1
|
Sakaguchi T, Iketani A, Ito K, Nishii Y, Katsuta K, Hataji O. Comparison of PD-L1 Expression Between Preoperative Biopsy Specimens and Surgical Specimens in Non-Small Cell Lung Cancer. Cancers (Basel) 2025; 17:398. [PMID: 39941767 PMCID: PMC11815912 DOI: 10.3390/cancers17030398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/18/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Recent advances in perioperative immunotherapies have led to a new era in the perioperative treatment of resectable, non-small cell lung cancer (NSCLC). Although the choice of neoadjuvant, adjuvant or perioperative immunotherapy remains controversial, few reports have compared programmed death ligand-1 (PD-L1) expression as a biomarker between preoperative biopsy specimens and surgical specimens. METHODS We retrospectively reviewed consecutive patients with NSCLC whose preoperative biopsy specimens and surgical specimens were tested for PD-L1 (22C3) and PD-L1 (SP263), respectively, from June 2022 to February 2024. The three categorical classifications of PD-L1 expression (negative [<1%], low [1-49%], and high [≥50%]) were compared between the two tests. RESULTS Of the 33 patients, 13 patients had negative PD-L1 expression, 9 patients had low PD-L1 expression and 11 patients had high PD-L1 expression with preoperative biopsy specimens, while 18 patients had negative PD-L1 expression, 10 patients had low PD-L1 expression and 5 patients had high PD-L1 expression with surgical specimens. The concordance rate for the three categorical classifications of PD-L1 expression between the preoperative biopsy specimens and surgical specimens was 57.6%. CONCLUSIONS PD-L1 expression may differ between preoperative biopsy specimens and surgical specimens. PD-L1 expression evaluated using small biopsy specimens may be largely influenced by chance due to intra-tumoral heterogeneity.
Collapse
Affiliation(s)
- Tadashi Sakaguchi
- Department of Respiratory Medicine, Matsusaka Municipal Hospital, 1550 Tonomachi, Matsusaka 515-0073, Mie, Japan
| | - Akemi Iketani
- Pathology Department, Matsusaka Municipal Hospital, 1550 Tonomachi, Matsusaka 515-0073, Mie, Japan (K.K.)
| | - Kentaro Ito
- Department of Respiratory Medicine, Matsusaka Municipal Hospital, 1550 Tonomachi, Matsusaka 515-0073, Mie, Japan
| | - Yoichi Nishii
- Department of Respiratory Medicine, Matsusaka Municipal Hospital, 1550 Tonomachi, Matsusaka 515-0073, Mie, Japan
| | - Koji Katsuta
- Pathology Department, Matsusaka Municipal Hospital, 1550 Tonomachi, Matsusaka 515-0073, Mie, Japan (K.K.)
| | - Osamu Hataji
- Department of Respiratory Medicine, Matsusaka Municipal Hospital, 1550 Tonomachi, Matsusaka 515-0073, Mie, Japan
| |
Collapse
|
2
|
Krishnamurthy K, Chai J, Liu X, Elsayad M, Goldstein DY. Intratumoral heterogeneity of oncogenic drivers in mixed histology lung adenocarcinomas: How tissue selection impacts molecular testing? Pathol Res Pract 2024; 263:155577. [PMID: 39265501 DOI: 10.1016/j.prp.2024.155577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/21/2024] [Accepted: 08/31/2024] [Indexed: 09/14/2024]
Abstract
Majority of the lung adenocarcinomas show a mixture of different histological patterns. The possibility of histologically heterogeneous areas of the adenocarcinoma showing genetic heterogeneity and harboring different driver mutations, with potentially significant clinical impact, has not been adequately addressed. Currently, there are no guidelines to suggest how to submit tumor tissue in adenocarcinomas with mixed histological features for molecular testing. The objective of this study is to assess intra-tumoral heterogeneity in prominent driver mutations among different morphological patterns of lung adenocarcinoma, its implications on the future of molecular testing as well as its potential impact on patient management. Twenty-three cases of mixed histology lung adenocarcinoma resected between 2018 and 2023 were retrieved from the archives. H&E slides were reviewed to identify the predominant and second most predominant histological patterns. The morphologically different tumor areas were manually macro-dissected for DNA extraction. Next-Generation Sequencing with Ion AmpliSeq™ Cancer Hotspot Panel v2 (Thermo Fisher Scientific, USA). Thirteen cases showed the same pathological variant in both histological components tested. Three cases (13 %) exhibited disparities in the variants detected across the different histological patterns tested (p=0.025). The discrepant findings had a direct therapeutic impact in 4.3 % cases. Seven cases showed no pathogenic variants detected on either of the histological components tested. This study elucidates the presence of infrequent yet significant intra-tumoral heterogeneity in the molecular profiles of mixed histology adenocarcinomas, highlighting the need for guidelines directing tissue selection for molecular testing to avoid missed therapeutic opportunities and mitigate disease relapse.
Collapse
Affiliation(s)
| | - Jiani Chai
- Montefiore medical Center, Bronx, NY, USA
| | | | | | - Doctor Y Goldstein
- Montefiore medical Center, Bronx, NY, USA; Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
3
|
Wang Y, Zhang Y, Chen Y, Wang S, Liu W, Liu Z, Hu M. [ 18F]AlF-NOTA-PCP2: a novel PET/CT tracer for enhanced PD-L1 heterogeneity imaging and comparative analysis with [ 18F]AlF-NOTA-WL12 in glioblastoma xenografts. Eur J Nucl Med Mol Imaging 2024; 51:3161-3175. [PMID: 38713298 DOI: 10.1007/s00259-024-06743-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE The unsatisfactory efficacy of PD-L1 antibodies in glioblastoma (GBM) is largely due to the temporal and spatial heterogeneity of PD-L1 expression. Molecular imaging can enhance understanding of the tumor immune microenvironment and guide immunotherapy. However, highly sensitive imaging agents capable of effectively visualizing PD-L1 heterogeneity are limited. This study introduces a novel PET tracer, offering improved imaging of PD-L1 heterogeneity in GBM xenografts, with a comparative analysis to [18F]AlF-NOTA-WL12. METHODS [18F]AlF-NOTA-PCP2 was synthesized with high purity and its affinity for PD-L1 was characterized using surface plasmon resonance (SPR) and cell binding assays. Its specificity for PD-L1 was evaluated both in vitro using various cell lines and in vivo with GBM xenograft models in NOD/SCID mice. PET/CT imaging was conducted to evaluate the tracer's biodistribution, pharmacokinetics, and ability to quantify tumoral spatial heterogeneity of PD-L1 expression. A focused comparative analysis between [18F]AlF-NOTA-PCP2 and [18F]AlF-NOTA-WL12 was conducted, examining binding affinity, biodistribution, pharmacokinetics, and imaging effectiveness in GBM xenografts. Additionally, human radiation dosimetry estimates compared the safety profiles of both tracers. RESULTS [18F]AlF-NOTA-PCP2 demonstrated high radiochemical purity (> 95%) and a strong affinity for PD-L1, comparable to [18F]AlF-NOTA-WL12. In vitro and in vivo studies confirmed its specificity for PD-L1, with increased uptake in PD-L1 expressing cells and tumors. Toxicological profiles indicated no significant abnormalities in serum biochemical indicators or major organ tissues. MicroPET/CT imaging showed [18F]AlF-NOTA-PCP2's effectiveness in visualizing PD-L1 expression levels and spatial heterogeneity in GBM xenografts. Comparative studies revealed [18F]AlF-NOTA-PCP2's improved pharmacokinetic properties, including higher tumor-to-blood ratios and lower nonspecific liver uptake, as well as reduced radiation exposure compared to [18F]AlF-NOTA-WL12. CONCLUSION [18F]AlF-NOTA-PCP2 distinguishes itself as an exceptionally sensitive PET/CT tracer, adept at non-invasively and accurately quantifying PD-L1 expression and its spatial heterogeneity in tumors, especially in GBM. Its favorable pharmacokinetic properties, safety profile, and high affinity for PD-L1 highlight its potential for enhancing the precision of cancer immunotherapy and guiding individualized treatment strategies. While promising, its clinical translation, especially in brain imaging, necessitates further validation in clinical trials.
Collapse
Affiliation(s)
- Yong Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Yang Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Yunhao Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Shijie Wang
- Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Wei Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Zhiguo Liu
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Man Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
4
|
Ufimtseva EG, Gileva MS, Kostenko RV, Kozlov VV, Gulyaeva LF. Development of Ex Vivo Analysis for Examining Cell Composition, Immunological Landscape, Tumor and Immune Related Markers in Non-Small-Cell Lung Cancer. Cancers (Basel) 2024; 16:2886. [PMID: 39199657 PMCID: PMC11352364 DOI: 10.3390/cancers16162886] [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: 06/05/2024] [Revised: 07/11/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
NSCLC is a very aggressive solid tumor, with a poor prognosis due to post-surgical recurrence. Analysis of the specific tumor and immune signatures of NSCLC samples is a critical step in prognostic evaluation and management decisions for patients after surgery. Routine histological assays have some limitations. Therefore, new diagnostic tools with the capability to quickly recognize NSCLC subtypes and correctly identify various markers are needed. We developed a technique for ex vivo isolation of cancer and immune cells from surgical tumor and lung tissue samples of patients with NSCLC (adenocarcinomas and squamous cell carcinomas) and their examination on ex vivo cell preparations and, parallelly, on histological sections after Romanovsky-Giemsa and immunofluorescent/immunochemical staining for cancer-specific and immune-related markers. As a result, PD-L1 expression was detected for some patients only by ex vivo analysis. Immune cell profiling in the tumor microenvironment revealed significant differences in the immunological landscapes between the patients' tumors, with smokers' macrophages with simultaneous expression of pro- and anti-inflammatory cytokines, neutrophils, and eosinophils being the dominant populations. The proposed ex vivo analysis may be used as an additional diagnostic tool for quick examination of cancer and immune cells in whole tumor samples and to avoid false negatives in histological assays.
Collapse
Affiliation(s)
- Elena G. Ufimtseva
- Federal Research Center of Fundamental and Translational Medicine, 2 Timakova Street, 630060 Novosibirsk, Russia;
| | - Margarita S. Gileva
- V. Zelman Institute for the Medicine and Psychology, Novosibirsk State University, 1 Pirogova Street, 630090 Novosibirsk, Russia;
| | - Ruslan V. Kostenko
- Novosibirsk Regional Clinical Oncology Dispensary, 2 Plakhotny Street, 630108 Novosibirsk, Russia; (R.V.K.); (V.V.K.)
| | - Vadim V. Kozlov
- Novosibirsk Regional Clinical Oncology Dispensary, 2 Plakhotny Street, 630108 Novosibirsk, Russia; (R.V.K.); (V.V.K.)
- Faculty of General Medicine, Novosibirsk State Medical University, 52 Krasny Prospect, 630091 Novosibirsk, Russia
| | - Lyudmila F. Gulyaeva
- Federal Research Center of Fundamental and Translational Medicine, 2 Timakova Street, 630060 Novosibirsk, Russia;
- V. Zelman Institute for the Medicine and Psychology, Novosibirsk State University, 1 Pirogova Street, 630090 Novosibirsk, Russia;
| |
Collapse
|
5
|
Hummelink K, van der Noort V, Muller M, Schouten RD, van den Heuvel MM, Thommen DS, Smit EF, Meijer GA, Monkhorst K. Head-to-head comparison of composite and individual biomarkers to predict clinical benefit to PD-1 blockade in non-small cell lung cancer. PLoS One 2024; 19:e0293707. [PMID: 39083541 PMCID: PMC11290656 DOI: 10.1371/journal.pone.0293707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/15/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND The efficacy of PD-1 blocking agents in advanced NSCLC has shown prolonged effectiveness, but only in a minority of patients. Multiple biomarkers have been explored to predict treatment benefit, yet their combined performance remains inadequately examined. In this study, we assessed the combined predictive performance of multiple biomarkers in NSCLC patients treated with nivolumab. METHODS Pretreatment samples from 135 patients receiving nivolumab were used to evaluate the predictive performance of CD8 tumor-infiltrating lymphocytes (TILs), intratumoral (IT) localization of CD8 TILs, PD-1 high expressing TILs (PD1T TILs), CD3 TILs, CD20 B-cells, tertiary lymphoid structures (TLS), PD-L1 tumor proportion score (TPS) and the Tumor Inflammation score (TIS). Patients were randomly assigned to a training (n = 55) and validation cohort (n = 80). The primary outcome measure was Disease Control at 6 months (DC 6m) and the secondary outcome measure was DC at 12 months (DC 12m). RESULTS In the validation cohort, the two best performing composite biomarkers (i.e. CD8+IT-CD8 and CD3+IT-CD8) demonstrated similar or lower sensitivity (64% and 83%) and NPV (76% and 85%) compared to individual biomarkers PD-1T TILs and TIS (sensitivity: 72% and 83%, NPV: 86% and 84%) for DC 6m, respectively. Additionally, at 12 months, both selected composite biomarkers (CD8+IT-CD8 and CD8+TIS) demonstrated inferior predictive performance compared to PD-1T TILs and TIS alone. PD-1T TILs and TIS showed high sensitivity (86% and 100%) and NPV (95% and 100%) for DC 12m. PD-1T TILs could more accurately discriminate patients with no long-term benefit, as specificity was substantially higher compared to TIS (74% versus 39%). CONCLUSION Composite biomarkers did not show improved predictive performance compared to PD-1T TILs and TIS alone for both the 6- and 12-month endpoints. PD-1T TILs and TIS identified patients with DC 12m with high sensitivity. Patients with no long-term benefit to PD-1 blockade were most accurately identified by PD-1T TILs.
Collapse
Affiliation(s)
- Karlijn Hummelink
- Department of Pathology, Division of Diagnostic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Thoracic Oncology, Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Mirte Muller
- Department of Thoracic Oncology, Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Robert D. Schouten
- Department of Thoracic Oncology, Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michel M. van den Heuvel
- Department of Thoracic Oncology, Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daniela S. Thommen
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Egbert F. Smit
- Department of Thoracic Oncology, Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Gerrit A. Meijer
- Department of Pathology, Division of Diagnostic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kim Monkhorst
- Department of Pathology, Division of Diagnostic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
6
|
Wu J, Sun W, Zhang Y, Mao L, Ding T, Huang X, Lin D. Impact of platinum-based chemotherapy on the tumor mutational burden and immune microenvironment in non-small cell lung cancer with postoperative recurrence. Clin Transl Oncol 2024; 26:1738-1747. [PMID: 38421562 DOI: 10.1007/s12094-024-03397-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE To investigate the impact of platinum-based adjuvant chemotherapy on the immunotherapeutic biomarkers of postoperative recurrent tumors in non-small cell lung cancer (NSCLC). METHODS This study involved twenty-two cases of NSCLC, all of which underwent postoperative platinum-based chemotherapy, with matched surgical samples obtained from both their primary tumors (PTs) and recurrent tumors (RTs). Multiplex immunofluorescence was performed to assess the tumor proportion score (TPS) and immune cells (IC) on whole sections. Whole exon sequencing (WES) was conducted to investigate the tumor mutational burden (TMB) and tumor neoantigen burden (TNB). RESULTS Compared to paired PTs, RTs exhibited higher PD-L1 expression, along with a slightly elevated density of intratumoral PD-L1+ cells (p = 0.082) and an increased tumor proportion score (mean TPS: 40.51% vs. 28.56%, p = 0.046). Regarding IC infiltration, RTs generally demonstrated significantly lower CD8+ cytotoxic T lymphocyte (CTL) density (p = 0.011) and lower CD68+ macrophage density (p = 0.005), with a loss of tertiary lymphoid structure (TLS). The comparison between RTs and PTs revealed no significant differences in TMB (p = 0.795), whereas the count of TNB in RTs was notably increased compared to PTs (p = 0.033). Prognosis analysis indicated that a higher density of CD8+ CTLs in RTs was positively correlated with improved overall survival (OS). CONCLUSIONS In NSCLC patients with a history of postoperative platinum-based chemotherapy, the RTs demonstrated a trend towards increased PD-L1 expression and TMB/TNB, but a state of immunosuppression characterized by decreased ICs and loss of TLS, which may potentially impact the therapeutic benefits of immunotherapy.
