51
|
Rui M, Wang Y, Li Y, Fei Z. Immunotherapy Guided by Immunohistochemistry PD-L1 Testing for Patients with NSCLC: A Microsimulation Model-Based Effectiveness and Cost-Effectiveness Analysis. BioDrugs 2024; 38:157-170. [PMID: 37792142 DOI: 10.1007/s40259-023-00628-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 10/05/2023]
Abstract
BACKGROUND On the basis of immunohistochemistry PD-L1 testing results, patients with advanced non-small cell lung cancer (NSCLC) are treated differently. Theoretically, patients with high PD-L1 expression (50% or 1%) should receive PD-1 monotherapy for fewer adverse reactions and cost savings from avoiding chemotherapy; however, there is controversy surrounding the cut-off criteria (1% or 50%) for immunohistochemistry testing and threshold for PD-1 monotherapy. OBJECTIVE This study aims to predict the effectiveness and cost-effectiveness of different immunotherapy strategies for patients with NSCLC in China from the healthcare system perspective. PATIENTS AND METHODS A microsimulation model was developed to evaluate the effectiveness and cost-effectiveness of three treatment strategies: PD-L1 testing (1%) (PD-1 monotherapy for those with PD-L1 expression at 1% threshold, and combination with chemotherapy for others with immunohistochemistry testing), PD-L1 testing (50%) (PD-1 monotherapy for those with PD-L1 expression at 50% threshold, and combination with chemotherapy for others with immunohistochemistry testing), and No PD-L1 testing (PD-1 combined with chemotherapy without immunohistochemistry testing). The model assumed 1000 patients per strategy, with each patient entering a unique clinical path prior to receiving treatment on the basis of PD-L1 test results. Clinical inputs were derived from clinical trials. Cost and utility parameters were obtained from the database and literature. One-way probabilistic sensitivity analyses (PSA) and six scenario analyses were used to test the model's robustness. RESULTS The study revealed a hierarchy of survival benefits across three strategies, with No PD-L1 testing demonstrating the most survival advantage, followed by PD-L1 testing (50%), and finally, PD-L1 testing (1%). The comparative analysis demonstrated that No PD-L1 testing significantly enhanced overall survival (OS) (HR 0.85, 95% CI 0.78-0.93), progression-free survival (HR 0.82, 95% CI 0.75-0.90), and progression-free2 survival (PFS2) (HR 0.91, 95% CI 0.83-0.99) when juxtaposed against PD-L1 testing (1%). However, these improvements were not as pronounced when compared with PD-L1 testing (50%), particularly in relation to PFS, PFS2, and OS. The cost-effectiveness analysis further unveiled incremental cost-utility ratios (ICUR), with No PD-L1 testing versus PD-L1 testing (50%) at $34,003 per quality-adjusted life year (QALY) and No PD-L1 testing versus PD-L1 testing (1%) at $34,804 per QALY. In parallel, the ICUR for PD-L1 testing (50%) versus PD-L1 testing (1%) stood at $35,713 per QALY. Remarkably, the PSA result under a willingness-to-pay (WTP) threshold of $10,144 per QALY, with a 100% probability, demonstrated PD-L1 testing (1%) as the most cost-effective option. CONCLUSIONS The survival benefits of PD-1 monotherapy for high expression with PD-L1 immunohistochemistry testing are inferior to those of PD-1 combined with chemotherapy without testing, but it is found to be more cost-effective at the WTP thresholds in China and holds great potential in increasing affordability and reducing the economic burden.
Collapse
Affiliation(s)
- Mingjun Rui
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Yingcheng Wang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yunfei Li
- Institute for Global Health, Department of Population Health Sciences, University College London, London, UK
| | - Zhengyang Fei
- Center for Pharmacoeconomics and Outcomes Research, China Pharmaceutical University, Nanjing, China
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
52
|
Salvia R, Rico LG, Ward MD, Bradford JA, Petriz J. Functional Flow Cytometry to Predict PD-L1 Conformational Changes. Curr Protoc 2023; 3:e944. [PMID: 38100257 DOI: 10.1002/cpz1.944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
The programmed cell death protein 1/programmed cell death protein ligand 1 (PD-1/PD-L1) axis is one of the most widely recognized targets for cancer immunotherapy. Importantly, PD-L1 conformational changes can hinder target binding when living cells are used. Antibody affinity, equilibrium binding, association and dissociation rates, and other affinity-related constants are fundamental to ensure target saturation. Here, PD-L1 changes in conformation and their potential impact on PD-L1 function and mutation are explored. Specifically, we present detailed flow cytometry procedures to analyze PD-L1 reactivity in myeloid-derived suppressor cells (MDSCs). This approach can also be used to study the contribution of protein conformational changes in living cells. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Sample preparation for PD-L1+ myeloid-derived suppressor cells detection by flow cytometry Basic Protocol 2: Protocol preparation, sample acquisition, and gating strategy for flow cytometric screening of PD-L1+ myeloid-derived suppressor cells in patients with lung cancer Support Protocol 1: Bioinformatic tools for the analysis of flow cytometric data.
Collapse
Affiliation(s)
- Roser Salvia
- Functional Cytomics Lab, Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona, Badalona, Barcelona, Spain
| | - Laura G Rico
- Functional Cytomics Lab, Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona, Badalona, Barcelona, Spain
| | | | | | - Jordi Petriz
- Functional Cytomics Lab, Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona, Badalona, Barcelona, Spain
| |
Collapse
|
53
|
Vryza P, Fischer T, Mistakidi E, Zaravinos A. Tumor mutation burden in the prognosis and response of lung cancer patients to immune-checkpoint inhibition therapies. Transl Oncol 2023; 38:101788. [PMID: 37776617 PMCID: PMC10542015 DOI: 10.1016/j.tranon.2023.101788] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/29/2023] [Accepted: 09/10/2023] [Indexed: 10/02/2023] Open
Abstract
Immune checkpoint inhibition (ICI) therapies have reshaped the therapeutic landscape in lung cancer management, providing first-time improvements in patient response, prognosis, and overall survival. Despite their clinical effectiveness, variability in treatment responsiveness, as well as drug resistance, have led to a compelling need for predictive biomarkers facilitating the individualized selection of the most efficient therapeutic approach. Significant progress has been made in the identification of such biomarkers, with tumor mutation burden (ΤΜΒ) appearing as the leading and most promising predictive biomarker for the efficacy of ICIs in non-small cell lung cancer (NSCLC) among other tumors. Anti-PD-1/PD-L1 and anti-CTLA-4 antibodies have been extensively studied and clinically utilized. However, the overall efficiency of these drugs remains unsatisfactory, urging for the investigation of novel inhibitors, such as those targeting LAG-3, TIM-3, TIGIT and VISTA, which could be used either as a monotherapy or synergistically with the PD-1/PD-L1 or CTLA-4 blockers. Here, we investigate the role of TMB and cancer neoantigens as predictive biomarkers in the response of lung cancer patients to different ICI therapies, specifically focusing on the most recent immune checkpoint inhibitors, against LAG-3, TIM-3, TIGIT and VISTA. We further discuss the new trends in immunotherapies, including CAR T-cell therapy and personalized tumor vaccines. We also review further potential biomarkers that could be used in lung cancer response to immunotherapy, such as PD-L1+ IHC, MSI/dMMR, tumor infiltrating lymphocytes (TILs), as well as the role of the microbiome and circulating tumor DNA (ctDNA). Finally, we discuss the limitations and challenges of each.
Collapse
Affiliation(s)
- Paraskevi Vryza
- School of Medicine, European University Cyprus, Nicosia 1516, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
| | - Timo Fischer
- School of Medicine, European University Cyprus, Nicosia 1516, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
| | - Elena Mistakidi
- School of Medicine, European University Cyprus, Nicosia 1516, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
| | - Apostolos Zaravinos
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus; Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus.
| |
Collapse
|
54
|
Miyakoshi J, Yazaki S, Shimoi T, Onishi M, Saito A, Kita S, Yamamoto K, Kojima Y, Sumiyoshi-Okuma H, Nishikawa T, Sudo K, Noguchi E, Murata T, Shiino S, Takayama S, Suto A, Fujiwara Y, Yoshida M, Yonemori K. Discordance in PD-L1 expression using 22C3 and SP142 assays between primary and metastatic triple-negative breast cancer. Virchows Arch 2023; 483:855-863. [PMID: 37668667 DOI: 10.1007/s00428-023-03634-2] [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: 05/19/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/06/2023]
Abstract
AIMS SP142 and 22C3 assays are approved companion diagnostic assays for anti-PD-1/PD-L1 therapy selection in metastatic triple-negative breast cancer (TNBC). The discordance in PD-L1 status between primary and metastatic tumors in the same patient has been poorly characterized. Here, we examined the concordance of PD-L1 status between the two assays and between primary tumors and metastases for each assay. METHODS We retrospectively evaluated tumor samples from 160 patients with TNBC, including 45 patients with paired primary and metastatic tumors. PD-L1 status was assessed using SP142 and 22C3 assays, to determine the immune cell (IC) score, tumor cell (TC) score (SP142 and 22C3), and combined proportion score (CPS: 22C3). RESULTS The concordance of PD-L1 positivity at diagnostic cutoffs for SP142 (IC ≥ 1) and 22C3 (CPS ≥ 10) was substantial (κ = 0.80) in primary tumors and moderate (κ = 0.60) in metastatic tumors. In comparison, between primary and metastatic tumors, the concordance with 22C3 was moderate (κ = 0.50), whereas that with SP142 was poor (κ = -0.03). Among patients who were PD-L1 negative for both assays in primary tumors, 7/30 (23.3%) were PD-L1 positive for both or either 22C3 or SP142 in the metastatic tumors. CONCLUSIONS The inter-assay concordance of PD-L1 positivity at diagnostic cutoffs was substantial in primary tumors and moderate in metastatic tumors. Discordance between PD-L1 status in primary and metastatic tumors was frequently observed, especially with SP142. Some patients with a PD-L1-negative status in primary tumors may still be candidates for immunotherapy, depending on the PD-L1 status in their metastatic tumors.
Collapse
Affiliation(s)
- Jun Miyakoshi
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shu Yazaki
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Tatsunori Shimoi
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Mai Onishi
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Ayumi Saito
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shosuke Kita
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Kasumi Yamamoto
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yuki Kojima
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Hitomi Sumiyoshi-Okuma
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Tadaaki Nishikawa
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Kazuki Sudo
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Emi Noguchi
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Takeshi Murata
- Department of Breast Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Sho Shiino
- Department of Breast Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shin Takayama
- Department of Breast Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Akihiko Suto
- Department of Breast Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yasuhiro Fujiwara
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Masayuki Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan.
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| |
Collapse
|
55
|
Catalano M, Iannone LF, Nesi G, Nobili S, Mini E, Roviello G. Immunotherapy-related biomarkers: Confirmations and uncertainties. Crit Rev Oncol Hematol 2023; 192:104135. [PMID: 37717881 DOI: 10.1016/j.critrevonc.2023.104135] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 08/18/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023] Open
Abstract
Immunotherapy profoundly changed oncology treatment, becoming one of the main therapeutical strategies. Remarkable improvement has been achieved in survival outcomes, but the percentage of patients who benefit from immunotherapy is still limited. Only one-third of patients receiving immune checkpoint inhibitors (ICIs) achieve long-term response. Several patients are not responsive to treatment or relapse after an initial response. To date, programmed death-ligand 1, microsatellite instability, and tumor mutational burden are the three biomarkers validated to predict the ICIs response, but a single variable seems still insufficient in the patient's selection. Considering the substantial and increasing use of these drugs, the identification of new predictive biomarkers of ICI response is of paramount importance. We summarize the state of the art and the clinical use of immune biomarkers in oncology, highlighting the strength and weaknesses of currently approved biomarkers, describing the emerging tissues and circulating biomarkers, and outlining future perspectives.
Collapse
Affiliation(s)
- Martina Catalano
- 1 Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Luigi Francesco Iannone
- 1 Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Gabriella Nesi
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Stefania Nobili
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, 50139 Florence, Italy
| | - Enrico Mini
- 1 Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Giandomenico Roviello
- 1 Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, 50139 Florence, Italy.
| |
Collapse
|
56
|
Bill R, Faquin WC, Pai SI. Assessing PD-L1 Expression in Head and Neck Squamous Cell Carcinoma: Trials and Tribulations. Head Neck Pathol 2023; 17:969-975. [PMID: 37930471 PMCID: PMC10739626 DOI: 10.1007/s12105-023-01590-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
Immune checkpoint inhibitors have improved the outcome of patients diagnosed with inoperable recurrent or metastatic head and neck squamous cell carcinoma. However, as only a subset of head and neck cancer patients benefit from this treatment, biomarkers predicting treatment response help guide physicians in their clinical decision-making. PD-L1 expression assessed by immunohistochemistry is the single most clinically relevant biomarker predicting response to PD-1-blocking antibodies. Here, we discuss in which clinical context assessment of PD-L1 expression is instrumental for the choice of therapy, how pathologists score it, and how it affects the approval of anti-PD-1 antibodies. Furthermore, we discuss the heterogeneity of PD-L1 expression and review technical aspects of determining this prominent biomarker-knowledge that might influence clinical decision-making.
Collapse
Affiliation(s)
- Ruben Bill
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - William C Faquin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara I Pai
- Department of Surgery, Yale School of Medicine, Yale University, 47 College Street, Suite 216, New Haven, CT, 06510, USA.
| |
Collapse
|
57
|
Zhang J, Zhao R, Xu H, Dong L, Chen X. The clinicopathological features of BRG1-deficient non-small cell lung cancer and its response to immunotherapy: A single-center retrospective study. Ann Diagn Pathol 2023; 67:152192. [PMID: 37639838 DOI: 10.1016/j.anndiagpath.2023.152192] [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: 08/01/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023]
Abstract
PURPOSE BRG1-deficient NSCLCs have been more intriguing recently for its highly aggressive clinical behavior and no effective therapies. This study characterized the clinical and pathological features of BRG1-deficient NSCLCs and investigated their response to immunotherapy. METHODS Forty-seven cases with BRG1-deficient NSCLC were included. Immunohistochemical markers such as BRG1, CK7, TTF-1, NapsinA, P40, HepPar-1, Ki-67, BRM, ARID1A and ARID1B were stained. Additionally, the PD-L1 expression level, overall survival, progression-free survival and disease control rate of patients received immunotherapy were evaluated. RESULTS This study revealed that: (1) Patients with BRG1-deficient NSCLC have a male predominance (89.4 %), smoker enrichment (76.6 %) and poor prognosis (median OS: 7.0 months for advanced stage). (2) Histologically, BRG1-deficient NSCLCs presented significant morphological diversity and no lepidic pattern. Inflammatory infiltration and tumor necrosis was a prominent feature. Immunohistochemical analyses showed a distinctive uniform immunophenotype (TTF-1-/NapsinA-/CK7+) in 60.9 % (28/46) of cases and HepPar-1 positive in 46.5 % (20/43) of cases. BRM loss or significant reduction coexisted in 11.8 % (4/34) of cases. No case (0/37) showed loss of ARID1A or ARID1B. (3) Eight patients with advanced tumor stage had received immunotherapy and 4 cases achieved a sustainable clinical response with the disease control rate of 50 %. CONCLUSION BRG1-deficient NSCLC showed diverse histopathological patterns and a unique immunohistochemical phenotype. ICIs-based immunotherapy is a promising therapy needs to be investigated further for BRG1- deficient NSCLC.
