1
|
Kay EJ, Zanivan S. The tumor microenvironment is an ecosystem sustained by metabolic interactions. Cell Rep 2025; 44:115432. [PMID: 40088447 DOI: 10.1016/j.celrep.2025.115432] [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: 07/24/2024] [Revised: 12/09/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) and immune cells make up two major components of the tumor microenvironment (TME), contributing to an ecosystem that can either support or restrain cancer progression. Metabolism is a key regulator of the TME, providing a means for cells to communicate with and influence each other, modulating tumor progression and anti-tumor immunity. Cells of the TME can metabolically interact directly through metabolite secretion and consumption or by influencing other aspects of the TME that, in turn, stimulate metabolic rewiring in target cells. Recent advances in understanding the subtypes and plasticity of cells in the TME both open up new avenues and create challenges for metabolically targeting the TME to hamper tumor growth and improve response to therapy. This perspective explores ways in which the CAF and immune components of the TME could metabolically influence each other, based on current knowledge of their metabolic states, interactions, and subpopulations.
Collapse
Affiliation(s)
- Emily Jane Kay
- Cancer Research UK Scotland Institute, Glasgow G61 1BD, UK.
| | - Sara Zanivan
- Cancer Research UK Scotland Institute, Glasgow G61 1BD, UK; School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
2
|
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
|
3
|
Molina-Castillo G, Monroy-García A, García-Rocha R, Weiss-Steider B, Montesinos-Montesinos JJ, Hernández-Montes J, Don-López CA, Castro-Manrreza ME, Escobar-Sánchez ML, Mora-García MDL. TGF-β Induces the Secretion of Extracellular Vesicles Enriched with CD39 and CD73 from Cervical Cancer Cells. Int J Mol Sci 2025; 26:2413. [PMID: 40141056 PMCID: PMC11942456 DOI: 10.3390/ijms26062413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
The presence of TGF-β in the tumor microenvironment of cervical cancer (CC) is important for tumor progression. In this study, we analyzed the effect of TGF-β on the expression of the ectonucleotidases CD39 and CD73, which are involved in the generation of adenosine (Ado), in CC cells and in extracellular vesicles (EVs) secreted by these cells. Treatment of HeLa and CaSki cells for 72 h with recombinant human TGF-β increased the expression of CD39 and CD73 by 20 and 30% and by 40 and 100%, respectively. The addition of SB505124, an inhibitor of the TGF-β1 receptor, or GW4869, an inhibitor of exosome formation and release, reduced the expression and release of both ectonucleotidases in CC cells. Furthermore, TGF-β promoted the secretion of medium-large EVs (>130 nm) in HeLa cells (HeLa + TGF-β/EVs) and CaSki cells (CaSki + TGF-β/EVs), which increased the expression of CD39 (>20%) and CD73 (>60%), and EVs obtained from cells treated with TGF-β had a greater capacity to generate Ado than did EVs obtained from cells cultured in the absence of this factor (HeLa/EVs and CaSki/EVs). These findings suggest that the production of TGF-β in the CC TME can promote neoplastic progression through the secretion of EVs enriched with CD39 and CD73. Therefore, the inhibition of CD39+ CD73+ EVs could be a strategy for the treatment of CC.
Collapse
Affiliation(s)
- Gabriela Molina-Castillo
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer-UMIEZ, FES-Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico; (G.M.-C.); (R.G.-R.); (B.W.-S.); (J.H.-M.); (C.A.D.-L.)
- Programa de Posgrado en Ciencias Biológicas, UNAM, Ciudad de México 09230, Mexico
| | - Alberto Monroy-García
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer-UMIEZ, FES-Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico; (G.M.-C.); (R.G.-R.); (B.W.-S.); (J.H.-M.); (C.A.D.-L.)
- Laboratorio de Inmunología y Cáncer, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México 06720, Mexico
| | - Rosario García-Rocha
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer-UMIEZ, FES-Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico; (G.M.-C.); (R.G.-R.); (B.W.-S.); (J.H.-M.); (C.A.D.-L.)
| | - Benny Weiss-Steider
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer-UMIEZ, FES-Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico; (G.M.-C.); (R.G.-R.); (B.W.-S.); (J.H.-M.); (C.A.D.-L.)
| | - Juan José Montesinos-Montesinos
- Laboratorio de Células Troncales Mesenquimales, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México 06720, Mexico;
| | - Jorge Hernández-Montes
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer-UMIEZ, FES-Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico; (G.M.-C.); (R.G.-R.); (B.W.-S.); (J.H.-M.); (C.A.D.-L.)
| | - Christian Azucena Don-López
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer-UMIEZ, FES-Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico; (G.M.-C.); (R.G.-R.); (B.W.-S.); (J.H.-M.); (C.A.D.-L.)
| | - Marta Elena Castro-Manrreza
- Laboratorio de Inmunología y Células Madre, Unidad Multidisciplinaria de Investigación Experimental Zaragoza, FES Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico;
| | - María Luisa Escobar-Sánchez
- Laboratorio de Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, Mexico;
| | - María de Lourdes Mora-García
- Laboratorio de Inmunobiología, Unidad de Investigación en Diferenciación Celular y Cáncer-UMIEZ, FES-Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico; (G.M.-C.); (R.G.-R.); (B.W.-S.); (J.H.-M.); (C.A.D.-L.)
| |
Collapse
|
4
|
Liu P, Guo J, Xie Z, Pan Y, Wei B, Peng Y, Hu S, Ding J, Chen X, Su J, Liu H, Zhou W. Co-Delivery of aPD-L1 and CD73 Inhibitor Using Calcium Phosphate Nanoparticles for Enhanced Melanoma Immunotherapy with Reduced Toxicity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410545. [PMID: 39716993 PMCID: PMC11831434 DOI: 10.1002/advs.202410545] [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: 08/30/2024] [Revised: 11/18/2024] [Indexed: 12/25/2024]
Abstract
Melanoma, a malignant skin tumor, presents significant treatment challenges, particularly in unresectable and metastatic cases. While immune checkpoint inhibitors (ICIs) targeting PD-1/PD-L1 have brought new hope, their efficacy is limited by low response rates and significant immune-mediated adverse events (irAEs). Through multi-omics data analysis, it is discovered that the spatial co-localization of CD73 and PD-L1 in melanoma correlates with improved progression-free survival (PFS), suggesting a synergistic potential of their inhibitors. Building on these insights, a novel therapeutic strategy using calcium phosphate (CaP) nanoparticles is developed for the co-delivery of aPD-L1 and APCP, a CD73 inhibitor. These nanoparticles, constructed via a biomineralization method, exhibit high drug-loading capacity and pH-responsive drug release. Compared to free aPD-L1, the CaP-delivered aPD-L1 effectively avoids systemic side effects while significantly enhancing anti-tumor efficacy, surpassing even a 20-fold dose of free aPD-L1. Furthermore, the co-delivery of aPD-L1 and APCP via CaP nanoparticles demonstrates a synergistic anti-tumor effect, with substantial immune activation and prevention of tumor recurrence through immune memory effects. These findings suggest that the co-delivery of aPD-L1 and APCP using CaP nanoparticles is a promising approach for improving melanoma immunotherapy, achieving enhanced efficacy and reduced toxicity.
Collapse
Affiliation(s)
- Peng Liu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Biological NanotechnologyChangshaHunan410008China
| | - Jia Guo
- Department of Dermatology, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaHunan410008China
- Hunan Key Laboratory of Skin Cancer and PsoriasisChangshaHunan410008China
| | - Zuozhong Xie
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya HospitalCentral South UniversityChangshaHunan410011China
| | - Yusheng Pan
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Benliang Wei
- Big Data InstituteCentral South UniversityChangshaHunan410083China
| | - Ying Peng
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Shuo Hu
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Biological NanotechnologyChangshaHunan410008China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Xiang Chen
- Department of Dermatology, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaHunan410008China
- Hunan Key Laboratory of Skin Cancer and PsoriasisChangshaHunan410008China
| | - Juan Su
- Department of Dermatology, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaHunan410008China
- Hunan Key Laboratory of Skin Cancer and PsoriasisChangshaHunan410008China
| | - Hong Liu
- Department of Dermatology, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaHunan410008China
- Hunan Key Laboratory of Skin Cancer and PsoriasisChangshaHunan410008China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
- Key Laboratory of Biological NanotechnologyChangshaHunan410008China
| |
Collapse
|
5
|
Carlomagno S, Setti C, Ortolani F, Sivori S. Pancreatic ductal adenocarcinoma microenvironment: Soluble factors and cancer associated fibroblasts as modulators of NK cell functions. Immunol Lett 2024; 269:106898. [PMID: 39019404 DOI: 10.1016/j.imlet.2024.106898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is the most frequent pancreatic cancer and represents one of the most aggressive human neoplasms. Typically identified at advance stage disease, most PDAC tumors are unresectable and resistant to standard therapies. The immunosuppressive microenvironment in PDAC impedes tumor control but a greater understanding of the complex stromal interactions within the tumor microenvironment (TME) and the development of strategies capable of restoring antitumor effector immune responses could be crucial to fight this aggressive tumor and its spread. Natural Killer (NK) cells play a crucial role in cancer immunosurveillance and represent an attractive target for immunotherapies, both as cell therapy and as a pharmaceutical target. This review describes some crucial components of the PDAC TME (collagens, soluble factors and fibroblasts) that can influence the presence, phenotype and function of NK cells in PDAC patients tumor tissue. This focused overview highlights the therapeutic relevance of dissecting the complex stromal composition to define new strategies for NK cell-based immunotherapies to improve the treatment of PDAC.