Collapse
Affiliation(s)
- Jianghua Wu
- Department of Pathology, Peking University Cancer Hospital & Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), No. 52, Fu-Cheng Road, Beijing, 100142, China
| | - Wei Sun
- Department of Pathology, Peking University Cancer Hospital & Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), No. 52, Fu-Cheng Road, Beijing, 100142, China
| | - Yanhui Zhang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Luning Mao
- Department of Pathology, Peking University Cancer Hospital & Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), No. 52, Fu-Cheng Road, Beijing, 100142, China
| | - Tingting Ding
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Xiaozheng Huang
- Department of Pathology, Peking University Cancer Hospital & Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), No. 52, Fu-Cheng Road, Beijing, 100142, China
| | - Dongmei Lin
- Department of Pathology, Peking University Cancer Hospital & Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), No. 52, Fu-Cheng Road, Beijing, 100142, China.
| |
Collapse
|
7
|
Ito H, Yoshizawa A, Terada K, Nakakura A, Rokutan-Kurata M, Sugimoto T, Nishimura K, Nakajima N, Sumiyoshi S, Hamaji M, Menju T, Date H, Morita S, Bise R, Haga H. A Deep Learning-Based Assay for Programmed Death Ligand 1 Immunohistochemistry Scoring in Non-Small Cell Lung Carcinoma: Does it Help Pathologists Score? Mod Pathol 2024; 37:100485. [PMID: 38588885 DOI: 10.1016/j.modpat.2024.100485] [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: 08/10/2023] [Revised: 02/08/2024] [Accepted: 04/01/2024] [Indexed: 04/10/2024]
Abstract
Several studies have developed various artificial intelligence (AI) models for immunohistochemical analysis of programmed death ligand 1 (PD-L1) in patients with non-small cell lung carcinoma; however, none have focused on specific ways by which AI-assisted systems could help pathologists determine the tumor proportion score (TPS). In this study, we developed an AI model to calculate the TPS of the PD-L1 22C3 assay and evaluated whether and how this AI-assisted system could help pathologists determine the TPS and analyze how AI-assisted systems could affect pathologists' assessment accuracy. We assessed the 4 methods of the AI-assisted system: (1 and 2) pathologists first assessed and then referred to automated AI scoring results (1, positive tumor cell percentage; 2, positive tumor cell percentage and visualized overlay image) for final confirmation, and (3 and 4) pathologists referred to the automated AI scoring results (3, positive tumor cell percentage; 4, positive tumor cell percentage and visualized overlay image) while determining TPS. Mixed-model analysis was used to calculate the odds ratios (ORs) with 95% CI for AI-assisted TPS methods 1 to 4 compared with pathologists' scoring. For all 584 samples of the tissue microarray, the OR for AI-assisted TPS methods 1 to 4 was 0.94 to 1.07 and not statistically significant. Of them, we found 332 discordant cases, on which the pathologists' judgments were inconsistent; the ORs for AI-assisted TPS methods 1, 2, 3, and 4 were 1.28 (1.06-1.54; P = .012), 1.29 (1.06-1.55; P = .010), 1.28 (1.06-1.54; P = .012), and 1.29 (1.06-1.55; P = .010), respectively, which were statistically significant. For discordant cases, the OR for each AI-assisted TPS method compared with the others was 0.99 to 1.01 and not statistically significant. This study emphasized the usefulness of the AI-assisted system for cases in which pathologists had difficulty determining the PD-L1 TPS.
Collapse
Affiliation(s)
- Hiroaki Ito
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan; Department of Diagnostic Pathology, Nara Medical University, Nara, Japan.
| | - Kazuhiro Terada
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Akiyoshi Nakakura
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Tatsuhiko Sugimoto
- Department of Advanced Information Technology, Kyushu University, Fukuoka, Japan
| | - Kazuya Nishimura
- Department of Advanced Information Technology, Kyushu University, Fukuoka, Japan
| | - Naoki Nakajima
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan; Department of Diagnostic Pathology, Toyooka Hospital, Hyogo, Japan
| | - Shinji Sumiyoshi
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan; Department of Diagnostic Pathology, Tenri Hospital, Nara, Japan
| | - Masatsugu Hamaji
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Toshi Menju
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryoma Bise
- Department of Advanced Information Technology, Kyushu University, Fukuoka, Japan
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
8
|
Shigeta N, Murakami S, Yokose T, Isaka T, Shinada K, Nagashima T, Adachi H, Shigefuku S, Murakami K, Miura J, Kikunishi N, Watabe K, Saito H, Ito H. Comparison of SP263 and 22C3 pharmDx assays to test programmed death ligand-1 (PD-L1) expression in surgically resected non-small cell lung cancer. Thorac Cancer 2024; 15:1343-1349. [PMID: 38698758 PMCID: PMC11168908 DOI: 10.1111/1759-7714.15319] [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: 03/14/2024] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Atezolizumab, one of the immune checkpoint inhibitors, has been approved as an adjuvant treatment following resection and platinum-based chemotherapy in patients with stage II-IIIA non-small cell lung cancer with 1% or more programmed death ligand-1 (PD-L1) expression. The Food and Drug Administration (FDA) has approved SP263 as a companion diagnostic assay for adjuvant treatment with atezolizumab; however, in clinical practice, the 22C3 assay is most commonly used for advanced non-small cell lung cancer. Therefore, our study aimed to compare two PD-L1 assays, SP263 and 22C3, to evaluate whether 22C3 could replace SP263 when deciding whether to administer adjuvant atezolizumab. METHODS We retrospectively and prospectively analyzed 98 patients who underwent surgical resection at Kanagawa Cancer Center (Japan). An immunohistochemistry assay was performed for all the cases with both SP263 and 22C3. We statistically analyzed the concordance of PD-L1 expression between SP263 and 22C3 assays. RESULTS The concordance between the two assays using Cohen's kappa was κ = 0.670 (95% CI: 0.522-0.818) at the 1% cutoff and κ = 0.796 (95% CI: 0.639-0.954) at the 50% cutoff. The Spearman correlation coefficient of 0.874 (p < 0.01) indicated high concordance. PD-L1 expression with 22C3 resulted slightly higher than that with SP263. CONCLUSIONS This study showed a high concordance of PD-L1 expression with the SP263 and 22C3 assays. Further studies examining the therapeutic effects of adjuvant atezolizumab are required.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/surgery
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Lung Neoplasms/surgery
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Male
- Female
- Aged
- B7-H1 Antigen/metabolism
- Middle Aged
- Retrospective Studies
- Aged, 80 and over
- Prospective Studies
- Adult
- Biomarkers, Tumor/metabolism
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
Collapse
Affiliation(s)
- Naoko Shigeta
- Department of Thoracic SurgeryKanagawa Cancer CenterYokohamaJapan
| | - Shuji Murakami
- Department of Thoracic OncologyKanagawa Cancer CenterYokohamaJapan
| | | | - Tetsuya Isaka
- Department of Thoracic SurgeryKanagawa Cancer CenterYokohamaJapan
| | - Kanako Shinada
- Department of Thoracic OncologyKanagawa Cancer CenterYokohamaJapan
| | - Takuya Nagashima
- Department of Thoracic SurgeryKanagawa Cancer CenterYokohamaJapan
| | - Hiroyuki Adachi
- Department of Thoracic SurgeryKanagawa Cancer CenterYokohamaJapan
| | | | - Kotaro Murakami
- Department of Thoracic SurgeryKanagawa Cancer CenterYokohamaJapan
| | - Jun Miura
- Department of Thoracic SurgeryKanagawa Cancer CenterYokohamaJapan
| | | | - Kozue Watabe
- Department of Thoracic SurgeryKanagawa Cancer CenterYokohamaJapan
| | - Haruhiro Saito
- Department of Thoracic OncologyKanagawa Cancer CenterYokohamaJapan
| | - Hiroyuki Ito
- Department of Thoracic SurgeryKanagawa Cancer CenterYokohamaJapan
| |
Collapse
|
9
|
Wu J, Mao L, Lei W, Sun W, Yang X, Zhang Y, Huang X, Lin D. Genomic discordances and heterogeneous mutational burden, PD-L1 expression and immune infiltrates of non-small cell lung cancer metastasis. J Clin Pathol 2024:jcp-2023-209328. [PMID: 38307721 DOI: 10.1136/jcp-2023-209328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/21/2024] [Indexed: 02/04/2024]
Abstract
AIMS To investigate the genomic discordances and heterogeneous mutational burden, PD-L1 expression and immune cell (IC) infiltrates of non-small cell lung cancer (NSCLC) metastasis. METHODS Surgical samples from 41 cases of NSCLC with metastatic tumours (MTs) and paired primary tumours (PTs) were collected. PD-L1 expression and ICs were quantified using image-based immunohistochemistry profiling. Whole exome sequencing was employed to explore discrepancies in genomic characteristics, tumour mutational burden (TMB) and tumour neoantigen burden (TNB) in 28 cases. RESULTS Non-synonymous mutations in MTs were slightly more than in PTs, with only 42.34% of mutations shared between paired PTs and MTs. The heterogeneity of TMB showed no significant difference (p=0.785) between MTs and PTs, while TNB significantly increased in MTs (p=0.013). MTs generally exhibited a higher density of PD-L1+ cells and a higher tumour proportion score with a lower density of IC infiltrates. Subgroup analysis considering clinicopathological factors revealed that the heterogeneity of immune biomarkers was closely associated with the histology of lung adenocarcinoma, metastatic sites of extrapulmonary, time intervals and treatment history. Prognosis analysis indicated that a high density of CD8+ T cells was a low-risk factor, whereas a high density of PD-L1+ cells in MTs was a high-risk factor for cancer-related death in metastatic NSCLC. CONCLUSIONS The mutational burden, PD-L1 expression and IC infiltrates undergo changes during NSCLC metastasis, which may impact the immunotherapeutic benefits in patients with NSCLC with metastatic progression and should be monitored according to clinical scenarios.
Collapse
Affiliation(s)
- Jianghua Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Luning Mao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wanjun Lei
- Tianjin Medical Laboratory, BGI-Tianjin, BGI-Shenzhen, Tianjin, China
| | - Wei Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanhui Zhang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center of Cancer; Key Laboratory of Cancer Prevention and Therapy; Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Xiaozheng Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Dongmei Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
10
|
Liao S, Zhou M, Wang Y, Lu C, Yin B, Zhang Y, Liu H, Yin X, Song G. Emerging biomedical imaging-based companion diagnostics for precision medicine. iScience 2023; 26:107277. [PMID: 37520706 PMCID: PMC10371849 DOI: 10.1016/j.isci.2023.107277] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023] Open
Abstract
The tumor heterogeneity, which leads to individual variations in tumor microenvironments, causes poor prognoses and limits therapeutic response. Emerging technology such as companion diagnostics (CDx) detects biomarkers and monitors therapeutic responses, allowing identification of patients who would benefit most from treatment. However, currently, most US Food and Drug Administration-approved CDx tests are designed to detect biomarkers in vitro and ex vivo, making it difficult to dynamically report variations of targets in vivo. Various medical imaging techniques offer dynamic measurement of tumor heterogeneity and treatment response, complementing CDx tests. Imaging-based companion diagnostics allow for patient stratification for targeted medicines and identification of patient populations benefiting from alternative therapeutic methods. This review summarizes recent developments in molecular imaging for predicting and assessing responses to cancer therapies, as well as the various biomarkers used in imaging-based CDx tests. We hope this review provides informative insights into imaging-based companion diagnostics and advances precision medicine.
Collapse
Affiliation(s)
- Shiyi Liao
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Mengjie Zhou
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Youjuan Wang
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Chang Lu
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Baoli Yin
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Ying Zhang
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Huiyi Liu
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Xia Yin
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Guosheng Song
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
11
|
König D, Savic Prince S, Hayoz S, Zens P, Berezowska S, Jochum W, Stauffer E, Braunersreuther V, Trachsel B, Thierstein S, Mark M, Schmid S, Curioni-Fontecedro A, Addeo A, Opitz I, Guckenberger M, Früh M, Betticher DC, Ris HB, Stupp R, Rothschild SI, Bubendorf L, Pless M. Neoadjuvant treatment does not influence PD-L1 expression in stage III non-small-cell lung cancer: a retrospective analysis of tumor samples from the trials SAKK 16/96, 16/00, 16/01, and 16/14. ESMO Open 2023; 8:101595. [PMID: 37441877 PMCID: PMC10515281 DOI: 10.1016/j.esmoop.2023.101595] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND The inclusion of immune checkpoint inhibitors (ICIs) in the treatment of operable stage III non-small-cell lung cancer is becoming a new standard. Programmed death-ligand 1 (PD-L1) protein expression on tumor cells has emerged as the most important biomarker for sensitivity to ICIs targeting the programmed cell death protein 1 (PD-1)-PD-L1 axis. Little is known about the impact of neoadjuvant treatment on PD-L1 expression. PATIENTS AND METHODS We assessed PD-L1 expression by immunohistochemistry (Ventana SP263 assay) on tumor cells in treatment-naive diagnostic tumor samples and matched lung resections from patients with stage III non-small-cell lung cancer included in the Swiss Group for Clinical Cancer Research (SAKK) trials 16/96, 16/00, 16/01, and 16/14. All patients received neoadjuvant chemotherapy (CT) with cisplatin/docetaxel, either as single modality (CT), with sequential radiotherapy [chemoradiation therapy (CRT)] or with the PD-L1 inhibitor durvalumab (CT + ICI). RESULTS Overall, 132 paired tumor samples were analyzed from patients with neoadjuvant CT (n = 69), CRT (n = 33) and CT + ICI (n = 30). For CT and CRT, PD-L1 expression before and after neoadjuvant treatment did not differ significantly (Wilcoxon test, P = 0.94). Likewise, no statistically significant difference was observed between CT and CRT for PD-L1 expression after neoadjuvant treatment (P = 0.97). For CT + ICI, PD-L1 expression before and after neoadjuvant treatment also did not differ significantly (Wilcoxon test, P > 0.99). Event-free survival and overall survival for patients with downregulation or upregulation of PD-L1 expression after neoadjuvant treatment were similar. CONCLUSIONS In our cohort of patients neoadjuvant treatment did not influence PD-L1 expression, irrespective of the specific neoadjuvant treatment protocol. Dynamic change of PD-L1 expression did not correlate with event-free survival or overall survival.