Collapse
Affiliation(s)
- Jing Zhang
- Respiratory and Critical Care Medicine Department, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Runze Zhao
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haimin Xu
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Dong
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Chen
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
58
|
Yamada T, Goto Y, Tanaka H, Kimura H, Minato K, Gyotoku H, Honda T, Watanabe S, Morimoto K, Kiyomi F, Uchino J, Takayama K. A phase 2 trial of durvalumab treatment following radiation monotherapy in patients with non-small cell lung cancer ineligible for stage III chemoradiotherapy: The SPIRAL-RT study. Eur J Cancer 2023; 195:113373. [PMID: 37890349 DOI: 10.1016/j.ejca.2023.113373] [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: 08/08/2023] [Revised: 09/30/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND Although concurrent chemoradiotherapy (CCRT) followed by durvalumab is the standard treatment for patients with stage III non-small cell lung cancer (NSCLC), only half of the patients are allowed to receive CCRT in real-world settings. We evaluated the efficacy and safety of durvalumab after radiation monotherapy for NSCLC patients who are ineligible for chemoradiotherapy. METHODS A single-arm, prospective, open-label, multicenter phase II trial was conducted in Japan. The patients received radiation (54-66 Gy) followed by durvalumab (10 mg/kg every 2 weeks for up to 12 months). The primary endpoint was the 1-year progression-free survival (PFS) rate. The secondary endpoints were the objective response rate (ORR), PFS, overall survival (OS), and safety. RESULTS Between September 2019 and April 2021, 33 patients were enroled from eight institutions. The median patient age was 79 years, and the majority of patients were male (78.8%). The 1-year PFS rate was 39.1% (90% confidence interval [CI]: 24.7-54.6%). Three patients (9.1%) had a performance status of 2. The ORR was 42.4% (95% CI: 27.2-59.2%). The median PFS and OS were 8.9 (95% CI: 7.4-19.4) and 20.8 (95% CI: 15.8-not estimable) months, respectively. The most common adverse event was radiation pneumonitis (51.5%). The median treatment duration was 6.4 (range: 0.50-12.0) months for durvalumab. At the endpoint, 30.3% (10/33) of the patients had completed 1 year of durvalumab therapy. CONCLUSIONS Durvalumab is an effective treatment with tolerable toxicity following radiation monotherapy in stage III NSCLC patients who are ineligible for chemoradiotherapy. TRIAL REGISTRATION JMA-IIA00434 (jRCT).
Collapse
Affiliation(s)
- Tadaaki Yamada
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuhiro Goto
- Department of Respiratory Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hiroshi Tanaka
- Department of Internal Medicine, Niigata Cancer Center Hospital, Niigata, Japan
| | - Hideharu Kimura
- Respiratory Medicine, Kanazawa University Hospital, Ishikawa, Japan
| | - Koichi Minato
- Division of Respiratory Medicine, Gunma Prefectural Cancer Center, Ota, Japan
| | - Hiroshi Gyotoku
- Department of Respiratory Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takeshi Honda
- Department of Internal Medicine Division of Medical Oncology, Teikyo University School of Medicine, Tokyo, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medicine and Dental Sciences, Niigata, Japan
| | - Kenji Morimoto
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Fumiaki Kiyomi
- Statistics and Data Center, Clinical Research Support Center Kyushu, Fukuoka, Japan
| | - Junji Uchino
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Koichi Takayama
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
59
|
Russell PA, Farrall AL, Prabhakaran S, Asadi K, Barrett W, Cooper C, Cooper W, Cotton S, Duhig E, Egan M, Fox S, Godbolt D, Gupta S, Hassan A, Leslie C, Leong T, Moffat D, Qiu MR, Sivasubramaniam V, Skerman J, Snell C, Walsh M, Whale K, Klebe S. Real-world prevalence of PD-L1 expression in non-small cell lung cancer: an Australia-wide multi-centre retrospective observational study. Pathology 2023; 55:922-928. [PMID: 37833206 DOI: 10.1016/j.pathol.2023.08.008] [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: 05/14/2023] [Revised: 07/30/2023] [Accepted: 08/17/2023] [Indexed: 10/15/2023]
Abstract
An investigator-initiated, Australia-wide multi-centre retrospective observational study was undertaken to investigate the real-world prevalence of programmed death ligand-1 (PD-L1) expression in non-small cell lung carcinoma (NSCLC). Multiple centres around Australia performing PD-L1 immunohistochemistry (IHC) were invited to participate. Histologically confirmed NSCLC of any stage with a PD-L1 IHC test performed for persons aged ≥18 years between 1 January 2018 and 1 January 2020, and eligible for review, were identified at each centre, followed by data extraction and de-identification, after which data were submitted to a central site for collation and analysis. In total data from 6690 eligible PD-L1 IHC tests from histologically (75%) or cytologically (24%) confirmed NSCLC of any stage were reviewed from persons with a median age of 70 years, 43% of which were female. The majority (81%) of tests were performed using the PD-L1 IHC SP263 antibody with the Ventana BenchMark Ultra platform and 19% were performed using Dako PD-L1 IHC 22C3 pharmDx assay. Reported PD-L1 tumour proportion score (TPS) was ≥50% for 30% of all tests, with 62% and 38% scoring PD-L1 ≥1% and <1%, respectively. Relative prevalence of clinicopathological features with PD-L1 scores dichotomised to <50% and ≥50%, or to <1% and ≥1%, were examined. Females scored ≥1% slightly more often than males (64% vs 61%, respectively, p=0.013). However, there was no difference between sexes or age groups (<70 or ≥70 years) where PD-L1 scored ≥50%. Specimens from patients with higher stage (III/IV) scored ≥1% or ≥50% marginally more often compared to specimens from patients with lower stage (I/II) (p≤0.002). Proportions of primary and metastatic specimens did not differ where PD-L1 TPS was ≥1%, however more metastatic samples scored TPS ≥50% than primary samples (metastatic vs primary; 34% vs 27%, p<0.001). Cytology and biopsy specimens were equally reported, at 63% of specimens, to score TPS ≥1%, whereas cytology samples scored TPS ≥50% slightly more often than biopsy samples (34% vs 30%, respectively, p=0.004). Resection specimens (16% of samples tested) were reported to score TPS ≥50% or ≥1% less often than either biopsy or cytology samples (p<0.001). There was no difference in the proportion of tests with TPS ≥1% between PD-L1 IHC assays used, however the proportion of tests scored at TPS ≥50% was marginally higher for 22C3 compared to SP263 (34% vs 29%, respectively, p<0.001). These real-world Australian data are comparable to some previously published global real-world data, with some differences noted.
Collapse
Affiliation(s)
- Prudence A Russell
- LifeStrands Genomics and, TissuPath Pathology, Mount Waverley, Vic, Australia
| | - Alexandra L Farrall
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Sarita Prabhakaran
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | | | - Wade Barrett
- Anatomical Pathology, St Vincent's Hospital Sydney, NSW, Australia
| | - Caroline Cooper
- Pathology Queensland, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - Wendy Cooper
- Anatomical Pathology, Royal Prince Alfred Hospital, NSW, Australia
| | - Samuel Cotton
- Anatomical Pathology, Royal Hobart Hospital, Tas, Australia
| | - Edwina Duhig
- Sullivan Nicolaides Pathology, Brisbane, Qld, Australia
| | - Matthew Egan
- Anatomical Pathology, St Vincent's Hospital Melbourne, Vic, Australia
| | - Stephen Fox
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - David Godbolt
- Pathology Queensland, Prince Charles Hospital, Brisbane, Qld, Australia
| | - Shilpa Gupta
- Pathology Queensland, Prince Charles Hospital, Brisbane, Qld, Australia
| | - Aniza Hassan
- SA Pathology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Connull Leslie
- Anatomical Pathology, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, WA, Australia
| | - Trishe Leong
- Anatomical Pathology, St Vincent's Hospital Melbourne, Vic, Australia
| | - David Moffat
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia; SA Pathology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Min Ru Qiu
- Anatomical Pathology, St Vincent's Hospital Sydney, NSW, Australia
| | - Vanathi Sivasubramaniam
- Anatomical Pathology, St Vincent's Hospital Sydney, NSW, Australia; Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Joanna Skerman
- Pathology Queensland, Prince Charles Hospital, Brisbane, Qld, Australia
| | - Cameron Snell
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Michael Walsh
- Sullivan Nicolaides Pathology, Brisbane, Qld, Australia
| | - Karen Whale
- Anatomical Pathology, Royal Hobart Hospital, Tas, Australia
| | - Sonja Klebe
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia; SA Pathology, Flinders Medical Centre, Bedford Park, SA, Australia.
| |
Collapse
|
60
|
Wu Y, Lv C, Lin M, Hong Y, Du B, Yao N, Zhu Y, Ji X, Li J, Lai J. Novel nomogram for predicting survival in advanced non-small cell lung cancer receiving anti-PD-1 plus chemotherapy with or without antiangiogenic therapy. Front Immunol 2023; 14:1297188. [PMID: 38022521 PMCID: PMC10663364 DOI: 10.3389/fimmu.2023.1297188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Background This study aimed to develop and validate a novel nomogram to predict survival in advanced non-small cell lung cancer (NSCLC) receiving programmed cell death 1 (PD-1) inhibitor plus chemotherapy with or without antiangiogenic therapy. Methods A total of 271 patients with advanced NSCLC who received anti-PD-1 plus chemotherapy with or without antiangiogenic therapy were enrolled in our center and randomized into the training cohort (n = 133) and the internal validation cohort (n = 138). Forty-five patients from another center were included as an independent external validation cohort. The nomogram was created based on the multivariate Cox regression analysis to predict overall survival (OS) and progression-free survival (PFS). The performance of the nomogram was assessed using the concordance index (C-index), the time-dependent area under the receiver operating (ROC) curves (AUCs), calibration curves, and decision curve analysis (DCA). Results Four factors significantly associated with OS were utilized to create a nomogram to predict OS: Eastern Cooperative Oncology Group performance status (ECOG PS), programmed cell death-ligand 1 (PD-L1) expression, chemotherapy cycle, and pretreatment lactate dehydrogenase-albumin ratio (LAR). Six variables significantly associated with PFS were incorporated into the development of a nomogram for predicting PFS: ECOG PS, histology, PD-L1 expression, chemotherapy cycle, pretreatment platelet to lymphocyte (PLR), and pretreatment LAR. The C-indexes of the nomogram for predicting OS and PFS were 0.750 and 0.747, respectively. The AUCs for predicting the 6-month, 12-month, and 18-month OS and PFS were 0.847, 0.791, and 0.776 and 0.810, 0.787, and 0.861, respectively. The calibration curves demonstrated a good agreement between predictions and actual observations. The DCA curves indicated that the nomograms had good net benefits. Furthermore, the nomogram model was well-validated in the internal and external cohorts. Conclusion The novel nomogram for predicting the prognosis of advanced NSCLC receiving anti-PD-1 plus chemotherapy with or without antiangiogenic therapy may help guide clinical treatment decisions.
Collapse
Affiliation(s)
- Yahua Wu
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chengliu Lv
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mingqian Lin
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Yaping Hong
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Bin Du
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Na Yao
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yingjiao Zhu
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaohui Ji
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jiancheng Li
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Jinhuo Lai
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
61
|
Kinoshita F, Takada K, Wakasu S, Saito S, Hashinokuchi A, Matsudo K, Nagano T, Akamine T, Kohno M, Takenaka T, Shimokawa M, Oda Y, Yoshizumi T. Granzyme B (GZMB)-Positive Tumor-Infiltrating Lymphocytes in Lung Adenocarcinoma: Significance as a Prognostic Factor and Association with Immunosuppressive Proteins. Ann Surg Oncol 2023; 30:7579-7589. [PMID: 37587364 DOI: 10.1245/s10434-023-14085-z] [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: 06/07/2023] [Accepted: 07/20/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Granzyme B (GZMB) is a serine protease produced by cytotoxic lymphocytes that reflects the activity of anti-tumor immune responses in tumor-infiltrating lymphocytes (TILs); however, the prognostic significance of GZMB+ TILs in lung adenocarcinoma is poorly understood. METHODS We analyzed 273 patients with pathological stage (pStage) I-IIIA lung adenocarcinoma who underwent surgery at Kyushu University from 2003 to 2012. We evaluated GZMB+ TIL counts by immunohistochemistry. We set the cut-off values at 12 cells/0.04 mm2 for GZMB+ TILs and divided the patients into GZMB-High (n = 171) and GZMB-Low (n = 102) groups. Then, we compared the clinicopathological characteristics of the two groups and clinical outcomes. Programmed cell death ligand-1 (PD-L1) and indoleamine 2,3-dioxygenase 1 (IDO1) expression in tumor cells was also evaluated, and combined prognostic analyses of GZMB+ TILs with PD-L1 or IDO1 were performed. RESULTS GZMB-Low was significantly associated with pStage II-III, PD-L1 positivity, and IDO1 positivity. Disease-free survival (DFS) and overall survival (OS) in the GZMB-Low group were significantly worse than in the GZMB-High group. In multivariable analysis, GZMB-Low was an independent prognostic factor for both DFS and OS. Furthermore, combined prognostic analyses of GZMB+ TILs with PD-L1 or IDO1 showed that GZMB-Low with high expression of these immunosuppressive proteins had the worst prognosis. CONCLUSIONS We analyzed GZMB+ TIL counts in lung adenocarcinoma and elucidated its prognostic significance and association with PD-L1 and IDO1. GZMB+ TIL counts might reflect the patient's immunity against cancer cells and could be a useful prognostic marker of lung adenocarcinoma.
Collapse
Affiliation(s)
- Fumihiko Kinoshita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuki Takada
- Department of Surgery, Saiseikai Fukuoka General Hospital, Fukuoka, Japan
| | - Sho Wakasu
- Department of Thoracic Surgery, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Shunichi Saito
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Asato Hashinokuchi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoto Matsudo
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taichi Nagano
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takaki Akamine
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mikihiro Kohno
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoyoshi Takenaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Mototsugu Shimokawa
- Department of Biostatistics, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
62
|
Yang X, Xiao Y, Hu H, Qiu ZB, Qi YF, Wang MM, Wu YL, Zhong WZ. Expression Changes in Programmed Death Ligand 1 from Precancerous Lesions to Invasive Adenocarcinoma in Subcentimeter Pulmonary Nodules: A Large Study of 2022 Cases in China. Ann Surg Oncol 2023; 30:7400-7411. [PMID: 37658270 DOI: 10.1245/s10434-023-14009-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/15/2023] [Indexed: 09/03/2023]
Abstract
PURPOSE This large-scale, multicenter, retrospective observational study aimed to evaluate the clinicopathological and molecular profiles associated with programmed death-ligand 1 (PD-L1) expression in precancerous lesions and invasive adenocarcinoma in subcentimeter pulmonary nodules. PATIENTS AND METHODS Patients with histologically confirmed atypical adenomatous hyperplasia (AAH), adenocarcinoma in situ (AIS), minimally invasive adenocarcinoma (MIA), and invasive adenocarcinoma (ADC) were included. PD-L1 expression was evaluated at each center using a PD-L1 immunohistochemistry 22C3 pharmDx kit (Agilent, Santa Clara, CA, USA). The tumor proportion score (TPS) cutoff values were set at ≥ 1% and ≥ 50%. RESULTS A total of 2022 nodules from 1844 patients were analyzed. Of these, 9 (0.45%) nodules had PD-L1 TPS ≥ 50%, 187 (9.25%) had PD-L1 TPS 1-49%, and 1826 (90.30%) had PD-L1 TPS < 1%. A total of 378 (18.69%), 1016 (50.25%), and 628 (31.06%) nodules were diagnosed as AAH/AIS, MIA, and ADC, respectively, by pathology. A total of 1377 (68.10%), 591 (25.67%), and 54 (2.67%) nodules were diagnosed as pure ground-glass opacity (GGO), mixed GGO, and solid nodules, respectively, by computed tomography. There was a significant difference between PD-L1 expression and anaplastic lymphoma kinase (ALK) mutation status (P < 0.001). PD-L1 expression levels were significantly different from those determined using the International Association for the Study of Lung Cancer (IASLC) grading system (P < 0.001). CONCLUSIONS PD-L1 expression was significantly associated with radiological and pathological invasiveness and driver mutation status in subcentimeter pulmonary nodules. The significance of PD-L1 expression in the evolution of early-stage lung adenocarcinoma requires further investigation.
Collapse
Affiliation(s)
- Xiongwen Yang
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yi Xiao
- Department of Cardio-Thoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hao Hu
- Department of Radiation Therapy, General Hospital of Southern Theater Command, Guangdong, China
| | - Zhen-Bin Qiu
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yi-Fan Qi
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Meng-Min Wang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wen-Zhao Zhong
- School of Medicine, South China University of Technology, Guangzhou, China.
