1
|
Long-Mira E, Bontoux C, Rignol G, Hofman V, Lassalle S, Benzaquen J, Boutros J, Lalvée-Moret S, Zahaf K, Lespinet-Fabre V, Bordone O, Maistre S, Bonnetaud C, Cohen C, Berthet JP, Marquette CH, Vouret-Craviari V, Ilié M, Hofman P. Exploring the Expression of CD73 in Lung Adenocarcinoma with EGFR Genomic Alterations. Cancers (Basel) 2025; 17:1034. [PMID: 40149368 PMCID: PMC11941413 DOI: 10.3390/cancers17061034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Immune checkpoint inhibitors (ICIs) benefit some lung cancer patients, but their efficacy is limited in advanced lung adenocarcinoma (LUAD) with EGFR mutations (EGFRm), largely due to a non-immunogenic tumour microenvironment (TME). Furthermore, EGFRm LUAD patients often experience increased toxicity with ICIs. CD73, an ectonucleotidase involved in adenosine production, promotes tumour immune evasion and could represent a novel therapeutic target. This study investigates CD73 expression in LUAD with EGFR alterations and its clinico-pathological correlations. METHODS CD73 expression in tumour (CD73TC) and stromal (CD73SC) cells was assessed in 76 treatment-naive LUAD patients using immunohistochemistry (IHC) (D7F9A clone) alongside IHC PD-L1 (22C3 clone). EGFR alterations were identified by molecular sequencing and FISH. Event-free survival (EFS) was analysed based on CD73TC expression. RESULTS CD73TC expression was observed in 66% of cases, with high expression (Tumour Proportion Score > 50%) correlating with improved EFS (p = 0.045). CD73TC and PD-L1 expression were not significantly correlated (p = 0.44), although a weak inverse trend was observed. CD73SC expression was detected in 18% of cases, predominantly in early-stage (p = 0.037), PD-L1-negative (p = 0.030), and non-EGFR-amplified (p = 0.0018) tumours. No significant associations were found with disease stage, histological subtype, EGFR mutation type, and amplification. CONCLUSIONS CD73 expression in EGFRm LUAD is heterogeneous and associated with diverse TME profiles. These findings support the potential of CD73 as a predictive biomarker and therapeutic target, highlighting its clinical relevance in EGFRm LUAD.
Collapse
Affiliation(s)
- Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Christophe Bontoux
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Guylène Rignol
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Jonathan Benzaquen
- Department of Thoracic Oncology, IHU RespirERA Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (J.B.); (J.B.); (C.-H.M.)
| | - Jacques Boutros
- Department of Thoracic Oncology, IHU RespirERA Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (J.B.); (J.B.); (C.-H.M.)
| | - Salomé Lalvée-Moret
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Katia Zahaf
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Virginie Lespinet-Fabre
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Olivier Bordone
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Sophia Maistre
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Christelle Bonnetaud
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Charlotte Cohen
- Department of Thoracic Surgery, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (C.C.); (J.-P.B.)
| | - Jean-Philippe Berthet
- Department of Thoracic Surgery, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (C.C.); (J.-P.B.)
| | - Charles-Hugo Marquette
- Department of Thoracic Oncology, IHU RespirERA Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (J.B.); (J.B.); (C.-H.M.)
| | - Valerie Vouret-Craviari
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| |
Collapse
|
2
|
Petruk N, Wood SL, Gregory W, Lopez-Guajardo A, Oliva M, Mella M, Sandholm J, Jukkola A, Brown JE, Selander KS. Increased primary breast tumor expression of CD73 is associated with development of bone metastases and is a potential biomarker for adjuvant bisphosphonate use. Sci Rep 2025; 15:9449. [PMID: 40108234 PMCID: PMC11923362 DOI: 10.1038/s41598-025-92841-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025] Open
Abstract
PURPOSE Increased CD73 expression has been associated with progression in various cancer types. Results of the AZURE and other trials suggest that, in postmenopausal breast cancer patients, adjuvant bisphosphonates inhibit bone relapses and prolong overall survival. Based on these findings, adjuvant bisphosphonates (typically zoledronic acid) are standard-of-care in postmenopausal patients with high-risk early breast cancer. However, biomarkers are needed for improved patient selection. The aim of this study was to investigate the association of primary tumor CD73 expression with later development of bone metastases. METHODS To determine whether CD73 levels correlated with tumor parameters (hormone receptor status, tumor stage and grade), patient outcomes (bone metastases and survival) or other patient characteristics (menopausal status, chemotherapy or statin use), we analyzed primary breast tumor CD73 expression immunohistochemically in tumor microarray samples from the AZURE (BIG01/04) trial. RESULTS In the AZURE control arm, high CD73 score are significantly prognostic for overall survival (p-value = 0.03, HR = 1.87, 95% CI = 1.06-3.29), disease-free survival (p-value = 0.06, HR = 1.66, 95% CI = 0.982-2.8) and time to first metastasis to bone (p-value = 0.04, HR = 2.23, 95% CI = 1.04-4.81), as compared with low CD73 scores. However, high CD73 score did not display an association with time to non-bone metastasis or first recurrence to a non-skeletal site. In the zoledronate arm, high CD73 score did not have association with patient outcomes, first metastasis to bone, nor with bone recurrence at any time (distant recurrence, including skeletal) or first non-skeletal recurrence. In multivariate testing, CD73 had no significant association with age, ER status, tumor stage, histological grade, menopausal status, chemotherapy or statin use in either arm. CONCLUSIONS High CD73 expression is associated with development of bone metastases. Zoledronate counteracts this effect. These results suggest that CD73 expression might serve as a biomarker for adjuvant zoledronic acid use.