Collapse
Affiliation(s)
- D König
- Department of Medical Oncology, University Hospital Basel, Basel.
| | - S Savic Prince
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel
| | - S Hayoz
- Swiss Group for Clinical Cancer Research, Bern
| | - P Zens
- Institute of Pathology, University of Bern, Bern; Graduate School for Health Science, University of Bern, Bern
| | - S Berezowska
- Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne
| | - W Jochum
- Institute of Pathology, Cantonal Hospital of St. Gallen, St. Gallen
| | | | | | - B Trachsel
- Swiss Group for Clinical Cancer Research, Bern
| | | | - M Mark
- Department of Oncology, Cantonal Hospital of Graubünden, Chur
| | - S Schmid
- Department of Medical Oncology, University Hospital of Bern (Inselspital), Bern
| | | | - A Addeo
- Department of Oncology/Hematology, University Hospital Geneva (HUG), Geneva
| | - I Opitz
- Department of Thoracic Surgery, University Hospital of Zurich, Zurich
| | - M Guckenberger
- Department of Radiation Oncology, University Hospital of Zurich, Zurich
| | - M Früh
- Department of Medical Oncology/Hematology, Cantonal Hospital of St. Gallen, St. Gallen; University of Bern, Bern
| | - D C Betticher
- Clinics of Medical Oncology, Cantonal Hospital of Fribourg (HFR), Fribourg
| | - H-B Ris
- Clinics for Thoracic Surgery, Hôpital du Valais, Sion, Switzerland
| | - R Stupp
- Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, USA; Department of Medical Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne
| | - S I Rothschild
- Department of Medical Oncology, University Hospital Basel, Basel; Department of Medical Oncology/Hematology, Cantonal Hospital Baden, Baden
| | - L Bubendorf
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel
| | - M Pless
- Department of Medical Oncology, Cantonal Hospital Winterthur, Winterthur, Switzerland
| |
Collapse
|
12
|
Anderson SA, Harbi D, Oramas Mogrovejo D, Floyd AD, Eltoum IE, Fatima H, Rosenblum F, Lora Gonzalez M, Lin D, Mackinnon AC, Siegal GP, Winokur T, Yalniz C, Huo L, Harada S, Huang X. PD-L1 (22C3) Expression Correlates with Clinical and Molecular Features of Lung Adenocarcinomas in Cytological Samples. Acta Cytol 2023; 67:507-518. [PMID: 37494911 DOI: 10.1159/000532036] [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: 05/18/2023] [Accepted: 07/16/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION PD-L1 expression is the most widely used predictive marker for immune checkpoint inhibitor (ICI) therapy in patients with lung adenocarcinoma. However, the current understanding of the association between PD-L1 expression and treatment response is suboptimal. A significant percentage of patients have only a cytological specimen available for clinical management. Therefore, it is relevant to examine the impact of molecular features on PD-L1 expression in cytological samples and how it might correlate with a therapeutic response. METHODS We evaluated patients diagnosed with adenocarcinoma of the lung who had both in-house targeted next-generation sequencing analysis and paired PD-L1 (22C3) immunohistochemical staining performed on the same cell blocks. We explored the association between molecular features and PD-L1 expression. In patients who underwent ICIs therapy, we assessed how a specific gene mutation impacted a therapeutic response. RESULTS 145 patients with lung adenocarcinoma were included in this study. PD-L1-high expression was found to be more common in pleural fluid than in other sample sites. Regional lymph node samples showed a higher proportion of PD-L1-high expression (29%) compared with lung samples (6%). The predictive value of PD-L1 expression was retained in cytological samples. Mutations in KRAS were also associated with a PD-L1-high expression. However, tumors with TP53 or KRAS mutations showed a lower therapy response rate regardless of the PD-L1 expression. CONCLUSION Cytological samples maintain a predictive value for PD-L1 expression in patients with lung adenocarcinoma as regards the benefit of ICI treatment. Specific molecular alterations additionally impact PD-L1 expression and its predictive value.
Collapse
Affiliation(s)
- Sarah A Anderson
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Djamel Harbi
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Diana Oramas Mogrovejo
- Department of Laboratory Medicine and Pathology, The University of Minnesota, Minneapolis, Minnesota, USA
| | - Antoinette D Floyd
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Isam-Eldin Eltoum
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Huma Fatima
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Frida Rosenblum
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Manuel Lora Gonzalez
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Diana Lin
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alexander C Mackinnon
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Gene P Siegal
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Thomas Winokur
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ceren Yalniz
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lei Huo
- Department of Pathology, Division of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shuko Harada
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xiao Huang
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
13
|
Zeng Y, Li QK, Roy S, Mills JC, Jin RU. Shared features of metaplasia and the development of adenocarcinoma in the stomach and esophagus. Front Cell Dev Biol 2023; 11:1151790. [PMID: 36994101 PMCID: PMC10040611 DOI: 10.3389/fcell.2023.1151790] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
Introduction: Plasticity is an inherent property of the normal gastrointestinal tract allowing for appropriate response to injury and healing. However, the aberrancy of adaptable responses is also beginning to be recognized as a driver during cancer development and progression. Gastric and esophageal malignancies remain leading causes of cancer-related death globally as there are limited early disease diagnostic tools and paucity of new effective treatments. Gastric and esophageal adenocarcinomas share intestinal metaplasia as a key precancerous precursor lesion.Methods: Here, we utilize an upper GI tract patient-derived tissue microarray that encompasses the sequential development of cancer from normal tissues to illustrate the expression of a set of metaplastic markers.Results: We report that in contrast to gastric intestinal metaplasia, which has traits of both incomplete and complete intestinal metaplasia, Barrett's esophagus (i.e., esophageal intestinal metaplasia) demonstrates hallmarks of incomplete intestinal metaplasia. Specifically, this prevalent incomplete intestinal metaplasia seen in Barrett's esophagus manifests as concurrent development and expression of both gastric and intestinal traits. Additionally, many gastric and esophageal cancers display a loss of or a decrease in these characteristic differentiated cell properties, demonstrating the plasticity of molecular pathways associated with the development of these cancers.Discussion: Further understanding of the commonalities and differences governing the development of upper GI tract intestinal metaplasias and their progression to cancer will lead to improved diagnostic and therapeutic avenues.
Collapse
Affiliation(s)
- Yongji Zeng
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Qing K. Li
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Sujayita Roy
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Jason C. Mills
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Departments of Medicine, Pathology and Immunology, and Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- *Correspondence: Jason C. Mills, ; Ramon U. Jin,
| | - Ramon U. Jin
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- *Correspondence: Jason C. Mills, ; Ramon U. Jin,
| |
Collapse
|
14
|
Layfield LJ, Zhang T, Esebua M. PD-L1 immunohistochemical testing: A review with reference to cytology specimens. Diagn Cytopathol 2023; 51:51-58. [PMID: 36053989 DOI: 10.1002/dc.25043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Immunotherapy based on disruption of the PD-1/PD-L1 axis is standard of care for many high stage malignancies including melanomas, non-small cell carcinomas of the lung, triple negative breast carcinomas, and squamous cell carcinomas of the head and neck. Eligibility for immunotherapy requires immunohistochemical assessment of PD-L1 expression. Currently, many high stage malignancies are diagnosed by cytology and cytologic material is the only specimen available for ancillary testing. Formal guidelines do not currently exist defining the optimal specimen type, antibody to be used or the best scoring system for cytologic material. Significant information has been published for PD-L1 testing of pulmonary specimens but much less data exists for the reproducibility, accuracy and best practices for material obtained from other body sites and types of malignancy. METHODS We searched the PubMed data base for manuscripts relating to PD-L1 testing of cytologic specimens. The search period was between 2016 and 2022. The search terms used were PD-L1, cytology, FNA, immunotherapy, immunohistochemistry, immunocytochemistry, cytology-histology correlation. Cross referencing techniques were used to screen for the most relevant manuscripts. The abstracts of these were then reviewed for final data collection and analysis. RESULTS A total of 86 studies were identified conforming to study relevancy. These were reviewed in their entirety by two authors (LJL, TZ) for extraction of data. The majority of studies involved pulmonary specimens (79) with three relating to PD-L1 testing of head and neck cytologic specimens and one each for PD-L1 testing of cytology specimens from melanomas, pancreas, pleural fluids, and triple negative breast carcinomas. While smears could be used, most studies found cell blocks optimal for testing. SUMMARY Currently, four drugs are approved for immunotherapy based on PD-L1 status. These drugs require specific antibody clones as well as scoring systems. Scoring systems and cut points vary with the type of neoplasm being treated. Cytology specimens from the lung, head and neck and melanomas can all be used for PD-L1 testing with good agreement with corresponding histology specimens.
Collapse
Affiliation(s)
- Lester J Layfield
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Tao Zhang
- Department of Surgical Pathology, M.D. Anderson, Houston, Texas, USA
| | - Magda Esebua
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
15
|
Hummelink K, van der Noort V, Muller M, Schouten RD, Lalezari F, Peters D, Theelen WS, Koelzer VH, Mertz KD, Zippelius A, van den Heuvel MM, Broeks A, Haanen JB, Schumacher TN, Meijer GA, Smit EF, Monkhorst K, Thommen DS. PD-1T TILs as a Predictive Biomarker for Clinical Benefit to PD-1 Blockade in Patients with Advanced NSCLC. Clin Cancer Res 2022; 28:4893-4906. [PMID: 35852792 PMCID: PMC9762332 DOI: 10.1158/1078-0432.ccr-22-0992] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/31/2022] [Accepted: 07/15/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Durable clinical benefit to PD-1 blockade in non-small cell lung cancer (NSCLC) is currently limited to a small fraction of patients, underlining the need for predictive biomarkers. We recently identified a tumor-reactive tumor-infiltrating T lymphocyte (TIL) pool, termed PD-1T TILs, with predictive potential in NSCLC. Here, we examined PD-1T TILs as biomarker in NSCLC. EXPERIMENTAL DESIGN PD-1T TILs were digitally quantified in 120 baseline samples from advanced NSCLC patients treated with PD-1 blockade. Primary outcome was disease control (DC) at 6 months. Secondary outcomes were DC at 12 months and survival. Exploratory analyses addressed the impact of lesion-specific responses, tissue sample properties, and combination with other biomarkers on the predictive value of PD-1T TILs. RESULTS PD-1T TILs as a biomarker reached 77% sensitivity and 67% specificity at 6 months, and 93% and 65% at 12 months, respectively. Particularly, a patient group without clinical benefit was reliably identified, indicated by a high negative predictive value (NPV) (88% at 6 months, 98% at 12 months). High PD-1T TILs related to significantly longer progression-free (HR 0.39, 95% CI, 0.24-0.63, P < 0.0001) and overall survival (HR 0.46, 95% CI, 0.28-0.76, P < 0.01). Predictive performance was increased when lesion-specific responses and samples obtained immediately before treatment were assessed. Notably, the predictive performance of PD-1T TILs was superior to PD-L1 and tertiary lymphoid structures in the same cohort. CONCLUSIONS This study established PD-1T TILs as predictive biomarker for clinical benefit to PD-1 blockade in patients with advanced NSCLC. Most importantly, the high NPV demonstrates an accurate identification of a patient group without benefit. See related commentary by Anagnostou and Luke, p. 4835.
Collapse
Affiliation(s)
- Karlijn Hummelink
- Department of Pathology, Division of Diagnostic Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Department of Thoracic Oncology, Division of Medical Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Vincent van der Noort
- Department of Biometrics, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mirte Muller
- Department of Thoracic Oncology, Division of Medical Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Robert D. Schouten
- Department of Thoracic Oncology, Division of Medical Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ferry Lalezari
- Department of Radiology, Division of Diagnostic Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Dennis Peters
- Core Facility Molecular Pathology and Biobanking, Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Willemijn S.M.E. Theelen
- Department of Thoracic Oncology, Division of Medical Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Viktor H. Koelzer
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Kirsten D. Mertz
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Alfred Zippelius
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Michel M. van den Heuvel
- Department of Thoracic Oncology, Division of Medical Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology and Biobanking, Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - John B.A.G. Haanen
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ton N. Schumacher
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Oncode Institute, Amsterdam, the Netherlands
| | - Gerrit A. Meijer
- Department of Pathology, Division of Diagnostic Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Egbert F. Smit
- Department of Thoracic Oncology, Division of Medical Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Kim Monkhorst
- Department of Pathology, Division of Diagnostic Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Corresponding Authors: Daniela S. Thommen, Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, the Netherlands. E-mail: ; and Kim Monkhorst,
| | - Daniela S. Thommen
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Corresponding Authors: Daniela S. Thommen, Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, the Netherlands. E-mail: ; and Kim Monkhorst,
| |
Collapse
|
16
|
Yang Z, Peng Y, Xu J, Chen P, Zhao Z, Cai Q, Li L, Tian H, Bai G, Liu L, Gao S, He J. PVR/TIGIT and PD-L1/PD-1 expression predicts survival and enlightens combined immunotherapy in lung squamous cell carcinoma. Transl Oncol 2022; 24:101501. [PMID: 35926369 PMCID: PMC9352965 DOI: 10.1016/j.tranon.2022.101501] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/03/2022] [Accepted: 07/27/2022] [Indexed: 11/25/2022] Open
Abstract
PVR/TIGIT and PD-L1/PD-1 axes play essential roles in tumor immune evasion and could be potential targets for combined immunotherapy. We aimed to evaluate the expression status of the above-mentioned immune markers in lung squamous cell carcinoma (LUSC), and investigate their survival impact and relevance with the immune microenvironment and clinicopathological features. We retrospectively collected specimens from 190 LUSC patients, who underwent pulmonary surgeries, and we performed immunohistochemistry assays of PVR, TIGIT, PD-L1, PD-1 and CD8. In our cohort, the positive rate of PVR was 85.8%, which was much higher than the positive rate of PD-L1 at 26.8%. A total of 32 (16.8%) patients demonstrated co-expression of PVR/PD-L1. High TIGIT density was correlated with positive PD-L1 expression, high PD-1 density, and high CD8 density (PD-L1, P=0.033; PD-1, P<0.001; CD8, P<0.001), and positive PVR expression was correlated with positive PD-L1 expression (P=0.046). High TIGIT density and high PVR/TIGIT expression were correlated with advanced TNM stage (TIGIT density, P=0.020; PVR/TIGIT expression, P=0.041). Patients with positive PVR expression, high TIGIT density, high PVR/TIGIT expression and PVR/PD-L1 co-expression exhibited a significantly worse prognosis (PVR, P=0.038; TIGIT, P=0.027; PVR/TIGIT, P=0.014; PVR/PD-L1, P=0.018). Multivariate analysis demonstrated that PVR/PD-L1 co-expression (Hazard ratio [HR], 1.756, 95% CI, 1.152-2.676, P=0.009) was an independent prognostic factor in LUSC patients. In conclusion, we demonstrated the expression status of PVR/TIGIT and PD-L1/PD-1 in LUSC. PVR/PD-L1 co-expression was an independent prognostic factor in LUSC patients and may serve as a potential predictive biomarker for dual-targeting immunotherapy.
Collapse
Affiliation(s)
- Zhenlin Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, China
| | - Yue Peng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, China; Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongtinanlu, Beijing, China
| | - Jiachen Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, China
| | - Ping Chen
- Department of Oncology, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, No. 166 Yulongxilu, Yancheng, Jiangsu Province, China
| | - Zhenshan Zhao
- Department of Thoracic Surgery, Kailuan General Hospital, No. 57 Xinhuadongdao, Tangshan, Hebei Province, China
| | - Qingyuan Cai
- School of Life Sciences, Peking University, No. 5 Yiheyuanlu, Beijing, China
| | - Lin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, China
| | - Guangyu Bai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, China
| | - Lei Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuannanli, Beijing, China.
| |
Collapse
|
17
|
Li X, Gu W, Liu Y, Wen X, Tian L, Yan S, Chen S. A novel quantitative prognostic model for initially diagnosed non-small cell lung cancer with brain metastases. Cancer Cell Int 2022; 22:251. [PMID: 35948974 PMCID: PMC9367158 DOI: 10.1186/s12935-022-02671-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/02/2022] [Indexed: 11/27/2022] Open
Abstract
Background The prognosis of non-small cell lung cancer (NSCLC) with brain metastases (BMs) had been researched in some researches, but the combination of clinical characteristics and serum inflammatory indexes as a noninvasive and more accurate model has not been described. Methods We retrospectively screened patients with BMs at the initial diagnosis of NSCLC at Sun Yat-Sen University Cancer Center. LASSO-Cox regression analysis was used to establish a novel prognostic model for predicting OS based on blood biomarkers. The predictive accuracy and discriminative ability of the prognostic model was compared to Adjusted prognostic Analysis (APA), Recursive Partition Analysis (RPA), and Graded Prognostic Assessment (GPA) using concordance index (C-index), time-dependent receiver operating characteristic (td-ROC) curve, Decision Curve Analysis(DCA), net reclassification improvement index (NRI), and integrated discrimination improvement index (IDI). Results 10-parameter signature's predictive model for the NSCLC patients with BMs was established according to the results of LASSO-Cox regression analysis. The C-index of the prognostic model to predict OS was 0.672 (95% CI = 0.609 ~ 0.736) which was significantly higher than APA,RPA and GPA. The td-ROC curve and DCA of the predictive model also demonstrated good predictive accuracy of OS compared to APA, RPA and GPA. Moreover, NRI and IDI analysis indicated that the prognostic model had improved prediction ability compared with APA, RPA and GPA. Conclusion The novel prognostic model demonstrated favorable performance than APA, RPA, and GPA for predicting OS in NSCLC patients with BMs. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02671-2.