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
63
|
Terada Y, Isaka M, Ono A, Kawata T, Serizawa M, Mori K, Muramatsu K, Tone K, Kenmotsu H, Ohshima K, Urakami K, Nagashima T, Kusuhara M, Akiyama Y, Sugino T, Takahashi T, Ohde Y. Prognostic significance of tumor microenvironment assessed by machine learning algorithm in surgically resected non-small cell lung cancer. Cancer Rep (Hoboken) 2023:e1926. [PMID: 37903603 DOI: 10.1002/cnr2.1926] [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: 03/31/2023] [Revised: 09/16/2023] [Accepted: 10/16/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND A methodology to assess the immune microenvironment (IME) of non-small cell lung cancer (NSCLC) has not been established, and the prognostic impact of IME factors is not yet clear. AIMS This study aimed to assess the IME factors and evaluate their prognostic values. METHODS AND RESULTS We assessed CD8+ tumor-infiltrating lymphocyte (TIL) density, forkhead box protein P3+ (Foxp3+ ) TIL density, and programmed death receptor ligand-1 (PD-L1) tumor proportion score (TPS) using a machine-learning algorithm in whole-slide imaging (WSI). We dichotomized patients according to TIL density or TPS and compared their clinical outcomes. Between September 2014 and September 2015, 165 patients with NSCLC were enrolled in the study. We assessed IME factors in the epithelium, stroma, and their combination. An improvement in disease-free survival (DFS) was observed in the high CD8+ TIL density group in the epithelium, stroma, and the combination of both. Moreover, the group with high PD-L1 TPS in the epithelium showed better DFS than that with low PD-L1 TPS. In the multivariate analysis, the CD8+ TIL density in the combination of epithelium and stroma and PD-L1 TPS in the epithelium were independent prognostic factors (hazard ratio [HR] = 0.43; 95% confidence interval [CI] = 0.26-0.72; p = .001, HR = 0.49; 95% CI = 0.30-0.81; p = .005, respectively). CONCLUSION Our approach demonstrated that the IME factors are related to survival in patients with NSCLC. The quantitative assessment of IME factors enables to discriminate patients with high risk of recurrence, who can be the candidates for adjuvant therapy. Assessing the CD8+ TIL density in the combination of epithelium and stroma might be more useful than their individual assessment because it is a simple and time-saving analysis of TILs in WSI.
Collapse
Affiliation(s)
- Yukihiro Terada
- Division of Thoracic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
- Division of Thoracic Surgery, Shinshu University School of Medicine, Nagano, Japan
| | - Mitsuhiro Isaka
- Division of Thoracic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Akira Ono
- Division of Thoracic Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Takuya Kawata
- Division of Pathology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Masakuni Serizawa
- Drug Discovery and Development Division, Research Institute, Shizuoka Cancer Center, Shizuoka, Japan
| | - Keita Mori
- Clinical Research Center, Shizuoka Cancer Center, Shizuoka, Japan
| | - Koji Muramatsu
- Division of Pathology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Kiyoshi Tone
- Division of Pathology, Shizuoka Cancer Center, Shizuoka, Japan
| | | | - Keiichi Ohshima
- Medical Genetics Division, Research Institute, Shizuoka Cancer Center, Shizuoka, Japan
| | - Kenichi Urakami
- Cancer Diagnostics Research Division, Research Institute, Shizuoka Cancer Center, Shizuoka, Japan
| | - Takeshi Nagashima
- Cancer Diagnostics Research Division, Research Institute, Shizuoka Cancer Center, Shizuoka, Japan
- SRL Inc, Tokyo, Japan
| | - Masatoshi Kusuhara
- Region Resources Division, Research Institute, Shizuoka Cancer Center, Shizuoka, Japan
| | - Yasuto Akiyama
- Immunotherapy Division, Research Institute, Shizuoka Cancer Center, Shizuoka, Japan
| | - Takashi Sugino
- Division of Pathology, Shizuoka Cancer Center, Shizuoka, Japan
| | | | - Yasuhisa Ohde
- Division of Thoracic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
64
|
Zhang J, Guan M, Lv M, Liu Y, Zhang H, Zhang Z, Zhang K. Localized Imaging of Programmed Death-Ligand 1 on Individual Tumor-Derived Extracellular Vesicles for Prediction of Immunotherapy Response. ACS NANO 2023; 17:20120-20134. [PMID: 37819165 DOI: 10.1021/acsnano.3c05799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Programmed death-ligand 1 (PD-L1) on tumor-derived small extracellular vesicles (EVs) is a biomarker for prediction of the immunotherapy response. However, conventional bulk measurement can hardly analyze the expression of PD-L1 on individual tumor-derived EVs. Herein, a method for localized imaging of tumor-derived individual EVs PD-L1 (LITIE) is developed. In this assay, EVs in plasma were directly captured on a biochip. Then the liposome-mediated membrane fusion strategy was used to image miR-21 in EVs to discriminate miR-21-positive EVs from the whole EVs populations. Subsequently, the primer exchange reaction (PER) is applied to generate localized and amplified fluorescent signals for imaging PD-L1 on identified tumor-derived EVs. When applied in clinical sample tests, the LITIE assay could effectively distinguish breast cancer patients from healthy donors or patients with benign tumors. Interestingly, in a mice melanoma model, the LITIE assay showed the ability to predict immunotherapy response even before drug treatment. Thus, we think the strategy of measuring individual tumor-derived EVs PD-L1 could serve as an alternative way for screening clinical responders suitable for immunotherapy.
Collapse
Affiliation(s)
- Junli Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Mengting Guan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Min Lv
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yingying Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hongling Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| |
Collapse
|
65
|
Cui Q, Li W, Wang D, Wang S, Yu J. Prognostic significance of blood-based PD-L1 analysis in patients with non-small cell lung cancer undergoing immune checkpoint inhibitor therapy: a systematic review and meta-analysis. World J Surg Oncol 2023; 21:318. [PMID: 37821941 PMCID: PMC10566159 DOI: 10.1186/s12957-023-03215-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/03/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND The main types of PD-L1 in the blood include soluble PD-L1 (sPD-L1), exosomal PD-L1 (exoPD-L1), and PD-L1 in circulating tumor cells (CTCs). However, the predictive and prognostic values of these three indicators in patients with non-small cell lung cancer (NSCLC) undergoing immune checkpoint inhibitor (ICI) therapy are unclear, warranting a systematic meta-analysis. METHODS A systematic literature search was performed in the PubMed, Cochrane Library, and Embase databases. The pooled hazard ratio (HR) and 95% confidence interval (CI) values were extracted from the included studies to investigate the correlation between the three PD-L1 indicators and overall survival (OS) or progression-free survival (PFS). The Newcastle-Ottawa Scale (NOS) was used to examine the quality of the included studies. Subgroup analyses were employed to investigate the heterogeneity. The publication bias of the included studies was assessed using Begg's and Egger's tests. P < 0.05 was regarded as significantly different. RESULTS The pooled results revealed that high pre-treatment sPD-L1 levels were significantly associated with inferior OS (HR = 2.32, 95% CI = 1.68-3.18, P < 0.001) and PFS (HR = 2.52, 95% CI = 1.72-3.68, P < 0.001). However, dynamic changes in sPD-L1 after immunotherapy were not statistically significant for OS (HR = 1.46, 95% CI = 0.65-3.26, P > 0.05) or PFS (HR = 1.62, 95% CI = 0.92-2.86, P > 0.05). Meanwhile, the upregulated pre-treatment exoPD-L1 levels were significantly associated with poor PFS (HR = 4.44, 95% CI = 2.87-6.89, P < 0.001), whereas the post-treatment dynamic upregulation of exoPD-L1 was significantly correlated with superior PFS (HR = 0.36, 95% CI = 0.24-0.54, P < 0.001) and OS (HR = 0.20, 95% CI = 0.07-0.53, P < 0.001). For PD-L1 in CTCs, the pooled results indicated that PD-L1 expression in CTCs was not significantly correlated with OS (HR = 0.75, 95% CI = 0.49-1.13, P = 0.170) and PFS (HR = 0.79, 95% CI = 0.59-1.06, P = 0.12). CONCLUSIONS Blood-based PD-L1 analysis is a potential strategy for predicting treatment efficacy and prognosis in patients with cancer.
Collapse
Affiliation(s)
- Qian Cui
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wentao Li
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Dong Wang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuangcui Wang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jianchun Yu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
66
|
Cai S, Deng Y, Wang Z, Zhu J, Huang C, Du L, Wang C, Yu X, Liu W, Yang C, Wang Z, Wang L, Ma K, Huang R, Zhou X, Zou H, Zhang W, Huang Y, Li Z, Qin T, Xu T, Guo X, Yu Z. Development and clinical validation of a microfluidic-based platform for CTC enrichment and downstream molecular analysis. Front Oncol 2023; 13:1238332. [PMID: 37849806 PMCID: PMC10578963 DOI: 10.3389/fonc.2023.1238332] [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: 06/11/2023] [Accepted: 08/29/2023] [Indexed: 10/19/2023] Open
Abstract
Background Although many CTC isolation and detection methods can provide information on cancer cell counts, downstream gene and protein analysis remain incomplete. Therefore, it is crucial to develop a technology that can provide comprehensive information on both the number and profile of CTC. Methods In this study, we developed a novel microfluidics-based CTC separation and enrichment platform that provided detailed information about CTC. Results This platform exhibits exceptional functionality, achieving high rates of CTC recovery (87.1%) and purification (∼4 log depletion of WBCs), as well as accurate detection (95.10%), providing intact and viable CTCs for downstream analysis. This platform enables successful separation and enrichment of CTCs from a 4 mL whole-blood sample within 15 minutes. Additionally, CTC subtypes, selected protein expression levels on the CTC surface, and target mutations in selected genes can be directly analyzed for clinical utility using immunofluorescence and real-time polymerase chain reaction, and the detected PD-L1 expression in CTCs is consistent with immunohistochemical assay results. Conclusion The microfluidic-based CTC enrichment platform and downstream molecular analysis together provide a possible alternative to tissue biopsy for precision cancer management, especially for patients whose tissue biopsies are unavailable.
Collapse
Affiliation(s)
- Songhua Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Youjun Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Zhe Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Junyu Zhu
- Institute of Cancer Control, Cancer Hospital of Xinjiang Medical University, Urumqi, China
| | - Chujian Huang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Longde Du
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Chunguang Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Xiangyang Yu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Wenyi Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Chenglin Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Zhe Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Lixu Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Kai Ma
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Rui Huang
- Shenzhen Futian Research Institute, City University of Hong Kong, Shenzhen, China
| | - Xiaoyu Zhou
- Department of Medical Affairs, Cellomics (ShenZhen) Limited, Shenzhen, China
| | - Heng Zou
- Department of Medical Affairs, Cellomics (ShenZhen) Limited, Shenzhen, China
| | - Wenchong Zhang
- Department of Medical Affairs, Cellomics (ShenZhen) Limited, Shenzhen, China
| | - Yan Huang
- Department of Medical Affairs, Cellomics (ShenZhen) Limited, Shenzhen, China
| | - Zhi Li
- Department of Medical Affairs, Cellomics (ShenZhen) Limited, Shenzhen, China
| | - Tiaoping Qin
- Department of Medical Affairs, Cellomics (ShenZhen) Limited, Shenzhen, China
| | - Tao Xu
- Department of Medical Affairs, Cellomics (ShenZhen) Limited, Shenzhen, China
| | - Xiaotong Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Zhentao Yu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| |
Collapse
|
67
|
Zhou C, Srivastava MK, Xu H, Felip E, Wakelee H, Altorki N, Reck M, Liersch R, Kryzhanivska A, Oizumi S, Tanaka H, Hamm J, McCune SL, Bennett E, Gitlitz B, McNally V, Ballinger M, McCleland M, Zou W, Das Thakur M, Novello S. Comparison of SP263 and 22C3 immunohistochemistry PD-L1 assays for clinical efficacy of adjuvant atezolizumab in non-small cell lung cancer: results from the randomized phase III IMpower010 trial. J Immunother Cancer 2023; 11:e007047. [PMID: 37903590 PMCID: PMC10619123 DOI: 10.1136/jitc-2023-007047] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Tumor samples from the phase III IMpower010 study were used to compare two programmed death-ligand 1 (PD-L1) immunohistochemistry assays (VENTANA SP263 and Dako 22C3) for identification of PD-L1 patient subgroups (negative, positive, low, and high expression) and their predictive value for adjuvant atezolizumab compared with best supportive care (BSC) in resectable early-stage non-small cell lung cancer (NSCLC). METHODS PD-L1 expression was assessed by the SP263 assay, which measured the percentage of tumor cells with any membranous PD-L1 staining, and the 22C3 assay, which scored the percentage of viable tumor cells showing partial or complete membranous PD-L1 staining. RESULTS When examining the concordance at the PD-L1-positive threshold (SP263: tumor cell (TC)≥1%; 22C3: tumor proportion score (TPS)≥1%), the results were concordant between assays for 83% of the samples. Similarly, at the PD-L1-high cut-off (SP263: TC≥50%; 22C3: TPS≥50%), the results were concordant between assays for 92% of samples. The disease-free survival benefit of atezolizumab over BSC was comparable between assays for PD-L1-positive (TC≥1% by SP263: HR, 0.58 (95% CI: 0.40 to 0.85) vs TPS≥1% by 22C3: HR, 0.65 (95% CI: 0.45 to 0.95)) and PD-L1-high (TC≥50% by SP263: HR, 0.27 (95% CI: 0.14 to 0.53) vs TPS≥50% by 22C3: HR, 0.31 (95% CI: 0.16 to 0.60)) subgroups. CONCLUSIONS The SP263 and 22C3 assays showed high concordance and a comparable clinical predictive value of atezolizumab at validated PD-L1 thresholds, suggesting that both assays can identify patients with early-stage NSCLC most likely to experience benefit from adjuvant atezolizumab. TRIAL REGISTRATION NUMBER NCT02486718.
Collapse
Affiliation(s)
- Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University Affiliated Shanghai Pulmonary Hospital, Shanghai, China
| | - Minu K Srivastava
- Oncology Biomarker Development, Genentech Inc, South San Francisco, California, USA
| | - Hao Xu
- F. Hoffman-La Roche Ltd, Mississauga, Ontario, Canada
| | - Enriqueta Felip
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Heather Wakelee
- Stanford University School of Medicine, Stanford Cancer Institute, Stanford, California, USA
| | - Nasser Altorki
- Department of Cardiothoracic Surgery, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany
| | - Rüdiger Liersch
- Practice for Hematology and Medical Oncology Clemenshospital Münster, Münster, Germany
| | - Anna Kryzhanivska
- Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine
| | - Satoshi Oizumi
- Department of Respiratory Medicine, National Hospital Organization Hokkaido Cancer Center, Sapporo, Japan
| | - Hiroshi Tanaka
- Department of Internal Medicine, Niigata Cancer Center Hospital, Niigata, Japan
| | - John Hamm
- Department of Medical Oncology, Norton Cancer Institute, Louisville, Kentucky, USA
| | | | - Elizabeth Bennett
- Product Development, Genentech Inc, South San Francisco, California, USA
| | - Barbara Gitlitz
- Product Development, Genentech Inc, South San Francisco, California, USA
| | | | - Marcus Ballinger
- Product Development, Genentech Inc, South San Francisco, California, USA
| | - Mark McCleland
- Oncology Biomarker Development, Genentech Inc, South San Francisco, California, USA
- Amunix, South San Francisco, California, USA
| | - Wei Zou
- Oncology Biomarker Development, Genentech Inc, South San Francisco, California, USA
| | - Meghna Das Thakur
- Oncology Biomarker Development, Genentech Inc, South San Francisco, California, USA
- Gilead Sciences, Foster City, CA, USA
| | - Silvia Novello
- University of Turin, AOU San Luigi Gonzaga, Orbassano, Turin, Italy
| |
Collapse
|
68
|
Martinez-Morilla S, Moutafi M, Fernandez AI, Jessel S, Divakar P, Wong PF, Garcia-Milian R, Schalper KA, Kluger HM, Rimm DL. Digital spatial profiling of melanoma shows CD95 expression in immune cells is associated with resistance to immunotherapy. Oncoimmunology 2023; 12:2260618. [PMID: 37781235 PMCID: PMC10540659 DOI: 10.1080/2162402x.2023.2260618] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 09/14/2023] [Indexed: 10/03/2023] Open
Abstract
Although immune checkpoint inhibitor (ICI) therapy has dramatically improved outcome for metastatic melanoma patients, many patients do not benefit. Since adverse events may be severe, biomarkers for resistance would be valuable, especially in the adjuvant setting. We performed high-plex digital spatial profiling (DSP) using the NanoString GeoMx® on 53 pre-treatment specimens from ICI-treated metastatic melanoma cases. We interrogated 77 targets simultaneously in four molecular compartments defined by S100B for tumor, CD68 for macrophages, CD45 for leukocytes, and nonimmune stromal cells defined as regions negative for all three compartment markers but positive for SYTO 13. For DSP validation, we confirmed the results obtained for some immune markers, such as CD8, CD4, CD20, CD68, CD45, and PD-L1, by quantitative immunofluorescence (QIF). In the univariable analysis, 38 variables were associated with outcome, 14 of which remained significant after multivariable adjustment. Among them, CD95 was further validated using multiplex immunofluorescence in the Discovery immunotherapy (ITX) Cohort and an independent validation cohort with similar characteristics, showing an association between high levels of CD95 and shorter progression-free survival. We found that CD95 in stroma was associated with resistance to ICI. With further validation, this biomarker could have value to select patients that will not benefit from immunotherapy.