Collapse
Affiliation(s)
- Nataliia Petruk
- Institute of Biomedicine, University of Turku, Turku, Finland
- Western Cancer Centre FICAN West, Turku, Finland
| | - Steven L Wood
- Division of Clinical Medicine, Medical School, Sheffield, UK
| | - Walter Gregory
- Division of Clinical Medicine, Medical School, Sheffield, UK
| | | | - Maria Oliva
- Division of Clinical Medicine, Medical School, Sheffield, UK
| | - Mikko Mella
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Oncology and Radiation Therapy, Oulu University Hospital, Oulu, Finland
| | - Jouko Sandholm
- Turku Bioscience Centre, University of Turku, Åbo Akademi University, Turku, Finland
| | - Arja Jukkola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Janet E Brown
- Division of Clinical Medicine, Medical School, Sheffield, UK
| | - Katri S Selander
- Department of Oncology and Radiation Therapy, Oulu University Hospital, Oulu, Finland.
- Department of Translational Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
3
|
Gao H, Zhang T, Li K, Li X. CD73: a new immune checkpoint for leukemia treatment. Front Immunol 2025; 16:1486868. [PMID: 40114928 PMCID: PMC11922907 DOI: 10.3389/fimmu.2025.1486868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
Recent studies on the pathogenesis of leukemia have led to remarkable advances in disease treatment. Numerous studies have shown the potential and viability of immune responses against leukemia. In the classical pathway, this process is often initiated by the upstream activity of CD39, which hydrolyzes extracellular adenosine triphosphate (ATP) and adenosine diphosphate (ADP) to AMP. Subsequently, CD73 acts on AMP to generate adenosine, contributing to an immunosuppressive microenvironment. However, CD73 can also utilize substrates derived from other molecules through the non-canonical NAD+ pathway, specifically via the CD38/CD203a/CD73 axis, further enhancing adenosine production and facilitating immune escape. Targeting CD73 has shown potential in disrupting these immunosuppressive pathways, thereby enhancing anti-leukemic immune responses and improving patient outcomes. Inhibiting CD73 not only reduces the levels of immunosuppressive adenosine but also increases the efficacy of existing immunotherapies, such as PD-1/PD-L1 inhibitors, making it a versatile therapeutic target in leukemia treatment. This review discusses the potential of CD73 as a therapeutic target and emphasizes its unique position in the immune escape mechanism of leukemia. Moreover, this review provides an overview of the current research progress and future trends, emphasizing the clinical significance of targeting CD73 and other potential therapeutic strategies in leukemia.
Collapse
Affiliation(s)
- Huan Gao
- Marine College, Shandong University, Weihai, China
| | - Tingting Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ke Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xia Li
- Marine College, Shandong University, Weihai, China
| |
Collapse
|
4
|
Deng XC, Liang JL, Zhang SM, Wang YZ, Lin YT, Meng R, Wang JW, Feng J, Chen WH, Zhang XZ. Interference of ATP-Adenosine Axis by Engineered Biohybrid for Amplifying Immunogenic Cell Death-Mediated Antitumor Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405673. [PMID: 39022876 DOI: 10.1002/adma.202405673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/07/2024] [Indexed: 07/20/2024]
Abstract
Immunogenic cell death (ICD) often results in the production and accumulation of adenosine (ADO), a byproduct that negatively impacts the therapeutic effect as well as facilitates tumor development and metastasis. Here, an innovative strategy is elaborately developed to effectively activate ICD while avoiding the generation of immunosuppressive adenosine. Specifically, ZIF-90, an ATP-responsive consumer, is synthesized as the core carrier to encapsulate AB680 (CD73 inhibitor) and then coated with an iron-polyphenol layer to prepare the ICD inducer (AZTF), which is further grafted onto prebiotic bacteria via the esterification reaction to obtain the engineered biohybrid (Bc@AZTF). Particularly, the designed Bc@AZTF can actively enrich in tumor sites and respond to the acidic tumor microenvironment to offload AZTF nanoparticles, which can consume intracellular ATP (iATP) content and simultaneously inhibit the ATP-adenosine axis to reduce the accumulation of adenosine, thereby alleviating adenosine-mediated immunosuppression and strikingly amplifying ICD effect. Importantly, the synergy of anti-PD-1 (αPD-1) with Bc@AZTF not only establishes a collaborative antitumor immune network to potentiate effective tumoricidal immunity but also activates long-lasting immune memory effects to manage tumor recurrence and rechallenge, presenting a new paradigm for ICD treatment combined with adenosine metabolism.
Collapse
Affiliation(s)
- Xin-Chen Deng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Jun-Long Liang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Shi-Man Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Yu-Zhang Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Yan-Tong Lin
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Ran Meng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Jia-Wei Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| |
Collapse
|
5
|
Saigí M, Mate JL, Carcereny E, Martínez-Cardús A, Esteve A, Andreo F, Centeno C, Cucurull M, Mesia R, Pros E, Sanchez-Cespedes M. HLA-I levels correlate with survival outcomes in response to immune checkpoint inhibitors in non-small cell lung cancer. Lung Cancer 2024; 189:107502. [PMID: 38359742 DOI: 10.1016/j.lungcan.2024.107502] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/16/2024] [Accepted: 02/04/2024] [Indexed: 02/17/2024]
Abstract
OBJECTIVES Immune checkpoint inhibitors (ICIs) have provided a breakthrough in the treatment of non-small cell lung cancer (NSCLC) patients, but only some patients benefit substantively. Identifying definitive predictive biomarkers could overcome this limitation. MATERIALS AND METHODS We selected 146 metastatic NSCLC patients treated with anti-PD-(L)1. Immunohistochemistry of HLA-I, PD-L1 and CD73 was performed in 122 tumor biopsies at diagnosis. The association with patients, tumor parameters, and the predictive value to ICI treatment were determined. RESULTS In our cohort, 42 %, 25 %, and 21 % of the tumors exhibited high levels of HLA-I, PD-L1, and CD73, respectively. Lung adenocarcinomas displayed elevated CD73 levels, compared with lung squamous cell carcinomas (P = 0.026). High PD-L1 was significantly correlated with high levels of HLA-I (P = 0.005) and of CD73 (P = 0.025). Patients with high-level HLA-I tumors exhibited more favorable clinical outcomes following ICI, with a median overall survival of 30.7 months (95 % confidence interval [CI]: 18.3 months-not reached), compared with 18.2 months (95 % CI: 12.4-25.2 months) in patients with low-level HLA-I tumors (P = 0.016). The median progression-free survival (PFS) for patients with high-level HLA-I tumors was 18.5 months (95 % CI: 11.1-57.1 months), longer than patients with low-level HLA-I tumors, whose median PFS was 9.2 months (95 % CI: 7.2-11.9 months) (P = 0.006). In a multivariable analysis, high-level HLA-I was independently associated with lower risk of progression to ICI (HR = 0.46, 95 % CI 0.24-0.87; P = 0.018). CONCLUSIONS High-level HLA-I were associated with better clinical outcomes to ICI in our cohort of NSCLC patients. Therefore, further investigations are warranted to refine this biomarker and validate its efficacy in prospective and larger set of patients.
