022; 13:956982. [PMID: 36389780 PMCID: PMC9659892 DOI: 10.3389/fimmu.2022.956982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 10/04/2022] [Indexed: 09/26/2023] Open
Abstract
Background Pulmonary sarcomatoid carcinoma (PSC) is a rare and aggressive disease without standardized treatment strategies. The efficacy of second-line or beyond immune checkpoint inhibitors (ICIs) has been proven in recent studies, whereas the evidence for first-line immunotherapy for PSC is still limited to case reports and remains poorly understood. Materials and methods This was a multicenter, retrospective analysis of 21 patients with a histological diagnosis of PSC who received ICI as first-line therapy from January 2019 to March 2022. The expression of PD-L1 was evaluated by immunohistochemistry (IHC) using the monoclonal antibody 22C3. Low and high PD-L1 expressions were defined using the tumor proportion score (TPS), with cutoffs of 1 and 50%, respectively. Results All eight patients had PD-L1 positivity who underwent PD-L1 expression assessment, and six patients (6/8, 75.0%) had high PD-L1 expression. Among the 21 PSC patients, seven received tislelizumab, six received camrelizumab, four received sintilimab, three received pembrolizumab, and one received durvalumab. Among them, 18 PSCs received combination therapy, whereas another three PSCs received immunotherapy alone. Out of the 21 PSC patients, 12 (57.1%) achieved a partial response (PR), and five patients had stable disease (SD) as the best response, whereas four PSCs experienced dramatic progressive disease (PD). The median progression-free survival (PFS) was 9.2 (95% CI [4.3, 14.1]) months, and the median OS was 22.8 (95% CI [4.0, 41.5]) months. Among the three treatment groups (immunotherapy alone, immunotherapy combined with anlotinib, and chemoimmunotherapy), the median PFS was 8.0, 9.4, and 9.6 months, and the median OS was 19.0, 22.8, and 30.6 months, respectively. There was no difference in PFS and OS between the three treatment regimen groups (P = 0.86 and P = 0.34, respectively) and different immunotherapies (P = 0.10 and P = 0.23, respectively). No serious adverse events (grade ≥ 3) were noted. Conclusion First-line immunotherapy has promising therapeutic potential in the treatment of PSC. More studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Zhimin Zeng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi, China
- Radiation Induced Heart Damage Institute of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaoying Qian
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fanrong Liu
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yong Wang
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yong Yuan
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chen Fang
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xinwei Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shangkun Yuan
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Renfang Chen
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Biao Yu
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tong Wang
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan Yin
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yong Li
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi, China
- Radiation Induced Heart Damage Institute of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
28
|
Liquid Biopsy and the Translational Bridge from the TIME to the Clinic. Cells 2022; 11:cells11193114. [PMID: 36231076 PMCID: PMC9563580 DOI: 10.3390/cells11193114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 09/09/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Research and advancing understanding of the tumor immune microenvironment (TIME) is vital to optimize and direct more effective cancer immune therapy. Pre-clinical bench research is vital to better understand the genomic interplay of the TIME and immune therapy responsiveness. However, a vital key to effective translational cancer research is having a bridge of translation to bring that understanding from the bench to the bedside. Without that bridge, research into the TIME will lack an efficient and effective translation into the clinic and cancer treatment decision making. As a clinical oncologist, the purpose of this commentary is to emphasize the importance of researching and improving clinical utility of the bridge, as well as the TIME research itself.
Collapse
|
29
|
Jiang D, Song Q, Wei X, Yu Z, Liu Y, Wang H, Wang X, Huang J, Su J, Hong Y, Xu Y, Xu C, Hou Y. PMS2 Expression With Combination of PD-L1 and TILs for Predicting Survival of Esophageal Squamous Cell Carcinoma. Front Oncol 2022; 12:897527. [PMID: 35865481 PMCID: PMC9294642 DOI: 10.3389/fonc.2022.897527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/02/2022] [Indexed: 11/17/2022] Open
Abstract
Background DNA mismatch repair (MMR) deficiency (dMMR) has been recognized as an important biomarker for immunotherapy in esophageal squamous cell carcinoma (ESCC), along with programmed death ligand 1 (PD-L1) expression and/or tumor-infiltrated lymphocytes (TILs). However, in ESCC, MMR protein assessment has not been well studied at present. Methods A total of 484 ESCC tissues treated between 2007 and 2010, in our hospital, were enrolled. Immunohistochemical expression of MLH1, MSH2, MSH6, PMS2, and PD-L1 on tissue microarray specimens and clinicopathological features, including TILs, were analyzed retrospectively. Results Out of the 484 studied cases, loss of MLH1, MSH2, MSH6, and PMS2 expression were found in 6.8%, 2.1%, 8.7%, and 4.8% patients, respectively. dMMR was found in 65 patients, 37 cases involved in one MMR protein, 17 cases involved in two proteins, 7 cases involved in three proteins, and 4 cases involved in four proteins. There was no significant survival difference between pMMR (MMR-proficient) and dMMR patients (P>0.05). However, 224 patients with low PMS2 expression had better DFS and OS than 260 patients with high PMS2 expression (P=0.006 for DFS and 0.008 for OS), which was identified as an independent prognostic factor in multivariate analyses. Positive PD-L1 expression was detected in 341 (70.5%) samples. In stage I-II disease, patients with PD-L1 expression had better DFS and OS than those without PD-L1 expression(P<0.05), which was not found in stage III-IV disease. With the ITWG system, 40.1% of cases were classified as high TILs. Patients in the high-TILs group tended to have better DFS (P=0.055) and OS (P=0.070) than those in the low-TILs group and the differences were statistically significant in pMMR, high MSH6, or PMS2 expression cases (P<0.05). Also, high PMS2 expression patients with both PD-L1 expression and high TILs, had similar DFS and OS compared with low PMS2 expression patients (P>0.05), which were much better than other high PMS2 expression patients. Conclusion The expression level of MMR proteins could also be used as a prognostic factor in ESCC and PMS2 expression outperformed other MMR proteins for predicting survival. The combination of PD-L1 expression and TILs may lead to more efficient risk stratification of ESCC.