Collapse
Affiliation(s)
| | - Myrto Moutafi
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | | | - Shlomit Jessel
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Pok Fai Wong
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Rolando Garcia-Milian
- Bioinformatics Support Program, Cushing/Whitney Medical Library, Yale School of Medicine, New Haven, CT, USA
| | - Kurt A. Schalper
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Harriet M. Kluger
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - David L. Rimm
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
69
|
Roque K, Ruiz R, Mas L, Pozza DH, Vancini M, Silva Júnior JA, de Mello RA. Update in Immunotherapy for Advanced Non-Small Cell Lung Cancer: Optimizing Treatment Sequencing and Identifying the Best Choices. Cancers (Basel) 2023; 15:4547. [PMID: 37760516 PMCID: PMC10526179 DOI: 10.3390/cancers15184547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/09/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
The introduction of immunotherapy has brought about a paradigm shift in the management of advanced non-small cell lung cancer (NSCLC). It has not only significantly improved the prognosis of patients but has also become a cornerstone of treatment, particularly in those without oncogenic driver mutations. Immune checkpoint inhibitors (ICIs) play a crucial role in the treatment of lung cancer and can be classified into two main groups: Anti-cytotoxic T lymphocyte antigen-4 (Anti-CTLA-4) and anti-T-cell receptor programmed cell death-1 or its ligand (Anti-PD-1 and Anti-PD-L1). Certainly, the landscape of approved first line immunotherapeutic approaches has expanded to encompass monotherapy, immunotherapy-exclusive protocols, and combinations with chemotherapy. The complexity of decision-making in this realm arises due to the absence of direct prospective comparisons. However, a thorough analysis of the long-term efficacy and safety data derived from pivotal clinical trials can offer valuable insights into optimizing treatment for different patient subsets. Moreover, ongoing research is investigating emerging biomarkers and innovative therapeutic strategies that could potentially refine the current treatment approach even further. In this comprehensive review, our aim is to highlight the latest advances in immunotherapy for advanced NSCLC, including the mechanisms of action, efficacy, safety profiles, and clinical significance of ICI.
Collapse
Affiliation(s)
- Katia Roque
- Discipline of Medical Oncology, Post-Graduation Programme in Medicine, Faculty of Medicine, Nine of July University (UNINOVE), São Paulo 04101-000, Brazil (J.A.S.J.)
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Angamos Este Av., 2520, Lima 15023, Peru; (R.R.); (L.M.)
- Faculty of Medicine, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
- Faculty of Medicine, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru
| | - Rossana Ruiz
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Angamos Este Av., 2520, Lima 15023, Peru; (R.R.); (L.M.)
- Escuela Profesional de Medicina Humana-Filial Ica, Universidad Privada San Juan Bautista, Ica 15067, Peru
| | - Luis Mas
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Angamos Este Av., 2520, Lima 15023, Peru; (R.R.); (L.M.)
- Faculty of Medicine, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
- Department of Medical Oncology, Oncosalud-AUNA, Av. Guardia Civil 571-San Borja, Lima 15036, Peru
| | - Daniel Humberto Pozza
- Experimental Biology Unit, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-319 Porto, Portugal
- i3S—Institute for Research and Innovation in Health and IBMC, University of Porto, 4200-319 Porto, Portugal
| | - Marina Vancini
- Discipline of Medical Oncology, Post-Graduation Programme in Medicine, Faculty of Medicine, Nine of July University (UNINOVE), São Paulo 04101-000, Brazil (J.A.S.J.)
| | - José Antônio Silva Júnior
- Discipline of Medical Oncology, Post-Graduation Programme in Medicine, Faculty of Medicine, Nine of July University (UNINOVE), São Paulo 04101-000, Brazil (J.A.S.J.)
| | - Ramon Andrade de Mello
- Discipline of Medical Oncology, Post-Graduation Programme in Medicine, Faculty of Medicine, Nine of July University (UNINOVE), São Paulo 04101-000, Brazil (J.A.S.J.)
- Oxford Cancer Centre, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Department of Oncology, University of Oxford, Oxford OX1 2JD, UK
| |
Collapse
|
70
|
Cekani E, Martorell C, Martucci F, Patella M, Cafarotti S, Valenti A, Freguia S, Molinari F, Froesch P, Frattini M, Stathis A, Wannesson L. Prognostic implication of PD-L1 in early-stage non-small cell lung cancer: a retrospective single-centre study. Swiss Med Wkly 2023; 153:40110. [PMID: 37769653 DOI: 10.57187/smw.2023.40110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND The prognostic role of programmed death-ligand 1 (PD-L1) expression in patients with localised and locally advanced non-small cell lung cancer has not been fully elucidated. This information could help to better interpret recent and upcoming results of phase III adjuvant or neoadjuvant anti-PD-1/PD-L1 immunotherapy studies. METHODS In a cohort of 146 patients with early or locally advanced non-small cell lung cancer treated with curative intent (by surgery or radiotherapy), we investigated the prognostic value of PD-L1 expression and its correlation with other biological and clinical features. PD-L1 expression was stratified by quartiles. Primary endpoints were overall and disease-free survival. We also analysed the prognostic impact of the presence of actionable mutations, implemented treatment modality and completion of the treatment plan. Neither type of patient received neoadjuvant or adjuvant immunotherapy or target therapy. RESULTS Of the 146 selected patients, 32 (21.9%) presented disease progression and 15 died (10.3%) at a median follow-up of 20 months. In a univariable analysis, PD-L1 expression ≥25% was associated with significantly lower disease-free survival (hazard ratio [HR]) 1.9, 95% confidence interval [CI] 1.0-3.9, p = 0.049). PD-L1 expression ≥50% did not lead to disease-free survival or overall survival benefits (HR 1.2 and 1.1, respectively; 95% CI 0.6-2.6 and 0.3-3.4, respectively; pnot significant). In a multivariate analysis, a stage >I (HR 2.7, 95% CI 1.2-6, p = 0.012) and having an inoperable tumour (HR 3.2, 95% CI 1.4-7.4, p = 0.005) were associated with lower disease-free survival. CONCLUSION The population of patients with early-stage non-small cell lung cancer and PD-L1 expression ≥25% who were treated with curative intent during the pre-immunotherapy era exhibited a worse prognosis. This finding provides justification for the utilisation of adjuvant immunotherapy in this subgroup of patients, based on the current evidence derived from disease-free survival outcomes. However, for patients with PD-L1 expression <25%, opting to wait for the availability of the overall survival results may be a prudent choice.
Collapse
Affiliation(s)
- Elona Cekani
- Istituto Oncologico della Svizzera Italiana (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | - Carolina Martorell
- Istituto Oncologico della Svizzera Italiana (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | - Francesco Martucci
- Istituto Oncologico della Svizzera Italiana (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | - Miriam Patella
- Thoracic Surgery Department, Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | - Stefano Cafarotti
- Thoracic Surgery Department, Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | - Antonio Valenti
- Pneumology Department, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
| | | | | | - Patrizia Froesch
- Istituto Oncologico della Svizzera Italiana (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | | | - Anastasios Stathis
- Istituto Oncologico della Svizzera Italiana (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | - Luciano Wannesson
- Istituto Oncologico della Svizzera Italiana (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| |
Collapse
|
71
|
Ohkuma R, Fujimoto Y, Ieguchi K, Onishi N, Watanabe M, Takayanagi D, Goshima T, Horiike A, Hamada K, Ariizumi H, Hirasawa Y, Ishiguro T, Suzuki R, Iriguchi N, Tsurui T, Sasaki Y, Homma M, Yamochi T, Yoshimura K, Tsuji M, Kiuchi Y, Kobayashi S, Tsunoda T, Wada S. Monocyte subsets associated with the efficacy of anti‑PD‑1 antibody monotherapy. Oncol Lett 2023; 26:381. [PMID: 37559573 PMCID: PMC10407861 DOI: 10.3892/ol.2023.13967] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/26/2023] [Indexed: 08/11/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are among the most notable advances in cancer immunotherapy; however, reliable biomarkers for the efficacy of ICIs are yet to be reported. Programmed death (PD)-ligand 1 (L1)-expressing CD14+ monocytes are associated with shorter overall survival (OS) time in patients with cancer treated with anti-PD-1 antibodies. The present study focused on the classification of monocytes into three subsets: Classical, intermediate and non-classical. A total of 44 patients with different types of cancer treated with anti-PD-1 monotherapy (pembrolizumab or nivolumab) were enrolled in the present study. The percentage of each monocyte subset was investigated, and the percentage of cells expressing PD-L1 or PD-1 within each of the three subsets was further analyzed. Higher pretreatment classical monocyte percentages were correlated with shorter OS (r=-0.32; P=0.032), whereas higher non-classical monocyte percentages were correlated with a favorable OS (r=0.39; P=0.0083). PD-L1-expressing classical monocytes accounted for a higher percentage of the total monocytes than non-classical monocytes with PD-L1 expression. In patients with non-small cell lung cancer (NSCLC), a higher percentage of PD-L1-expressing classical monocytes was correlated with shorter OS (r=-0.60; P=0.012), which is similar to the observation for the whole patient cohort. Comparatively, higher percentages of non-classical monocytes expressing PD-L1 were significantly associated with better OS, especially in patients with NSCLC (r=0.60; P=0.010). Moreover, a higher percentage of non-classical monocytes contributed to prolonged progression-free survival in patients with NSCLC (r=0.50; P=0.042), with similar results for PD-L1-expressing non-classical monocytes. The results suggested that the percentage of monocyte subsets in patients with cancer before anti-PD-1 monotherapy may predict the treatment efficacy and prognosis. Furthermore, more classical monocytes and fewer non-classical monocytes, especially those expressing PD-L1, are involved in shortening OS time, which may indicate the poor efficiency of anti-PD-1 treatment approaches.
Collapse
Affiliation(s)
- Ryotaro Ohkuma
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
| | - Yuki Fujimoto
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
| | - Katsuaki Ieguchi
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
| | - Nobuyuki Onishi
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
| | - Makoto Watanabe
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo 142-8555, Japan
| | - Daisuke Takayanagi
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
| | - Tsubasa Goshima
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
| | - Atsushi Horiike
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Kazuyuki Hamada
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Hirotsugu Ariizumi
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Yuya Hirasawa
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Tomoyuki Ishiguro
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Risako Suzuki
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Nana Iriguchi
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Toshiaki Tsurui
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Yosuke Sasaki
- Department of Pathology, Showa University School of Medicine, Tokyo 157-8577, Japan
| | - Mayumi Homma
- Department of Pathology, Showa University School of Medicine, Tokyo 157-8577, Japan
| | - Toshiko Yamochi
- Department of Pathology, Showa University School of Medicine, Tokyo 157-8577, Japan
| | - Kiyoshi Yoshimura
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
- Department of Clinical Immuno-oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
| | - Mayumi Tsuji
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo 142-8555, Japan
| | - Yuji Kiuchi
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo 142-8555, Japan
| | - Shinichi Kobayashi
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
| | - Takuya Tsunoda
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Satoshi Wada
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo 157-8577, Japan
| |
Collapse
|
72
|
Zhang K, Huang Z, Wang Y, Xue J, Liang N, Wei Z. Rapid determination of the presence of EGFR mutations with DNA-based nanocalipers. NANOSCALE 2023; 15:13834-13841. [PMID: 37580989 DOI: 10.1039/d3nr02665e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
Selecting 1st-line treatment for lung cancer is currently a binary choice, either chemotherapy or targeted medicine, depending on whether EGFR mutations exist. Next-generation sequencing is fully capable of accurately identifying EGFR mutations and guiding the usage of tyrosine kinase inhibitors, but it is highly expensive. Moreover, as the sequencing is not helpful for patients with wild-type EGFR, the long wait for sequencing may delay the chemotherapy and correspondingly increase the risks of cancer progression. To address this issue, a new method for rapidly determining the presence of EGFR mutations is developed in this study. A series of DNA origami-engineered nanocalipers are designed and constructed to determine the EGFR spatial distribution of either mutated EGFR or wild-type EGFR lung cancer cells. The experimental results on cancer cell lines and 9 clinical tissue samples show that compared with wild-type EGFR cells, mutated EGFR cells have narrower EGFR spacing. Hence, the DNA nanocalipers are demonstrated to be capable of determining the presence of EGFR mutations and shrinking the detection period from weeks to hours, compared with sequencing. For determining EGFR mutation status in 9 clinical samples, DNA nanocalipers show 100% consistency with next-generation sequencing.
Collapse
Affiliation(s)
- Kexin Zhang
- Department of Biomedical Engineering, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Zhicheng Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yadong Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianchao Xue
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Zewen Wei
- Department of Biomedical Engineering, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
73
|
Jia K, Chen Y, Xie Y, Chong X, Li Y, Wu Y, Yuan J, Li Y, Feng X, Hu Y, Sun Y, Gong J, Zhang X, Li J, Shen L. Multidimensional immune profiling in Gastric Cancer Multiplex Immunohistochemistry Atlas from Peking University Cancer Hospital project informs PD-1/PD-L1 blockade efficacy. Eur J Cancer 2023; 189:112931. [PMID: 37343324 DOI: 10.1016/j.ejca.2023.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND Immunotherapy has resulted in impressive objective response rates and durable tumour remission, but only in a subset of gastric cancer (GC) patients. The PD-L1 combined positive score is the most widely used tissue-based biomarker for anti-PD-1/PD-L1 therapy; however, this unidimensional method has limitations. Next-generation exploration of tissue-based biomarkers for GC requires characterisation of various cellular markers and key immunoregulatory molecule expression in situ. Thus, a complete, stepwise solution covering the entire process from staining samples to cross-site utilisation of pathomics data is urgently needed. METHODS With the advanced multispectral imaging analysis method, web-based data repository, and interactive sharing technology, we conducted a project entitled Gastric Cancer Multiplex Immunohistochemistry Atlas from Peking University Cancer Hospital (GMAP). We propose a standard pipeline covering sample collection, staining, scanning multispectral images, constructing a spectral library, identifying and phenotyping cells, positioning each element, and quantitatively extracting immune features. We designed an open-access relational database to explore tissue-based biomarkers to determine PD-1/PD-L1 blockade efficacy. RESULTS The GMAP project detected the functional status and spatial location of more than 50 million cells using 15 markers in 80 GC patients, based on which billions of cell pairs were recognised, highlighting the rich spatial arrangement information and the fine tumour microenvironment structure. We generated a tumour-immune atlas using the count and spatial features of 65 immune cell types. We eventually selected the indicators and built a comprehensive risk-scoring system. Patients with higher risk score showed superior immunotherapy-related progression-free survival (irPFS) (hazard ratio [HR]: 3.19; P < 0.001; median irPFS: 4.87 versus 19.87months, respectively) and immunotherapy-related overall survival (HR: 3.10; P = 0.001; median irPFS: 10.03 versus 24.87months, respectively) compared with lower risk patients, demonstrating their potential for guiding anti-PD-1/PD-L1-based immunotherapy. Importantly, an easy-to-use and versatile web server was built to promote tissue-based biomarker exploration in GC. CONCLUSION The GMAP project highlighted the clinical value of tissue-based immune features as biomarkers for immunotherapeutic decision-making. We present a well-designed, detailed workflow for the orderly generation and use of a high-quality, spatially resolved pathological database.
Collapse
Affiliation(s)
- Keren Jia
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yi Xie
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoyi Chong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yilin Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yingcheng Wu
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanyan Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xujiao Feng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yajie Hu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yu Sun
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
74
|
Porta FM, Sajjadi E, Venetis K, Frascarelli C, Cursano G, Guerini-Rocco E, Fusco N, Ivanova M. Immune Biomarkers in Triple-Negative Breast Cancer: Improving the Predictivity of Current Testing Methods. J Pers Med 2023; 13:1176. [PMID: 37511789 PMCID: PMC10381494 DOI: 10.3390/jpm13071176] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Triple-negative breast cancer (TNBC) poses a significant challenge in terms of prognosis and disease recurrence. The limited treatment options and the development of resistance to chemotherapy make it particularly difficult to manage these patients. However, recent research has been shifting its focus towards biomarker-based approaches for TNBC, with a particular emphasis on the tumor immune landscape. Immune biomarkers in TNBC are now a subject of great interest due to the presence of tumor-infiltrating lymphocytes (TILs) in these tumors. This characteristic often coincides with the presence of PD-L1 expression on both neoplastic cells and immune cells within the tumor microenvironment. Furthermore, a subset of TNBC harbor mismatch repair deficient (dMMR) TNBC, which is frequently accompanied by microsatellite instability (MSI). All of these immune biomarkers hold actionable potential for guiding patient selection in immunotherapy. To fully capitalize on these opportunities, the identification of additional or complementary biomarkers and the implementation of highly customized testing strategies are of paramount importance in TNBC. In this regard, this article aims to provide an overview of the current state of the art in immune-related biomarkers for TNBC. Specifically, it focuses on the various testing methodologies available and sheds light on the immediate future perspectives for patient selection. By delving into the advancements made in understanding the immune landscape of TNBC, this study aims to contribute to the growing body of knowledge in the field. The ultimate goal is to pave the way for the development of more personalized testing strategies, ultimately improving outcomes for TNBC patients.