Collapse
Affiliation(s)
- Maria Saigí
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Badalona, Barcelona, Spain; Badalona·Applied Research Group in Oncology (B·ARGO), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Badalona, Barcelona, Spain.
| | - Jose L Mate
- Department of Pathology, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain
| | - Enric Carcereny
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Badalona, Barcelona, Spain; Badalona·Applied Research Group in Oncology (B·ARGO), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Badalona, Barcelona, Spain
| | - Anna Martínez-Cardús
- Badalona·Applied Research Group in Oncology (B·ARGO), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Badalona, Barcelona, Spain
| | - Anna Esteve
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Badalona, Barcelona, Spain; Statistics Department, Catalan Institute of Oncology (ICO)-Badalona, Badalona, Barcelona, Spain
| | - Felipe Andreo
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Carmen Centeno
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Marc Cucurull
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Badalona, Barcelona, Spain; Badalona·Applied Research Group in Oncology (B·ARGO), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Badalona, Barcelona, Spain
| | - Ricard Mesia
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Badalona, Barcelona, Spain; Badalona·Applied Research Group in Oncology (B·ARGO), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Badalona, Barcelona, Spain
| | - Eva Pros
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Montse Sanchez-Cespedes
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain.
| |
Collapse
|
6
|
Cho U, Im S, Park HS. Exploring histological predictive biomarkers for immune checkpoint inhibitor therapy response in non-small cell lung cancer. J Pathol Transl Med 2024; 58:49-58. [PMID: 38389279 PMCID: PMC10948248 DOI: 10.4132/jptm.2024.01.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/16/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
Treatment challenges persist in advanced lung cancer despite the development of therapies beyond the traditional platinum-based chemotherapy. The early 2000s marked a shift to tyrosine kinase inhibitors targeting epidermal growth factor receptor, ushering in personalized genetic-based treatment. A further significant advance was the development of immune checkpoint inhibitors (ICIs), especially for non-small cell lung cancer. These target programmed death-ligand 1 (PD-L1) and cytotoxic T lymphocyte antigen 4, which enhanced the immune response against tumor cells. However, not all patients respond, and immune-related toxicities arise. This review emphasizes identifying biomarkers for ICI response prediction. While PD-L1 is a widely used, validated biomarker, its predictive accuracy is imperfect. Investigating tumor-infiltrating lymphocytes, tertiary lymphoid structure, and emerging biomarkers such as high endothelial venule, Human leukocyte antigen class I, T-cell immunoreceptors with Ig and ITIM domains, and lymphocyte activation gene-3 counts is promising. Understanding and exploring additional predictive biomarkers for ICI response are crucial for enhancing patient stratification and overall care in lung cancer treatment.
Collapse
Affiliation(s)
- Uiju Cho
- Department of Pathology, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Soyoung Im
- Department of Pathology, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Hyung Soon Park
- Division of Medical Oncology, Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| |
Collapse
|
7
|
Wang R, Liu Z, Wang T, Zhang J, Liu J, Zhou Q. Landscape of adenosine pathway and immune checkpoint dual blockade in NSCLC: progress in basic research and clinical application. Front Immunol 2024; 15:1320244. [PMID: 38348050 PMCID: PMC10859755 DOI: 10.3389/fimmu.2024.1320244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/10/2024] [Indexed: 02/15/2024] Open
Abstract
Lung cancer poses a global threat to human health, while common cancer treatments (chemotherapy and targeted therapies) have limited efficacy. Immunotherapy offers hope of sustained remission for many patients with lung cancer, but a significant proportion of patients fail to respond to treatment owing to immune resistance. There is extensive evidence to suggest the immunosuppressive microenvironment as the cause of this treatment failure. Numerous studies have suggested that the adenosine (ADO) pathway plays an important role in the formation of an immunosuppressive microenvironment and may be a key factor in the development of immune resistance in EGFR-mutant cell lung cancer. Inhibition of this pathway may therefore be a potential target to achieve effective reversal of ADO pathway-mediated immune resistance. Recently, an increasing number of clinical trials have begun to address the broad prospects of using the ADO pathway as an immunotherapeutic strategy. However, few researchers have summarized the theoretical basis and clinical rationale of the ADO pathway and immune checkpoint dual blockade in a systematic and detailed manner, particularly in lung cancer. As such, a timely review of the potential value of the ADO pathway in combination with immunotherapy strategies for lung cancer is warranted. This comprehensive review first describes the role of ADO in the formation of a lung tumor-induced immunosuppressive microenvironment, discusses the key mechanisms of ADO inhibitors in reversing lung immunosuppression, and highlights recent evidence from preclinical and clinical studies of ADO inhibitors combined with immune checkpoint blockers to improve the lung cancer immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Rulan Wang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenkun Liu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Wang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiabi Zhang