Collapse
Affiliation(s)
- Simona Carlomagno
- Department of Medicine (DMED), University of Udine, Piazzale Kolbe 4, Udine 33100, Italy.
| | - Chiara Setti
- Department of Experimental Medicine (DIMES), University of Genoa, Via Leon Battista Alberti 2, Genoa 16132, Italy
| | - Fulvia Ortolani
- Department of Medicine (DMED), University of Udine, Piazzale Kolbe 4, Udine 33100, Italy
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), University of Genoa, Via Leon Battista Alberti 2, Genoa 16132, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
6
|
Figueroa L, Rosas M, Alvarez M, Aguilar E, Mateu V, Bonilla E. Interaction of Purine and its Derivatives with A1, A2-Adenosine Receptors and Vascular Endothelial Growth Factor Receptor-1 (Vegf-R1) as a Therapeutic Alternative to Treat Cancer. Drug Res (Stuttg) 2024; 74:379-393. [PMID: 39173673 DOI: 10.1055/a-2376-5771] [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: 08/24/2024]
Abstract
BACKGROUND There are several studies that indicate that cancer development may be conditioned by the activation of some biological systems that involve the interaction of different biomolecules, such as adenosine and vascular endothelial growth factor. These biomolecules have been targeted of some drugs for treat of cancer; however, there is little information on the interaction of purine derivatives with adenosine and vascular endothelial growth factor receptor (VEGF-R1). OBJECTIVE The aim of this research was to determine the possible interaction of purine (1: ) and their derivatives (2-31: ) with A1, A2-adenosine receptors, and VEGF-R1. METHODS Theoretical interaction of purine and their derivatives with A1, A2-adenosine receptors and VEGF-R1 was carried out using the 5uen, 5mzj and 3hng proteins as theoretical tools. Besides, adenosine, cgs-15943, rolofylline, cvt-124, wrc-0571, luf-5834, cvt-6883, AZD-4635, cabozantinib, pazopanib, regorafenib, and sorafenib drugs were used as controls. RESULTS The results showed differences in the number of aminoacid residues involved in the interaction of purine and their derivatives with 5uen, 5mzj and 3hng proteins compared with the controls. Besides, the inhibition constants (Ki) values for purine and their derivatives 5: , 9: , 10: , 14: , 15: , 16: , and 20: were lower compared with the controls CONCLUSIONS: Theoretical data suggest that purine and their derivatives 5: , 9: , 10: , 14: , 15: , 16: , and 20: could produce changes in cancer cell growth through inhibition of A1, A2-adenosine receptors and VEGFR-1 inhibition. These data indicate that these purine derivatives could be a therapeutic alternative to treat some types of cancer.
Collapse
Affiliation(s)
- Lauro Figueroa
- Laboratory of Pharmaco-Chemistry, Faculty of Chemical Biological Sciences, University Autonomous of Campeche, Campeche, Camp., México
| | - Marcela Rosas
- Faculty of Nutrition, University Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| | - Magdalena Alvarez
- Faculty of Nutrition, University Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| | - Emilio Aguilar
- Facultad de Medicina, Universidad Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| | - Virginia Mateu
- Faculty of Nutrition, University Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| | - Enrique Bonilla
- Faculty of Nutrition, University Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| |
Collapse
|
7
|
Gardani CFF, Diz FM, Dondé LB, Rockenbach L, Laufer S, Morrone FB. The potential role of purinergic signaling in cancer therapy: perspectives on anti-CD73 strategies for prostate cancer. Front Immunol 2024; 15:1455469. [PMID: 39355246 PMCID: PMC11442216 DOI: 10.3389/fimmu.2024.1455469] [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: 06/26/2024] [Accepted: 08/12/2024] [Indexed: 10/03/2024] Open
Abstract
Purines and pyrimidines are signaling molecules in the tumor microenvironment that affect cancer immunity. The purinergic signaling pathways have been shown to play an important role in the development and progression of cancer. CD39 and CD73 are ectonucleotidases responsible for breaking down ATP or ADP into adenosine, which regulates immunosuppression in various types of cancer. These enzymes have been studied as a potential therapeutic target in immunotherapy, and recent research suggests a correlation between ectonucleotidases and clinical outcomes in cancer.Prostate cancer is the most diagnosed cancer in men, after non-melanoma skin tumors, and is the second leading cause of death in men in the world. Despite having long survival periods, patients often receive excessive or insufficient treatment. Within this complex landscape, the adenosine/CD73 pathway plays a crucial role. Therefore, this review aims to highlight new findings on the potential role of purinergic signaling in cancer treatment and emphasizes the importance of anti-CD73 as a pharmacological strategy for prostate cancer therapy.
Collapse
Affiliation(s)
- Carla Fernanda Furtado Gardani
- Escola de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernando Mendonça Diz
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Centro de Pesquisa Pré-Clínica, Instituto do Cerebro do Rio Grande do Sul (InsCer), Pontíficia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luísa Brandalise Dondé
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Liliana Rockenbach
- Escola de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Stefan Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Fernanda Bueno Morrone
- Escola de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Escola de Ciências da Saúde e da Vida, Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
8
|
Xu Y, Lau P, Chen X, Zhao S, He Y, Jiang Z, Chen X, Zhang G, Liu H. Integrated multiomics revealed adenosine signaling predict immunotherapy response and regulate tumor ecosystem of melanoma. Hum Genomics 2024; 18:101. [PMID: 39278925 PMCID: PMC11404024 DOI: 10.1186/s40246-024-00651-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/28/2024] [Indexed: 09/18/2024] Open
Abstract
Extracellular adenosine is extensively involved in regulating the tumor microenvironment. Given the disappointing results of adenosine-targeted therapy trials, personalized treatment might be necessary, tailored to the microenvironment status of individual patients. Here, we introduce the adenosine signaling score (ADO-score) model using non-negative matrix fraction identified patient subtypes using publicly available melanoma dataset, which aimed to profile adenosine signaling-related genes and construct a model to predict prognosis. We analyzed 580 malignant melanoma samples and demonstrated its robust value for prognosis. Further investigation in immune checkpoint inhibitor dataset suggests its potential as a stratified factor of immune checkpoint inhibitor efficacy. We validated the power of the ADO-score at the protein level immunofluorescence in a melanoma cohort from Xiangya Hospital. More importantly, single-cell and spatial transcriptomic data highlighted the cell-specific expression patterns of adenosine signaling-related genes and the existence of adenosine signaling-mediated crosstalk between tumor cells and immune cells in melanoma. Our study reveals a robust connection between adenosine signaling and clinical benefits in melanoma patients and proposes a universally applicable adenosine signaling model, the ADO-score, in gene expression profiles and histological sections. This model enables us to more precisely and conveniently select patients who are likely to benefit from immunotherapy.
Collapse
Affiliation(s)
- Yantao Xu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Poyee Lau
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Shuang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Yi He
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Zixi Jiang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China.
- Xiangya School of Medicine, Central South University, Changsha, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China.
- Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China.
| | - Guanxiong Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China.
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China.
- Xiangya School of Medicine, Central South University, Changsha, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China.
- Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
9
|
Yu Y, Zhang C, Dong B, Zhang Z, Li X, Huang S, Tang D, Jing X, Yu S, Zheng T, Wu D, Tai S. Neutrophil extracellular traps promote immune escape in hepatocellular carcinoma by up-regulating CD73 through Notch2. Cancer Lett 2024; 598:217098. [PMID: 38969159 DOI: 10.1016/j.canlet.2024.217098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
Immune escape is the main reason that immunotherapy is ineffective in hepatocellular carcinoma (HCC). Here, this study illustrates a pathway mediated by neutrophil extracellular traps (NETs) that can promote immune escape of HCC. Mechanistically, we demonstrated that NETs up-regulated CD73 expression through activating Notch2 mediated nuclear factor kappa B (NF-κB) pathway, promoting regulatory T cells (Tregs) infiltration to mediate immune escape of HCC. In addition, we found the similar results in mouse HCC models by hydrodynamic plasmid transfection. The treatment of deoxyribonuclease I (DNase I) could inhibit the action of NETs and improve the therapeutic effect of anti-programmed cell death protein 1 (PD-1). In summary, our results revealed that targeting of NETs was a promising treatment to improve the therapeutic effect of anti-PD-1.