Collapse
Affiliation(s)
- Xiaohui Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People's Republic of China.,Department of Clinical Laboratory Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Wenshen Gu
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, 510120, People's Republic of China
| | - Yijun Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People's Republic of China.,Department of Clinical Laboratory Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Xiaoyan Wen
- Department of Central Sterile Supply, Guanghua School of Stomatology, Affiliated Stomatological Hospital, Guangdong Province Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, 510055, People's Republic of China
| | - Liru Tian
- Research Center for Translational Medicine, the First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road 2, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Shumei Yan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People's Republic of China. .,Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Shulin Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People's Republic of China. .,Department of Clinical Laboratory Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China. .,Guangdong Esophageal Cancer Institute, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
18
|
Punekar SR, Shum E, Grello CM, Lau SC, Velcheti V. Immunotherapy in non-small cell lung cancer: Past, present, and future directions. Front Oncol 2022; 12:877594. [PMID: 35992832 PMCID: PMC9382405 DOI: 10.3389/fonc.2022.877594] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Many decades in the making, immunotherapy has demonstrated its ability to produce durable responses in several cancer types. In the last decade, immunotherapy has shown itself to be a viable therapeutic approach for non-small cell lung cancer (NSCLC). Several clinical trials have established the efficacy of immune checkpoint blockade (ICB), particularly in the form of anti-programmed death 1 (PD-1) antibodies, anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibodies and anti-programmed death 1 ligand (PD-L1) antibodies. Many trials have shown progression free survival (PFS) and overall survival (OS) benefit with either ICB alone or in combination with chemotherapy when compared to chemotherapy alone. The identification of biomarkers to predict response to immunotherapy continues to be evaluated. The future of immunotherapy in lung cancer continues to hold promise with the development of combination therapies, cytokine modulating therapies and cellular therapies. Lastly, we expect that innovative advances in technology, such as artificial intelligence (AI) and machine learning, will begin to play a role in the future care of patients with lung cancer.
Collapse
|
19
|
Li W, Bi Z, Wu J, Duan X, Pang L, Jing Y, Yin X, Cheng H. Effect of depression disorder on the efficacy and quality of life of first-line chemotherapy combined with immunotherapy in oncogene-driver negative NSCLC patients. Front Oncol 2022; 12:772102. [PMID: 35957880 PMCID: PMC9359314 DOI: 10.3389/fonc.2022.772102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 06/30/2022] [Indexed: 11/21/2022] Open
Abstract
Objective The current research was to assess the relevance between depression disorder and first-line chemotherapy combined with immunotherapy, quality of life in patients with oncogene-driver negative non-small cell cancer (NSCLC). Methods NSCLC patients (33 with depression disorder and 34 with no depression disorder) who was received first-line chemotherapy combined with immunotherapy performed Zung Self-rating Depression Scale (SDS) and European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30). Results The Progression-Free Survival (PFS) of depression disorder group survivors were lower than these of no depression disorder group survivors (HR, 0.352; 95% CI, 0.201-0.617; P<0.05). The statistical significant was revealed about the Objective Response Rate (ORR) and Disease Control Rate (DCR) in two groups (P<0.05). The quality of life scores of NSCLC patients in no depression disorder group was significantly higher after chemotherapy combined with immunotherapy, and manifested as 92.7 ± 28 vs. 76.3 ± 23.3 (t=8.317, P<0.05), and had a significant difference. Conclusion Depression disorder in oncogene-driver negative NSCLC patients influence the curative effect of chemotherapy combined with immunotherapy, and depression disorder was significantly negatively associated with quality of life following chemotherapy combined with immunotherapy.
Collapse
Affiliation(s)
- Wen Li
- Cancer Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ziran Bi
- Cancer Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Junxu Wu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xu Duan
- Cancer Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lulian Pang
- Cancer Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanyan Jing
- Cancer Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiangxiang Yin
- Cancer Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huaidong Cheng
- Cancer Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Huaidong Cheng,
| |
Collapse
|
20
|
Peng Y, Qiu B, Tan F, Xu J, Bie F, He H, Liu L, Tian H, Bai G, Zhou B, Li Y, Huai Q, Yang Z, Gao S. TIGIT/CD47 dual high expression predicts prognosis and is associated with immunotherapy response in lung squamous cell carcinoma. Thorac Cancer 2022; 13:2014-2023. [PMID: 35611464 PMCID: PMC9284170 DOI: 10.1111/1759-7714.14478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/01/2022] [Accepted: 05/02/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Recent studies indicated that T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT) and cluster of differentiation 47 (CD47) have emerged as new potential immunotherapy targets. However, the roles of TIGIT and CD47 in lung squamous cell carcinoma (LUSC) have not been fully illustrated. METHODS The specimens and clinicopathological information from 190 LUSC patients who underwent surgeries in our center were retrospectively collected. Immunohistochemical staining for TIGIT and CD47 was conducted. Transcriptional and clinical data of 479 LUSC were downloaded from The Cancer Genome Atlas (TCGA). RESULTS In the TCGA LUSC cohort, 142 (29.6%) cases were TIGIT/CD47 dual high expression at RNA level. The expression levels of TIGIT and CD47 were significantly correlated (p < 0.001). The proportions of patients with high TIGIT expression (p = 0.001) and high TIGIT/CD47 dual high expression (p = 0.049) were both higher in female cases. Advanced TNM stage (p = 0.006) and TIGIT/CD47 dual high expression (p = 0.047) were independent prognostic factors for LUSC. In the 190 LUSC cohort of our center, 75 (39.5%) cases were TIGIT/CD47 dual high expression at protein level. Cross-table analysis showed a correlation between TIGIT and CD47 expression. Older age (p = 0.001), advanced TNM stage (p < 0.001) and TIGIT/CD47 dual high expression (p = 0.046) were independent prognostic factors in our cohort. CONCLUSION We found that TIGIT and CD47 dual high expression was associated with poor prognosis in LUSC. We speculated that patients with dual high expression of CD47/TIGIT might be suitable for new target immunotherapy in the future.
Collapse
Affiliation(s)
- Yue Peng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Bin Qiu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Jiachen Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
- Guangdong Provincial People's Hospital/Guangdong Provincial Academy of Medical SciencesGuangdong Provincial Key Lab of Translational Medicine in Lung CancerGuangzhouChina
| | - Fenglong Bie
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Huayu He
- Department of Thoracic SurgeryPeking University Third HospitalBeijingChina
| | - Lei Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Guangyu Bai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Bolun Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Yuan Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Qilin Huai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Zhenlin Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| |
Collapse
|
21
|
SP142 PD-L1 Assays in Multiple Samples from the Same Patients with Early or Advanced Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:cancers14133042. [PMID: 35804813 PMCID: PMC9265054 DOI: 10.3390/cancers14133042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/15/2022] [Accepted: 06/19/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The IMpassion130 trial suggests that metastatic triple-negative breast cancer (TNBC) patients with PD-L1+ derived a clinical benefit from atezolizumab-combined treatment regardless of the sample collection time or origin. Therefore, if the PD-L1 test is positive at least once in multiple samples, the patient could have an opportunity to receive atezolizumab-based treatments. We aimed to know whether multiple PD-L1 testing might increase a rate of PD-L1+ in patients with TNBC. SP142 PD-L1 assays were performed in multiple samples from 77 patients in early TNBC. Multiple PD-L1 test using multiple samples raised the PD-L1+ rate more than a single biopsied sample test (68.8% vs. 37.6%, p = 0.00002). Among the group with metastatic TNBC treated with atezolizumab and nab-paclitaxel, PD-L1 assays were performed at least twice in 8/12 patients; 5/8 had heterogeneous results of PD-L1 assays. Consequently, a vigorous PD-L1 test using multiple samples was considered necessary in TNBC because a single test might be insufficient to represent the PD-L1 status. Abstract Purpose: The discernible PD-L1 staining of tumor-infiltrating lymphocytes occupying ≥ 1% of the tumor area is considered SP142 PD-L1 positive for atezolizumab, and the PD-L1 status of multiple samples within a single patient could be discrepant. In this study, we evaluated the PD-L1 status by using the SP142 clone in serially collected matched samples from the same individuals with early or metastatic triple-negative breast cancer (TNBC). Method: the SP142 PD-L1 assay was performed using biopsies and surgical specimens from 77 patients with early TNBC. Among these patients, 47 underwent upfront surgery, and 30 underwent neoadjuvant chemotherapy (NAC) between biopsy and surgery. PD-L1 assays were performed at least twice in 8/12 (66.7%) patients with metastatic TNBC treated with atezolizumab and nab-paclitaxel. Results: Of the 47 patients who underwent upfront surgery, 15/47 (31.9%) had PD-L1+ on biopsied samples. PD-L1+ rates in the biopsy and surgical specimens increased to 66.0% (33 of 47) after subsequent surgery. Similarly, in the 30 patients with residual invasive cancer who underwent neoadjuvant chemotherapy, the PD-L1+ rate increased from 46.6% at baseline to 74.2% after surgery. In the 77 patients with early TNBC, multiple PD-L1 testing in the biopsies and surgical specimens significantly increased the number of patients with PD-L1+ compared with the number of patients with PD-L1+ assessed with initial biopsy samples alone (68.8% vs. 37.6%; p = 0.00002). Among the metastatic TNBC patients, those with constant PD-L1+ over 1% positivity in multiple samples showed a response which was longer than 12 months. Conclusions: Our findings reveal the heterogeneous SP142 PD-L1 expression in TNBC and suggest that PD-L1 evaluation in baseline biopsy might be insufficient to represent the PD-L1 status of whole tumors. In TNBC, vigorous PD-L1 examination using multiple available tumor samples could identify more patients eligible for immune checkpoint blockade.
Collapse
|
22
|
Staudte S, Klinghammer K, Jurmeister PS, Jank P, Blohmer JU, Liebs S, Rhein P, Hauser AE, Tinhofer I. Multiparametric Phenotyping of Circulating Tumor Cells for Analysis of Therapeutic Targets, Oncogenic Signaling Pathways and DNA Repair Markers. Cancers (Basel) 2022; 14:cancers14112810. [PMID: 35681790 PMCID: PMC9179910 DOI: 10.3390/cancers14112810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Detection of circulating tumor cells (CTCs) has been established as an independent prognostic marker in solid cancer. In order to expand the clinical utility of this blood–based minimally invasive biomarker we established a protocol allowing multiparametric phenotyping of CTCs to analyze the expression levels of therapeutic target proteins. By applying this assay, we demonstrated intratumoral heterogeneity of PD–L1 expression in CTCs from head and neck cancer patients, an observation previously reported in tumor tissue specimens. We further verified the feasibility of applying the protocol to analyze the activation status of important oncogenic pathways and the extent of DNA repair following radiation. These promising preliminary results warrant further study and may lead to the implementation of this assay in clinical routine for improved treatment selection and monitoring. Abstract Detection of circulating tumor cells (CTCs) has been established as an independent prognostic marker in solid cancer. Multiparametric phenotyping of CTCs could expand the area of application for this liquid biomarker. We evaluated the Amnis® brand ImageStream®X MkII (ISX) (Luminex, Austin, TX, USA) imaging flow cytometer for its suitability for protein expression analysis and monitoring of treatment effects in CTCs. This was carried out using blood samples from patients with head and neck squamous cell carcinoma (n = 16) and breast cancer (n = 8). A protocol for negative enrichment and staining of CTCs was established, allowing quantitative analysis of the therapeutic targets PD–L1 and phosphorylated EGFR (phospho–EGFR), and the treatment response marker γH2AX as an indicator of radiation–induced DNA damage. Spiking experiments revealed a sensitivity of 73% and a specificity of 100% at a cut–off value of ≥3 CTCs, and thus confirmed the suitability of the ISX-based protocol to detect phospho–EGFR and γH2AX foci in CTCs. Analysis of PD–L1/–L2 in both spiked and patient blood samples further showed that assessment of heterogeneity in protein expression within the CTC population was possible. Further validation of the diagnostic potential of this ISX protocol for multiparametric CTC analysis in larger clinical cohorts is warranted.
Collapse
Affiliation(s)
- Stephanie Staudte
- Department of Radiooncology and Radiotherapy, Charité University Hospital, 10117 Berlin, Germany;
- German Cancer Consortium (DKTK) Partner Site, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence:
| | - Konrad Klinghammer
- Department of Hematology and Oncology, Charité University Hospital, 10117 Berlin, Germany;
- Charité Comprehensive Cancer Center (CCCC), Charité University Hospital, 10117 Berlin, Germany;
| | - Philipp Sebastian Jurmeister
- Institute of Pathology, Charité University Hospital, 10117 Berlin, Germany;
- Institute of Pathology, Ludwig Maximilians University Hospital Munich, 80337 Munich, Germany
| | - Paul Jank
- Institute of Pathology, Philipps-University Marburg and University-Hospital Marburg (UKGM), 35039 Marburg, Germany;
| | - Jens-Uwe Blohmer
- Breast Cancer Center, Charité University Hospital, 10117 Berlin, Germany;
| | - Sandra Liebs
- Charité Comprehensive Cancer Center (CCCC), Charité University Hospital, 10117 Berlin, Germany;
| | - Peter Rhein
- Luminex B.V., A DiaSorin Company, 5215 MV‘s-Hertogenbosch, The Netherlands;
| | - Anja E. Hauser
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, 10117 Berlin, Germany;
- Deutsches Rheuma-Forschungszentrum (DRFZ), Leibniz Association, 10117 Berlin, Germany
| | - Ingeborg Tinhofer
- Department of Radiooncology and Radiotherapy, Charité University Hospital, 10117 Berlin, Germany;
- German Cancer Consortium (DKTK) Partner Site, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
23
|
Ben Dori S, Aizic A, Zubkov A, Tsuriel S, Sabo E, Hershkovitz D. The risk of PD-L1 expression misclassification in triple-negative breast cancer. Breast Cancer Res Treat 2022; 194:297-305. [PMID: 35622241 PMCID: PMC9239943 DOI: 10.1007/s10549-022-06630-3] [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: 01/11/2022] [Accepted: 05/09/2022] [Indexed: 11/29/2022]
Abstract
Purpose Stratification of patients with triple-negative breast cancer (TNBC) for anti-PD-L1 therapy is based on PD-L1 expression in tumor biopsies. This study sought to evaluate the risk of PD-L1 misclassification. Methods We conducted a high-resolution analysis on ten surgical specimens of TNBC. First, we determined PD-L1 expression pattern distribution via manual segmentation and measurement of 6666 microscopic clusters of positive PD-L1 immunohistochemical staining. Then, based on these results, we generated a computer model to calculate the effect of the positive PD-L1 fraction, aggregate size, and distribution of PD-L1 positive cells on the diagnostic accuracy. Results Our computer-based model showed that larger aggregates of PD-L1 positive cells and smaller biopsy size were associated with higher fraction of false results (P < 0.001, P < 0.001, respectively). Additionally, our model showed a significant increase in error rate when the fraction of PD-L1 expression was close to the cut-off (error rate of 12.1%, 0.84%, and 0.65% for PD-L1 positivity of 0.5–1.5%, ≤ 0.5% ,and ≥ 1.5%, respectively, P < 0.0001). Interestingly, false positive results were significantly higher than false negative results (0.51–22.62%, with an average of 6.31% versus 0.11–11.36% with an average of 1.58% for false positive and false negative results, respectively, P < 0.05). Furthermore, heterogeneous tumors with different aggregate sizes in the same tumor, were associated with increased rate of false results in comparison to homogenous tumors (P < 0.001). Conclusion Our model can be used to estimate the risk of PD-L1 misclassification in biopsies, with potential implications for treatment decisions. Supplementary Information The online version contains supplementary material available at 10.1007/s10549-022-06630-3.