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, United States
| | - Jiewei Liu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qinghua Zhou
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Lima CF, Tamegnon A, Rodriguez S, Maru D, Martin PL, Cooper ZA, Rodriguez-Canales J, Parra ER. Exploring the Expression of Adenosine Pathway-Related Markers CD73 and CD39 in Colorectal and Pancreatic Carcinomas Characterized by Multiplex Immunofluorescence: A Pilot Study. Pathobiology 2023; 91:205-218. [PMID: 37926083 PMCID: PMC11524541 DOI: 10.1159/000534677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/30/2023] [Indexed: 11/07/2023] Open
Abstract
INTRODUCTION Generating high levels of immunosuppressive adenosine (ADO) in the tumor microenvironment contributes to cancer immune evasion. CD39 and CD73 hydrolyze adenosine triphosphate into ADO; thus, efforts have been made to target this pathway for cancer immunotherapy. Our objective was optimizing a multiplex immunofluorescence (mIF) panel to explore the role of CD39 and CD73 within the tumor microenvironment. MATERIALS AND METHODS In three-time points, a small cohort (n = 8) of colorectal and pancreatic adenocarcinomas were automated staining using an mIF panel against CK, CD3, CD8, CD20, CD39, CD73, and CD68 to compare them with individual markers immunohistochemistry (IHC) for internal panel validation. Densities of immune cells and distances from different tumor-associated immune cells to tumor cells were exploratory assessment and compared with clinicopathologic variables and outcomes. RESULTS Comparing the three-time points and individual IHC staining results, we demonstrated high reproducibility of the mIF panel. CD39 and CD73 expression was low in malignant cells; the exploratory analysis showed higher densities of CD39 expression by various cells, predominantly stromal cells, followed by T cells, macrophages, and B cells. No expression of CD73 by B cells or macrophages was detected. Distance analysis revealed proximity of cytotoxic T cells, macrophages, and T cells expressing CD39 to malignant cells, suggesting a close regulatory signal driven by this ADO marker. CONCLUSIONS We optimized an mIF panel for detection of markers in the ADO pathway, an emerging clinically relevant pathway. The densities and spatial distribution demonstrated that this pathway may modulate aspects of the tumor immune microenvironment.
Collapse
Affiliation(s)
- Cibelle Freitas Lima
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA,
| | - Auriole Tamegnon
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Saxon Rodriguez
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dipen Maru
- Departments of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Philip L Martin
- Department of Translational Medicine, Oncology R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Zachary A Cooper
- Department of Translational Medicine, Oncology R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Jaime Rodriguez-Canales
- Department of Translational Medicine, Oncology R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Edwin Roger Parra
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
9
|
Bendell J, LoRusso P, Overman M, Noonan AM, Kim DW, Strickler JH, Kim SW, Clarke S, George TJ, Grimison PS, Barve M, Amin M, Desai J, Wise-Draper T, Eck S, Jiang Y, Khan AA, Wu Y, Martin P, Cooper ZA, Elgeioushi N, Mueller N, Kumar R, Patel SP. First-in-human study of oleclumab, a potent, selective anti-CD73 monoclonal antibody, alone or in combination with durvalumab in patients with advanced solid tumors. Cancer Immunol Immunother 2023; 72:2443-2458. [PMID: 37016126 PMCID: PMC10264501 DOI: 10.1007/s00262-023-03430-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/19/2023] [Indexed: 04/06/2023]
Abstract
BACKGROUND CD73 upregulation in tumors leads to local immunosuppression. This phase I, first-in-human study evaluated oleclumab (MEDI9447), an anti-CD73 human IgG1λ monoclonal antibody, alone or with durvalumab in patients with advanced colorectal cancer (CRC), pancreatic ductal adenocarcinoma (PDAC), or epidermal growth factor receptor-mutant non-small-cell lung cancer (NSCLC). METHODS Patients received oleclumab 5-40 mg/kg (dose-escalation) or 40 mg/kg (dose-expansion) intravenously every 2 weeks (Q2W), alone (escalation only) or with durvalumab 10 mg/kg intravenously Q2W. RESULTS 192 patients were enrolled, 66 during escalation and 126 (42 CRC, 42 PDAC, 42 NSCLC) during expansion. No dose-limiting toxicities occurred during escalation. In the monotherapy and combination therapy escalation cohorts, treatment-related adverse events (TRAEs) occurred in 55 and 54%, respectively, the most common being fatigue (17 and 25%). In the CRC, PDAC, and NSCLC expansion cohorts, 60, 57, and 45% of patients had TRAEs, respectively; the most common were fatigue (15%), diarrhea (9%), and rash (7%). Free soluble CD73 and CD73 expression on peripheral T cells and tumor cells showed sustained decreases, accompanied by reduced CD73 enzymatic activity in tumor cells. Objective response rate during escalation was 0%. Response rates in the CRC, PDAC, and NSCLC expansion cohorts were 2.4% (1 complete response [CR]), 4.8% (1 CR, 1 partial response [PR]), and 9.5% (4 PRs), respectively; 6-month progression-free survival rates were 5.4, 13.2, and 16.0%. CONCLUSIONS Oleclumab ± durvalumab had a manageable safety profile, with pharmacodynamic activity reflecting oleclumab's mechanism of action. Evidence of antitumor activity was observed in tumor types that are generally immunotherapy resistant. CLINICAL TRIAL REGISTRATION Clinicaltrials.gov, NCT02503774; date of registration, July 17, 2015.
Collapse
Affiliation(s)
- Johanna Bendell
- Sarah Cannon Research Institute, Nashville, TN, USA.