Collapse
Affiliation(s)
- Francesca Maria Porta
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
| | - Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
| | - Chiara Frascarelli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
| |
Collapse
|
75
|
LIAM CK, YEW CY, PANG YK, WONG CK, POH ME, TAN JL, SOO CI, LOH TC, CHIN KK, MUNUSAMY V, LIAM YS, IBRAHIM NH. Common driver mutations and programmed death-ligand 1 expression in advanced non-small cell lung cancer in smokers and never smokers. BMC Cancer 2023; 23:659. [PMID: 37452277 PMCID: PMC10347799 DOI: 10.1186/s12885-023-11156-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 07/06/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND In non-small cell lung cancer (NSCLC), there may be a relationship between programmed death-ligand 1 (PD-L1) expression, driver mutations and cigarette smoking. METHODS In this single-center retrospective study, the relationship between common driver mutations (EGFR mutation and ALK rearrangement) and PD-L1 expression in advanced NSCLC according to the patients' smoking history was examined. Light, moderate and heavy smokers had smoked < 20, 20-39, and ≥ 40 pack-years, respectively. The level of PD-L1 expression, assessed using Ventana SP263 monoclonal antibody assay, was defined by the tumor proportion score (TPS) as high expression (TPS ≥ 50%), low expression (TPS 1%-49%) and no expression (TPS < 1%). RESULTS 101 (52.9%) of 191 advanced NSCLC patients were never smokers. EGFR mutations were more common in never smokers (64.4%) than in smokers (17.8%) with advanced NSCLC (P < 0.0001). A higher proportion of smokers (26.7%) had high PD-L1 expression compared to never smokers (13.9%) (P = 0.042). There was a trend for a higher proportion of male NSCLC patients [28 of 115 (24.3%)] than female patients [10 of 76 (13.2%)] to have high PD-L1 expression (P = 0.087]. High PD-L1 expression was seen in 32 of 110 (29.1%) patients with EGFR wild-type NSCLC but only in 6 of 81 (7.4%) patients with EGFR-mutant tumors (P < 0.0001). Among the 90 smokers with NSCLC, a higher proportion of heavy smokers (35.8%) than non-heavy smokers (13.5%) had high PD-L1 expression (P = 0.034). In patients with adenocarcinoma, high PD-L1 expression was seen in 25 of 77 (32.5%) patients with EGFR wild-type tumors but only in 4 of 70 (5.7%) patients with EGFR-mutant tumors (P < 0.0001). Among patients with adenocarcinoma, a significantly higher proportion of ever smokers (29.3%) than never smokers (13.5%) had high PD-L1 expression (P = 0.032). Among smokers with adenocarcinoma, a significantly higher proportion of heavy smokers (44.1%) than non-heavy smokers (8.3%) had high PD-L1 expression (P = 0.004). On multivariate analysis, after adjusting for gender and smoking status, heavy smoking and EGFR wild-type tumors remained significantly associated with high PD-L1 expression in NSCLCs and also in adenocarcinoma. CONCLUSIONS Heavy smoking and EGFR wild-type tumors were significantly associated with high PD-L1 expression in NSCLCs and also in adenocarcinoma.
Collapse
Affiliation(s)
- Chong Kin LIAM
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Department of Medicine, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| | - Chian Yih YEW
- Department of Medicine, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| | - Yong Kek PANG
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Department of Medicine, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| | - Chee Kuan WONG
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Department of Medicine, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| | - Mau Ern POH
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Department of Medicine, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| | - Jiunn Liang TAN
- Department of Medicine, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| | - Chun Ian SOO
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Department of Medicine, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| | - Thian Chee LOH
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Department of Medicine, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| | - Ka Kiat CHIN
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Department of Medicine, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| | - Vijayan MUNUSAMY
- Department of Medicine, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| | - Yong Sheng LIAM
- Clinical Investigation Centre, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| | - Nur Husna IBRAHIM
- Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Department of Medicine, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| |
Collapse
|
76
|
Harms PW, Frankel TL, Moutafi M, Rao A, Rimm DL, Taube JM, Thomas D, Chan MP, Pantanowitz L. Multiplex Immunohistochemistry and Immunofluorescence: A Practical Update for Pathologists. Mod Pathol 2023; 36:100197. [PMID: 37105494 DOI: 10.1016/j.modpat.2023.100197] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 03/07/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023]
Abstract
Our understanding of the biology and management of human disease has undergone a remarkable evolution in recent decades. Improved understanding of the roles of complex immune populations in the tumor microenvironment has advanced our knowledge of antitumor immunity, and immunotherapy has radically improved outcomes for many advanced cancers. Digital pathology has unlocked new possibilities for the assessment and discovery of the tumor microenvironment, such as quantitative and spatial image analysis. Despite these advances, tissue-based evaluations for diagnosis and prognosis continue to rely on traditional practices, such as hematoxylin and eosin staining, supplemented by the assessment of single biomarkers largely using chromogenic immunohistochemistry (IHC). Such approaches are poorly suited to complex quantitative analyses and the simultaneous evaluation of multiple biomarkers. Thus, multiplex staining techniques have significant potential to improve diagnostic practice and immuno-oncology research. The different approaches to achieve multiplexed IHC and immunofluorescence are described in this study. Alternatives to multiplex immunofluorescence/IHC include epitope-based tissue mass spectrometry and digital spatial profiling (DSP), which require specialized platforms not available to most clinical laboratories. Virtual multiplexing, which involves digitally coregistering singleplex IHC stains performed on serial sections, is another alternative to multiplex staining. Regardless of the approach, analysis of multiplexed stains sequentially or simultaneously will benefit from standardized protocols and digital pathology workflows. Although this is a complex and rapidly advancing field, multiplex staining is now technically feasible for most clinical laboratories and may soon be leveraged for routine diagnostic use. This review provides an update on the current state of the art for tissue multiplexing, including the capabilities and limitations of different techniques, with an emphasis on potential relevance to clinical diagnostic practice.
Collapse
Affiliation(s)
- Paul W Harms
- Department of Pathology, Michigan Medicine/University of Michigan, Ann Arbor, Michigan; Department of Dermatology, Michigan Medicine/University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, Michigan Medicine/University of Michigan, Ann Arbor, Michigan.
| | - Timothy L Frankel
- Rogel Cancer Center, Michigan Medicine/University of Michigan, Ann Arbor, Michigan; Department of Surgery, Michigan Medicine/University of Michigan, Ann Arbor, Michigan
| | - Myrto Moutafi
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, Michigan Medicine/University of Michigan, Ann Arbor, Michigan; Department of Radiation Oncology, Michigan Medicine/University of Michigan, Ann Arbor, Michigan; Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Janis M Taube
- Department of Oncology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center, and Johns Hopkins Bloomberg-Kimmel Institute for Cancer Immunotherapy, Baltimore, Maryland; Department of Dermatology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center, and Johns Hopkins Bloomberg-Kimmel Institute for Cancer Immunotherapy, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center, and Johns Hopkins Bloomberg-Kimmel Institute for Cancer Immunotherapy, Baltimore, Maryland
| | - Dafydd Thomas
- Department of Pathology, Michigan Medicine/University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, Michigan Medicine/University of Michigan, Ann Arbor, Michigan
| | - May P Chan
- Department of Pathology, Michigan Medicine/University of Michigan, Ann Arbor, Michigan; Department of Dermatology, Michigan Medicine/University of Michigan, Ann Arbor, Michigan
| | - Liron Pantanowitz
- Department of Pathology, Michigan Medicine/University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
77
|
Bulen BJ, Khazanov NA, Hovelson DH, Lamb LE, Matrana M, Burkard ME, Yang ESH, Edenfield WJ, Claire Dees E, Onitilo AA, Buchschacher GL, Miller AM, Parsons BM, Wassenaar TR, Suga JM, Siegel RD, Irvin W, Nair S, Slim JN, Misleh J, Khatri J, Masters GA, Thomas S, Safa MM, Anderson DM, Mowers J, Dusenbery AC, Drewery S, Plouffe K, Reeder T, Vakil H, Patrias L, Falzetta A, Hamilton R, Kwiatkowski K, Johnson DB, Rhodes DR, Tomlins SA. Validation of Immunotherapy Response Score as Predictive of Pan-solid Tumor Anti-PD-1/PD-L1 Benefit. CANCER RESEARCH COMMUNICATIONS 2023; 3:1335-1349. [PMID: 37497337 PMCID: PMC10367935 DOI: 10.1158/2767-9764.crc-23-0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/16/2023] [Accepted: 06/29/2023] [Indexed: 07/28/2023]
Abstract
Immunotherapy response score (IRS) integrates tumor mutation burden (TMB) and quantitative expression biomarkers to predict anti-PD-1/PD-L1 [PD-(L)1] monotherapy benefit. Here, we evaluated IRS in additional cohorts. Patients from an observational trial (NCT03061305) treated with anti-PD-(L)1 monotherapy were included and assigned to IRS-High (-H) versus -Low (-L) groups. Associations with real-world progression-free survival (rwPFS) and overall survival (OS) were determined by Cox proportional hazards (CPH) modeling. Those with available PD-L1 IHC treated with anti-PD-(L)1 with or without chemotherapy were separately assessed. Patients treated with PD-(L)1 and/or chemotherapy (five relevant tumor types) were assigned to three IRS groups [IRS-L divided into IRS-Ultra-Low (-UL) and Intermediate-Low (-IL), and similarly assessed]. In the 352 patient anti-PD-(L)1 monotherapy validation cohort (31 tumor types), IRS-H versus IRS-L patients had significantly longer rwPFS and OS. IRS significantly improved CPH associations with rwPFS and OS beyond microsatellite instability (MSI)/TMB alone. In a 189 patient (10 tumor types) PD-L1 IHC comparison cohort, IRS, but not PD-L1 IHC nor TMB, was significantly associated with anti-PD-L1 rwPFS. In a 1,103-patient cohort (from five relevant tumor types), rwPFS did not significantly differ in IRS-UL patients treated with chemotherapy versus chemotherapy plus anti-PD-(L)1, nor in IRS-H patients treated with anti-PD-(L)1 versus anti-PD-(L)1 + chemotherapy. IRS associations were consistent across subgroups, including both Europeans and non-Europeans. These results confirm the utility of IRS utility for predicting pan-solid tumor PD-(L)1 monotherapy benefit beyond available biomarkers and demonstrate utility for informing on anti-PD-(L)1 and/or chemotherapy treatment. Significance This study confirms the utility of the integrative IRS biomarker for predicting anti-PD-L1/PD-1 benefit. IRS significantly improved upon currently available biomarkers, including PD-L1 IHC, TMB, and MSI status. Additional utility for informing on chemotherapy, anti-PD-L1/PD-1, and anti-PD-L1/PD-1 plus chemotherapy treatments decisions is shown.
Collapse
Affiliation(s)
| | | | | | | | - Marc Matrana
- Ochsner Cancer Institute, New Orleans, Louisiana
| | - Mark E. Burkard
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Eddy Shih-Hsin Yang
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | | | | | - Adedayo A. Onitilo
- Cancer Care and Research Center, Marshfield Clinic Research Institute, Marshfield, Wisconsin
| | | | | | | | | | | | | | | | - Suresh Nair
- Lehigh Valley Topper Cancer Institute, Allentown, Pennsylvania
| | | | | | - Jamil Khatri
- ChristianaCare Oncology Hematology, Newark, Delaware
| | - Gregory A. Masters
- Medical Oncology Hematology Consultants, Helen F Graham Cancer Center and Research Institute, Newark, Delaware
| | - Sachdev Thomas
- Kaiser Permanente Northern California, Oakland, California
| | | | - Daniel M. Anderson
- Metro-Minnesota Community Oncology Research Consortium, St. Louis Park, Minnesota
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Fernandez AI, Gaule P, Rimm DL. Tissue Age Affects Antigenicity and Scoring for the 22C3 Immunohistochemistry Companion Diagnostic Test. Mod Pathol 2023; 36:100159. [PMID: 36925070 PMCID: PMC10502188 DOI: 10.1016/j.modpat.2023.100159] [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: 11/14/2022] [Revised: 02/14/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023]
Abstract
Programmed death-ligand 1 (PD-L1) antibody 22C3 is the approved companion diagnostic immunohistochemistry test for treatment with pembrolizumab and cemiplimab in multiple cancer types. The 22C3 and 28-8 antibodies target the extracellular domain (ECD) of PD-L1, which is known to contain N-glycosylation sites. We hypothesize that antigenicity could be affected by the degradation of the glycan part of the epitope and thus change the scoring of the assay over time. Here, we test samples over time and assess the effects of time and deglycosylation on PD-L1 signal by comparing an antibody with an ECD antigen to an antibody with an intracellular domain (ICD) antigen. Ten whole-tissue sections of non-small-cell lung cancer (NSCLC) from 2018 were selected for testing. Fresh-cut serial sections for each case were stained on DAKO Link48 for 22C3 according to the label. In parallel, a previously described laboratory-developed test using E1L3N (an ICD antibody) was performed on the Leica BondRX. Tumor proportion scores for 22C3 and E1L3N were read by a pathologist and compared to the previous clinical diagnoses. To determine the effect using a quantitative approach, a tissue microarray (TMA) cohort with 90 NSCLC cases was similarly assessed. Finally, to determine whether the possible effect of epitope glycosylation, antibodies were tested before and after enzymatic deglycosylation of specimens. We found that 6 of 7 archival positive samples showed a significant reduction in positive staining with 22C3 compared to the original diagnostic sample assessed 3 years earlier. In an older archival TMA cohort, a quantitative significant difference in signal intensity was noted when staining with 22C3 was compared to E1L3N. This loss of signal was not noted in the fresh cell line TMA consistent with a time-dependent degradation of staining. Finally, quantitative assessment of the fresh TMA showed a significant loss of signal after a deglycosylation procedure when stained with 22C3, which was not seen when stained with E1L3N. We believe that these data show that the glycan part of the 22C3 epitope is not stable over time, and that this issue should be considered when assessing archival tissue for diagnostic or research purposes.
Collapse
Affiliation(s)
- Aileen I Fernandez
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Patricia Gaule
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut; Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
79
|
Yang J, Wei M, Liu X, Shao X, Yan J, Liu J, Wen J, Zhang X, Dong R, Min M. PD-L1 expression downregulation by RNF43 in gastric carcinoma enhances antitumour activity of T cells. Scand J Immunol 2023; 97:e13268. [PMID: 39007965 DOI: 10.1111/sji.13268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/10/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023]
Abstract
Ring finger protein 43 (RNF43), a transmembrane E3 ubiquitin ligase, has been indicated to be a potential biomarker for gastric cancer treatment, as this protein increases tumour cell apoptosis and suppresses cellular proliferation. The role of RNF43 in cellular immunotherapy remains unclear. Herein, we aimed to explore the expression level of RNF43 in gastric cancer cell lines and its role in cellular immunotherapy. The expression level of RNF43 and PD-L1 and their correlation in gastric cancer cell lines were analysed. The expression of PD-L1 was negatively correlated with that of RNF43 in gastric cancer cell lines. RNF43 interacted with PD-L1 to augment both K48- and K63-linked ubiquitination of PD-L1 in gastric cancer cell lines. In addition, RNF43 expression in gastric cancer cell lines could enhance the antitumour activity of T cells. In conclusion, this study reveals that RNF43 can inhibit PD-L1 expression to enhance the antitumour activity of cellular immunotherapy.