Collapse
Affiliation(s)
- Yang Yu
- Department of Hepatic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Congyi Zhang
- Key Laboratory of Precision Nutrition and Health of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Bowen Dong
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Zhihua Zhang
- Department of Hepatic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Xiaoqing Li
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Shizhuan Huang
- Department of Hepatic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Daowei Tang
- Department of Hepatic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Xiaowei Jing
- Department of Hepatic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Shan Yu
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China.
| | - Tongsen Zheng
- Heilongjiang Province Key Laboratory of Molecular Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China.
| | - Dehai Wu
- Department of Hepatic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China.
| | - Sheng Tai
- Department of Hepatic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China.
| |
Collapse
|
10
|
Zhang H, Yang L, Han M, Han Y, Jiang Z, Zheng Q, Dong J, Wang T, Li Z. Boost Infiltration and Activity of T Cells via Inhibiting Ecto-5'-nucleotidase (CD73) Immune Checkpoint to Enhance Glioblastoma Immunotherapy. ACS NANO 2024; 18:23001-23013. [PMID: 39150454 DOI: 10.1021/acsnano.4c04553] [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: 08/17/2024]
Abstract
The currently available immune checkpoint therapy shows a disappointing therapeutic efficacy for glioblastoma multiforme (GBM), and it is of great importance to discover better immune checkpoints and develop innovative targeting strategies. The discovered metabolic immune checkpoint ecto-5-nucleotidase (CD73) in a tumor contributes to its immune evasion due to the dysregulation of extracellular adenosine (ADO), which significantly inhibits the function of antitumor T cells and increases the activity of immunosuppressive cells. Herein, we drastically inhibit the expression of CD73 to reduce the production of ADO by using versatile Au@Cu2-xSe nanoparticles (ACS NPs). ACS NPs can decrease the expression of CD73 by alleviating the tumor hypoxia through their Fenton-like reaction to weaken the ADO-driven immunosuppression for enhancing antitumor T cell infiltration and activity of GBM. The copper ions (Cu2+) released from ACS NPs can chelate with disulfide, leading to the formation of cytotoxic bis(N,N-diethyldithiocarbamate)-copper complex (CuET), which can be combined with radiotherapy to recruit more antitumor T cells to infiltrate into the tumor site. Based on the inhibition of CD73 to promote the infiltration and activity of antitumor T cells, a cascade of enhancing GBM immunotherapy effects can be achieved. The significant increase in CD8+ T and CD4+ T cells within the tumor and the memory T cells in the spleen effectively reduces tumor size by 92%, which demonstrates the excellent efficacy of immunotherapy achieved by a combination of metabolic immune checkpoint CD73 inhibition with chemoradiotherapy. This work demonstrates that modulation of CD73-mediated tumor immunosuppression is an important strategy of improving the outcome of GBM immunotherapy.
Collapse
Affiliation(s)
- Hao Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Li Yang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Mengxiao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Zhilin Jiang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Qing Zheng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Jun Dong
- Department of Neurosurgery, Second Affiliated Hospital of Soochow University, Suzhou 215025, China
| | - Tingting Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou 215123, China
| |
Collapse
|
11
|
Zhan J, Huang L, Niu L, Lu W, Sun C, Liu S, Ding Z, Li E. Regulation of CD73 on NAD metabolism: Unravelling the interplay between tumour immunity and tumour metabolism. Cell Commun Signal 2024; 22:387. [PMID: 39090604 PMCID: PMC11292923 DOI: 10.1186/s12964-024-01755-y] [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: 06/05/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024] Open
Abstract
CD73, a cell surface-bound nucleotidase, serves as a crucial metabolic and immune checkpoint. Several studies have shown that CD73 is widely expressed on immune cells and plays a critical role in immune escape, cell adhesion and migration as a costimulatory molecule for T cells and a factor in adenosine production. However, recent studies have revealed that the protumour effects of CD73 are not limited to merely inhibiting the antitumour immune response. Nicotinamide adenine dinucleotide (NAD+) is a vital bioactive molecule in organisms that plays essential regulatory roles in diverse biological processes within tumours. Accumulating evidence has demonstrated that CD73 is involved in the transport and metabolism of NAD, thereby regulating tumour biological processes to promote growth and proliferation. This review provides a holistic view of CD73-regulated NAD + metabolism as a complex network and further highlights the emerging roles of CD73 as a novel target for cancer therapies.
Collapse
Affiliation(s)
- Jianhao Zhan
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- HuanKui Academy, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Le Huang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- HuanKui Academy, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Liyan Niu
- HuanKui Academy, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Wenhui Lu
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Chengpeng Sun
- HuanKui Academy, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Shanshan Liu
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi province, China
| | - Zijun Ding
- School of Ophthalmology and Optometry, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Enliang Li
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China.
| |
Collapse
|
12
|
Delorme AS, Laguide A, Tamagne M, Pinheiro MK, Cagnet L, Neyrinck-Leglantier D, Khelfa M, Cleophax S, Pirenne F, Vingert B. Immune interactions and regulation with CD39 + extracellular vesicles from platelet concentrates. Front Immunol 2024; 15:1397967. [PMID: 38947317 PMCID: PMC11211594 DOI: 10.3389/fimmu.2024.1397967] [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: 03/08/2024] [Accepted: 05/27/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction CD39 plays an important role in the immunoregulation and inhibition of effector cells. It is expressed on immune cells, including Tregs, and on extracellular vesicles (EVs) budding from the plasma membrane. Platelet transfusion may induce alloimmunization against HLA-I antigens, leading to refractoriness to platelet transfusion with severe consequences for patients. Tregs may play a key role in determining whether alloimmunization occurs in patients with hematologic disorders. We hypothesized that CD39+ EVs might play an immunoregulatory role, particularly in the context of platelet transfusions in patients with hematologic disorders. Such alloimmunization leads to the production of alloantibodies and is sensitive to the regulatory action of CD39. Methods We characterized CD39+ EVs in platelet concentrates by flow cytometry. The absolute numbers and cellular origins of CD39+ EVs were evaluated. We also performed functional tests to evaluate interactions with immune cells and their functions. Results We found that CD39+ EVs from platelet concentrates had an inhibitory phenotype that could be transferred to the immune cells with which they interacted: CD4+ and CD8+ T lymphocytes (TLs), dendritic cells, monocytes, and B lymphocytes (BLs). Moreover, the concentration of CD39+ EVs in platelet concentrates varied and was very high in 10% of concentrates. The number of these EVs present was determinant for EV-cell interactions. Finally, functional interactions were observed with BLs, CD4+ TLs and CD39+ EVs for immunoglobulin production and lymphoproliferation, with potential implications for the immunological management of patients.
Collapse
Affiliation(s)
- Adèle Silane Delorme
- Univ Paris Est Creteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Alexandra Laguide
- Univ Paris Est Creteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Marie Tamagne
- Univ Paris Est Creteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Marion Klea Pinheiro
- Univ Paris Est Creteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Léonie Cagnet
- Univ Paris Est Creteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Deborah Neyrinck-Leglantier
- Univ Paris Est Creteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Mehdi Khelfa
- Univ Paris Est Creteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | | | - France Pirenne
- Univ Paris Est Creteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Benoît Vingert
- Univ Paris Est Creteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| |
Collapse
|
13
|
Molina OE, LaRue H, Simonyan D, Hovington H, Vittrant B, Têtu B, Fradet V, Lacombe L, Bergeron A, Fradet Y. Regulatory and memory T lymphocytes infiltrating prostate tumors predict long term clinical outcomes. Front Immunol 2024; 15:1372837. [PMID: 38887294 PMCID: PMC11180786 DOI: 10.3389/fimmu.2024.1372837] [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: 01/18/2024] [Accepted: 05/13/2024] [Indexed: 06/20/2024] Open
Abstract
Introduction The localization, density but mostly the phenotype of tumor infiltrating lymphocytes (TIL) provide important information on the initial interaction between the host immune system and the tumor. Our objective was to assess the prognostic significance of T (CD3+), T regulatory (Treg) (FoxP3+) and T memory (Tmem) (CD45RO+) infiltrating lymphocytes and of genes associated with TIL in prostate cancer (PCa). Methods Immunohistochemistry (IHC) was used to assess the infiltration of CD3+, FoxP3+ and CD45RO+ cells in the tumor area, tumor margin and adjacent normal-like epithelium of a series of 98 PCa samples with long clinical follow-up. Expression of a panel of 31 TIL-associated genes was analyzed by Taqman Low-Density Array (TLDA) technology in another series of 50 tumors with long clinical follow-up. Kaplan-Meier and Cox proportional hazards regression analyses were performed to determine association of these markers with biochemical recurrence (BCR), need for definitive androgen deprivation therapy (ADT) or lethal PCa. Results TIL subtypes were present at different densities in the tumor, tumor margin and adjacent normal-like epithelium, but their density and phenotype in the tumor area were the most predictive of clinical outcomes. In multivariate analyses, a high density of Treg (high FoxP3+/CD3+ cell ratio) predicted a higher risk for need of definitive ADT (HR=7.69, p=0.001) and lethal PCa (HR=4.37, p=0.04). Conversely, a high density of Tmem (high CD45RO+/CD3+ cell ratio) predicted a reduced risk of lethal PCa (HR=0.06, p=0.04). TLDA analyses showed that a high expression of FoxP3 was associated with a higher risk of lethal PCa (HR=5.26, p=0.02). Expression of CTLA-4, PD-1, TIM-3 and LAG-3 were correlated with that of FoxP3. Amongst these, only a high expression of TIM-3 was associated with a significant higher risk for definitive ADT in univariate Cox regression analysis (HR=3.11, p=0.01). Conclusion These results show that the proportion of Treg and Tmem found within the tumor area is a strong and independent predictor of late systemic progression of PCa. Our results also suggest that inhibition of TIM-3 might be a potential approach to counter the immunosuppressive functions of Treg in order to improve the anti-tumor immune response against PCa.