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | | | - Michael Overman
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anne M Noonan
- Ohio State University, Wexner Medical Center, James Comprehensive Cancer Center, Columbus, OH, USA
| | - Dong-Wan Kim
- Seoul National University Hospital, Seoul, South Korea
| | | | | | - Stephen Clarke
- Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Thomas J George
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | | | - Minal Barve
- Mary Crowley Cancer Research, Dallas, TX, USA
| | - Manik Amin
- Washington University School of Medicine, St. Louis, MO, USA
- Dartmouth-Hitchcock Medical Center, Norris Cotton Cancer Center, Lebanon, NH, USA
| | - Jayesh Desai
- Royal Melbourne Hospital, Parkville, VIC, Australia
| | | | | | - Yu Jiang
- AstraZeneca, Gaithersburg, MD, USA
| | | | | | | | | | | | | | | | - Sandip Pravin Patel
- Moores Cancer Center, University of California San Diego, La Jolla, San Diego, CA, USA
| |
Collapse
|
10
|
Kurago Z, Guo G, Shi H, Bollag RJ, Groves MW, Byrd JK, Cui Y. Inhibitors of the CD73-adenosinergic checkpoint as promising combinatory agents for conventional and advanced cancer immunotherapy. Front Immunol 2023; 14:1212209. [PMID: 37435071 PMCID: PMC10330720 DOI: 10.3389/fimmu.2023.1212209] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/31/2023] [Indexed: 07/13/2023] Open
Abstract
The cell surface enzyme CD73 is increasingly appreciated as a pivotal non-redundant immune checkpoint (IC) in addition to PD-1/PD-L1 and CTLA-4. CD73 produces extracellular adenosine (eADO), which not only inhibits antitumor T cell activity via the adenosine receptor (AR) A2AR, but also enhances the immune inhibitory function of cancer-associated fibroblasts and myeloid cells via A2BR. Preclinical studies show that inhibition of the CD73-adenosinergic pathway in experimental models of many solid tumors either as a monotherapy or, more effectively, in combination with PD-1/PD-L1 or CTLA-4 IC blockades, improves antitumor immunity and tumor control. Consequently, approximately 50 ongoing phase I/II clinical trials targeting the CD73-adenosinergic IC are currently listed on https://clinicaltrials.gov. Most of the listed trials employ CD73 inhibitors or anti-CD73 antibodies alone, in combination with A2AR antagonists, and/or with PD-1/PD-L1 blockade. Recent evidence suggests that the distribution of CD73, A2AR and A2BR in tumor microenvironments (TME) is heterogeneous, and this distribution affects CD73-adenosinergic IC function. The new insights have implications for the optimally effective, carefully tailored approaches to therapeutic targeting of this essential IC. In the mini-review, we briefly discuss the cellular and molecular mechanisms of CD73/eADO-mediated immunosuppression during tumor progression and therapy in the spatial context of the TME. We include preclinical data regarding therapeutic CD73-eADO blockade in tumor models as well as available clinical data from completed trials that targeted CD73-adenosinergic IC with or without PD-1/PD-L1 inhibitors and discuss factors that are potentially important for optimal therapeutic outcomes in cancer patients.
Collapse
Affiliation(s)
- Zoya Kurago
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Gang Guo
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Huidong Shi
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Roni J. Bollag
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Michael W. Groves
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Otolaryngology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - J. Kenneth Byrd
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Otolaryngology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Yan Cui
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
11
|
Zhang M, Dai X, Xiang Y, Xie L, Sun M, Shi J. Advances in CD73 inhibitors for immunotherapy: Antibodies, synthetic small molecule compounds, and natural compounds. Eur J Med Chem 2023; 258:115546. [PMID: 37302340 DOI: 10.1016/j.ejmech.2023.115546] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/20/2023] [Accepted: 06/04/2023] [Indexed: 06/13/2023]
Abstract
Tumors, a disease with a high mortality rate worldwide, have become a serious threat to human health. Exonucleotide-5'-nucleotidase (CD73) is an emerging target for tumor therapy. Its inhibition can significantly reduce adenosine levels in the tumor microenvironment. It has a better therapeutic effect on adenosine-induced immunosuppression. In the immune response, extracellular ATP exerts immune efficacy by activating T cells. However, dead tumor cells release excess ATP, overexpress CD39 and CD73 on the cell membrane and catabolize this ATP to adenosine. This leads to further immunosuppression. There are a number of inhibitors of CD73 currently under investigation. These include antibodies, synthetic small molecule inhibitors and a number of natural compounds with prominent roles in the anti-tumor field. However, only a small proportion of the CD73 inhibitors studied to date have successfully reached the clinical stage. Therefore, effective and safe inhibition of CD73 in oncology therapy still holds great therapeutic potential. This review summarizes the currently reported CD73 inhibitors, describes their inhibitory effects and pharmacological mechanisms, and provides a brief review of them. It aims to provide more information for further research and development of CD73 inhibitors.
Collapse
Affiliation(s)
- Mingxue Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Xiaoqin Dai
- Department of Traditional Chinese Medicine, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan Province, China
| | - Yu Xiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Linshen Xie
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
| | - Minghan Sun
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
12
|
Wu S, Sun Z, Guo Z, Li P, Mao Q, Tang Y, Chen H, Peng H, Wang S, Cao Y. The effectiveness of blood-activating and stasis-transforming traditional Chinese medicines (BAST) in lung cancer progression-a comprehensive review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116565. [PMID: 37172918 DOI: 10.1016/j.jep.2023.116565] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/20/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Blood-activating and stasis-transforming traditional Chinese medicines (BAST) are a class of herbs that have the effect of dilating blood vessels and dispersing stagnation. Modern pharmaceutical research has demonstrated that they are capable of improving hemodynamics and micro-flow, resist thrombosis and promote blood flow. BAST contain numerous active ingredients, which can theoretically regulate multiple targets at the same time and have a wide range of pharmacological effects in the treatment of diseases including human cancers. Clinically, BAST have minimal side effects and can be used in combination with Western medicine to improve patients' quality of life, lessen adverse effects and minimize the risk of recurrence and metastasis of cancers. AIM OF THE REVIEW We aimed to summarize the research progression of BAST on lung cancer in the past five years and present a prospect for the future. Particularly, this review further analyzes the effects and molecular mechanisms that BAST inhibit the invasion and metastasis of lung cancer. MATERIALS AND METHODS Relevant studies about BSAT were collected from PubMed and Web of science. RESULTS Lung cancer is one of the malignant tumors with the highest mortality rate. Most patients with lung cancer are diagnosed at an advanced stage and are highly susceptible to metastasis. Recent studies have shown that BAST, a class of traditional Chinese medicine (TCM) with the function of opening veins and dispersing blood stasis, significantly improve hemodynamics and microcirculation, prevent thrombosis and promote blood flow, and thereby inhibiting the invasion and metastasis of lung cancer. In the current review, we analyzed 51 active ingredients extracted from BAST. It was found that BAST and their active ingredients contribute to the prevention of invasion and metastasis of lung cancer through multiple mechanisms, such as regulation of EMT process, specific signaling pathway and metastasis-related genes, tumor blood vessel formation, immune microenvironment and inflammatory response of tumors. CONCLUSIONS BSAT and its active ingredients have showed promising anticancer activity and significantly inhibit the invasion and metastasis of lung cancer. A growing number of studies have realized their potential clinical significance in the therapy of lung cancer, which will provide substantial evidences for the development of new TCM for lung cancer therapy.