Collapse
Affiliation(s)
- Shani Ben Dori
- B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Asaf Aizic
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Dafna 5, 6492601, Tel Aviv, Israel
| | - Asia Zubkov
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Dafna 5, 6492601, Tel Aviv, Israel
| | - Shlomo Tsuriel
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Dafna 5, 6492601, Tel Aviv, Israel
| | - Edmond Sabo
- B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Institute of Pathology, Carmel Medical Center, Haifa, Israel
| | - Dov Hershkovitz
- Institute of Pathology, Tel-Aviv Sourasky Medical Center, Dafna 5, 6492601, Tel Aviv, Israel. .,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
24
|
Wu J, Sun W, Yang X, Wang H, Liu X, Chi K, Zhou L, Huang X, Mao L, Zhao S, Ding T, Meng B, Lin D. Heterogeneity of programmed death-ligand 1 expression and infiltrating lymphocytes in paired resected primary and metastatic non-small cell lung cancer. Mod Pathol 2022; 35:218-227. [PMID: 34493824 DOI: 10.1038/s41379-021-00903-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 11/09/2022]
Abstract
Metastatic tumors (MTs) may show different characteristics of the immune microenvironment from primary tumors (PTs) in non-small cell lung cancer (NSCLC). The heterogeneity of immune markers in metastatic NSCLC and its associated factors has not been well demonstrated. In this study, 64 surgically resected specimens of paired PTs and MTs were obtained from 28 patients with NSCLC. Multiplex immunofluorescence (mIF; panel including programmed death-ligand 1 (PD-L1), Cytokeratin, CD8, and CD68) was performed on whole sections. The heterogeneity of the immune contexture of PD-L1 expression, infiltrating lymphocytes, and immune-to-tumor cell distances was quantified via digital image analysis. In a quantitative comparison of MTs and corresponding PTs, MTs showed higher PD-L1 expression levels, lower density of CD8+ cytotoxic T lymphocytes (CTLs), and longer spatial distance between CTLs and tumor cells. Subgroup analysis, which associated clinical factors, revealed that the heterogeneity of immune markers was more obvious in extrapulmonary, metachronous, and treated MTs, while fewer differences were observed in intrapulmonary, synchronous, and untreated MTs. In particular, MTs showed significantly higher PD-L1 expression and lower lymphocyte infiltration in metastatic NSCLC with EGFR mutations. Prognosis analysis showed that an increased density of CD8+ CTLs in MTs was associated with better overall survival (OS). Therefore, significant discrepancies in PD-L1 expression and lymphocyte infiltration in metastatic NSCLC are most likely associated with temporal heterogeneity with a history of anti-treatment and correlated with EGFR mutations. The detection of immune markers in re-obtained metastatic specimens may be required for immunotherapy prediction in these patients with metastatic NSCLC.
Collapse
Affiliation(s)
- Jianghua Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China.,Department of Pathology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center of Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Wei Sun
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xin Yang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Haiyue Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xinying Liu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Kaiwen Chi
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lixin Zhou
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaozheng Huang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Luning Mao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shuai Zhao
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center of Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Tingting Ding
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center of Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Bin Meng
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center of Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Dongmei Lin
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
25
|
Li J, Ge S, Sang S, Hu C, Deng S. Evaluation of PD-L1 Expression Level in Patients With Non-Small Cell Lung Cancer by 18F-FDG PET/CT Radiomics and Clinicopathological Characteristics. Front Oncol 2021; 11:789014. [PMID: 34976829 PMCID: PMC8716940 DOI: 10.3389/fonc.2021.789014] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/30/2021] [Indexed: 12/22/2022] Open
Abstract
PURPOSE In the present study, we aimed to evaluate the expression of programmed death-ligand 1 (PD-L1) in patients with non-small cell lung cancer (NSCLC) by radiomic features of 18F-FDG PET/CT and clinicopathological characteristics. METHODS A total 255 NSCLC patients (training cohort: n = 170; validation cohort: n = 85) were retrospectively enrolled in the present study. A total of 80 radiomic features were extracted from pretreatment 18F-FDG PET/CT images. Clinicopathologic features were compared between the two cohorts. The least absolute shrinkage and selection operator (LASSO) regression was used to select the most useful prognostic features in the training cohort. Radiomics signature and clinicopathologic risk factors were incorporated to develop a prediction model by using multivariable logistic regression analysis. The receiver operating characteristic (ROC) curve was used to assess the prognostic factors. RESULTS A total of 80 radiomic features were extracted in the training dataset. In the univariate analysis, the expression of PD-L1 in lung tumors was significantly correlated with the radiomic signature, histologic type, Ki-67, SUVmax, MTV, and TLG (p< 0.05, respectively). However, the expression of PD-L1 was not correlated with age, TNM stage, and history of smoking (p> 0.05). Moreover, the prediction model for PD-L1 expression level over 1% and 50% that combined the radiomic signature and clinicopathologic features resulted in an area under the curve (AUC) of 0.762 and 0.814, respectively. CONCLUSIONS A prediction model based on PET/CT images and clinicopathological characteristics provided a novel strategy for clinicians to screen the NSCLC patients who could benefit from the anti-PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Jihui Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shushan Ge
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shibiao Sang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chunhong Hu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shengming Deng
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Nuclear Medicine, Suqian First Hospital, Suqian, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
26
|
Choi B, Kim DH. Multifunctional Nanocarriers-Mediated Synergistic Combination of Immune Checkpoint Inhibitor Cancer Immunotherapy and Interventional Oncology Therapy. ADVANCED NANOBIOMED RESEARCH 2021; 1:2100010. [PMID: 35663354 PMCID: PMC9162439 DOI: 10.1002/anbr.202100010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitor (ICI) cancer immunotherapies are becoming one of the standard therapies for cancer patients. However, ICI cancer immunotherapy's overall response rate is still moderate and even combinational ICI cancer immunotherapies are not showing significant improvement in therapeutic outcomes. Only a subset of patients responds to the therapy due to the resistance and ignorance to the ICI cancer immunotherapy. Following immune-related adverse events (irAEs) are also limiting the whole therapeutic regimens. New approaches that can increase the immunotherapeutic efficacy and reduce systemic irAEs are required. Recently, multifunctional nanocarriers, which can extend the half-life of ICIs and modulate tumor microenvironment (TME), have shown a substantial opportunity to enhance ICI cancer immunotherapies. Interventional oncology (IO) allowing simultaneous diagnosis, immunogenic loco-regional therapeutic delivery, and real-time monitoring of the treatment efficacy have advanced to demonstrate the effective conversion of TME. The use of multifunctional nanocarriers with the IO therapies amplify the image guidance capability and immunogenic therapeutic localization for the potential combinational ICI cancer immunotherapy. This article will discuss the emerging opportunity of multifunctional nanocarriers mediated synergistic combination of ICI cancer immunotherapy and IO local therapy.
Collapse
Affiliation(s)
- Bongseo Choi
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
- Department of Biomedical Engineering, McCormick School of Engineering, Evanston, IL 60208, USA
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
27
|
Fang Q, Wu Y, Du W, Zhang X, Chen D. Incidence and Prognostic Significance of PD-L1 Expression in High-Grade Salivary Gland Carcinoma. Front Oncol 2021; 11:701181. [PMID: 34513680 PMCID: PMC8427307 DOI: 10.3389/fonc.2021.701181] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/10/2021] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE PD-L1 is one of the predictors of immunotherapy efficacy. Our goal was to analyze its expression and prognostic significance in high-grade salivary gland carcinoma (SGC). METHODS PD-L1 expression was evaluated using paraffin-embedded specimens from patients with surgically treated high-grade SGC, and it was scored by the tumor proportion score (TPS), combined positive score (CPS), and immune cell (IC) score. Associations between clinicopathological variables, disease-free survival (DFS), overall survival (OS) and PD-L1 expression were assessed. RESULTS TPS≥1% occurred in 47 patients with an incidence of 43.1%, and it was significantly related to an advanced tumor stage. In patients with TPS<1%, TPS ranging from 1% to 20%, and TPS≥20%, the 5-year DFS rates were 36%, 26%, and 13%, respectively, and the difference was significant. In patients with TPS<1%, TPS ranging from 1% to 20%, and TPS≥20%, the 5-year OS rates were 49%, 24%, and 13%, respectively, and the difference was significant. CPS≥1 occurred in 87 patients with an incidence of 79.8%. IC scores of 0, 1, 2, and 3 were noted in 24 (22.0%), 37 (33.9%), 31 (28.4%), and 17 (15.6%) patients, respectively. Both CPS and IC scores had no impact on DFS or OS. CONCLUSIONS The expression of PD-L1 in tumor cells of high-grade SGCs was not uncommon, and it was significantly associated with tumor stage. PD-L1 expression in tumor cells rather than in immune cells indicated a poor prognosis.
Collapse
Affiliation(s)
- Qigen Fang
- Department of Head Neck and Thyroid, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | | | | | | | | |
Collapse
|
28
|
Raunig DL, Schmid AM, Miller CG, Walovitch RC, O'Connor M, Noever K, Hristova I, O'Neal M, Brueggenwerth G, Ford RR. Radiologists and Clinical Trials: Part 2: Practical Statistical Methods for Understanding and Monitoring Independent Reader Performance. Ther Innov Regul Sci 2021; 55:1122-1138. [PMID: 34244987 DOI: 10.1007/s43441-021-00317-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 06/18/2021] [Indexed: 01/02/2023]
Abstract
Though many clinical trials rely on medical image evaluations for primary or key secondary endpoints, the methods to monitor reader performance are all too often mired in the legacy use of adjudication rates. If misused, this simple metric can be misleading and sometimes entirely contradictory. Furthermore, attempts to overcome the limitations of adjudication rates using de novo or ad hoc methods often ignore well-established research conducted over the last half-century and can lead to inaccurate conclusions or variable interpretations. Underperforming readers can be missed, expert readers retrained, or worse, replaced. This paper aims to standardize reader performance evaluations using proven statistical methods. Additionally, these methods will describe how to discriminate between scenarios of concern and normal medical interpretation variability. Statistical methods are provided for inter-reader and intra-reader variability and bias, including the adjudicator's bias. Finally, we have compiled guidelines for calculating correct sample sizes, considerations for intra-reader memory recall, and applying alternative designs for independent readers.
Collapse
Affiliation(s)
- David L Raunig
- Takeda, 300 Massachusetts Ave, Cambridge, MA, 02139, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Yang Z, Peng Y, Guo W, Xu J, Li L, Tian H, Li R, Liu L, Tan F, Gao S, He J. PD-L1 and CD47 co-expression predicts survival and enlightens future dual-targeting immunotherapy in non-small cell lung cancer. Thorac Cancer 2021; 12:1743-1751. [PMID: 33979899 PMCID: PMC8169290 DOI: 10.1111/1759-7714.13989] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/17/2021] [Accepted: 04/17/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUD Recent studies have indicated that programmed cell death-ligand 1 (PD-L1) and cluster of differentiation 47 (CD47) play an essential role in tumor immune evasion and may serve as potential targets for combined immunotherapy. The aim of our study was to evaluate the PD-L1/CD47 expression status in lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD), and explore its survival impact and relevance with the immune microenvironment. METHODS The specimens from 190 LUSC and 240 LUAD patients who underwent intent-to-treat surgeries were retrospectively collected for immunohistochemistry assays of PD-L1, CD47, cluster of differentiation 8 (CD8), and cluster of differentiation 68 (CD68). RESULTS A total of 96 (22.3%) and 296 (68.8%) cases were positive for PD-L1 and CD47 expression, respectively, and 80 (18.6%) of them demonstrated the co-expression of PD-L1/CD47. The rate of PD-L1/CD47 co-expression was 23.7% in LUSC, significantly higher than the 14.6% in LUAD (p = 0.018). The median overall survival (OS) for all patients was 55.9 months (range 2.0-146.0 months). The univariate analysis showed that patients with positive CD47 expression (LUSC p = 0.003, LUAD p = 0.036) and PD-L1/CD47 co-expression (LUSC p = 0.023, LUAD p = 0.004) exhibited significantly worse prognosis. The multivariate analysis demonstrated that PD-L1/CD47 co-expression was an independent prognostic factor for OS (LUSC hazard ratio [HR] 1.922, 95% CI 1.245-2.969, p = 0.003; LUAD HR 1.549, 95% CI 1.015-2.364, p = 0.043). PD-L1/CD47 co-expression was associated with high CD8-positive T-lymphocyte density in LUSC (p = 0.004) and LUAD (p = 0.043), and with high CD68-positive macrophage density in LUSC (p = 0.026). CONCLUSIONS PD-L1/CD47 co-expression was an independent prognostic factor for LUSC and LUAD patients and may serve as a potential predictive biomarker for combined dual-targeting immunotherapy.
Collapse
Affiliation(s)
- Zhenlin Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yue Peng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiachen Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Renda Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lei Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
30
|
Tejerina E, Garca Tobar L, Echeveste JI, de Andrea CE, Vigliar E, Lozano MD. PD-L1 in Cytological Samples: A Review and a Practical Approach. Front Med (Lausanne) 2021; 8:668612. [PMID: 34026795 PMCID: PMC8139418 DOI: 10.3389/fmed.2021.668612] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/09/2021] [Indexed: 12/25/2022] Open
Abstract
With a growing number of predictive biomarkers needed to manage patients with non-small cell lung cancer (NSCLC), there has been a paradigm shift in care and handling of diagnostic samples. Among the various testing methods, immunohistochemistry (IHC) is the most cost- effective and widely available. Furthermore, over the past decade immunotherapy has emerged as one of the most promising cancer treatments. In this scenario IHC is the most used testing method available for PDL-1/PD1 immunotherapy. Several monoclonal antibodies targeting programmed death 1 (PD-1)/programmed death ligand-1 (PD-L1) pathways have been integrated into standard-of-care treatments of a wide range of cancer types, once provided evidence of PD-L1 expression in tumor cells by immunohistochemistry (IHC). Since currently available PD-L1 assays have been developed on formalin-fixed paraffin embedded (FFPE) histological specimens, a growing body of research is being dedicated to confirm the feasibility of applying PDL-1 assays also to cytological samples. Albeit promising results have been reported, several important issues still need to be addressed. Among these are the type of cytological samples, pre-analytical issues, cyto-histological correlation, and inter-observer agreement. This review briefly summarizes the knowledge of the role of cytopathology in the analysis of PD-L1 by immunocytochemistry (ICC) and future directions of cytopathology in the immunotherapy setting.