Collapse
Affiliation(s)
- Oscar Eduardo Molina
- Axe oncologie, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec, QC, Canada
| | - Hélène LaRue
- Axe oncologie, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec, QC, Canada
| | - David Simonyan
- Plateforme de recherche clinique et évaluative, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Hélène Hovington
- Axe oncologie, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec, QC, Canada
| | - Benjamin Vittrant
- Axe oncologie, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec, QC, Canada
| | - Bernard Têtu
- Axe oncologie, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec, QC, Canada
- Département de pathologie, CHU de Québec-Université Laval, Québec, QC, Canada
| | - Vincent Fradet
- Axe oncologie, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec, QC, Canada
- Département de chirurgie, Université Laval, Québec, QC, Canada
| | - Louis Lacombe
- Axe oncologie, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec, QC, Canada
- Département de chirurgie, Université Laval, Québec, QC, Canada
| | - Alain Bergeron
- Axe oncologie, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec, QC, Canada
- Département de chirurgie, Université Laval, Québec, QC, Canada
| | - Yves Fradet
- Axe oncologie, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Québec, QC, Canada
- Département de chirurgie, Université Laval, Québec, QC, Canada
| |
Collapse
|
14
|
Alonso-Gordoa T, Goodman M, Vulsteke C, Roubaud G, Zhang J, Parikh M, Piulats JM, Azaro A, James GD, Cavazzina R, Gangl ET, Thompson J, Pouliot G, Kumar R, Sweeney C. A phase II study (AARDVARC) of AZD4635 in combination with durvalumab and cabazitaxel in patients with progressive, metastatic, castration-resistant prostate cancer. ESMO Open 2024; 9:103446. [PMID: 38838502 PMCID: PMC11190476 DOI: 10.1016/j.esmoop.2024.103446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND This phase II nonrandomized study evaluated the efficacy and safety of AZD4635 in combination with durvalumab (Arm A) or durvalumab plus cabazitaxel (Arm B) in patients with metastatic castration-resistant prostate cancer (mCRPC) previously treated with docetaxel and ≥1 novel hormonal agent. PATIENTS AND METHODS The primary endpoint was radiographic progression-free survival (rPFS) per RECIST v1.1 (soft tissue) or the Prostate Cancer Clinical Trials Working Group 3 (bone). Secondary endpoints included safety, tolerability, overall survival, confirmed prostate-specific antigen (PSA50) response, pharmacokinetics, and objective response rate. Enrollment in Arm A was stopped following a sponsor decision unrelated to safety. The study was stopped based on the planned futility analysis due to low PSA50 response in Arm B. RESULTS In the final analysis (1 November 2021), 30 patients were treated (Arm A, n = 2; Arm B, n = 28). The median rPFS in Arm B was 5.8 months (95% confidence interval 4.2-not calculable). Median rPFS was 5.8 months versus 4.2 months for patients with high versus low blood-based adenosine signature. The most common treatment-related adverse events in Arm B were nausea (50.0%), diarrhea (46.4%), anemia and neutropenia (both 35.7%), asthenia (32.1%), and vomiting (28.6%). Overall, AZD4635 in combination with durvalumab or AZD4635 in combination with cabazitaxel and durvalumab showed limited efficacy in patients with mCRPC. CONCLUSIONS Although the safety profile of both combinations was consistent with known safety data of the individual agents, the results of this trial do not support further development of the combinations.
Collapse
Affiliation(s)
| | - M Goodman
- Atrium Health Wake Forest Baptist, Winston-Salem, USA
| | - C Vulsteke
- Integrated Cancer Center, Maria Middelares General Hospital, Ghent; Center for Oncological Research, University of Antwerp, Antwerp, Belgium
| | | | - J Zhang
- H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - M Parikh
- Hematology/Oncology Clinic, UC Davis Comprehensive Cancer Center, Sacramento, USA
| | - J M Piulats
- Catalan Institute of Oncology, Barcelona, Spain
| | - A Azaro
- Oncology R&D, AstraZeneca, Cambridge
| | - G D James
- Medical Statistics Consultancy Ltd, London, UK
| | | | | | | | | | - R Kumar
- Oncology R&D, AstraZeneca, Gaithersburg
| | - C Sweeney
- Dana-Farber Cancer Institute, Boston, USA
| |
Collapse
|
15
|
Coban B, Wang Z, Liao CY, Beslmüller K, Timmermans MA, Martens JW, Hundscheid JH, Slutter B, Zweemer AJ, Neubert E, Danen EH. GRHL2 suppression of NT5E/CD73 in breast cancer cells modulates CD73-mediated adenosine production and T cell recruitment. iScience 2024; 27:109738. [PMID: 38706844 PMCID: PMC11068632 DOI: 10.1016/j.isci.2024.109738] [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: 10/10/2023] [Revised: 03/03/2024] [Accepted: 04/10/2024] [Indexed: 05/07/2024] Open
Abstract
Tumor tissues often contain high extracellular adenosine, promoting an immunosuppressed environment linked to mesenchymal transition and immune evasion. Here, we show that loss of the epithelial transcription factor, GRHL2, triggers NT5E/CD73 ecto-enzyme expression, augmenting the conversion of AMP to adenosine. GRHL2 binds an intronic NT5E sequence and is negatively correlated with NT5E/CD73 in breast cancer cell lines and patients. Remarkably, the increased adenosine levels triggered by GRHL2 depletion in MCF-7 breast cancer cells do not suppress but mildly increase CD8 T cell recruitment, a response mimicked by a stable adenosine analog but prevented by CD73 inhibition. Indeed, NT5E expression shows a positive rather than negative association with CD8 T cell infiltration in breast cancer patients. These findings reveal a GRHL2-regulated immune modulation mechanism in breast cancers and show that extracellular adenosine, besides its established role as a suppressor of T cell-mediated cytotoxicity, is associated with enhanced T cell recruitment.