Collapse
Affiliation(s)
- Siqi Wu
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Zhe Sun
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Zehuai Guo
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Peiqin Li
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Qianqian Mao
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yang Tang
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Hongyu Chen
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Huiting Peng
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Sisi Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yang Cao
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
13
|
CD73: Friend or Foe in Lung Injury. Int J Mol Sci 2023; 24:ijms24065545. [PMID: 36982618 PMCID: PMC10056814 DOI: 10.3390/ijms24065545] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023] Open
Abstract
Ecto-5′-nucleotidase (CD73) plays a strategic role in calibrating the magnitude and chemical nature of purinergic signals that are delivered to immune cells. Its primary function is to convert extracellular ATP to adenosine in concert with ectonucleoside triphosphate diphosphohydrolase-1 (CD39) in normal tissues to limit an excessive immune response in many pathophysiological events, such as lung injury induced by a variety of contributing factors. Multiple lines of evidence suggest that the location of CD73, in proximity to adenosine receptor subtypes, indirectly determines its positive or negative effect in a variety of organs and tissues and that its action is affected by the transfer of nucleoside to subtype-specific adenosine receptors. Nonetheless, the bidirectional nature of CD73 as an emerging immune checkpoint in the pathogenesis of lung injury is still unknown. In this review, we explore the relationship between CD73 and the onset and progression of lung injury, highlighting the potential value of this molecule as a drug target for the treatment of pulmonary disease.
Collapse
|
14
|
Yoshida R, Saigi M, Tani T, Springer BF, Shibata H, Kitajima S, Mahadevan NR, Campisi M, Kim W, Kobayashi Y, Thai TC, Haratani K, Yamamoto Y, Sundararaman SK, Knelson EH, Vajdi A, Canadas I, Uppaluri R, Paweletz CP, Miret JJ, Lizotte PH, Gokhale PC, Jänne PA, Barbie DA. MET-Induced CD73 Restrains STING-Mediated Immunogenicity of EGFR-Mutant Lung Cancer. Cancer Res 2022; 82:4079-4092. [PMID: 36066413 PMCID: PMC9627131 DOI: 10.1158/0008-5472.can-22-0770] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/27/2022] [Accepted: 08/29/2022] [Indexed: 12/14/2022]
Abstract
Immunotherapy has shown limited efficacy in patients with EGFR-mutated lung cancer. Efforts to enhance the immunogenicity of EGFR-mutated lung cancer have been unsuccessful to date. Here, we discover that MET amplification, the most common mechanism of resistance to third-generation EGFR tyrosine kinase inhibitors (TKI), activates tumor cell STING, an emerging determinant of cancer immunogenicity (1). However, STING activation was restrained by ectonucleosidase CD73, which is induced in MET-amplified, EGFR-TKI-resistant cells. Systematic genomic analyses and cell line studies confirmed upregulation of CD73 in MET-amplified and MET-activated lung cancer contexts, which depends on coinduction of FOSL1. Pemetrexed (PEM), which is commonly used following EGFR-TKI treatment failure, was identified as an effective potentiator of STING-dependent TBK1-IRF3-STAT1 signaling in MET-amplified, EGFR-TKI-resistant cells. However, PEM treatment also induced adenosine production, which inhibited T-cell responsiveness. In an allogenic humanized mouse model, CD73 deletion enhanced immunogenicity of MET-amplified, EGFR-TKI-resistant cells, and PEM treatment promoted robust responses regardless of CD73 status. Using a physiologic antigen recognition model, inactivation of CD73 significantly increased antigen-specific CD8+ T-cell immunogenicity following PEM treatment. These data reveal that combined PEM and CD73 inhibition can co-opt tumor cell STING induction in TKI-resistant EGFR-mutated lung cancers and promote immunogenicity. SIGNIFICANCE MET amplification upregulates CD73 to suppress tumor cell STING induction and T-cell responsiveness in TKI-resistant, EGFR-mutated lung cancer, identifying a strategy to enhance immunogenicity and improve treatment.