Collapse
Affiliation(s)
- Eva Tejerina
- Department of Pathology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Laura Garca Tobar
- Department of Pathology, Clinica University of Navarra, Pamplona, Spain
| | - Jos I Echeveste
- Department of Pathology, Clinica University of Navarra, Pamplona, Spain
| | | | - Elena Vigliar
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Mara D Lozano
- Department of Pathology, Clinica University of Navarra, Pamplona, Spain
| |
Collapse
|
31
|
Conde E, Hernandez S, Benito A, Caminoa A, Garrido P, Lopez-Rios F. Screening for ROS1 fusions in patients with advanced non-small cell lung carcinomas using the VENTANA ROS1 (SP384) Rabbit Monoclonal Primary Antibody. Expert Rev Mol Diagn 2021; 21:437-444. [PMID: 33899645 DOI: 10.1080/14737159.2021.1919512] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: The development of several ROS1 inhibitors means that the importance of accurately identifying ROS1-positive lung cancer patients has never been greater. Therefore, it is crucial that ROS1 testing assays become more standardized.Areas covered: Based on primary literature, combined with personal diagnostic and research experience, this review provide a pragmatic update on the use of the recently released VENTANA ROS1 (SP384) Rabbit Monoclonal Primary Antibody.Expert opinion: This assay provides high sensitivity, so it is an excellent analytical option when screening for ROS1 fusions in patients with advanced non-small cell lung carcinomas.
Collapse
Affiliation(s)
- Esther Conde
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HM Hospitales, CIBERONC, Madrid, Spain
| | - Susana Hernandez
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HM Hospitales, Madrid, Spain
| | - Amparo Benito
- Pathology, Ramon Y Cajal University Hospital, Madrid, Spain
| | | | - Pilar Garrido
- Medical Oncology, Ramon Y Cajal University Hospital, CIBERONC, Madrid, Spain
| | - Fernando Lopez-Rios
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HM Hospitales, CIBERONC, Madrid, Spain
| |
Collapse
|
32
|
Naso JR, Banyi N, Al-Hashami Z, Zhu J, Wang G, Ionescu DN, Ho C. Discordance in PD-L1 scores on repeat testing of non-small cell lung carcinomas. Cancer Treat Res Commun 2021; 27:100353. [PMID: 33770662 DOI: 10.1016/j.ctarc.2021.100353] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 12/11/2022]
Abstract
INTRODUCTION PD-L1 expression may be used as a biomarker predictive of non-small cell lung carcinoma (NSCLC) response to PD-L1 inhibitor treatment. Spatial and temporal heterogeneity in PD-L1 expression and variation in PD-L1 test interpretation may contribute to differences in PD-L1 test results between samples of the same patient's disease. METHODS Retrospective chart review identified 77 NSCLC patients with 22C3 PharmDx PD-L1 assays performed on two different tumor samples. Patients clinically suspected to have two separate primaries were excluded. PD-L1 test results in different score categories (<1%, 1-49% and ≥50%) were considered discordant. Clinical and pathologic factors associated with discordance were assessed. RESULTS 28 (36%) of the 77 cases had discordant PD-L1 scores between samples. Patients with an initial test result of 1-49% were most likely to have a discordant second test result. Specimen type (cytology, small biopsy or resection), specimen site (lung, lymph node, pleura/pleural effusion or distant metastasis), time between specimen collection, and treatment between specimen collection were not significantly associated with the rate of discordance. CONCLUSIONS Repeat PD-L1 testing of the same patient's NSCLC results frequently resulted in discordant test results, independent of whether the samples differed in clinical or pathologic factors. This discordance rate underscores the extent to which PD-L1 levels are heterogeneous and difficult to accurately represent with a single test value. Further study of the predictive value of PD-L1 scores in cases with discordant results is needed.
Collapse
Affiliation(s)
- Julia R Naso
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC, Canada, V6T 1Z7
| | - Norbert Banyi
- Department of Pathology, BC Cancer, 600 W 10th Ave, Vancouver, BC, Canada, V5Z 4E6
| | - Zamzam Al-Hashami
- Department of Medical Oncology, BC Cancer, 600 W 10th Ave, Vancouver, BC, Canada V5Z 4E6; Department of Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z7
| | - Jingyao Zhu
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Gang Wang
- Department of Pathology, BC Cancer, 600 W 10th Ave, Vancouver, BC, Canada, V5Z 4E6
| | - Diana N Ionescu
- Department of Pathology, BC Cancer, 600 W 10th Ave, Vancouver, BC, Canada, V5Z 4E6
| | - Cheryl Ho
- Department of Medical Oncology, BC Cancer, 600 W 10th Ave, Vancouver, BC, Canada V5Z 4E6; Department of Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z7.
| |
Collapse
|
33
|
PD-L1 as a biomarker of response to immune-checkpoint inhibitors. Nat Rev Clin Oncol 2021; 18:345-362. [PMID: 33580222 DOI: 10.1038/s41571-021-00473-5] [Citation(s) in RCA: 859] [Impact Index Per Article: 214.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 02/07/2023]
Abstract
Immune-checkpoint inhibitors targeting PD-1 or PD-L1 have already substantially improved the outcomes of patients with many types of cancer, although only 20-40% of patients derive benefit from these new therapies. PD-L1, quantified using immunohistochemistry assays, is currently the most widely validated, used and accepted biomarker to guide the selection of patients to receive anti-PD-1 or anti-PD-L1 antibodies. However, many challenges remain in the clinical use of these assays, including the necessity of using different companion diagnostic assays for specific agents, high levels of inter-assay variability in terms of both performance and cut-off points, and a lack of prospective comparisons of how PD-L1+ disease diagnosed using each assay relates to clinical outcomes. In this Review, we describe the current role of PD-L1 immunohistochemistry assays used to inform the selection of patients to receive anti-PD-1 or anti-PD-L1 antibodies, we discuss the various technical and clinical challenges associated with these assays, including regulatory issues, and we provide some perspective on how to optimize PD-L1 as a selection biomarker for the future treatment of patients with solid tumours.
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Radiogenomics is a growing field that has garnered immense interest over the past decade, owing to its numerous applications in the field of oncology and its potential value in improving patient outcomes. Current applications have only begun to delve into the potential of radiogenomics, and particularly in interventional oncology, there is room for development and increased value of these applications. RECENT FINDINGS The field of interventional oncology (IO) has seen valuable radiogenomic applications, from prediction of response to locoregional therapies in hepatocellular carcinoma to identification of genetic mutations in non-small cell lung cancer. Future directions that can increase the value of radiogenomics include applications that address tumor heterogeneity, predict immune responsiveness of tumors, and differentiate between oligoprogression and early widespread progression, among others. Radiogenomics, whether in terms of methodologies or applications, is still in the early stages of development and far from maturation. Future applications, particularly in the field of interventional oncology, will allow realization of its full potential.
Collapse
|
35
|
Nicoś M, Krawczyk P, Crosetto N, Milanowski J. The Role of Intratumor Heterogeneity in the Response of Metastatic Non-Small Cell Lung Cancer to Immune Checkpoint Inhibitors. Front Oncol 2020; 10:569202. [PMID: 33344229 PMCID: PMC7746867 DOI: 10.3389/fonc.2020.569202] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) represent one of the most promising therapeutic approaches in metastatic non-small cell lung cancer (M-NSCLC). Unfortunately, approximately 50–75% of patients do not respond to this treatment modality. Intratumor heterogeneity (ITH) at the genetic and phenotypic level is considered as a major cause of anticancer therapy failure, including resistance to ICIs. Recent observations suggest that spatial heterogeneity in the composition and spatial organization of the tumor microenvironment plays a major role in the response of M-NSCLC patients to ICIs. In this mini review, we first present a brief overview of the use of ICIs in M-NSCLC. We then discuss the role of genetic and non-genetic ITH on the efficacy of ICIs in patients with M-NSCLC.
Collapse
Affiliation(s)
- Marcin Nicoś
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland.,Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| | - Nicola Crosetto
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Janusz Milanowski
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
36
|
Nimmagadda S. Quantifying PD-L1 Expression to Monitor Immune Checkpoint Therapy: Opportunities and Challenges. Cancers (Basel) 2020; 12:cancers12113173. [PMID: 33137949 PMCID: PMC7692040 DOI: 10.3390/cancers12113173] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Malignant cells hijack the regulatory roles of immune checkpoint proteins for immune evasion and survival. Therapeutics blocking those proteins can restore the balance of the immune system and lead to durable responses in cancer patients. Although a subset of patients derive benefit, there are few non-invasive technologies to guide and monitor those therapies to improve success rates. This is a review of the advancements in non-invasive methods for quantification of immune checkpoint protein programmed death ligand 1 expression, a biomarker detected by immunohistochemistry and widely used for guiding immune checkpoint therapy. Abstract Therapeutics targeting programmed death ligand 1 (PD-L1) protein and its receptor PD-1 are now dominant players in restoring anti-tumor immune responses. PD-L1 detection by immunohistochemistry (IHC) is emerging as a reproducible biomarker for guiding patient stratification for those therapies in some cancers. However, PD-L1 expression in the tumor microenvironment is highly complex. It is upregulated by aberrant genetic alterations, and is highly regulated at the transcriptional, posttranscriptional, and protein levels. Thus, PD-L1 IHC is inadequate to fully understand the relevance of PD-L1 levels in the whole body and their dynamics to improve therapeutic outcomes. Imaging technologies could potentially assist in meeting that need. Early clinical investigations show promising results in quantifying PD-L1 expression in the whole body by positron emission tomography (PET). Within this context, this review summarizes advancements in regulation of PD-L1 expression and imaging agents, and in PD-L1 PET for drug development, and discusses opportunities and challenges presented by these innovations for guiding immune checkpoint therapy (ICT).
Collapse
Affiliation(s)
- Sridhar Nimmagadda
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; ; Tel.: +1-410-502-6244; Fax: +1-410-614-3147
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Bloomberg–Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
37
|
Hua P, Zhang Y, Jin C, Zhang G, Wang B. Integration of gene profile to explore the hub genes of lung adenocarcinoma: A quasi-experimental study. Medicine (Baltimore) 2020; 99:e22727. [PMID: 33120770 PMCID: PMC7581154 DOI: 10.1097/md.0000000000022727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Lung cancer is a leading cause of morbidity diseases worldwide, but the key mechanisms of lung cancer remain elusive. This study aims to integrate of GSE 118370 and GSE 32863 profile and identify the key genes and pathway involved in human lung adenocarcinoma. METHODS R software (RStudio, Version info: R 3.2.3, Forrester, USA) were utilized to find the differentially expressed genes. All the differentially expressed genes were analyzed by gene ontology, kyoto encyclopedia of genes and genomes. Protein-protein interaction networks were constructed by STRING database and analyzed by Cytohubber and Module. The cancer genome atlas database was used to verification the expression of hub genes. Quantitative reverse transcription-PCR was used to verify the bio-information results. RESULTS Sixty-four lung adenocarcinoma and 64 adjacent normal tissues were used for integration analysis. Five hundred ninety-nine co-expression genes were locked. Biological processes mainly enriched in angiogenesis. Cellular component focused on extracellular exosome and molecular function aimed on protein disulfide isomerase activity. Cytohubber analysis showed that GNG11, FPR2, P4HB, PIK3R1, CDC20, ADCY4, TIMP1, IL6, CXC chemokine ligand (CXCL)12, and GAS6 acted as the hub genes during lung adenocarcinoma. Module analysis presented Chemokine signaling pathway was a key pathway. Quantitative reverse transcription-PCR showed that the expression level of GNG11, FPR2, PIK3R1, ADCY4, IL6, CXCL12, and GAS6 were significantly decreased and P4HB, CDC20 and TIMP1 were increased in human adenocarcinoma tissues (P < .05). The cancer genome atlas online analysis showed GNG11 was not associated with survival. CONCLUSIONS This study firstly reported GNG11 acting as a hub gene in adenocarcinoma. GNG11 could be used as a biomarker for human adenocarcinoma. Chemokine signaling pathway might play important roles in lung adenocarcinoma.
Collapse
|
38
|
Ye M, Huang D, Zhang Q, Weng W, Tan C, Qin G, Jiang W, Sheng W, Wang L. Heterogeneous programmed death-ligand 1 expression in gastric cancer: comparison of tissue microarrays and whole sections. Cancer Cell Int 2020; 20:186. [PMID: 32489322 PMCID: PMC7247123 DOI: 10.1186/s12935-020-01273-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 05/16/2020] [Indexed: 12/11/2022] Open
Abstract
Background Programmed death-ligand 1 (PD-L1) expression determines the eligibility for anti-PD-1 treatment in patients with advanced gastric cancer, but evidence indicates that PD-L1 staining is heterogeneous. Patients who are ineligible for radical surgery could be tested for PD-L1 expression with biopsy staining, but it is unclear if a small biopsy is representative of the PD-L1 status of the whole tumor. The aim of our study was to determine how many biopsy specimens are needed to accurately reflect the objective status of PD-L1 expression in whole sections. Methods We built tissue microarrays (TMAs) as substitutes for core biopsies, collecting 6 cores per case from 152 gastric cancer specimens. All of the slides and TMAs underwent PD-L1 immunohistochemical staining, and PD-L1 expression in at least 1% of tumor cells or immune cells was defined as positive. Results It was necessary to randomly select multiple cores from TMAs to reach a suitable agreement rate (> 90%) and Cohen’s κ value (> 0.8) between TMAs and whole sections. We defined the PD-L1 staining status from the whole section as the standard. The evaluation of five randomly selected cores from TMAs agreed well with the evaluation of whole sections. The sensitivity, specificity and the area under the curve (AUC) of the receiver-operating characteristic (ROC) were 0.93, 0.92, and 0.922 (95% confidence interval (CI) 0.863–0.982), respectively. Conclusions We conclude that PD-L1 expression among TMA samples had different degrees of relevance to the corresponding surgical specimens, which indicates that at least five biopsies might be necessary to characterize patients taking anti-PD-1 treatment.
Collapse
Affiliation(s)
- Min Ye
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Qiongyan Zhang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China
| | - Weiwei Weng
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Cong Tan
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Guangqi Qin
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China
| | - Wenhua Jiang
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Lei Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| |
Collapse
|
39
|
Association of tumour and stroma PD-1, PD-L1, CD3, CD4 and CD8 expression with DCB and OS to nivolumab treatment in NSCLC patients pre-treated with chemotherapy. Br J Cancer 2020; 123:392-402. [PMID: 32433601 PMCID: PMC7403301 DOI: 10.1038/s41416-020-0888-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 03/30/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors are most beneficial in patients with high tumour PD-L1 expression. However, the use of PD-L1 expression is not straightforward. We investigated PD-L1 expression and immune cell (IC) infiltrates in non-small-cell lung cancer (NSCLC) patients treated with nivolumab. METHODS Tumour tissue specimens of 139 NSCLC patients were scored for tumour/stromal PD-L1 and various IC expression markers, and associated with durable clinical benefit (DCB) and overall survival (OS). RESULTS Median OS was higher for patients with high stromal infiltration of CD8+ ICs (9.0 months) compared with patients with low and intermediate infiltration (both 5.0 months, p = 0.035) and for patients with high infiltration of stromal CD4+ ICs (9.0 months) compared with patients with low and intermediate infiltration (both 5.0 months, p = 0.010) and this was confirmed in the validation cohort. Post hoc analyses showed that biopsies taken after the last line of chemotherapy (ACT) were predictive for DCB and OS, whereas samples obtained before the last line of chemotherapy (BCT) were not. CONCLUSIONS Stromal infiltration of ICs can predict response to PD-1-directed immunotherapy in NSCLC patients. Interestingly, we found differences in the predictive value of IC markers between the ACT and BCT biopsies, suggesting that chemotherapy might influence the immune microenvironment.