Collapse
Affiliation(s)
- Bircan Coban
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | - Zi Wang
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
- Department of clinical laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Chen-yi Liao
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | - Klara Beslmüller
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | - Mieke A.M. Timmermans
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - John W.M. Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Bram Slutter
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | - Annelien J.M. Zweemer
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | - Elsa Neubert
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | - Erik H.J. Danen
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| |
Collapse
|
16
|
Falchook GS, Reeves J, Gandhi S, Spigel DR, Arrowsmith E, George DJ, Karlix J, Pouliot G, Hattersley MM, Gangl ET, James GD, Thompson J, Russell DL, Patel B, Kumar R, Lim E. A phase 2 study of AZD4635 in combination with durvalumab or oleclumab in patients with metastatic castration-resistant prostate cancer. Cancer Immunol Immunother 2024; 73:72. [PMID: 38430405 PMCID: PMC10908633 DOI: 10.1007/s00262-024-03640-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/22/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Inhibition of the adenosine 2A receptor (A2AR) diminishes the immunosuppressive effects of adenosine and may complement immune-targeting drugs. This phase 2 study evaluated the A2AR antagonist AZD4635 in combination with durvalumab or oleclumab in patients with metastatic castration-resistant prostate cancer. METHODS Patients with histologically/cytologically confirmed disease progressing within 6 months on ≥ 2 therapy lines were randomly assigned to either Module 1 (AZD4635 + durvalumab) or Module 2 (AZD4635 + oleclumab). Primary endpoints were objective response rate per RECIST v1.1 and prostate-specific antigen (PSA) response rate. Secondary endpoints included radiological progression-free survival (rPFS), overall survival, safety, and pharmacokinetics. RESULTS Fifty-nine patients were treated (Module 1, n = 29; Module 2, n = 30). Median number of prior therapies was 4. One confirmed complete response by RECIST (Module 1) and 2 confirmed PSA responses (1 per module) were observed. The most frequent adverse events (AEs) possibly related to AZD4635 were nausea (37.9%), fatigue (20.7%), and decreased appetite (17.2%) in Module 1; nausea (50%), fatigue (30%), and vomiting (23.3%) in Module 2. No dose-limiting toxicities or treatment-related serious AEs were observed. In Module 1, AZD4635 geometric mean trough concentration was 124.9 ng/mL (geometric CV% 69.84; n = 22); exposures were similar in Module 2. In Modules 1 and 2, median (95% CI) rPFS was 2.3 (1.6 -3.8) and 1.5 (1.3- 4.0) months, respectively. Median PFS was 1.7 versus 2.3 months for patients with high versus low blood-based adenosine signature. CONCLUSION In this heavily pretreated population, AZD4635 with durvalumab or oleclumab demonstrated minimal antitumor activity with a manageable safety profile. CLINICAL TRIAL gov identifier: NCT04089553.
Collapse
Affiliation(s)
- Gerald S Falchook
- Drug Development Unit, Sarah Cannon Research Institute at HealthONE, Denver, CO, USA.
| | - James Reeves
- Florida Cancer Specialists South, Sarah Cannon Research Institute, Fort Meyers, FL, USA
| | - Sunil Gandhi
- Florida Cancer Specialists South, Sarah Cannon Research Institute, St. Petersberg, FL, USA
| | - David R Spigel
- Tennessee Oncology, Sarah Cannon Research Institute, Nashville, TN, USA
| | - Edward Arrowsmith
- Tennessee Oncology, Sarah Cannon Research Institute, Nashville, TN, USA
| | | | - Janet Karlix
- Sarah Cannon Research Institute, Gainesville, FL, USA
| | | | | | | | | | | | | | | | - Rakesh Kumar
- Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Emerson Lim
- Medical Oncology & Hematology-LHCP, Corewell Health Medical Group, Grand Rapids, MI, USA
| |
Collapse
|
17
|
Chuang YM, Tzeng SF, Ho PC, Tsai CH. Immunosurveillance encounters cancer metabolism. EMBO Rep 2024; 25:471-488. [PMID: 38216787 PMCID: PMC10897436 DOI: 10.1038/s44319-023-00038-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 12/02/2023] [Accepted: 12/12/2023] [Indexed: 01/14/2024] Open
Abstract
Tumor cells reprogram nutrient acquisition and metabolic pathways to meet their energetic, biosynthetic, and redox demands. Similarly, metabolic processes in immune cells support host immunity against cancer and determine differentiation and fate of leukocytes. Thus, metabolic deregulation and imbalance in immune cells within the tumor microenvironment have been reported to drive immune evasion and to compromise therapeutic outcomes. Interestingly, emerging evidence indicates that anti-tumor immunity could modulate tumor heterogeneity, aggressiveness, and metabolic reprogramming, suggesting that immunosurveillance can instruct cancer progression in multiple dimensions. This review summarizes our current understanding of how metabolic crosstalk within tumors affects immunogenicity of tumor cells and promotes cancer progression. Furthermore, we explain how defects in the metabolic cascade can contribute to developing dysfunctional immune responses against cancers and discuss the contribution of immunosurveillance to these defects as a feedback mechanism. Finally, we highlight ongoing clinical trials and new therapeutic strategies targeting cellular metabolism in cancer.
Collapse
Affiliation(s)
- Yu-Ming Chuang
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Sheue-Fen Tzeng
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
| | - Chin-Hsien Tsai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.
- Department and Graduate Institute of Biochemistry, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
18
|
Rastin F, Javid H, Oryani MA, Rezagholinejad N, Afshari AR, Karimi-Shahri M. Immunotherapy for colorectal cancer: Rational strategies and novel therapeutic progress. Int Immunopharmacol 2024; 126:111055. [PMID: 37992445 DOI: 10.1016/j.intimp.2023.111055] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 11/24/2023]
Abstract
There are increasing incidences and mortality rates for colorectal cancer in the world. It is common for chemotherapy and radiation given to patients with colorectal cancer to cause toxicities that limit their effectiveness and cause cancer cells to become resistant to these treatments. Additional targeted treatments are needed to improve patient's quality of life and outcomes. Immunotherapy has rapidly emerged as an incredibly exciting and promising avenue for cancer treatment in recent years. This innovative approach provides novel options for tackling solid tumors, effectively establishing itself as a new cornerstone in cancer treatment. Specifically, in the realm of colorectal cancer (CRC), there is great promise in developing new drugs that target immune checkpoints, offering a hopeful and potentially transformative solution. While immunotherapy of CRC has made significant advances, there are still obstacles and limitations. CRC patients have a poor response to treatment because of the immune-suppressing function of their tumor microenvironment (TME). In addition to blocking inhibitory immune checkpoints, checkpoint-blocking antibodies may also boost immune responses against tumors. The review summarizes recent advances in immune checkpoint inhibitors (ICIs) for CRC, including CTLA-4, PD-1, PD-L1, LAG-3, and TIM-3.
Collapse
Affiliation(s)
- Farangis Rastin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mahsa Akbari Oryani
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amir-R Afshari
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
19
|
Garapati K, Ding H, Charlesworth MC, Kim Y, Zenka R, Saraswat M, Mun DG, Chavan S, Shingade A, Lucien F, Zhong J, Kandasamy RK, Pandey A. sBioSITe enables sensitive identification of the cell surface proteome through direct enrichment of biotinylated peptides. Clin Proteomics 2023; 20:56. [PMID: 38053024 PMCID: PMC10696767 DOI: 10.1186/s12014-023-09445-6] [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: 08/21/2023] [Accepted: 11/17/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND Cell surface proteins perform critical functions related to immune response, signal transduction, cell-cell interactions, and cell migration. Expression of specific cell surface proteins can determine cell-type identity, and can be altered in diseases including infections, cancer and genetic disorders. Identification of the cell surface proteome remains a challenge despite several enrichment methods exploiting their biochemical and biophysical properties. METHODS Here, we report a novel method for enrichment of proteins localized to cell surface. We developed this new approach designated surface Biotinylation Site Identification Technology (sBioSITe) by adapting our previously published method for direct identification of biotinylated peptides. In this strategy, the primary amine groups of lysines on proteins on the surface of live cells are first labeled with biotin, and subsequently, biotinylated peptides are enriched by anti-biotin antibodies and analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS By direct detection of biotinylated lysines from PC-3, a prostate cancer cell line, using sBioSITe, we identified 5851 peptides biotinylated on the cell surface that were derived from 1409 proteins. Of these proteins, 533 were previously shown or predicted to be localized to the cell surface or secreted extracellularly. Several of the identified cell surface markers have known associations with prostate cancer and metastasis including CD59, 4F2 cell-surface antigen heavy chain (SLC3A2) and adhesion G protein-coupled receptor E5 (CD97). Importantly, we identified several biotinylated peptides derived from plectin and nucleolin, both of which are not annotated in surface proteome databases but have been shown to have aberrant surface localization in certain cancers highlighting the utility of this method. CONCLUSIONS Detection of biotinylation sites on cell surface proteins using sBioSITe provides a reliable method for identifying cell surface proteins. This strategy complements existing methods for detection of cell surface expressed proteins especially in discovery-based proteomics approaches.