Collapse
Affiliation(s)
- Ryohei Yoshida
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Respiratory Center, Asahikawa Medical University, Hokkaido, Japan.,Corresponding authors: David A. Barbie, M.D., Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, LC4115, Boston, Massachusetts, 02215, USA, , Tel: 617-632-6036; Pasi A Jänne, M.D. Ph.D., Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, LC4114, Boston, Massachusetts, 02215, USA, , Tel: 617-632-6036; Ryohei Yoshida, M.D. Ph.D., Respiratory Center, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan, , Tel: 81-166-69-3290
| | - Maria Saigi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Department of Medical Oncology, Catalan Institute of Oncology (ICO), Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Tetsuo Tani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Benjamin F Springer
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Hirofumi Shibata
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Shunsuke Kitajima
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Navin R Mahadevan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115 USA
| | - Marco Campisi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - William Kim
- Jong Wook Kim Ph.D., University of California San Diego, School of Medicine, Moores Cancer Center
| | - Yoshihisa Kobayashi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Division of Molecular Pathology, National Cancer Center Research Institute, Tokyo, Japan
| | - Tran C Thai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Koji Haratani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Yurie Yamamoto
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Shriram K Sundararaman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Erik H Knelson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Amir Vajdi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Israel Canadas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Ravindra Uppaluri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Cloud P Paweletz
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Juan J Miret
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Patrick H Lizotte
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Prafulla C Gokhale
- Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Pasi A Jänne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Corresponding authors: David A. Barbie, M.D., Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, LC4115, Boston, Massachusetts, 02215, USA, , Tel: 617-632-6036; Pasi A Jänne, M.D. Ph.D., Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, LC4114, Boston, Massachusetts, 02215, USA, , Tel: 617-632-6036; Ryohei Yoshida, M.D. Ph.D., Respiratory Center, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan, , Tel: 81-166-69-3290
| | - David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.,Corresponding authors: David A. Barbie, M.D., Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, LC4115, Boston, Massachusetts, 02215, USA, , Tel: 617-632-6036; Pasi A Jänne, M.D. Ph.D., Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, LC4114, Boston, Massachusetts, 02215, USA, , Tel: 617-632-6036; Ryohei Yoshida, M.D. Ph.D., Respiratory Center, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan, , Tel: 81-166-69-3290
| |
Collapse
|
15
|
Akhave N, Zhang J, Bayley E, Frank M, Chiou SH, Behrens C, Chen R, Hu X, Parra ER, Lee WC, Swisher S, Solis L, Weissferdt A, Moran C, Kalhor N, Zhang J, Scheet P, Vaporciyan AA, Sepesi B, Gibbons DL, Heymach JV, Lee JJ, Wistuba II, Andrew Futreal P, Zhang J, Fujimoto J, Reuben A. Immunogenomic profiling of lung adenocarcinoma reveals poorly differentiated tumors are associated with an immunogenic tumor microenvironment. Lung Cancer 2022; 172:19-28. [PMID: 35973335 DOI: 10.1016/j.lungcan.2022.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 10/15/2022]
Abstract
OBJECTIVES Pathologists have routinely observed distinct histologic patterns of growth in early-stage lung adenocarcinoma (LUAD), which have been suggested to be associated with prognosis. Herein, we investigated the relationship between LUAD patterns of growth, as defined by the updated international association for the study of lung cancer (IASLC) grading criteria, and differences in the tumor immune microenvironment to identify predictors of response to immunotherapy. METHODS 174 resected stage I-III LUAD tumors were classified by histologic pattern of growth (i.e. solid, micropapillary, acinar, papillary, and lepidic) and then grouped as well differentiated, moderately differentiated, and poorly differentiated. Comprehensive multiplatform analysis including whole exome sequencing, gene expression profiling, immunohistochemistry, CIBERSORT, and T-cell receptor sequencing was performed and groups were compared for differences in genomic drivers, immune cell infiltrate, clonality, and survival. Finally, multivariate analysis was performed adjusting for pathologic stage and smoking status. RESULTS Poorly differentiated tumors demonstrated a strong association with smoking relative to moderately differentiated or well differentiated tumors. However, unlike in prior reports, poorly differentiated tumors were not associated with a worse survival after curative-intent resection. Genomic analysis revealed that poorly differentiated tumors are associated with high tumor mutation burden but showed no association with oncogenic drivers. Immune analyses revealed that poorly differentiated tumors are associated with increased T-cell clonality, expression of PD-L1, and infiltration by cytotoxic CD8 T-cells, activated CD4 T-cells, and pro-inflammatory (M1) macrophages. Finally, multivariate analysis controlling for stage and smoking status confirmed independence of immune differences between IASLC grade groups. CONCLUSIONS Poorly differentiated tumors, as defined by the updated IASLC grading criteria, are associated with a distinct immunogenic tumor microenvironment that predicts for therapeutic response to immune agents, including checkpoint inhibitors, and should be included in the clinical trial design of immunotherapy studies in early-stage lung adenocarcinoma.
Collapse
Affiliation(s)
- Neal Akhave
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Jiexin Zhang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Erin Bayley
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Meredith Frank
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Shin-Heng Chiou
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, 195 Little Albany St, New Brunswick, NJ 08901, USA
| | - Carmen Behrens
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA; Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Runzhe Chen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Xin Hu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Edwin Roger Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Won-Chul Lee
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Stephen Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Luisa Solis
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Annikka Weissferdt
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA; Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Cesar Moran
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA; Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Neda Kalhor
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA; Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Paul Scheet
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA; Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Ara A Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Jack J Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Ignacio I Wistuba
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA; Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - P Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA.
| | - Junya Fujimoto
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA.
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA.
| |
Collapse
|
16
|
Bauer A, Gebauer N, Knief J, Tharun L, Arnold N, Riecke A, Steinestel K, Witte HM. The expression of the adenosine pathway markers CD39 and CD73 in salivary gland carcinomas harbors the potential for novel immune checkpoint inhibition. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04211-x. [PMID: 35902382 DOI: 10.1007/s00432-022-04211-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/13/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND In salivary gland carcinomas (SGC), there is only a small fraction of entities that appears to profit from immune checkpoint inhibition (ICI). Recent findings connected the activation of adenosine-signaling with a tolerogenic microenvironment. Therefore, the inhibition of adenosine pathway markers (CD39 and/or CD73) can augment ICI and/or display a novel immunotherapeutic strategy beyond ICI. Here, we assessed the immuno-histochemical expression of CD39 and CD73 across a wide spectrum of SGCs. METHODS In total, 114 patients with SGCs consecutively diagnosed between 2001 and 2021 were assessed for clinicopathological baseline characteristics and underwent confirmatory histopathological review. Immunohistochemical expression levels of CD39 and CD73 were assessed by applying the tumor proportion score (TPS) and the immune proportional score (IPS) comparable to PD-L1 expression analysis in routine clinical practice. Additionally, findings were correlated with PD-L1 expression levels. RESULTS The median age was 60.6 and 51.8% patients were female. The cohort covered a spectrum of eight distinct entities. Advanced-stage disease (UICC/AJCC III/IVA-IVC) at initial diagnosis was present in the majority of patients (64/114). Immunohistochemical staining revealed positivity for CD39 and CD73 in 48.2% and 21.1% on tumor cells (TPS ≥ 1%) as well as 46.4% and 42.9% within the immune cell infiltrate (IPS ≥ 1%), respectively. Further comparative analyses revealed immune-cold entities such adenoid cystic carcinoma (AdCC), immune-hot tumors such as adenocarcinoma, not otherwise specified (AC (NOS)) and entities with intermediate immunologic features such as acinic cell carcinoma (ACC). CONCLUSION Current results indicate entity-specific adenosine signaling signatures. These findings suggest that the adenosine pathway plays a decisive role in tumor immunity among the major spectrum of SGCs. Targeting the adenosine pathway might pose a promising therapeutic option for selected entities.