Collapse
|
40
|
Xu Y, Xiao Y, Luo C, Liu Q, Wei A, Yang Y, Zhao L, Wang Y. Blocking PD-1/PD-L1 by an ADCC enhanced anti-B7-H3/PD-1 fusion protein engages immune activation and cytotoxicity. Int Immunopharmacol 2020; 84:106584. [PMID: 32422527 DOI: 10.1016/j.intimp.2020.106584] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/28/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022]
Abstract
Antibody therapy based on PD-1/PD-L1 blocking or ADCC effector has produced significant clinical benefit for cancer patients. We generated a novel anti-B7-H3 antibody (07B) and engineered the Fc fragment to enhance ADCC. To improve efficacy and tumor selectivity, we developed anti-B7-H3/PD-1 bispecific fusion proteins that simultaneously engaged tumor associate marker B7-H3 and immune suppressing ligand PD-L1 as well as enhanced ADCC to promote potent and highly selective tumor killing. Fusion proteins were designed by fusing human PD-1 extra domain to 07B in four different formats and showed good binding capacity to both targets. Indeed, the affinity of fusion proteins to B7-H3 is over 10,000 fold higher compared to that of the analogous PD-L1 and the blocking of fusion proteins to PD-L1 was worse but it greatly enhanced when bound to B7-H3, thus achieving directly PD-L1-blockade to B7-H3-expressing tumor cells. Importantly, IL-2 production was enhanced by fusion proteins from staphylococcal enterotoxin B (SEB) stimulated PBMC. Similarly, cytokines induced by fusion proteins was enhanced when co-cultured with stimulated CD8+ T cells and B7-H3/PD-L1 transfected raji cells. Additionally, fusion proteins improved activation to CD16a by Fc modification and delivered selective cytotoxicity to B7-H3 expressing tumor cells. In conclusion, fusion proteins blocked the PD-1/PD-L1 signal pathway and significantly increased potency of ADCC in a B7-H3-directed manner, thereby selectively activating CD8+ T cells and enhancing natural killing towards tumor. This novel fusion protein with its unique targeting preference may be useful to enhance efficacy and safety of immunotherapy for B7-H3-overexpressing malignancies.
Collapse
Affiliation(s)
- Yao Xu
- Sanhome-CPU Joint Laboratory, China Pharmaceutical University, Nanjing 211198, PR China; Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yang Xiao
- Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China
| | - Cheng Luo
- Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China
| | - Qingxia Liu
- Sanhome-CPU Joint Laboratory, China Pharmaceutical University, Nanjing 211198, PR China; Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China
| | - Aiqi Wei
- Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China
| | - Yang Yang
- Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China
| | - Liwen Zhao
- Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China
| | - Yong Wang
- Sanhome-CPU Joint Laboratory, China Pharmaceutical University, Nanjing 211198, PR China; Sanhome R&D Centre, Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing 221116, PR China.
| |
Collapse
|
41
|
Stovgaard ES, Bokharaey M, List-Jensen K, Roslind A, Kümler I, Høgdall E, Nielsen D, Balslev E. PD-L1 diagnostics in the neoadjuvant setting: implications of intratumoral heterogeneity of PD-L1 expression in triple negative breast cancer for assessment in small biopsies. Breast Cancer Res Treat 2020; 181:553-560. [PMID: 32358635 DOI: 10.1007/s10549-020-05655-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 04/25/2020] [Indexed: 02/04/2023]
Abstract
PURPOSE PD-L1 expression is a predictive biomarker for anti-PD-L1 immunotherapy in triple negative breast cancer (TNBC). In the neoadjuvant setting, immunohistochemical (IHC) evaluation of PD-L1 expression can only be performed on small tissue biopsies. In our study we investigated heterogeneity of PD-L1 expression in TNBC, and how reliably PD-L1 expression in small tissue samples reflects PD-L1 expression in larger tumor sections in TNBC. METHODS Tissue microarrays (TMAs) were constructed from surgical specimens of 110 patients with TNBC. TMAs contained 4 cores (1 mm in diameter) per patient. To evaluate PD-L1 expression, TMAs were stained with PD-L1 IHC 22C3 PharmDx. Single-core PD-L1 expression was compared to overall PD-L1 expression of each patient's tumor, to ascertain how often small samples of tumor tissue show the same PD-L1 expression as larger tumor samples. RESULTS Our study found substantial heterogeneity of PD-L1 expression between different TMA cores from the same patient. Heterogeneity was greater in immune cells (ICs) than in tumor cells, in large part due to the uneven distribution of ICs in the tumor. For IC PD-L1 expression, we found that sensitivity can be as low as 0.81 for detecting PD-L1 expression at the 1% threshold most commonly used in breast cancer. Negative predictive value for ICs was 0.7. CONCLUSIONS There is substantial heterogeneity of PD-L1 expression between small tissue samples from the same TNBC tumor, especially for IC expression. This poses challenges for evaluation of PD-L1 expression in the neoadjuvant setting. Negative biopsies should prompt further investigation, and multiple biopsies might be necessary.
Collapse
MESH Headings
- B7-H1 Antigen/metabolism
- Biomarkers, Tumor/metabolism
- Carcinoma, Ductal, Breast/diagnosis
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/surgery
- Carcinoma, Lobular/diagnosis
- Carcinoma, Lobular/metabolism
- Carcinoma, Lobular/surgery
- Female
- Follow-Up Studies
- Humans
- Neoadjuvant Therapy
- Patient Selection
- Prognosis
- Retrospective Studies
- Triple Negative Breast Neoplasms/classification
- Triple Negative Breast Neoplasms/diagnosis
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/surgery
Collapse
Affiliation(s)
- E S Stovgaard
- Department of Pathology, Herlev and Gentofte Hospital, University of Copenhagen, Borgmester Ib Juuls vej 1, 2730, Herlev, Denmark.
| | - M Bokharaey
- Department of Pathology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - K List-Jensen
- Department of Pathology, Hvidovre Hospital, Kettegård Allé 30, 2650, Hvidovre, Denmark
| | - A Roslind
- Department of Pathology, Herlev and Gentofte Hospital, University of Copenhagen, Borgmester Ib Juuls vej 1, 2730, Herlev, Denmark
| | - I Kümler
- Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Borgmester Ib Juuls vej 1, 2730, Herlev, Denmark
| | - E Høgdall
- Department of Pathology, Herlev and Gentofte Hospital, University of Copenhagen, Borgmester Ib Juuls vej 1, 2730, Herlev, Denmark
| | - D Nielsen
- Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Borgmester Ib Juuls vej 1, 2730, Herlev, Denmark
| | - E Balslev
- Department of Pathology, Herlev and Gentofte Hospital, University of Copenhagen, Borgmester Ib Juuls vej 1, 2730, Herlev, Denmark
| |
Collapse
|
42
|
Thunnissen E, Kerr KM, Dafni U, Bubendorf L, Finn SP, Soltermann A, Biernat W, Cheney R, Verbeken E, Warth A, Marchetti A, Speel EJM, Pokharel S, Quinn AM, Monkhorst K, Navarro A, Madsen LB, Tsourti Z, Geiger T, Kammler R, Peters S, Stahel RA. Programmed death-ligand 1 expression influenced by tissue sample size. Scoring based on tissue microarrays' and cross-validation with resections, in patients with, stage I-III, non-small cell lung carcinoma of the European Thoracic Oncology Platform Lungscape cohort. Mod Pathol 2020; 33:792-801. [PMID: 31740722 DOI: 10.1038/s41379-019-0383-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 11/08/2022]
Abstract
PD-L1, as assessed by immunohistochemistry, is a predictive biomarker for immuno-oncology treatment in lung cancer. Different scoring methods have been used to assess its status, resulting in a wide range of positivity rates. We use the European Thoracic Oncology Platform Lungscape non-small cell lung carcinoma cohort to explore this issue. PD-L1 expression was assessed via immunohistochemistry on tissue microarrays (up to four cores per case), using the DAKO 28-8 immunohistochemistry assay, following a two-round external quality assessment procedure. All samples were analyzed under the same protocol. Cross-validation of scoring between tissue microarray and whole sections was performed in 10% randomly selected samples. Cutoff points considered: ≥1, 50 (primarily), and 25%. At the two external quality assessment rounds, tissue microarray scoring agreement rates between pathologists were: 73% and 81%. There were 2008 cases with valid immunohistochemistry tissue microarray results (50% all cores evaluable). Concordant cases at 1, 25, and 50% were: 85, 91, and 93%. Tissue microarray core results were identical for 70% of cases. Sensitivity of the tissue microarray method for 1, 25, and 50% was: 80, 78, and 79% (specificity: 90, 95, 98%). Complete agreement between tissue microarrays and whole sections was achieved for 60% of the cases. Highest sensitivity rates for 1% and 50% cutoffs were detected for higher number of cores. Underestimation of PD-L1 expression on small samples is more common than overestimation. We demonstrated that classification of PD-L1 on small biopsy samples does not represent the overall expression of PD-L1 in all non-small cell cancer carcinoma cases, although the majority of cases are 'correctly' classified. In future studies, sampling more and larger biopsies, recording the biopsy size and tumor load may permit further refinement, increasing predictive accuracy.
Collapse
Affiliation(s)
- Erik Thunnissen
- Department of Pathology, Amsterdam University Medical Center, location VU University Medical Center, Amsterdam, Netherlands.
| | - Keith M Kerr
- Department of Pathology, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Urania Dafni
- Froniter Science Foundation-Hellas & National and Kapodistrian University of Athens, Athens, Greece
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Stephen P Finn
- Department of Histopathology, St James's Hospital and Trinity College, Dublin, Ireland
| | - Alex Soltermann
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Wojciech Biernat
- Department of Pathomorphology, Medical University of Gdansk, Gdansk, Poland
| | - Richard Cheney
- Department of Pathology, State University of New York at Buffalo, Buffalo, NY, USA
| | - Erik Verbeken
- Department of Pathology, University Hospital KU Leuven, Leuven, Belgium
| | - Arne Warth
- Department of Pathology, Universitätsklinikum Heidelberg, Heidelberg, Germany
- Institute of Pathology, Cytopathology, and Molecular Pathology MVZ UEGP Giessen, Wetzlar, Limburg, Germany
| | - Antonio Marchetti
- Center of Predicitve Predictive Molecular Medicine, CeSI, University of Chieti-Pescara, Chieti, Italy
| | - Ernst-Jan M Speel
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Saraswati Pokharel
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Anne Marie Quinn
- Wythenshawe Hospital, Department of Histopathology, Manchester University NHS Foundation Trust, Manchester, UK
| | - Kim Monkhorst
- Division of Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Atilio Navarro
- Department of Pathology, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Line Bille Madsen
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Zoi Tsourti
- Frontier Science Foundation-Hellas & University of Athens, Athens, Greece
| | - Thomas Geiger
- Translational Research Coordination, European Thoracic Oncology Platform Coordinating Office, Bern, Switzerland
| | - Roswitha Kammler
- Translational Research Coordination, European Thoracic Oncology Platform Coordinating Office, Bern, Switzerland
| | - Solange Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Rolf A Stahel
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
43
|
Rossi G, Russo A, Tagliamento M, Tuzi A, Nigro O, Vallome G, Sini C, Grassi M, Dal Bello MG, Coco S, Longo L, Zullo L, Tanda ET, Dellepiane C, Pronzato P, Genova C. Precision Medicine for NSCLC in the Era of Immunotherapy: New Biomarkers to Select the Most Suitable Treatment or the Most Suitable Patient. Cancers (Basel) 2020; 12:E1125. [PMID: 32365882 PMCID: PMC7281184 DOI: 10.3390/cancers12051125] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
In recent years, the evolution of treatments has made it possible to significantly improve the outcomes of patients with non-small cell lung cancer (NSCLC). In particular, while molecular targeted therapies are effective in specific patient sub-groups, immune checkpoint inhibitors (ICIs) have greatly influenced the outcomes of a large proportion of NSCLC patients. While nivolumab activity was initially assessed irrespective of predictive biomarkers, subsequent pivotal studies involving other PD-1/PD-L1 inhibitors in pre-treated advanced NSCLC (atezolizumab within the OAK study and pembrolizumab in the Keynote 010 study) reported the first correlations between clinical outcomes and PD-L1 expression. However, PD-L1 could not be sufficient on its own to select patients who may benefit from immunotherapy. Many studies have tried to discover more precise markers that are derived from tumor tissue or from peripheral blood. This review aims to analyze any characteristics of the immunogram that could be used as a predictive biomarker for response to ICIs. Furthermore, we describe the most important genetic alteration that might predict the activity of immunotherapy.
Collapse
Affiliation(s)
- Giovanni Rossi
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
| | | | - Marco Tagliamento
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Alessandro Tuzi
- UO Oncologia, ASST Sette Laghi, 21100 Varese, Italy; (A.T.); (O.N.)
| | - Olga Nigro
- UO Oncologia, ASST Sette Laghi, 21100 Varese, Italy; (A.T.); (O.N.)
| | - Giacomo Vallome
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Claudio Sini
- Oncologia Medica e CPDO, ASSL di Olbia-ATS Sardegna, 07026 Olbia, Italy;
| | - Massimiliano Grassi
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Maria Giovanna Dal Bello
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Simona Coco
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Luca Longo
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Lodovica Zullo
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Enrica Teresa Tanda
- Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Chiara Dellepiane
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Paolo Pronzato
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| | - Carlo Genova
- Lung Cancer Unit, Medical Oncology 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.T.); (G.V.); (M.G.); (M.G.D.B.); (S.C.); (L.L.); (L.Z.); (C.D.); (P.P.); (C.G.)
| |
Collapse
|
44
|
Zito Marino F, Rossi G, Montella M, Botti G, De Cecio R, Morabito A, La Manna C, Ronchi A, Micheli M, Salatiello G, Micheli P, Rocco D, Accardo M, Franco R. Heterogeneity of PD-L1 Expression in Lung Mixed Adenocarcinomas and Adenosquamous Carcinomas. Am J Surg Pathol 2020; 44:378-386. [DOI: 10.1097/pas.0000000000001400] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immune checkpoint inhibitors against programmed cell death protein 1/programmed death-ligand 1 (PD-L1) have proven to be remarkably effective in non–small cell lung cancer. PD-L1 represents a predictive biomarker in lung cancer, although its heterogenous expression represents an emerging challenge for accurate biomarker-based patient selection. Lung adenocarcinomas (ADCs) show a high rate of intratumor morphologic heterogeneity that may reflect a heterogenous molecular and immunophenotypic profile. The aim of our study was to analyze the expression of PD-L1 in different intratumor subtypes and/or growth patterns in a series of mixed adenocarcinomas (mADCs) and adenosquamous lung carcinomas (AdSqLCs). As many as 73 mADCs and 6 AdSqLCs were selected. Comprehensive histologic subtyping was performed, and PD-L1 expression was assessed by immunohistochemistry assay using different primary antibodies and automated immunostainers. Overall, PD-L1 expression was observed in 37 of 79 cases (39.2%) (31 mADCs and all AdSqLCs). PD-L1 expression was heterogenous in 22 of 37 PD-L1-positive cases (23.2% mADC and 83% AdSqLC). PD-L1 expression was observed more frequently in ADC with solid pattern. Heterogeneity of PD-L1 expression was significantly related to the presence of micropapillary (P=0.028) and solid (P=0.017) patterns. All PD-L1-positive cases were epidermal growth factor receptor wild-type, 2 cases harbored concomitantly PD-L1 expression and ALK rearrangement. Our data suggest that PD-L1 expression is quite heterogenous in mADCs and AdSqLCs, partly contributing to explaining the discrepant results between biopsy and surgical resections and discordant clinical effectiveness in regard to PD-L1-positive or negative ADC diagnosed on cytology/small biopsy.