Collapse
Affiliation(s)
- Kishore Garapati
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
- Institute of Bioinformatics, International Technology Park, Bangalore, Karnataka, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Husheng Ding
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Yohan Kim
- Department of Urology, Mayo Clinic, Rochester, MN, USA
| | - Roman Zenka
- Proteomics Core, Mayo Clinic, Rochester, MN, USA
| | - Mayank Saraswat
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Dong-Gi Mun
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Sandip Chavan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ashish Shingade
- Institute of Bioinformatics, International Technology Park, Bangalore, Karnataka, India
| | - Fabrice Lucien
- Department of Urology, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Jun Zhong
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Richard K Kandasamy
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
20
|
Gardani CFF, Pedrazza EL, Paz VS, Zanirati GG, da Costa JC, Andrejew R, Ulrich H, Scholl JN, Figueiró F, Rockenbach L, Morrone FB. Exploring CD39 and CD73 Expression as Potential Biomarkers in Prostate Cancer. Pharmaceuticals (Basel) 2023; 16:1619. [PMID: 38004484 PMCID: PMC10675019 DOI: 10.3390/ph16111619] [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: 09/01/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Prostate cancer (PC) is the most diagnosed tumor in males and ranks as the second leading cause of male mortality in the western world. The CD39 and CD73 enzymes play a crucial role in cancer regulation by degrading nucleotides and forming nucleosides. This study aimed to investigate the expression of the CD39 and CD73 enzymes as potential therapeutic targets for PC. The initial part of this study retrospectively analyzed tissue samples from 23 PC patients. Using the TissueFAXSTM cytometry platform, we found significantly higher levels of CD39-labeling its intensity compared to CD73. Additionally, we observed a correlation between the Gleason score and the intensity of CD39 expression. In the prospective arm, blood samples were collected from 25 patients at the time of diagnosis and after six months of treatment to determine the expression of CD39 and CD73 in the serum extracellular vesicles (EVs) and to analyze nucleotide hydrolysis. Notably, the expression of CD39 in the EVs was significantly increased compared to the CD73 and/or combined CD39/CD73 expression levels at initial collection. Furthermore, our results demonstrated positive correlations between ADP hydrolysis and the transurethral resection and Gleason score. Understanding the role of ectonucleotidases is crucial for identifying new biomarkers in PC.
Collapse
Affiliation(s)
- Carla Fernanda Furtado Gardani
- Escola de Medicina, Programa de Pós-Graduaҫão em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre 90619-900, RS, Brazil; (C.F.F.G.); (L.R.)
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Partenon, Porto Alegre 90619-900, RS, Brazil; (E.L.P.); (V.S.P.)
| | - Eduardo Luiz Pedrazza
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Partenon, Porto Alegre 90619-900, RS, Brazil; (E.L.P.); (V.S.P.)
| | - Victória Santos Paz
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Partenon, Porto Alegre 90619-900, RS, Brazil; (E.L.P.); (V.S.P.)
| | - Gabriele Goulart Zanirati
- Instituto do Cérebro da PUCRS, InsCer, Avenida Ipiranga, 6690, Jardim Botânico, Porto Alegre 906010-000, RS, Brazil; (G.G.Z.); (J.C.d.C.)
| | - Jaderson Costa da Costa
- Instituto do Cérebro da PUCRS, InsCer, Avenida Ipiranga, 6690, Jardim Botânico, Porto Alegre 906010-000, RS, Brazil; (G.G.Z.); (J.C.d.C.)
| | - Roberta Andrejew
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Prof. Lineu Prestes, 748, Butantã, São Paulo 05508-000, SP, Brazil; (R.A.); (H.U.)
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Prof. Lineu Prestes, 748, Butantã, São Paulo 05508-000, SP, Brazil; (R.A.); (H.U.)
| | - Juliete Nathali Scholl
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre 90035-003, RS, Brazil; (J.N.S.); (F.F.)
| | - Fabrício Figueiró
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre 90035-003, RS, Brazil; (J.N.S.); (F.F.)
| | - Liliana Rockenbach
- Escola de Medicina, Programa de Pós-Graduaҫão em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre 90619-900, RS, Brazil; (C.F.F.G.); (L.R.)
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Partenon, Porto Alegre 90619-900, RS, Brazil; (E.L.P.); (V.S.P.)
| | - Fernanda Bueno Morrone
- Escola de Medicina, Programa de Pós-Graduaҫão em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre 90619-900, RS, Brazil; (C.F.F.G.); (L.R.)
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Partenon, Porto Alegre 90619-900, RS, Brazil; (E.L.P.); (V.S.P.)
- Instituto do Cérebro da PUCRS, InsCer, Avenida Ipiranga, 6690, Jardim Botânico, Porto Alegre 906010-000, RS, Brazil; (G.G.Z.); (J.C.d.C.)
- Escola de Ciências da Saúde e da Vida, Programa de Pós-Graduaҫão em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre 90619-900, RS, Brazil
| |
Collapse
|
21
|
Mougola Bissiengou P, Montcho Comlan JG, Atsame Ebang G, Sylla Niang M, Djoba Siawaya JF. Prostate malignant tumor and benign prostatic hyperplasia microenvironments in black African men: Limited infiltration of CD8+ T lymphocytes, NK-cells, and high frequency of CD73+ stromal cells. Cancer Rep (Hoboken) 2023; 6 Suppl 1:e1817. [PMID: 37092584 PMCID: PMC10440842 DOI: 10.1002/cnr2.1817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Anti-cancerous immunology has yet to be investigated in the African black population, despite being the dawn of precision medicine. AIM Here we investigated the tumor microenvironment of prostate cancer and benign prostatic hyperplasia (BPH) in black Africans. METHODS Through immunohistochemistry analysis of prostate cancer and BPH patients' biopsies, we investigated the expression and distribution of CD73, CCD8 T-lymphocytes, and natural killer cells. In addition, we looked at tumor-infiltrating features CD8 T-lymphocytes and natural killer cells. RESULTS We show for the first time in black Africans a high expression of CD73 in epithelial-stromal cells and virtually no infiltration of CD8 T lymphocytes and natural killer cells in the tumoral area. In addition, CD73 was seven (7) times more likely to be expressed in prostate cancer stromal tissues than in benign prostatic hyperplasia tissues (odds ratio = 7.2; χ2 = 21; p < .0001). In addition, PSA concentration was significantly higher in prostate cancer patients than in BPH patients (p < .001). Also, the PSA-based ROC. analysis showed an area under the curve of 0.87 (p < .0001). CONCLUSION CD73 expression is more likely expressed in prostate cancer stromal tissues than in benign prostatic hyperplasia tissues. The features of prostate cancer in Black Africans suggest CD73 expression as a possible target for immunotherapy in this population.
Collapse
Affiliation(s)
- Pélagie Mougola Bissiengou
- Service d'Immunologie, Département des Sciences Fondamentales, Faculté de MédecineUniversité des Sciences de la SantéLibrevilleGabon
- Service d'Immunologie, Département des Sciences Biologiques et Pharmaceutiques Appliquées, Faculté de Médecine, de Pharmacie et d'OdontostomatologieUniversité Cheikh Anta DiopDakarSenegal
| | - Jérôme Gaston Montcho Comlan
- Service d'Immunologie, Département des Sciences Biologiques et Pharmaceutiques Appliquées, Faculté de Médecine, de Pharmacie et d'OdontostomatologieUniversité Cheikh Anta DiopDakarSenegal
| | - Gabrielle Atsame Ebang
- Unité d'anatomie‐Cytologie‐PathologieCentre Hospitalier Universitaire de LibrevilleLibrevilleGabon
| | - Maguette Sylla Niang
- Service d'Immunologie, Département des Sciences Biologiques et Pharmaceutiques Appliquées, Faculté de Médecine, de Pharmacie et d'OdontostomatologieUniversité Cheikh Anta DiopDakarSenegal
| | - Joel Fleury Djoba Siawaya
- Service LaboratoireCentre Hospitalier Universitaire Mère‐Enfant Fondation Jeanne EBORILibrevilleGabon
| |
Collapse
|
22
|
Jin Z, Zhou Q, Cheng JN, Jia Q, Zhu B. Heterogeneity of the tumor immune microenvironment and clinical interventions. Front Med 2023; 17:617-648. [PMID: 37728825 DOI: 10.1007/s11684-023-1015-9] [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: 02/15/2023] [Accepted: 06/24/2023] [Indexed: 09/21/2023]
Abstract
The tumor immune microenvironment (TIME) is broadly composed of various immune cells, and its heterogeneity is characterized by both immune cells and stromal cells. During the course of tumor formation and progression and anti-tumor treatment, the composition of the TIME becomes heterogeneous. Such immunological heterogeneity is not only present between populations but also exists on temporal and spatial scales. Owing to the existence of TIME, clinical outcomes can differ when a similar treatment strategy is provided to patients. Therefore, a comprehensive assessment of TIME heterogeneity is essential for developing precise and effective therapies. Facilitated by advanced technologies, it is possible to understand the complexity and diversity of the TIME and its influence on therapy responses. In this review, we discuss the potential reasons for TIME heterogeneity and the current approaches used to explore it. We also summarize clinical intervention strategies based on associated mechanisms or targets to control immunological heterogeneity.