Collapse
Affiliation(s)
- Jing Yang
- Department of Gastroenterology, the First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Meng Wei
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xin Liu
- Department of Laboratory Medicine, the Eighth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao Shao
- Department of Pharmacology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jingshuang Yan
- Department of Gastroenterology, the First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jialong Liu
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Jing Wen
- Department of Gastroenterology, Chinese PLA 984 Hospital, Beijing, China
| | - Xueting Zhang
- Department of Gastroenterology, the First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ruihua Dong
- Department of Research Ward, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Min Min
- Department of Gastroenterology, the First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
80
|
Lu X, Deng S, Xu J, Green BL, Zhang H, Cui G, Zhou Y, Zhang Y, Xu H, Zhang F, Mao R, Zhong S, Cramer T, Evert M, Calvisi DF, He Y, Liu C, Chen X. Combination of AFP vaccine and immune checkpoint inhibitors slows hepatocellular carcinoma progression in preclinical models. J Clin Invest 2023; 133:e163291. [PMID: 37040183 PMCID: PMC10231990 DOI: 10.1172/jci163291] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 04/04/2023] [Indexed: 04/12/2023] Open
Abstract
Many patients with hepatocellular carcinoma (HCC) do not respond to the first-line immune checkpoint inhibitor treatment. Immunization with effective cancer vaccines is an attractive alternative approach to immunotherapy. However, its efficacy remains insufficiently evaluated in preclinical studies. Here, we investigated HCC-associated self/tumor antigen, α-fetoprotein-based (AFP-based) vaccine immunization for treating AFP (+) HCC mouse models. We found that AFP immunization effectively induced AFP-specific CD8+ T cells in vivo. However, these CD8+ T cells expressed exhaustion markers, including PD1, LAG3, and Tim3. Furthermore, the AFP vaccine effectively prevented c-MYC/Mcl1 HCC initiation when administered before tumor formation, while it was ineffective against full-blown c-MYC/Mcl1 tumors. Similarly, anti-PD1 and anti-PD-L1 monotherapy showed no efficacy in this murine HCC model. In striking contrast, AFP immunization combined with anti-PD-L1 treatment triggered significant inhibition of HCC progression in most liver tumor nodules, while in combination with anti-PD1, it induced slower tumor progression. Mechanistically, we demonstrated that HCC-intrinsic PD-L1 expression was the primary target of anti-PD-L1 in this combination therapy. Notably, the combination therapy had a similar therapeutic effect in the cMet/β-catenin mouse HCC model. These findings suggest that combining the AFP vaccine and immune checkpoint inhibitors may be effective for AFP (+) HCC treatment.
Collapse
Affiliation(s)
- Xinjun Lu
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shanshan Deng
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, USA
| | - Jiejie Xu
- Department of Hearing and Speech Science, Guangzhou Xinhua University, Guangzhou, China
| | | | - Honghua Zhang
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guofei Cui
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, USA
| | - Yi Zhou
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, USA
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yi Zhang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, USA
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Hongwei Xu
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, USA
- Department of Liver Surgery, Center of Liver Transplantation, West China Hospital of Sichuan University, Chengdu, China
| | - Fapeng Zhang
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rui Mao
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Sheng Zhong
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, USA
| | - Thorsten Cramer
- Department of General, Visceral and Transplantation Surgery, RWTH University Hospital, Aachen, Germany
- Department of Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Diego F. Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Yukai He
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Chao Liu
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, USA
- University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| |
Collapse
|
81
|
Chen Y, Ding X, Bai X, Zhou Z, Liu Y, Zhang X, Yu J, Hu M. The current advances and future directions of PD-1/PD-L1 blockade in head and neck squamous cell carcinoma (HNSCC) in the era of immunotherapy. Int Immunopharmacol 2023; 120:110329. [PMID: 37207445 DOI: 10.1016/j.intimp.2023.110329] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 05/21/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have previously demonstrated their efficacy and safety in various solid tumors, and with the growing interest in the application of ICIs in head and neck squamous cell carcinoma (HNSCC), various data have been reported. Mechanistically, HNSCC cells express programmed death ligand 1 (PD-L1), which binds to its receptor programmed death 1 (PD-1). Immune escape plays a key role in disease initiation and progression. Studying the abnormal activation of related pathways of PD-1/PD-L1 will help to understand the way of immunotherapy and find the advantageous population of immunotherapy. How to reduce HNSCC-related mortality and morbidity in this process has promoted the search for new therapeutic strategies, especially in the era of immunotherapy. PD-1 inhibitors have demonstrated significant prolongation of survival in recurrent/metastatic (R/M) HNSCC with a favorable safety profile. It also holds great promise in locally advanced (LA) HNSCC, where numerous studies are underway. Although immunotherapy has made great progress in HNSCC research, there are still many challenges. Therefore, in the review, we conducted an in-depth study on the expression of PD-L1 and the regulatory, immunosuppressive mechanisms caused by PD-L1, especially in head and neck squamous cell carcinoma, which is different from other tumors. And further summarize the situation, challenges and development trends of PD-1 and PD-L1 blockade in clinical practice.
Collapse
Affiliation(s)
- Yunhao Chen
- Department of Radiation Oncology, Shandong University Cancer Center, Jinan, Shandong 250117, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xingchen Ding
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xinbin Bai
- Department of Radiation Oncology, Tumor Hospital of Jining, Jining, Shandong 272007, China
| | - Zihan Zhou
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yu Liu
- Department of Oncology, Weifang Medical University, Weifang, Shandong 261053, China
| | - Xianbin Zhang
- Department of General Surgery and Integrated Chinese and Western Medicine, Institute of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong University Cancer Center, Jinan, Shandong 250117, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong 250117, China.
| | - Man Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| |
Collapse
|
82
|
Berner M, Hartmann A, Erber R. Role of Surgical Pathologist for Detection of Predictive Immuno-oncological Factors in Breast Cancer. Adv Anat Pathol 2023; 30:195-202. [PMID: 36418243 DOI: 10.1097/pap.0000000000000382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have changed therapy strategies in breast cancer (BC) patients suffering from triple-negative breast cancer (TNBC). For example, in Europe the anti-programmed cell death 1 ligand 1 (PD-L1) ICI Azetolizumab is approved for adult patients with locally advanced or metastasized TNBC (mTNBC), depending on the immunohistochemical (IHC) PD-L1 expression of immune cells in the tumor area [immune cell (IC) score ≥1%); the anti-programmed cell death 1 (PD-1) ICI pembrolizumab is approved for mTNBC if PD-L1 Combined Positive Score (CPS), that is PD-L1 expression on tumor and/or immune cells, is ≥10. For early TNBC, in contrast, neoadjuvant use of pembrolizumab is approved in the United States and Europe independent from PD-L1 IHC expression. The determination of PD-L1 expression in tumor tissue to predict response to ICI therapy requires sensitive immunostaining with appropriate primary antibodies and staining protocols and a standardized and meticulous assessment of PD-L1 IHC stained breast cancer tissue slides. For the selection of the test material and continuous quality control of the dyeing, high standards must be applied. The evaluation is carried out according to various evaluation algorithms (scores). Here, the role of PD-L1 in BC and the currently most relevant PD-L1 assays and scores for TNBC will be explained. Furthermore, other tissue-based biomarkers potentially predictive for ICI therapy response in BC, for example, tumor mutational burden (TMB), will be presented in this review.
Collapse
Affiliation(s)
- Mandy Berner
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | | | | |
Collapse
|
83
|
Galsky MD, Bajorin DF, Witjes JA, Gschwend JE, Tomita Y, Nasroulah F, Li J, Collette S, Valderrama BP, Grimm MO, Appleman L, Gravis G, Necchi A, Ye D, Stenner F, Wind-Rotolo M, Zhang J, Ünsal-Kaçmaz K. Disease-free Survival Analysis for Patients with High-risk Muscle-invasive Urothelial Carcinoma from the Randomized CheckMate 274 Trial by PD-L1 Combined Positive Score and Tumor Cell Score. Eur Urol 2023; 83:432-440. [PMID: 36868932 PMCID: PMC10520464 DOI: 10.1016/j.eururo.2023.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 12/09/2022] [Accepted: 01/03/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND The CheckMate 274 trial demonstrated improved disease-free survival (DFS) with adjuvant nivolumab versus placebo in patients with muscle-invasive urothelial carcinoma at high risk of recurrence after radical surgery in both the intent-to-treat population and the subset with tumor programmed death ligand 1 (PD-L1) expression ≥1%. OBJECTIVE To analyze DFS by combined positive score (CPS), which is based on PD-L1 expression in both tumor and immune cells. DESIGN, SETTING, AND PARTICIPANTS We randomized a total of 709 patients 1:1 to nivolumab 240 mg or placebo every 2 wk intravenously for ≤1 yr of adjuvant treatment. INTERVENTION Nivolumab 240 mg. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Primary endpoints were DFS in the intent-to-treat population and patients with tumor PD-L1 expression ≥1% using the tumor cell (TC) score. CPS was determined retrospectively from previously stained slides. Tumor samples with both quantifiable CPS and TC were analyzed. RESULTS AND LIMITATIONS Of 629 patients evaluable for CPS and TC, 557 (89%) had CPS ≥1, 72 (11%) had CPS <1, 249 (40%) had TC ≥1%, and 380 (60%) had TC <1%. Among patients with TC <1%, 81% (n = 309) had CPS ≥1. DFS was improved with nivolumab versus placebo for patients with TC ≥1% (hazard ratio [HR] 0.50, 95% confidence interval [CI] 0.35-0.71), those with CPS ≥1 (HR 0.62, 95% CI 0.49-0.78), and patients with both TC <1% and CPS ≥1 (HR 0.73, 95% CI 0.54-0.99). CONCLUSION More patients had CPS ≥1 than TC ≥1%, and most patients who had TC <1% had CPS ≥1. In addition, patients with CPS ≥1 experienced improved DFS with nivolumab. These results may, in part, explain the mechanisms underlying a benefit with adjuvant nivolumab even in patients who had both TC <1% and CPS ≥1. PATIENT SUMMARY We studied survival time without cancer recurrence (disease-free survival; DFS) for patients treated with nivolumab versus placebo after surgery to remove the bladder or components of the urinary tract for bladder cancer in the CheckMate 274 trial. We assessed the impact of levels of the protein PD-L1 expressed either on tumor cells (tumor cell score; TC) or on both tumor cells and immune cells surrounding the tumor (combined positive score; CPS). DFS was impoved with nivolumab versus placebo for patients with TC ≥1%, CPS ≥1, and for patients with both TC <1% and CPS ≥1. This analysis may help physicians understand which patients would benefit most from treatment with nivolumab.
Collapse
Affiliation(s)
| | - Dean F Bajorin
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Yoshihiko Tomita
- Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | - Jun Li
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | - Leonard Appleman
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Gwenaelle Gravis
- Institut Paoli-Calmettes Aix-Marseille Université, Marseille, France
| | - Andrea Necchi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Dingwei Ye
- Fudan University Shanghai Cancer Center, Shanghai, China
| | | | | | | | | |
Collapse
|
84
|
Zhang YJ, Xiong SP, Yang YZ, Fu S, Wang TM, Suster DI, Jiang GY, Zhang XF, Xiang J, Wu YX, Zhang WL, Cao Y, Huang YH, Yun JP, Liu QW, Sun Q, Chen Y, Yang X, Li Y, Wang EH, Liu JL, Zhang JB. Clinicopathologic features, tumor immune microenvironment and genomic landscape of EBV-related and EBV-unrelated poorly differentiated nonkeratinizing squamous cell carcinoma of the thymus. Lung Cancer 2023; 179:107178. [PMID: 37004385 DOI: 10.1016/j.lungcan.2023.107178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/20/2023] [Accepted: 03/19/2023] [Indexed: 04/03/2023]
Abstract
OBJECTIVES Knowledge regarding thymic EBV-related poorly differentiated nonkeratinizing squamous cell carcinoma (PDNKSCC), also known as lymphoepithelial carcinoma (LEC), is extremely limited due to its rarity. MATERIALS AND METHODS This multi-institutional study enrolled 85 patients with thymic PDNKSCC. DNA in situ hybridization was performed to evaluate the EBV status of all 85 cases. Immunohistochemistry and next generation sequencing were performed to compare the differences in the clinicopathological and molecular features between EBV-related and EBV-unrelated PDNKSCC. Tumor-infiltrating lymphocytes (TILs) were also analyzed by these methods. RESULTS The 85 cases were classified into 27 EBV-related PDNKSCCs (31.8 %) and 58 EBV-unrelated PDNKSCCs (68.2 %) according to the EBV status, and 35 Lymphoepithelioma pattern (LP) (41.2 %) and 50 desmoplastic pattern (DP) (58.8 %) according to the histological characteristics. Compared to the EBV-unrelated PDNKSCC, EBV-related PDNKSCC showed a younger patient predominance and more commonly displayed a LP subtype. Additionally, LP-type cases were divided into two groups: Group 1 (EBV-related, 20/85) and Group 2 (EBV-unrelated, 15/85); the DP-type cases were divided into Group 3 (EBV-unrelated, 43/85) and Group 4 (EBV-related, 7/85). The four Groups showed a significant association with patients' OS and PFS. EBV-related PDNKSCC had significantly higher PD-L1 + tumor cells (TCs) and PD-L1 + and CD8 + immune cells (ICs) than EBV-unrelated PDNKSCC. The tumor microenvironment immune type (TMIT) I (PDL1-Tumor+/CD8-High) was more common in EBV-related PDNKSCC, especially in Group 1(LP and EBV related) with more than 90 % cases belonged to TMIT I. Molecular analysis demonstrated that EBV-related PDNKSCC had a significantly higher tumour mutational burden and frequency of somatic mutations than EBV-unrelated cases. CONCLUSIONS EBV-related PDNKSCC, especially the Group 1, could be a candidate for immunotherapy and EBV positivity may provide an indication for the selection of targeted therapy due to their high tumour mutational burden.
Collapse
|
85
|
Mohanty SK, Lobo A, Mishra SK, Cheng L. Precision Medicine in Bladder Cancer: Present Challenges and Future Directions. J Pers Med 2023; 13:jpm13050756. [PMID: 37240925 DOI: 10.3390/jpm13050756] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Bladder cancer (BC) is characterized by significant histopathologic and molecular heterogeneity. The discovery of molecular pathways and knowledge of cellular mechanisms have grown exponentially and may allow for better disease classification, prognostication, and development of novel and more efficacious noninvasive detection and surveillance strategies, as well as selection of therapeutic targets, which can be used in BC, particularly in a neoadjuvant or adjuvant setting. This article outlines recent advances in the molecular pathology of BC with a better understanding and deeper focus on the development and deployment of promising biomarkers and therapeutic avenues that may soon make a transition into the domain of precision medicine and clinical management for patients with BC.
Collapse
Affiliation(s)
- Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute and CORE Diagnostics, Gurgaon 122016, India
| | - Anandi Lobo
- Department of Pathology and Laboratory Medicine, Kapoor Center for Pathology and Urology, Raipur 490042, India
| | - Sourav K Mishra
- Department of Medical Oncology, All India Institute of Medical Sciences, Bhubaneswar 750017, India
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Brown University Warren Alpert Medical School, Lifespan Academic Medical Center, and the Legorreta Cancer Center at Brown University, 593 Eddy Street, APC 12-105, Providence, RI 02903, USA
| |
Collapse
|
86
|
Shahidi M, Abazari O, Dayati P, Reza JZ, Modarressi MH, Tofighi D, Haghiralsadat BF, Oroojalian F. Using chitosan-stabilized, hyaluronic acid-modified selenium nanoparticles to deliver CD44-targeted PLK1 siRNAs for treating bladder cancer. Nanomedicine (Lond) 2023; 18:259-277. [PMID: 37125618 DOI: 10.2217/nnm-2022-0198] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Aims: Achieving an effective biocompatible system for siRNAs delivery to the tumor site remains a significant challenge. Materials & methods: Selenium nanoparticles (SeNPs) modified by chitosan (CS) and hyaluronic acid (HA) were fabricated for PLK1 siRNAs (siPLK1) delivery to the bladder cancer cells. The HA-CS-SeNP@siPLK1 efficacy was evaluated using in vitro and in vivo models. Results: HA-CS-SeNP@siPLK1 was selectively internalized into T24 cells through clathrin-mediated endocytosis. Treatment with HA-CS-SeNP@siPLK1 successfully silenced the PLK1 gene, inhibited cell proliferation and induced cell cycle arrest in vitro. HA-CS-SeNP@siPLK1 could also inhibit tumor growth in vivo without causing systemic toxicity. Conclusion: Our results suggest that HA-CS-SeNPs may provide a good vehicle for delivering siPLK1 to the bladder tumor site.