Collapse
Affiliation(s)
- Federica Zito Marino
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”
| | - Giulio Rossi
- Pathology Unit, S. Maria delle Croci Hospital, Ravenna, Italy
| | - Marco Montella
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”
| | | | | | | | - Carmine La Manna
- Thoracic Department, National Cancer Institute, IRCCS—Fondazione G. Pascale
| | - Andrea Ronchi
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”
| | | | | | | | | | - Marina Accardo
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”
| |
Collapse
|
45
|
Ou YC, Wen X, Johnson CA, Shae D, Ayala OD, Webb JA, Lin EC, DeLapp RC, Boyd KL, Richmond A, Mahadevan-Jansen A, Rafat M, Wilson JT, Balko JM, Tantawy MN, Vilgelm AE, Bardhan R. Multimodal Multiplexed Immunoimaging with Nanostars to Detect Multiple Immunomarkers and Monitor Response to Immunotherapies. ACS NANO 2020; 14:651-663. [PMID: 31851488 PMCID: PMC7391408 DOI: 10.1021/acsnano.9b07326] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The overexpression of immunomarker programmed cell death protein 1 (PD-1) and engagement of PD-1 to its ligand, PD-L1, are involved in the functional impairment of cluster of differentiation 8+ (CD8+) T cells, contributing to cancer progression. However, heterogeneities in PD-L1 expression and variabilities in biopsy-based assays render current approaches inaccurate in predicting PD-L1 status. Therefore, PD-L1 screening alone is not predictive of patient response to treatment, which motivates us to simultaneously detect multiple immunomarkers engaged in immune modulation. Here, we have developed multimodal probes, immunoactive gold nanostars (IGNs), that accurately detect PD-L1+ tumor cells and CD8+ T cells simultaneously in vivo, surpassing the limitations of current immunoimaging techniques. IGNs integrate the whole-body imaging of positron emission tomography with high sensitivity and multiplexing of Raman spectroscopy, enabling the dynamic tracking of both immunomarkers. IGNs also monitor response to immunotherapies in mice treated with combinatorial PD-L1 and CD137 agonists and distinguish responders from those nonresponsive to treatment. Our results showed a multifunctional nanoscale probe with capabilities that cannot be achieved with either modality alone, allowing multiplexed immunologic tumor profiling critical for predicting early response to immunotherapies.
Collapse
Affiliation(s)
- Yu-Chuan Ou
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Xiaona Wen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Christopher A. Johnson
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee 37212, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Daniel Shae
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Oscar D. Ayala
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Joseph A. Webb
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Eugene C. Lin
- Radiology and Radiological Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt University Institute of Imaging Science, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi 62106, Taiwan
| | - Rossane C. DeLapp
- Civil and Environmental Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Kelli L. Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Ann Richmond
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee 37212, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Anita Mahadevan-Jansen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Marjan Rafat
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - John T. Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Justin M. Balko
- Vanderbilt Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Mohammed N. Tantawy
- Radiology and Radiological Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt University Institute of Imaging Science, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Anna E. Vilgelm
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee 37212, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Pathology, Ohio State University, Columbus, Ohio 43210, United States
| | - Rizia Bardhan
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Vanderbilt University Institute of Imaging Science, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50012, United States
| |
Collapse
|
46
|
High degree of heterogeneity of PD-L1 and PD-1 from primary to metastatic endometrial cancer. Gynecol Oncol 2020; 157:260-267. [PMID: 31973911 DOI: 10.1016/j.ygyno.2020.01.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/07/2020] [Accepted: 01/13/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE PD-L1 and PD-1 are predictive markers for immunotherapy and increasingly relevant in endometrial cancer. The reported fraction of positive primary tumors has been inconsistent. We investigated the expression of PD-L1 and PD-1 in primary tumors, also stratified by MSI. As immunotherapy is foremost relevant for metastatic disease, PD-L1 and PD-1 expression was also assessed in corresponding metastatic lesions. METHODS PD-L1 and PD-1 was investigated in a prospective, population based endometrial cancer cohort of 700 patients with corresponding metastatic lesions from 68 and 74 patients respectively. Fresh tissue was used for gene expression analysis. RESULTS In primary tumors, PD-L1 and PD-1 are expressed in 59% and 63%, respectively, but with no impact on survival, nor when stratified for MSS and MSI. Expression patterns of PD-L1 and PD-1 are similar in MSI and MSS tumors. Available metastatic lesions show heterogeneous expression of PD-L1 and PD-1. In gene expression analysis several genes related to immunological activity, including CD274 (encoding for PD-L1), were upregulated in PD-1 positive tumors. CONCLUSION PD-L1 and PD-1 are frequently expressed in endometrial cancer and expression patterns are similar across MSS and MSI tumors. Expression in corresponding metastatic lesions is discordant compared to primary tumors. These findings are in particular relevant for treatment decisions in advanced and recurrent disease.
Collapse
|
47
|
Dong Z, Liu Y, Jiang T, Hou L, Wu F, Gao G, Li X, Zhao C, Wang Y, Yang S, Mao S, Liu Q, Li Y, Xu C, Wu C, Ren S, Zhou C, Zhang J, Hirsch FR. Cell Block as a Surrogate for Programmed Death-Ligand 1 Staining Testing in Patients of Non-Small Cell Lung Cancer. J Cancer 2020; 11:551-558. [PMID: 31942178 PMCID: PMC6959032 DOI: 10.7150/jca.35810] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 09/19/2019] [Indexed: 12/26/2022] Open
Abstract
Introduction: Programmed death-ligand 1 (PD-L1) staining is used in clinical practice to guide the proper use of immune checkpoint inhibitors. This study aimed to investigate the accuracy of PD-L1 staining of non-small cell lung cancer (NSCLC) cytological cell block samples. Methods: Paired cytological cell block and surgical resection samples were consecutively collected from January 2016 to February 2017 in Shanghai Pulmonary Hospital, Tongji University. Two trial-validated PD-L1 assays (28-8 and SP142) were used to quantify PD-L1 expression. Results: A total of 112 pairs of specimens were collected, including 68(60.7%) adenocarcinomas and 28(25.0%) squamous cell carcinomas. Based on a tumor proportion score (TPS) cutoff of 1% for the 28-8 and SP142 assays, PD-L1 expression was positive in 78.6% and 58.9% of surgical samples respectively, while PD-L1 expression was positive in 67.9% and 25.0% of cytological cell block samples. Based on staining by each antibody, fair to substantial concordance of PD-L1 expression was observed for cytological cell block specimens as compared to surgical resection (𝛋 ranges from 0.377 to 0.686). However, as the tumor cells in the cell block specimen increased, the consistency of PD-L1 expression increased. The concordance of PD-L1 expression in cell blocks with abundant cellularity was nearly perfect with various cutoffs (28-8: tumor cells over 400; SP142: tumor cells over 500). Conclusion: Cytological cell block specimens may serve as a surrogate for PD-L1 staining in patients of NSCLC when more than 400-500 cancer cells were contained (over 400 cancer cells for 28-8, over 500 cancer cells for SP142).
Collapse
Affiliation(s)
- Zhengwei Dong
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 200433, Shanghai, P.R. China
| | | | | | - Likun Hou
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 200433, Shanghai, P.R. China
| | | | | | - Xuefei Li
- Department of Lung Cancer and Immunology
| | - Chao Zhao
- Department of Lung Cancer and Immunology
| | | | | | | | | | - Yumei Li
- Department of Oncology and Nursing
| | - Chuan Xu
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, PR China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 200433, Shanghai, P.R. China
| | | | | | - Jun Zhang
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Fred R Hirsch
- Department of Medicine, Division of Medical Oncology, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA.,Clinical Institute for Lung Cancer, Mount Sinai Cancer, Mount Sinai Health System, Tisch Cancer Institute, Icahn School of Medicine, New York, New York
| |
Collapse
|
48
|
Naso JR, Wang G, Pender A, Wong SK, Zhu J, Ho C, Ionescu DN, Zhou C. Intratumoral heterogeneity in programmed death-ligand 1 immunoreactivity is associated with variation in non-small cell lung carcinoma histotype. Histopathology 2019; 76:394-403. [PMID: 31454429 DOI: 10.1111/his.13983] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/24/2019] [Indexed: 01/10/2023]
Abstract
AIMS Accurate assessment of programmed death-ligand 1 (PD-L1) levels in non-small cell lung carcinoma (NSCLC) samples is complicated by intratumoral heterogeneity. We aimed to: (i) establish whether intratumoral PD-L1 variation is associated with differences in local histotype; (ii) identify histotypes associated with a tendency for there to be higher or lower PD-L1 scores; and (iii) estimate the frequency of clinically significant discordance in PD-L1 levels between intratumoral histotype areas. METHODS AND RESULTS We reviewed 166 NSCLC resection specimens clinically tested for PD-L1 with the 22C3 pharmDx assay. Multiple histotypes were present in 55% (68/123) of non-mucinous adenocarcinoma samples. Solid histotypes had significantly higher PD-L1 levels than other histotypes, both when samples were grouped by predominant histotype, and when histotype areas within a tumour were compared (P < 0.02). Lepidic areas had significantly lower PD-L1 levels than other histotype areas within the same tumour (P < 0.02). Discordance between intratumoral histotype areas at a clinically relevant threshold (PD-L1 tumour proportion score of 1% or 50%) was present in 32% (22/68) of non-mucinous adenocarcinoma specimens with multiple histotype areas. The lepidic histotype was most frequently involved in discordance. CONCLUSIONS Intratumoral heterogeneity in PD-L1 is associated with variation in histotype. Over-representation of solid areas may increase the PD-L1 score assigned to a tumour, whereas over-representation of lepidic areas may decrease the PD-L1 score. Evaluation of how histotype representation impacts on the predictive value of PD-L1 testing is warranted.
Collapse
Affiliation(s)
- Julia R Naso
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gang Wang
- Department of Pathology, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Alexandra Pender
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Selina K Wong
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Jingyao Zhu
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Cheryl Ho
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Diana N Ionescu
- Department of Pathology, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Chen Zhou
- Department of Pathology, British Columbia Cancer Agency, Vancouver, BC, Canada
| |
Collapse
|
49
|
Gosney JR, Haragan A, Chadwick C, Giles TE, Grundy S, Tippett V, Gumparthy KP, Wight A, Tan HG. Programmed death ligand 1 expression in EBUS aspirates of non-small cell lung cancer: Is interpretation affected by type of fixation? Cancer Cytopathol 2019; 128:100-106. [PMID: 31851430 DOI: 10.1002/cncy.22216] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/11/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Much of the reluctance about using cytology specimens rather than histology specimens to assess programmed death ligand 1 (PD-L1) expression for guiding the use of immune modulating drugs in the management of non-small cell lung cancer (NSCLC) is based on the belief that the alcohol-based fixatives favored by cytopathologists might reduce the antigenicity of PD-L1 and lead to artifactually low expression levels and false-negative reporting. Therefore, this study was performed to determine whether there is any difference in PD-L1 expression between endobronchial ultrasound (EBUS)-guided aspirates of NSCLC fixed in alcohol-based fixatives and those fixed in neutral buffered formalin (NBF), the standard laboratory fixative for histology specimens. METHODS The expression of PD-L1 was compared in 50 paired EBUS aspirates of NSCLC taken from the same lymph node during the same procedure. One aspirate of each pair was fixed in an alcohol-based fixative, and the other was fixed in NBF. RESULTS In none of the 50 pairs was there any significant difference, qualitative or quantitative, in the strength, pattern, or extent of PD-L1 expression. In the great majority, the expression was identical, regardless of fixation. CONCLUSIONS There is no evidence from this study showing that the use of alcohol-based fixatives has any effect on the expression of PD-L1 or its interpretation. Notwithstanding the general challenges in accurately assessing such expression in cytology specimens, pathologists should feel able to interpret them with confidence, and clinicians should feel able to rely on the results.
Collapse
Affiliation(s)
- John R Gosney
- Department of Cellular Pathology, Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - Alexander Haragan
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Claire Chadwick
- Department of Cellular Pathology, Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - Tom E Giles
- Department of Cellular Pathology, Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - Seamus Grundy
- Department of Respiratory Medicine, Salford Royal National Health Service Foundation Trust, Salford, United Kingdom
| | - Victoria Tippett
- Department of Respiratory Medicine, Aintree University Hospital, Liverpool, United Kingdom
| | - Krishna P Gumparthy
- Department of Histopathology, Wirral University Teaching Hospital, Birkenhead, United Kingdom
- Department of Respiratory Medicine, Wirral University Teaching Hospital, Birkenhead, United Kingdom
| | - Andrew Wight
- Department of Histopathology, Wirral University Teaching Hospital, Birkenhead, United Kingdom
- Department of Respiratory Medicine, Wirral University Teaching Hospital, Birkenhead, United Kingdom
| | - Hock G Tan
- Department of Histopathology, Wirral University Teaching Hospital, Birkenhead, United Kingdom
- Department of Respiratory Medicine, Wirral University Teaching Hospital, Birkenhead, United Kingdom
| |
Collapse
|
50
|
Song JS, Kim D, Kwon JH, Kim HR, Choi CM, Jang SJ. Clinicopathologic Significance and Immunogenomic Analysis of Programmed Death-Ligand 1 (PD-L1) and Programmed Death 1 (PD-1) Expression in Thymic Epithelial Tumors. Front Oncol 2019; 9:1055. [PMID: 31681591 PMCID: PMC6803548 DOI: 10.3389/fonc.2019.01055] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 09/27/2019] [Indexed: 12/26/2022] Open
Abstract
Objectives: Thymic epithelial tumors (TETs) are rare malignant tumors that exhibit heterogeneous histology and clinical behavior. As immune check point inhibitors, drugs targeting anti-programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) have shown remarkable results against many cancers; thus, the importance of PD-1/PD-L1 immunohistochemistry as a predictive or prognostic biomarker has grown. However, limited data on PD-L1 and PD-1 expression in TETs have been reported; moreover, these results have been variable. Here, we examined the expression of PD-1/PD-L1 proteins in TETs and analyzed the clinicopathologic significance of this expression. Patients and Methods: A tissue microarray was constructed using 368 samples of TETs, each in triplicate. Immunohistochemistry for PD-L1 (SP263 assay) and PD-1 in TETs and CD8 in thymic carcinoma (TC) was performed; next, correlations with clinicopathologic characteristics were analyzed. PD-L1high was designated as ≥50% of tumor proportion score; PD-1high and CD8high were defined as ≥5% and 1% of tumoral immune cells, respectively. Results: The cohort consisted of 308 patients with thymomas and 60 patients with TC. PD-L1 positivity was identified in 90.6% (328/362, ≥1%) of TETs, PD-1 expression of intra-/peritumoral T cells was identified in 53.6% (194/362) of TETs and CD8 positivity was identified in 11% (7/60, ≥1%) of TC. Of the 362 patients, 141 (39.0%) exhibited high PD-L1 expression (PD-L1high). The PD-L1high thymoma group was correlated with high Masaoka-Koga stage (p < 0.001), type B3 histology (p < 0.001), and myasthenia gravis (p < 0.001). This group exhibited poor overall survival (OS, p = 0.003, log-rank) and worse disease-free survival (DFS, p = 0.042, log-rank). No survival differences were detected between PD-L1high and PD-L1low groups in TC. Additionally, there was no correlation between PD-1 expression and survival in patients with TETs. Multivariate analysis revealed that PD-L1high expression was an independent poor prognostic factor (p = 0.047, HR 2.087, 95% CI, 1.009–4.318) in thymomas. Conclusions: To our knowledge, this is the largest study on TETs published in English literature. This study provides useful information regarding the prognosis of and potential therapeutic options for patients with TETs.
Collapse
Affiliation(s)
- Joon Seon Song
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Deokhoon Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Center for Cancer Genome Discovery, Asan Medical Center, Asan Institute for Life Sciences, Seoul, South Korea
| | - Ji Hyun Kwon
- Samkwang Medical Laboratories, Department of Pathology, Seoul, South Korea
| | - Hyeong Ryul Kim
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Chang-Min Choi
- Department of Pulmonology and Medical Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Se Jin Jang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Center for Cancer Genome Discovery, Asan Medical Center, Asan Institute for Life Sciences, Seoul, South Korea
| |
Collapse
|