Collapse
Affiliation(s)
- Zheng Jin
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China
- Research Institute, GloriousMed Clinical Laboratory (Shanghai) Co. Ltd., Shanghai, 201318, China
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Qin Zhou
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Jia-Nan Cheng
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China.
| | - Qingzhu Jia
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China.
| | - Bo Zhu
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
- Key Laboratory of Tumor Immunotherapy, Chongqing, 400037, China.
| |
Collapse
|
23
|
Saber R, Henault D, Messaoudi N, Rebolledo R, Montagnon E, Soucy G, Stagg J, Tang A, Turcotte S, Kadoury S. Radiomics using computed tomography to predict CD73 expression and prognosis of colorectal cancer liver metastases. J Transl Med 2023; 21:507. [PMID: 37501197 PMCID: PMC10375693 DOI: 10.1186/s12967-023-04175-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/30/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Finding a noninvasive radiomic surrogate of tumor immune features could help identify patients more likely to respond to novel immune checkpoint inhibitors. Particularly, CD73 is an ectonucleotidase that catalyzes the breakdown of extracellular AMP into immunosuppressive adenosine, which can be blocked by therapeutic antibodies. High CD73 expression in colorectal cancer liver metastasis (CRLM) resected with curative intent is associated with early recurrence and shorter patient survival. The aim of this study was hence to evaluate whether machine learning analysis of preoperative liver CT-scan could estimate high vs low CD73 expression in CRLM and whether such radiomic score would have a prognostic significance. METHODS We trained an Attentive Interpretable Tabular Learning (TabNet) model to predict, from preoperative CT images, stratified expression levels of CD73 (CD73High vs. CD73Low) assessed by immunofluorescence (IF) on tissue microarrays. Radiomic features were extracted from 160 segmented CRLM of 122 patients with matched IF data, preprocessed and used to train the predictive model. We applied a five-fold cross-validation and validated the performance on a hold-out test set. RESULTS TabNet provided areas under the receiver operating characteristic curve of 0.95 (95% CI 0.87 to 1.0) and 0.79 (0.65 to 0.92) on the training and hold-out test sets respectively, and outperformed other machine learning models. The TabNet-derived score, termed rad-CD73, was positively correlated with CD73 histological expression in matched CRLM (Spearman's ρ = 0.6004; P < 0.0001). The median time to recurrence (TTR) and disease-specific survival (DSS) after CRLM resection in rad-CD73High vs rad-CD73Low patients was 13.0 vs 23.6 months (P = 0.0098) and 53.4 vs 126.0 months (P = 0.0222), respectively. The prognostic value of rad-CD73 was independent of the standard clinical risk score, for both TTR (HR = 2.11, 95% CI 1.30 to 3.45, P < 0.005) and DSS (HR = 1.88, 95% CI 1.11 to 3.18, P = 0.020). CONCLUSIONS Our findings reveal promising results for non-invasive CT-scan-based prediction of CD73 expression in CRLM and warrant further validation as to whether rad-CD73 could assist oncologists as a biomarker of prognosis and response to immunotherapies targeting the adenosine pathway.
Collapse
Affiliation(s)
- Ralph Saber
- MedICAL Laboratory, Polytechnique Montréal, Montréal, H3T 1J4, Canada
- Imaging and Engineering Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis R10.430, Montréal, QC, H2X 0A9, Canada
| | - David Henault
- Cancer Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis, Room R10.430, Montréal, QC, H2X 0A9, Canada
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Service, Centre hospitalier de l'Université de Montréal, 1000, rue Saint-Denis, Montréal, QC, H2X 0C1, Canada
| | - Nouredin Messaoudi
- Cancer Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis, Room R10.430, Montréal, QC, H2X 0A9, Canada
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Service, Centre hospitalier de l'Université de Montréal, 1000, rue Saint-Denis, Montréal, QC, H2X 0C1, Canada
- Department of Surgery, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel) and Europe Hospitals, Brussels, Belgium
| | - Rolando Rebolledo
- Cancer Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis, Room R10.430, Montréal, QC, H2X 0A9, Canada
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Service, Centre hospitalier de l'Université de Montréal, 1000, rue Saint-Denis, Montréal, QC, H2X 0C1, Canada
| | - Emmanuel Montagnon
- Imaging and Engineering Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis R10.430, Montréal, QC, H2X 0A9, Canada
| | - Geneviève Soucy
- Pahology Department, Centre hospitalier de l'Université de Montréal, 1000, rue Saint-Denis, Montréal, QC, H2X 0C1, Canada
| | - John Stagg
- Cancer Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis, Room R10.430, Montréal, QC, H2X 0A9, Canada
| | - An Tang
- Imaging and Engineering Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis R10.430, Montréal, QC, H2X 0A9, Canada
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montréal, Montréal, H3T 1J4, Canada
| | - Simon Turcotte
- Cancer Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis, Room R10.430, Montréal, QC, H2X 0A9, Canada.
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation Service, Centre hospitalier de l'Université de Montréal, 1000, rue Saint-Denis, Montréal, QC, H2X 0C1, Canada.
| | - Samuel Kadoury
- MedICAL Laboratory, Polytechnique Montréal, Montréal, H3T 1J4, Canada.
- Imaging and Engineering Axis, Centre de recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal, 900 rue Saint-Denis R10.430, Montréal, QC, H2X 0A9, Canada.
- Department of Computer and Software Engineering, Institute of Biomedical Engineering, Polytechnique Montréal, Montréal, H3T 1J4, Canada.
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montréal, Montréal, H3T 1J4, Canada.
| |
Collapse
|
24
|
Bach N, Winzer R, Tolosa E, Fiedler W, Brauneck F. The Clinical Significance of CD73 in Cancer. Int J Mol Sci 2023; 24:11759. [PMID: 37511518 PMCID: PMC10380759 DOI: 10.3390/ijms241411759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/04/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The search for new and effective treatment targets for cancer immunotherapy is an ongoing challenge. Alongside the more established inhibitory immune checkpoints, a novel potential target is CD73. As one of the key enzymes in the purinergic signalling pathway CD73 is responsible for the generation of immune suppressive adenosine. The expression of CD73 is higher in tumours than in the corresponding healthy tissues and associated with a poor prognosis. CD73, mainly by the production of adenosine, is critical in the suppression of an adequate anti-tumour immune response, but also in promoting cancer cell proliferation, tumour growth, angiogenesis, and metastasis. The upregulation of CD73 and generation of adenosine by tumour or tumour-associated immune cells is a common resistance mechanism to many cancer treatments such as chemotherapy, radiotherapy, targeted therapy, and immunotherapy. Therefore, the inhibition of CD73 represents a new and promising approach to increase therapy efficacy. Several CD73 inhibitors have already been developed and successfully demonstrated anti-cancer activity in preclinical studies. Currently, clinical studies evaluate CD73 inhibitors in different therapy combinations and tumour entities. The initial results suggest that inhibiting CD73 could be an effective option to augment anti-cancer immunotherapeutic strategies. This review provides an overview of the rationale behind the CD73 inhibition in different treatment combinations and the role of CD73 as a prognostic marker.
Collapse
Affiliation(s)
- Niklas Bach
- Department of Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Riekje Winzer
- Department of Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Eva Tolosa
- Department of Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Franziska Brauneck
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
25
|
Lin YS, Chiang SF, Chen CY, Hong WZ, Chen TW, Chen WTL, Ke TW, Yang PC, Liang JA, Shiau AC, Chao KSC, Huang KCY. Targeting CD73 increases therapeutic response to immunogenic chemotherapy by promoting dendritic cell maturation. Cancer Immunol Immunother 2023; 72:2283-2297. [PMID: 36881132 PMCID: PMC10991491 DOI: 10.1007/s00262-023-03416-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 02/22/2023] [Indexed: 03/08/2023]
Abstract
The CD39-CD73-adenosinergic pathway converts adenosine triphosphate (ATP) to adenosine for inhibiting anti-tumor immune responses. Therefore, targeting CD73 to reinvigorate anti-tumor immunity is considered the novel cancer immunotherapy to eradicate tumor cells. To fully understand the critical role of CD39/CD73 in colon adenocarcinoma (COAD), this study aims to comprehensive investigate the prognostic significance of CD39 and CD73 in stage I-IV COAD. Our data demonstrated that CD73 staining strongly marked malignant epithelial cells and CD39 was highly expressed in stromal cells. Attractively, tumor CD73 expression was significantly associated with tumor stage and the risk of distant metastasis, which suggested CD73 was as an independent factor for colon adenocarcinoma patients in univariate COX analysis [HR = 1.465, 95%CI = 1.084-1.978, p = 0.013]; however, high stromal CD39 in COAD patients was more likely to have favorable survival outcome [HR = 1.458, p = 1.103-1.927, p = 0.008]. Notably, high CD73 expression in COAD patients showed poor response to adjuvant chemotherapy and high risk of distant metastasis. High CD73 expression was inversely associated with less infiltration of CD45+ and CD8+ immune cells. However, administration with anti-CD73 antibodies significantly increased the response to oxaliplatin (OXP). Blockade of CD73 signaling synergistically enhanced OXP-induced ATP release, which is a marker of immunogenic cell death (ICD), promotes dendritic cell maturation and immune cell infiltration. Moreover, the risk of colorectal cancer lung metastasis was also decreased. Taken together, the present study revealed tumor CD73 expression inhibited the recruitment of immune cells and correlated with a poor prognosis in COAD patients, especially patients received adjuvant chemotherapy. Targeting CD73 to markedly increased the therapeutic response to chemotherapy and inhibited lung metastasis. Therefore, tumor CD73 may be an independent prognostic factor as well as the potential of therapeutic target for immunotherapy to benefit colon adenocarcinoma patients.