Collapse
Affiliation(s)
- Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Qi Song
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaojun Wei
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zixiang Yu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yufeng Liu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haixing Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xingxing Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jieakesu Su
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Hong
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifan Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Yingyong Hou, ; Chen Xu,
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- *Correspondence: Yingyong Hou, ; Chen Xu,
| |
Collapse
|
30
|
Ushio R, Murakami S, Saito H. Predictive Markers for Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer. J Clin Med 2022; 11:1855. [PMID: 35407463 PMCID: PMC9000007 DOI: 10.3390/jcm11071855] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically improved the outcomes of non-small cell lung cancer patients and have increased the possibility of long-term survival. However, few patients benefit from ICIs, and no predictive biomarkers other than tumor programmed cell death ligand 1 (PD-L1) expression have been established. Hence, the identification of biomarkers is an urgent issue. This review outlines the current understanding of predictive markers for the efficacy of ICIs, including PD-L1, tumor mutation burden, DNA mismatch repair deficiency, microsatellite instability, CD8+ tumor-infiltrating lymphocytes, human leukocyte antigen class I, tumor/specific genotype, and blood biomarkers such as peripheral T-cell phenotype, neutrophil-to-lymphocyte ratio, interferon-gamma, and interleukin-8. A tremendous number of biomarkers are in development, but individual biomarkers are insufficient. Tissue biomarkers have issues in reproducibility and accuracy because of intratumoral heterogeneity and biopsy invasiveness. Furthermore, blood biomarkers have difficulty in reflecting the tumor microenvironment and therefore tend to be less predictive for the efficacy of ICIs than tissue samples. In addition to individual biomarkers, the development of composite markers, including novel technologies such as machine learning and high-throughput analysis, may make it easier to comprehensively analyze multiple biomarkers.
Collapse
Affiliation(s)
| | - Shuji Murakami
- Kanagawa Cancer Center, Department of Thoracic Oncology, 2-3-2 Nakao, Asahi, Yokohama 241-8515, Japan; (R.U.); (H.S.)
| | | |
Collapse
|
31
|
Daily Practice Assessment of KRAS Status in NSCLC Patients: A New Challenge for the Thoracic Pathologist Is Right around the Corner. Cancers (Basel) 2022; 14:cancers14071628. [PMID: 35406400 PMCID: PMC8996900 DOI: 10.3390/cancers14071628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/16/2022] Open
Abstract
Simple Summary RAS mutation is the most frequent oncogenic alteration in human cancers and KRAS is the most frequently mutated, notably in non-small cell lung carcinomas (NSCLC). Various attempts to inhibit KRAS in the past were unsuccessful in these latter tumors. However, recently, several small molecules (AMG510, MRTX849, JNJ-74699157, and LY3499446) have been developed to specifically target KRAS G12C-mutated tumors, which seems promising for patient treatment and should soon be administered in daily practice for non-squamous (NS)-NSCLC. In this context, it will be mandatory to systematically assess the KRAS status in routine clinical practice, at least in advanced NS-NSCLC, leading to new challenges for thoracic oncologists. Abstract KRAS mutations are among the most frequent genomic alterations identified in non-squamous non-small cell lung carcinomas (NS-NSCLC), notably in lung adenocarcinomas. In most cases, these mutations are mutually exclusive, with different genomic alterations currently known to be sensitive to therapies targeting EGFR, ALK, BRAF, ROS1, and NTRK. Recently, several promising clinical trials targeting KRAS mutations, particularly for KRAS G12C-mutated NSCLC, have established new hope for better treatment of patients. In parallel, other studies have shown that NSCLC harboring co-mutations in KRAS and STK11 or KEAP1 have demonstrated primary resistance to immune checkpoint inhibitors. Thus, the assessment of the KRAS status in advanced-stage NS-NSCLC has become essential to setting up an optimal therapeutic strategy in these patients. This stimulated the development of new algorithms for the management of NSCLC samples in pathology laboratories and conditioned reorganization of optimal health care of lung cancer patients by the thoracic pathologists. This review addresses the recent data concerning the detection of KRAS mutations in NSCLC and focuses on the new challenges facing pathologists in daily practice for KRAS status assessment.