Collapse
Affiliation(s)
- Arthur Bauer
- Department of Hematology and Oncology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany.,Institute of Pathology and Molecular Pathology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany
| | - Niklas Gebauer
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Juliana Knief
- Institute of Pathology, Marienkrankenhaus Hamburg, Alfredstraße 9, 22087, Hamburg, Germany
| | - Lars Tharun
- Institute of Pathology, University Hospital of Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Nele Arnold
- Department of ENT, Federal Armed Forces Hospital Hamburg, Lesserstraße 180, 22049, Hamburg, Germany
| | - Armin Riecke
- Department of Hematology and Oncology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany
| | - Konrad Steinestel
- Institute of Pathology and Molecular Pathology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany
| | - Hanno M Witte
- Department of Hematology and Oncology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany. .,Institute of Pathology and Molecular Pathology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany. .,Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
| |
Collapse
|
17
|
Liu T, Guo L, Liu G, Xie F, Zhang J, Dai Z, Wang J, Zhang J. Identification of necroptosis-related signature and tumor microenvironment infiltration characteristics in lung adenocarcinoma. Lung Cancer 2022; 172:75-85. [DOI: 10.1016/j.lungcan.2022.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/11/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022]
|
18
|
Rocha P, Zhang J, Laza-Briviesca R, Cruz-Bermúdez A, Bota-Rabassedas N, Sanchez-Espiridion B, Yoshimura K, Behrens C, Lu W, Tang X, Pataer A, Parra ER, Haymaker C, Fujimoto J, Swisher SG, Heymach JV, Gibbons DL, Lee JJ, Sepesi B, Cascone T, Solis LM, Provencio M, Wistuba II, Kadara H. Distinct immune gene programs associated with host tumor immunity, neoadjuvant chemotherapy and chemoimmunotherapy in resectable NSCLC. Clin Cancer Res 2022; 28:2461-2473. [PMID: 35394499 DOI: 10.1158/1078-0432.ccr-21-3207] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 02/12/2022] [Accepted: 03/30/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Our understanding of the immunopathology of resectable NSCLC is still limited. Here, we explore immune programs that inform of tumor immunity and response to neoadjuvant chemotherapy and chemoimmunotherapy in localized NSCLC. EXPERIMENTAL DESIGN Targeted immune gene sequencing using the HTG Precision Immuno-Oncology panel was performed in localized NSCLCs from three cohorts based on treatment: naïve (n=190), neoadjuvant chemotherapy (n=38) and neoadjuvant chemoimmunotherapy (n=21). Tumor immune microenvironment (TIME) phenotypes based on the location of CD8+ T cells (inflamed, cold, excluded), tumoral PD-L1 expression (<1% and {greater than or equal to}1%), and tumor infiltrating lymphocytes (TILs). Immune programs and signatures were statistically analyzed based on tumoral PD-L1 expression, immune phenotypes, pathological response and were cross-compared across the three cohorts. RESULTS PD-L1 positive tumors exhibited increased signature scores for various lymphoid and myeloid cell subsets (p<0.05). TIME phenotypes exhibited disparate frequencies by stage, PD-L1 expression, and mutational burden. Inflamed and PD-L1+/TILs+ NSCLCs displayed overall significantly heightened levels of immune signatures, with the excluded group representing an intermediate state. A cytotoxic T cell signature was associated with favorable survival in neoadjuvant chemotherapy-treated NSCLCs (p<0.05). Pathological response to chemoimmunotherapy was positively associated with higher expression of genes involved in immune activation, chemotaxis, as well as T and NK cells (p<0.05 for all). Among the three cohorts, chemoimmunotherapy-treated NSCLCs exhibited highest scores for various immune cell subsets including T effector and B cells (p<0.05). CONCLUSIONS Our findings highlight immune gene programs that may underlie host tumor immunity and response to neoadjuvant chemotherapy and chemoimmunotherapy in resectable NSCLC.
Collapse
Affiliation(s)
- Pedro Rocha
- The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Jiexin Zhang
- The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | | | - Alberto Cruz-Bermúdez
- Servicio de Oncología Médica, Instituto de Investigación Sanitaria Puerta de Hierro, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain., Madrid, Spain
| | | | | | - Katsuhiro Yoshimura
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Carmen Behrens
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Wei Lu
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ximing Tang
- The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Apar Pataer
- The University of Texas MD Anderson Cancer Center, houston, Texas, United States
| | - Edwin R Parra
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cara Haymaker
- The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Junya Fujimoto
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stephen G Swisher
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John V Heymach
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Don L Gibbons
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - J Jack Lee
- The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Boris Sepesi
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Tina Cascone
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Luisa M Solis
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mariano Provencio
- Medical Oncology Department, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain., Majadahonda, Madrid, Spain
| | - Ignacio I Wistuba
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Humam Kadara
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|