Collapse
Affiliation(s)
- Maryamsadat Shahidi
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences & Health Services, Yazd, 89151, Iran
| | - Omid Abazari
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences & Health Services, Yazd, 89151, Iran
| | - Parisa Dayati
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14115, Iran
| | - Javad Zavar Reza
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences & Health Services, Yazd, 89151, Iran
| | - Mohammad Hossein Modarressi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, 14176, Iran
| | - Davood Tofighi
- Department of Psychology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Bibi Fatemeh Haghiralsadat
- Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, 89151, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnūrd, 94149, Iran
- Natural Products & Medicinal Plants Research Center, North Khorasan University of Medical Sciences Bojnūrd, 94149, Iran
| |
Collapse
|
87
|
Isla D, Lozano MD, Paz-Ares L, Salas C, de Castro J, Conde E, Felip E, Gómez-Román J, Garrido P, Belén Enguita A. [New update to the guidelines on testing predictive biomarkers in non-small-cell lung cancer: a National Consensus of the Spanish Society of Pathology and the Spanish Society of Medical Oncology]. REVISTA ESPANOLA DE PATOLOGIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ANATOMIA PATOLOGICA Y DE LA SOCIEDAD ESPANOLA DE CITOLOGIA 2023; 56:97-112. [PMID: 37061248 DOI: 10.1016/j.patol.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 02/08/2023] [Indexed: 04/17/2023]
Abstract
Non-small cell lung cancer (NSCLC) presents the greatest number of identified therapeutic targets, some of which have therapeutic utility. Currently, detecting EGFR, BRAF, KRAS and MET mutations, ALK, ROS1, NTRK and RET translocations, and PD-L1 expression in these patients is considered essential. The use of next-generation sequencing (NGS) facilitates precise molecular diagnosis and allows the detection of other emerging mutations, such as the HER2 mutation and predictive biomarkers for immunotherapy responses. In this consensus, a group of experts in the diagnosis and treatment of NSCLC selected by the Spanish Society of Pathology (SEAP) and the Spanish Society of Medical Oncology (SEOM) have evaluated currently available information and propose a series of recommendations to optimize the detection and use of biomarkers in daily clinical practice.
Collapse
Affiliation(s)
- Dolores Isla
- Hospital Clínico Universitario Lozano Blesa, IIS Aragón, Sociedad Española de Oncología Médica (SEOM), Zaragoza, España
| | - María D Lozano
- Clínica Universidad de Navarra, Sociedad Española de Citología (SEC), Sociedad Española de Anatomía Patológica (SEAP), Pamplona, España
| | - Luis Paz-Ares
- Hospital Universitario 12 de Octubre, Sociedad Española de Oncología Médica (SEOM), Madrid, España
| | - Clara Salas
- Hospital Universitario Puerta de Hierro, Sociedad Española de Anatomía Patológica (SEAP), Madrid, España
| | - Javier de Castro
- Hospital Universitario La Paz, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Sociedad Española de Oncología Médica (SEOM), Madrid, España
| | - Esther Conde
- Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital Universitario 12 de Octubre (i+12), Sociedad Española de Anatomía Patológica (SEAP), Madrid, España
| | - Enriqueta Felip
- Hospital Universitario Vall d'Hebron, Sociedad Española de Oncología Médica (SEOM), Barcelona, España
| | - Javier Gómez-Román
- Universidad de Cantabria, Hospital Universitario Marqués de Valdecilla, Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Sociedad Española de Anatomía Patológica (SEAP), Santander, España
| | - Pilar Garrido
- Hospital Universitario Ramón y Cajal, Sociedad Española de Oncología Médica (SEOM), Madrid, España
| | - Ana Belén Enguita
- Hospital Universitario 12 de Octubre, Sociedad Española de Anatomía Patológica (SEAP), Madrid, España.
| |
Collapse
|
88
|
Dacic S. State of the Art of Pathologic and Molecular Testing. Hematol Oncol Clin North Am 2023; 37:463-473. [PMID: 36964109 DOI: 10.1016/j.hoc.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
Abstract
Advances in the treatment of non-small cell lung carcinoma have resulted in improved histologic classification and the implementation of molecular testing for predictive biomarkers into the routine diagnostic workflow. Over the past decade, molecular testing has evolved from single-gene assays to high-thoroughput comprehensive next-generation sequencing. Economic barriers, suboptimal turnaround time to obtain the results, and limited tissue available for molecular assays resulted in adoption of liquid biopsies (ctDNA) into clinical practice. Multiplex immunohistochemical/immunofluorescence assays evaluating tumor microenvironment together with the AI approaches are anticipated to translate from research into clinical care.
Collapse
Affiliation(s)
- Sanja Dacic
- Department of Pathology, Yale School of Medicine, 200 South Frontage Road, EP2-631, New Haven, CT 06510, USA.
| |
Collapse
|
89
|
Wu HX, Pan YQ, He Y, Wang ZX, Guan WL, Chen YX, Yao YC, Shao NY, Xu RH, Wang F. Clinical Benefit of First-Line Programmed Death-1 Antibody Plus Chemotherapy in Low Programmed Cell Death Ligand 1-Expressing Esophageal Squamous Cell Carcinoma: A Post Hoc Analysis of JUPITER-06 and Meta-Analysis. J Clin Oncol 2023; 41:1735-1746. [PMID: 36473145 PMCID: PMC10022847 DOI: 10.1200/jco.22.01490] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/10/2022] [Accepted: 10/13/2022] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Pembrolizumab or nivolumab plus chemotherapy was approved as a first-line treatment for high programmed cell death ligand 1 (PD-L1)-expressing esophageal squamous cell carcinoma (ESCC) by the European Medicines Agency, whereas the US Food and Drug Administration approved this regimen regardless of PD-L1 expression. The superiority of programmed death-1 (PD-1) antibody plus chemotherapy over chemotherapy alone in patients with low PD-L1-expressing ESCC remains debatable. METHODS Post hoc analysis of the Chinese JUPITER-06 study focusing on efficacy stratified by PD-L1 tumor proportion score (TPS; using JS311 antibody) was conducted. Electronic databases were searched to identify eligible randomized controlled trials for meta-analysis. Study-level pooled analyses of hazard ratios (HRs) for overall survival and progression-free survival and odds ratios for objective response rate according to PD-L1 expression were performed. RESULTS The post hoc analysis of JUPITER-06 showed more prominent clinical benefit with PD-1 antibody plus chemotherapy than with chemotherapy alone in both the high and low PD-L1-expressing subgroups. Five randomized controlled trials were included in the meta-analysis, and two PD-L1 expression scoring criteria, TPS (≥ 1%/< 1%) and combined positive score (CPS, ≥ 10/< 10), were analyzed. Significant overall survival benefit by adding PD-1 antibody to chemotherapy was observed in both the TPS < 1% (HR, 0.74; 95% CI, 0.56 to 0.97) and CPS < 10 (HR, 0.77; 95% CI, 0.66 to 0.89) subgroups. Similarly, significantly prolonged progression-free survival was observed in both the TPS < 1% (HR, 0.66; 95% CI, 0.50 to 0.86) and CPS < 10 (HR, 0.63; 95% CI, 0.47 to 0.84) subgroups. In addition, the objective response rate of the TPS < 1% subgroup was significantly improved (odds ratio, 1.71; 95% CI, 1.27 to 2.29). In all high PD-L1-expressing subgroups, the pooled benefit of PD-1 antibody plus chemotherapy was significantly better than that of chemotherapy. CONCLUSION This study provided novel evidence supporting the superiority of PD-1 antibody plus chemotherapy to chemotherapy alone in patients with advanced ESCC with low PD-L1 expression. Further studies of predictive biomarkers are warranted.
Collapse
Affiliation(s)
- Hao-Xiang Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Yi-Qian Pan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Ye He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Zi-Xian Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Wen-Long Guan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Yan-Xing Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Yi-Chen Yao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Ning-Yi Shao
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China
| | - Rui-Hua Xu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Feng Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
90
|
Gillison ML, Ferris RL, Harris J, Colevas AD, Mell LK, Kong C, Jordan RC, Moore KL, Truong MT, Kirsch C, Chakravarti A, Blakaj DM, Clump DA, Ohr JP, Deeken JF, Gensheimer MF, Saba NF, Dorth JA, Rosenthal DI, Leidner RS, Kimple RJ, Machtay M, Curran WJ, Torres-Saavedra P, Le QT. Safety of Nivolumab Added to Chemoradiation Therapy Platforms for Intermediate and High-Risk Locoregionally Advanced Head and Neck Squamous Cell Carcinoma: RTOG Foundation 3504. Int J Radiat Oncol Biol Phys 2023; 115:847-860. [PMID: 36228746 PMCID: PMC11189668 DOI: 10.1016/j.ijrobp.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/10/2022] [Accepted: 10/04/2022] [Indexed: 11/08/2022]
Abstract
PURPOSE Programmed death-1 immune checkpoint blockade improves survival of patients with recurrent/metastatic head and neck squamous cell carcinoma (HNSCC), but the benefits of addition to (chemo)radiation for newly diagnosed patients with HNSCC remain unknown. METHODS AND MATERIALS We evaluated the safety of nivolumab concomitant with 70 Gy intensity modulated radiation therapy and weekly cisplatin (arm 1), every 3-week cisplatin (arm 2), cetuximab (arm 3), or alone for platinum-ineligible patients (arm 4) in newly diagnosed intermediate- or high-risk locoregionally advanced HNSCC. Patients received nivolumab from 2 weeks prior to radiation therapy until 3 months post-radiation therapy. The primary endpoint was dose-limiting toxicity (DLT). If ≤2 of the first 8 evaluable patients experienced a DLT, an arm was considered safe. Secondary endpoints included toxicity and feasibility of adjuvant nivolumab to 1 year, defined as all 7 additional doses received by ≥4 of the first 8 evaluable patients across arms. RESULTS Of 39 patients (10 in arms 1, 3, 4 and 9 in arm 2), 72% had T3-4 tumors, 85% had N2-3 nodal disease, and 67% had >10 pack-years of smoking. There were no DLTs in arms 1 and 2, 1 in arm 3 (mucositis), and 2 in arm 4 (lipase elevation and mucositis in 1 and fatigue in another). The most common grade ≥3 nivolumab-related adverse events were lipase increase, mucositis, diarrhea, lymphopenia, hyponatremia, leukopenia, fatigue, and serum amylase increase. Adjuvant nivolumab was feasible as defined in the protocol. CONCLUSIONS Concomitant nivolumab with the 4 tested regimens was safe for patients with intermediate- and high-risk HNSCC, and subsequent adjuvant nivolumab was feasible as defined (NCT02764593).
Collapse
Affiliation(s)
| | | | - Jonathan Harris
- RTOG Foundation Statistics and Data Management Center, American College of Radiology, Philadelphia, Pennsylvania
| | | | - Loren K Mell
- UC San Diego Moores Cancer Center, La Jolla, California
| | - Christina Kong
- Stanford Cancer Institute, Palo Alto, Stanford, California
| | | | - Kevin L Moore
- UC San Diego Moores Cancer Center, La Jolla, California
| | | | | | | | | | - David A Clump
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - James P Ohr
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | | | | | - Nabil F Saba
- Emory University Hospital/Winship Cancer Institute, Atlanta, Georgia
| | | | | | - Rom S Leidner
- Providence Portland Medical Center, Portland, Oregon
| | - Randall J Kimple
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Mitchell Machtay
- Penn State Milton S Hershey Medical Center, Hershey, Pennsylvania
| | | | - Pedro Torres-Saavedra
- RTOG Foundation Statistics and Data Management Center, American College of Radiology, Philadelphia, Pennsylvania
| | - Quynh Thu Le
- Stanford Cancer Institute, Palo Alto, Stanford, California.
| |
Collapse
|
91
|
Robert ME, Rüschoff J, Jasani B, Graham RP, Badve SS, Rodriguez-Justo M, Kodach LL, Srivastava A, Wang HL, Tang LH, Troncone G, Rojo F, Van Treeck BJ, Pratt J, Shnitsar I, Kumar G, Karasarides M, Anders RA. High Interobserver Variability among Pathologists Using Combined Positive Score to Evaluate PD-L1 Expression in Gastric, Gastroesophageal Junction and Esophageal Adenocarcinoma. Mod Pathol 2023; 36:100154. [PMID: 36925069 DOI: 10.1016/j.modpat.2023.100154] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/17/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023]
Abstract
Reliable, reproducible methods to interpret programmed death ligand-1 (PD-L1) expression on tumor cells (TC) and immune cells (IC) are needed for pathologists to inform decisions associated with checkpoint inhibitor therapies. Our international study compared interpathologist agreement of PD-L1 expression using combined positive score (CPS) under standardized conditions on samples from patients with gastric/gastroesophageal junction/esophageal adenocarcinoma. Tissue sections from 100 adenocarcinoma pre-treatment biopsies were stained in a single laboratory using the PD-L1 IHC 28-8 and 22C3(Agilent) pharmDx immunohistochemical assays. PD-L1 CPS was evaluated by 12 pathologists on scanned whole slide images of these biopsies before and after a 2-hour CPS training session by Agilent. Additionally, pathologists determined PD-L1 positive TC, IC, and total viable TC on a single tissue fragment from 35 of 100 biopsy samples. Scoring agreement among pathologists was assessed using the intraclass correlation coefficient (ICC). Interobserver variability for CPS for 100 biopsies was high with only fair agreement among pathologists both pre- (range 0.45 to 0.55) and post-training (range 0.56 to 0.57) for both assays. For the 35 single biopsy samples, poor/fair agreement was also observed for the total number viable TC (ICC 0.09), number of PD-L1 positive IC (ICC 0.19), number of PD-L1 positive TC (ICC 0.54), and calculated CPS (ICC 0.14), while calculated TC score (positive TC/Total TC) showed excellent agreement (ICC 0.82). Retrospective histologic review of samples with the poorest interpathologist agreement revealed (1) ambiguous identification of positively staining stromal cells, (2) faint or variable intensity of staining, (3) difficulty in distinguishing membranous from cytoplasmic tumor staining, and (4) cautery and crush artifact, as possible confounding factors. These results emphasize the need for objective techniques to standardize the interpretation of PD-L1 expression when using the CPS methodology on gastric/gastroesophageal junction cancer biopsies to accurately identify patients most likely to benefit from immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
| | | | | | | | - Sunil S Badve
- Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | - Hanlin L Wang
- University of California Los Angeles, Los Angeles, CA, USA
| | - Laura H Tang
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Federico Rojo
- IIS-Fundacion Jimenez Diaz CIBERONC (Madrid), Madrid, Spain
| | | | | | | | | | | | - Robert A Anders
- John Hopkins University,; Convergence Institute,; Bloomberg∼Kimmel Intitute for Cancer Immunotherapy Baltimore, MD, USA.
| |
Collapse
|
92
|
Si K, Ye Z, Ali DJ, Ding B, He C, Dai Z, Li Z, Sun B, Shen Y, Xiao Z. Co-delivery of PDL1-blocking scFv and chemotherapeutics using engineered exosomes for cancer therapy. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
93
|
Marmarelis ME, Sun L, Aggarwal C. Adjuvant Atezolizumab Should Be Administered to All Patients With Programmed Death-Ligand 1 Expressing Surgically Resected Stage II to III NSCLC After Chemotherapy: In Favor. J Thorac Oncol 2023; 18:265-267. [PMID: 36842810 DOI: 10.1016/j.jtho.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 02/28/2023]
Affiliation(s)
- Melina E Marmarelis
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lova Sun
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Charu Aggarwal
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Penn Center for Cancer Care Innovation, University of Pennsylvania, Philadelphia, Pennsylvania; Penn Center for Precision Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
94
|
Stefanini B, Ielasi L, Chen R, Abbati C, Tonnini M, Tovoli F, Granito A. TKIs in combination with immunotherapy for hepatocellular carcinoma. Expert Rev Anticancer Ther 2023; 23:279-291. [PMID: 36794716 DOI: 10.1080/14737140.2023.2181162] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
INTRODUCTION The treatment landscape of hepatocellular carcinoma (HCC) has significantly changed over the last 5 years with multiple options in the frontline, second line, and beyond. Tyrosine kinase inhibitors (TKIs) were the first approved systemic treatments for the advanced stage of HCC; however, thanks to the increasing knowledge and characterization of the immunological features of the tumor microenvironment, the systemic treatment of HCC has been further expanded with the immune checkpoint inhibitor (ICI) approach and the following evidence of the higher efficacy obtained with combined treatment with atezolizumab plus bevacizumab over sorafenib. AREAS COVERED In this review, we look at rationale, efficacy, and safety profiles of current and emerging ICI/TKI combination treatments and discuss the available results from other clinical trials using similar combinatorial therapeutic approaches. EXPERT OPINION Angiogenesis and immune evasion are the two key pathogenic hallmarks of HCC. While the pioneering regimen of atezolizumab/bevacizumab is consolidating as the first-line treatment of advanced HCC, it will be essential, in the near future, to determine the best second-line treatment options and how to optimize the selection of the most effective therapies. These points still need to be addressed by future studies that are largely warranted to enhance the treatment's effectiveness and ultimately to tackle down HCC lethality.