Collapse
|
32
|
Pan Y, Fu Y, Zeng Y, Liu X, Peng Y, Hu C, Deng C, Qiu Z, Zou J, Liu Y, Wu F. The key to immunotherapy: how to choose better therapeutic biomarkers for patients with non-small cell lung cancer. Biomark Res 2022; 10:9. [PMID: 35255999 PMCID: PMC8900392 DOI: 10.1186/s40364-022-00355-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 02/18/2022] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy has become the standard of care for non-small cell lung cancer (NSCLC), either in combination or monotherapy. However, there are still some patients who cannot benefit from it. Immunization strategies for NSCLC are based on the expression of PD-L1 on tumor cells and TMB, and although these indicators have a certain predictive effect, their predictive performance is not good. Therefore, clinicians must make adjustments to recognize markers. This is a review article that summarized immunotherapeutic biomarkers according to the "seed-soil-environment", generalizes primary resistance to immunotherapy, and summarizes the integration of markers.
Collapse
Affiliation(s)
- Yue Pan
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yucheng Fu
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, People's Republic of China
| | - Yue Zeng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xiaohan Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yurong Peng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Chunhong Hu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Chao Deng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Zhenhua Qiu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jian Zou
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Yuxuan Liu
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Fang Wu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
- Hunan Cancer Mega-Data Intelligent Application and Engineering Research Centre, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
- Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
33
|
Wang L, Xu X, Shang B, Sun J, Liang B, Wang X, You W, Jiang S. High farnesoid X receptor expression predicts favorable clinical outcomes in PD‑L1 low/negative non‑small cell lung cancer patients receiving anti‑PD‑1‑based chemo‑immunotherapy. Int J Oncol 2022; 60:40. [PMID: 35211760 PMCID: PMC8923651 DOI: 10.3892/ijo.2022.5330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
Anti-programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1)-directed immunotherapy has revolutionized the treatment of advanced non-small cell lung cancer (NSCLC). However, predictive biomarkers are still lacking, particularly in identifying PD-L1low/negative patients who will benefit from immunotherapy. It was previously reported that farnesoid X receptor (FXR) downregulated PD-L1 expression in NSCLC, and that FXRhighPD-L1low mouse Lewis lung carcinoma tumors showed an increased susceptibility to PD-1 blockade compared with mock tumors. At present, whether the FXRhighPD-L1low phenotype predicts clinical response to immunotherapy in patients with NSCLC remains unclear. Herein, a retrospective study was conducted to examine the expression levels of FXR, PD-L1 and CD8+ T cells by immunohistochemistry in a cohort of 149 patients with NSCLC receiving anti-PD-1-based chemo-immunotherapy. The results revealed that high FXR and PD-L1 expression levels were associated with higher objective response rates (ORR) in all patients. High PD-L1 expression also indicated superior progression-free survival (PFS). Interestingly, an inverse correlation was identified between FXR and PD-L1 expression in specimens with NSCLC. Subgroup analysis revealed that high FXR expression was associated with a higher ORR, as well as longer PFS and overall survival (OS) in PD-L1low patients. Cox multivariate analysis revealed that high FXR expression was an independent predictor for PFS and OS in PD-L1low patients. Tumor microenvironment evaluation revealed a statistically significant decrease of infiltrating CD8+ T cells in FXRhigh specimens with NSCLC. Overall, the present study proposed an FXRhighPD-L1low signature as a candidate predictor of response to anti-PD-1-based chemo-immunotherapy in PD-L1low/negative patients with NSCLC, providing evidence that could be used to broaden the patients benefitting from immunotherapy.
Collapse
Affiliation(s)
- Lina Wang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Xiaolong Xu
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Bin Shang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Jian Sun
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Bin Liang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Xingguang Wang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Wenjie You
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Shujuan Jiang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
34
|
Baseline PD-L1 expression and tumour-infiltrated lymphocyte status predict the efficacy of durvalumab consolidation therapy after chemoradiotherapy in unresectable locally advanced patients with non-small-cell lung cancer. Eur J Cancer 2022; 162:1-10. [DOI: 10.1016/j.ejca.2021.11.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/18/2022]
|
35
|
Analytical validation of automated multiplex chromogenic immunohistochemistry for diagnostic and predictive purpose in non-small cell lung cancer. Lung Cancer 2022; 166:1-8. [DOI: 10.1016/j.lungcan.2022.01.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/24/2022] [Accepted: 01/30/2022] [Indexed: 12/12/2022]
|