Collapse
Affiliation(s)
- Bernardo Stefanini
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Luca Ielasi
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Rusi Chen
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Chiara Abbati
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Matteo Tonnini
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Francesco Tovoli
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Granito
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
95
|
Yatabe Y. Comments on the Article by Miura et al.: "Comparable Clinical Benefit Between Different Subsets Selected by PD-L1 22C3 and SP142 Tests". JTO Clin Res Rep 2023; 4:100492. [PMID: 36942129 PMCID: PMC10023938 DOI: 10.1016/j.jtocrr.2023.100492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 02/19/2023] [Indexed: 03/16/2023] Open
Affiliation(s)
- Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
96
|
Licata L, Mariani M, Rossari F, Viale G, Notini G, Naldini MM, Bosi C, Piras M, Dugo M, Bianchini G. Tissue- and liquid biopsy-based biomarkers for immunotherapy in breast cancer. Breast 2023; 69:330-341. [PMID: 37003065 PMCID: PMC10070181 DOI: 10.1016/j.breast.2023.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 03/29/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy and now represent the mainstay of treatment for many tumor types, including triple-negative breast cancer and two agnostic registrations. However, despite impressive durable responses suggestive of an even curative potential in some cases, most patients receiving ICIs do not derive a substantial benefit, highlighting the need for more precise patient selection and stratification. The identification of predictive biomarkers of response to ICIs may play a pivotal role in optimizing the therapeutic use of such compounds. In this Review, we describe the current landscape of tissue and blood biomarkers that could serve as predictive factors for ICI treatment in breast cancer. The integration of these biomarkers in a "holistic" perspective aimed at developing comprehensive panels of multiple predictive factors will be a major step forward towards precision immune-oncology.
Collapse
Affiliation(s)
- Luca Licata
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Marco Mariani
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Federico Rossari
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy; San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Viale
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Giulia Notini
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Matteo Maria Naldini
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy; San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carlo Bosi
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Marta Piras
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy
| | - Matteo Dugo
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
97
|
de Miguel-Perez D, Russo A, Gunasekaran M, Buemi F, Hester L, Fan X, Carter-Cooper BA, Lapidus RG, Peleg A, Arroyo-Hernández M, Cardona AF, Naing A, Hirsch FR, Mack PC, Kaushal S, Serrano MJ, Adamo V, Arrieta O, Rolfo C. Baseline extracellular vesicle TGF-β is a predictive biomarker for response to immune checkpoint inhibitors and survival in non-small cell lung cancer. Cancer 2023; 129:521-530. [PMID: 36484171 DOI: 10.1002/cncr.34576] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/09/2022] [Accepted: 10/24/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Immune-checkpoint inhibitors (ICIs) are an effective therapeutic strategy, improving the survival of patients with lung cancer compared with conventional treatments. However, novel predictive biomarkers are needed to stratify which patients derive clinical benefit because the currently used and highly heterogenic histological PD-L1 has shown low accuracy. Liquid biopsy is the analysis of biomarkers in body fluids and represents a minimally invasive tool that can be used to monitor tumor evolution and treatment effects, potentially reducing biases associated with tumor heterogeneity associated with tissue biopsies. In this context, cytokines, such as transforming growth factor-β (TGF-β), can be found free in circulation in the blood and packaged into extracellular vesicles (EVs), which have a specific delivery tropism and can affect in tumor/immune system interaction. TGF-β is an immunosuppressive cytokine that plays a crucial role in tumor immune escape, treatment resistance, and metastasis. Thus, we aimed to evaluate the predictive value of circulating and EV TGF-β in patients with non-small-cell lung cancer receiving ICIs. METHODS Plasma samples were collected in 33 patients with advanced non-small-cell lung cancer before and during treatment with ICIs. EV were isolated from plasma by serial ultracentrifugation methods and circulating and EV TGF-β expression levels were evaluated by enzyme-linked immunosorbent assay. RESULTS Baseline high expression of TGF-β in EVs was associated with nonresponse to ICIs as well as shorter progression-free survival and overall survival, outperforming circulating TGF-β levels and tissue PD-L1 as a predictive biomarker. CONCLUSION If validated, EV TGF-β could be used to improve patient stratification, increasing the effectiveness of treatment with ICIs and potentially informing combinatory treatments with TGF-β blockade. PLAIN LANGUAGE SUMMARY Treatment with immune-checkpoint inhibitors (ICIs) has improved the survival of some patients with lung cancer. However, the majority of patients do not benefit from this treatment, making it essential to develop more reliable biomarkers to identify patients most likely to benefit. In this pilot study, the expression of transforming growth factor-β (TGF-β) in blood circulation and in extracellular vesicles was analyzed. The levels of extracellular vesicle TGF-β before treatment were able to determine which patients would benefit from treatment with ICIs and have a longer survival with higher accuracy than circulating TGF-β and tissue PD-L1, which is the currently used biomarker in clinical practice.
Collapse
Affiliation(s)
- Diego de Miguel-Perez
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, Mount Sinai, New York, New York, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alessandro Russo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Messina, Italy
| | - Muthukumar Gunasekaran
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Departments of Surgery and Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Francesco Buemi
- Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Messina, Italy
| | - Lisa Hester
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Xiaoxuan Fan
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Brandon A Carter-Cooper
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Rena G Lapidus
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ariel Peleg
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, Mount Sinai, New York, New York, USA
| | | | - Andres F Cardona
- Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC)/Foundation for Clinical and Applied Cancer Research (FICMAC)/Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogota, Colombia
| | - Aung Naing
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fred R Hirsch
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, Mount Sinai, New York, New York, USA
| | - Philip C Mack
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, Mount Sinai, New York, New York, USA
| | - Sunjay Kaushal
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Departments of Surgery and Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Maria Jose Serrano
- GENYO Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain
| | - Vincenzo Adamo
- Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Messina, Italy
| | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, Mount Sinai, New York, New York, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
98
|
Tomlins SA, Khazanov NA, Bulen BJ, Hovelson DH, Shreve MJ, Lamb LE, Matrana MR, Burkard ME, Yang ESH, Edenfield WJ, Dees EC, Onitilo AA, Thompson M, Buchschacher GL, Miller AM, Menter A, Parsons B, Wassenaar T, Hwang LC, Suga JM, Siegel R, Irvin W, Nair S, Slim JN, Misleh J, Khatri J, Masters G, Thomas S, Safa M, Anderson DM, Kwiatkowski K, Mitchell K, Hu-Seliger T, Drewery S, Fischer A, Plouffe K, Czuprenski E, Hipp J, Reeder T, Vakil H, Johnson DB, Rhodes DR. Development and validation of an integrative pan-solid tumor predictor of PD-1/PD-L1 blockade benefit. COMMUNICATIONS MEDICINE 2023; 3:14. [PMID: 36750617 PMCID: PMC9905474 DOI: 10.1038/s43856-023-00243-7] [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: 01/25/2022] [Accepted: 01/12/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Anti-PD-1 and PD-L1 (collectively PD-[L]1) therapies are approved for many advanced solid tumors. Biomarkers beyond PD-L1 immunohistochemistry, microsatellite instability, and tumor mutation burden (TMB) may improve benefit prediction. METHODS Using treatment data and genomic and transcriptomic tumor tissue profiling from an observational trial (NCT03061305), we developed Immunotherapy Response Score (IRS), a pan-tumor predictive model of PD-(L)1 benefit. IRS real-world progression free survival (rwPFS) and overall survival (OS) prediction was validated in an independent cohort of trial patients. RESULTS Here, by Cox modeling, we develop IRS-which combines TMB with CD274, PDCD1, ADAM12 and TOP2A quantitative expression-to predict pembrolizumab rwPFS (648 patients; 26 tumor types; IRS-High or -Low groups). In the 248 patient validation cohort (248 patients; 24 tumor types; non-pembrolizumab PD-[L]1 monotherapy treatment), median rwPFS and OS are significantly longer in IRS-High vs. IRS-Low patients (rwPFS adjusted hazard ratio [aHR] 0.52, p = 0.003; OS aHR 0.49, p = 0.005); TMB alone does not significantly predict PD-(L)1 rwPFS nor OS. In 146 patients treated with systemic therapy prior to pembrolizumab monotherapy, pembrolizumab rwPFS is only significantly longer than immediately preceding therapy rwPFS in IRS-High patients (interaction test p = 0.001). In propensity matched lung cancer patients treated with first-line pembrolizumab monotherapy or pembrolizumab+chemotherapy, monotherapy rwPFS is significantly shorter in IRS-Low patients, but is not significantly different in IRS-High patients. Across 24,463 molecularly-evaluable trial patients, 7.6% of patients outside of monotherapy PD-(L)1 approved tumor types are IRS-High/TMB-Low. CONCLUSIONS The validated, predictive, pan-tumor IRS model can expand PD-(L)1 monotherapy benefit outside currently approved indications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mark E Burkard
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Eddy Shih-Hsin Yang
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | | | - E Claire Dees
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Adedayo A Onitilo
- Cancer Care and Research Center, Marshfield Clinic Research Institute, Marshfield, WI, USA
| | - Michael Thompson
- Aurora Cancer Care, Advocate Aurora Health, Milwaukee, WI, USA
- Tempus Labs, Chicago, IL, USA
| | | | - Alan M Miller
- SCL Health-CO, Broomfield, CO, USA
- Translational Drug Development, Scottsdale, USA
| | | | | | | | - Leon C Hwang
- Kaiser Permanente of the Mid-Atlantic States, Rockville, MD, USA
| | - J Marie Suga
- Kaiser Permanente Northern California, Vallejo, CA, USA
| | - Robert Siegel
- Bon Secours St. Francis Cancer Center, Greenville, SC, USA
| | | | - Suresh Nair
- Lehigh Valley Topper Cancer Institute, Allentown, PA, USA
| | | | | | - Jamil Khatri
- ChristianaCare Oncology Hematology, Newark, DE, USA
| | - Gregory Masters
- Medical Oncology Hematology Consultants, Helen F Graham Cancer Center and Research Institute,, Newark, DE, USA
| | - Sachdev Thomas
- Kaiser Permanente - Northern California, Oakland, CA, USA
| | | | - Daniel M Anderson
- Metro-Minnesota Community Oncology Research Consortium, St. Louis Park, MN, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Sadeghirad H, Bahrami T, Layeghi SM, Yousefi H, Rezaei M, Hosseini-Fard SR, Radfar P, Warkiani ME, O'Byrne K, Kulasinghe A. Immunotherapeutic targets in non-small cell lung cancer. Immunology 2023; 168:256-272. [PMID: 35933597 DOI: 10.1111/imm.13562] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 08/02/2022] [Indexed: 01/17/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common types of cancer in the world and has a 5-year survival rate of ~20%. Immunotherapies have shown promising results leading to durable responses, however, they are only effective for a subset of patients. To determine the best therapeutic approach, a thorough and in-depth profiling of the tumour microenvironment (TME) is required. The TME is a complex network of cell types that form an interconnected network, promoting tumour cell initiation, growth and dissemination. The stroma, immune cells and endothelial cells that comprise the TME generate a plethora of cytotoxic or cytoprotective signalling pathways. In this review, we discuss immunotherapeutic targets in NSCLC tumours and how the TME may influence patients' response to immunotherapy.
Collapse
Affiliation(s)
- Habib Sadeghirad
- University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Tayyeb Bahrami
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Sepideh M Layeghi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, Louisiana, USA
| | - Meysam Rezaei
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Seyed R Hosseini-Fard
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Payar Radfar
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Majid E Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Ken O'Byrne
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Arutha Kulasinghe
- University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
100
|
Yu SL, Hsiao YJ, Cooper WA, Choi YL, Avilés-Salas A, Chou TY, Coudry R, Raskin GA, Fox SB, Huang CC, Jeon YK, Ko YH, Ku WH, Kwon GY, Leslie C, Lin MC, Lou PJ, Scapulatempo-Neto C, Mendoza Ramírez S, Savelov N, Shim HS, Lara Torres CO, Cunha IW, Zavalishina L, Chen YM. The Ring Study: an international comparison of PD-L1 diagnostic assays and their interpretation in non-small cell lung cancer, head and neck squamous cell cancer and urothelial cancer. Pathology 2023; 55:19-30. [PMID: 36319485 DOI: 10.1016/j.pathol.2022.07.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 07/11/2022] [Accepted: 07/21/2022] [Indexed: 01/11/2023]
Abstract
PD-L1 immunohistochemistry has been approved as a diagnostic assay for immunotherapy. However, an international comparison across multiple cancers is lacking. This study aimed to assess the performance of PD-L1 diagnostic assays in non-small cell lung cancer (NSCLC), head and neck squamous cell cancer (HNSCC) and urothelial cancer (UC). The excisional specimens of NSCLC, HNSCC and UC were assayed by Ventana SP263 and scored at three sites in each country, including Australia, Brazil, Korea, Mexico, Russia and Taiwan. All slides were rotated to two other sites for interobserver scoring. The same cohort of NSCLC was assessed with Dako 22C3 pharmDx PD-L1 for comparison. The PD-L1 immunopositivity was scored according to the approved PD-L1 scoring algorithms which were the percentage of PD-L1-expressing tumour cell (TC) and tumour proportion score (TPS) by Ventana SP263 and Dako 22C3 staining, respectively. In NSCLC, the comparison demonstrated the comparability of the SP263 and 22C3 assays (cut-off of 1%, κ=0.71; 25%, κ=0.75; 50%, κ=0.81). The interobserver comparisons showed moderate to almost perfect agreement for SP263 in TC staining at 25% cut-off (NSCLC, κ=0.72 to 0.86; HNSCC, κ=0.60 to 0.82; UC, κ=0.68 to 0.91) and at 50% cut-off for NSCLC (κ=0.64 to 0.90). Regarding the immune cell (IC) scoring in UC, there was a lower correlation (concordance correlation coefficient=0.10 to 0.68) and poor to substantial agreements at the 1%, 5%, 10% and 25% cut-offs (κ= -0.04 to 0.76). The interchangeability of SP263 and 22C3 in NSCLC might be acceptable, especially at the 50% cut-off. In HNSCC, the performance of SP263 is comparable across five countries. In UC, there was low concordance of IC staining, which may affect treatment decisions. Overall, the study showed the reliability and reproducibility of SP263 in NSCLC, HNSCC and UC.
Collapse
Affiliation(s)
- Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - Yi-Jing Hsiao
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan.
| | - Wendy A Cooper
- Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia; School of Medicine, Western Sydney University, Sydney, NSW, Australia.
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | | | - Teh-Ying Chou
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Renata Coudry
- Department of Pathology, Sirio Libanes Hospital and United Health Group Brazil, Sao Paulo, Brazil.
| | - Grigory A Raskin
- A.M. Granov Russian Scientific Center of Radiological and Surgical Technologies, St Petersburg, Russia.
| | - Stephen B Fox
- Molecular Pathology Laboratory, Peter MacCallum Cancer Centre and University of Melbourne, Melbourne, Vic, Australia
| | - Chao-Cheng Huang
- Biobank and Tissue Bank and Department of Pathology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea; Cancer Research Institute, Seoul National University, Seoul, South Korea
| | - Young-Hyeh Ko
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Wen-Hui Ku
- Taipei Institute of Pathology, Taipei, Taiwan
| | - Ghee-Young Kwon
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - Mei-Chun Lin
- National Taiwan University Cancer Center, Taipei, Taiwan; Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Pei-Jen Lou
- Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Cristovam Scapulatempo-Neto
- Pathology and Molecular Diagnostics, Diagnósticos da América, DASA, São Paulo, Brazil; Molecular Oncology Research Center, Hospital de Amor de Barretos, Barretos, Brazil
| | | | | | - Hyo-Sup Shim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | - Isabela Werneck Cunha
- Institute of Anatomical Pathology, Rede D'Or São Luiz Hospitals Network, Rio de Janeiro and São Paulo, Brazil; D'Or Institute for Research and Education, Rio de Janeiro and São Paulo, Brazil
| | - Larisa Zavalishina
- Pathology Department of the Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - Yan-Ming Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|