Collapse
Affiliation(s)
- Yun-Shan Lin
- Department of Pathology, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Shu-Fen Chiang
- Lab of Precision Medicine, Feng-Yuan Hospital, Ministry of Health and Welfare, Taichung, 42055, Taiwan, ROC
| | - Chia-Yi Chen
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Wei-Ze Hong
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Tsung-Wei Chen
- Graduate Institute of Biomedical Science, China Medical University, Taichung, 40402, Taiwan, ROC
- Department of Pathology, Asia University Hospital, Asia University, Taichung, 41354, Taiwan, ROC
| | - William Tzu-Liang Chen
- Department of Colorectal Surgery, China Medical University HsinChu Hospital, China Medical University, HsinChu, 302, Taiwan, ROC
- Department of Colorectal Surgery, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
- Department of Surgery, School of Medicine, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Tao-Wei Ke
- Department of Colorectal Surgery, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
- School of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Pei-Chen Yang
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Ji-An Liang
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
- Department of Radiotherapy, School of Medicine, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
| | - An-Cheng Shiau
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, 40402, Taiwan, ROC
| | - K S Clifford Chao
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC.
- Graduate Institute of Biomedical Science, China Medical University, Taichung, 40402, Taiwan, ROC.
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC.
- Department of Radiotherapy, School of Medicine, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC.
| | - Kevin Chih-Yang Huang
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, 40402, Taiwan, ROC.
- Translation Research Core, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC.
| |
Collapse
|
26
|
Yu Y, Wang Y, Xi D, Wang N, Gao L, Shi Q, Yu R, Li H, Xiang L, Maswikiti EP, Chen H. A novel adenosine signalling-based prognostic signature in gastric cancer and its association with cancer immune features and immunotherapy response. Cell Biol Int 2023. [PMID: 37366248 DOI: 10.1002/cbin.12053] [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: 12/11/2022] [Revised: 04/10/2023] [Accepted: 05/21/2023] [Indexed: 06/28/2023]
Abstract
Reliable prognostic signatures that can reflect the intrinsic characteristics of gastric cancer (GC) are still rare. Here, we developed an adenosine-based prognostic signature and explored its association with the tumour immune in GC patients, aiming at confirming the prognostic value of adenosine-related genes and guiding the GC risk stratification and immunotherapeutic response prediction. We collected adenosine pathway-related genes from STRING websites and manual searching. We enrolled the The Cancer Genome Atlas cohort and four gene expression omnibus cohorts of GC for generating and validating the adenosine pathway-based signature using the Cox regression method. Gene expression in the signature was verified using polymerase chain reaction. We also performed gene set enrichment analysis, immune infiltration assessment and immunotherapy response prediction based on this signature. Our study resulted in a six-gene adenosine signature (GNAS, CXCR4, PPP1R1B, ADCY6, NT5E and NOS3) for risk stratification of GC prognosis, with the highest area under the receiver operating characteristic curve up to 0.767 for predicting 10-year overall survival (OS). In the training cohort, patients with signature-defined high risk had significantly poorer OS than those with low risk (p < .001). Multivariate analysis identified the signature as an independent prognostic factor (hazard ratio 2.863, 95% confidence interval [1.871-4.381], p < .001). These findings were confirmed in four independent cohorts. Expression detection showed that all signature genes were upregulated in both GC tissues and cell lines. Further analysis revealed that the signature-defined high-risk patients were characterised by immunosuppressive states and associated with a poor immunotherapy response. In conclusion, the adenosine pathway-based signature represents a promising risk stratification tool for GC in guiding individualised prognostication and immunotherapy.
Collapse
Affiliation(s)
- Yang Yu
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yidian Wang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Dayong Xi
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Na Wang
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Lei Gao
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Qianling Shi
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Rong Yu
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Haiyuan Li
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Lin Xiang
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Ewetse Paul Maswikiti
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Hao Chen
- The Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
27
|
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
|
28
|
Jain D, Somasundaram DB, Aravindan S, Yu Z, Baker A, Esmaeili A, Aravindan N. Prognostic significance of NT5E/CD73 in neuroblastoma and its function in CSC stemness maintenance. Cell Biol Toxicol 2023; 39:967-989. [PMID: 34773529 DOI: 10.1007/s10565-021-09658-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/10/2021] [Indexed: 12/22/2022]
Abstract
Cluster of differentiation 73 (CD73), a cell surface enzyme that catalyzes adenosine monophosphate (AMP) breakdown to adenosine, is differentially expressed in cancers and has prognostic significance. We investigated its expression profile in neuroblastoma (NB), its association with NB clinical outcomes, and its influence in the regulation of cancer stem cells' (CSCs) stemness maintenance. RNA-Seq data mining (22 independent study cohorts, total n = 3836) indicated that high CD73 can predict good NB prognosis. CD73 expression (immunohistochemistry) gauged in an NB patient cohort (n = 87) showed a positive correlation with longer overall survival (OS, P = 0.0239) and relapse-free survival (RFS, P = 0.0242). Similarly, high CD73 correlated with longer OS and RFS in advanced disease stages, MYCN non-amplified (MYCN-na), and Stage-4-MYCN-na subsets. Despite no definite association in children < 2 years old (2Y), high CD73 correlated with longer OS (P = 0.0294) and RFS (P = 0.0315) in children > 2Y. Consistently, high CD73 was associated with better OS in MYCN-na, high-risk, and stage-4 subsets of children > 2Y. Multivariate analysis identified CD73 as an independent (P = 0.001) prognostic factor for NB. Silencing CD73 in patient-derived (stage 4, progressive disease) CHLA-171 and CHLA-172 cells revealed cell-line-independent activation of 58 CSC stemness maintenance molecules (QPCR profiling). Overexpressing CD73 in CHLA-20 and CHLA-90 cells with low CD73 and silencing in CHLA-171 and CHLA-172 cells with high CD73 showed that CD73 regulates epithelial to mesenchymal transition (E-Cadherin, N-Cadherin, Vimentin), stemness maintenance (Sox2, Nanog, Oct3/4), self-renewal capacity (Notch), and differentiation inhibition (leukemia inhibitory factor, LIF) proteins (confocal-immunofluorescence). These results demonstrate that high CD73 can predict good prognosis in NB, and further suggest that CD73 regulates stemness maintenance in cells that defy clinical therapy.
Collapse
Affiliation(s)
- Drishti Jain
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Dinesh Babu Somasundaram
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Zhongxin Yu
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ashley Baker
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Azadeh Esmaeili
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Natarajan Aravindan
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Stephenson Cancer Center, Oklahoma City, OK, USA.
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
29
|
Yu X, Liu R, Gao W, Wang X, Zhang Y. Single-cell omics traces the heterogeneity of prostate cancer cells and the tumor microenvironment. Cell Mol Biol Lett 2023; 28:38. [PMID: 37161356 PMCID: PMC10170780 DOI: 10.1186/s11658-023-00450-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/17/2023] [Indexed: 05/11/2023] Open
Abstract
Prostate cancer is one of the more heterogeneous tumour types. In recent years, with the rapid development of single-cell sequencing and spatial transcriptome technologies, researchers have gained a more intuitive and comprehensive understanding of the heterogeneity of prostate cancer. Tumour-associated epithelial cells; cancer-associated fibroblasts; the complexity of the immune microenvironment, and the heterogeneity of the spatial distribution of tumour cells and other cancer-promoting molecules play a crucial role in the growth, invasion, and metastasis of prostate cancer. Single-cell multi-omics biotechnology, especially single-cell transcriptome sequencing, reveals the expression level of single cells with higher resolution and finely dissects the molecular characteristics of different tumour cells. We reviewed the recent literature on prostate cancer cells, focusing on single-cell RNA sequencing. And we analysed the heterogeneity and spatial distribution differences of different tumour cell types. We discussed the impact of novel single-cell omics technologies, such as rich omics exploration strategies, multi-omics joint analysis modes, and deep learning models, on future prostate cancer research. In this review, we have constructed a comprehensive catalogue of single-cell omics studies in prostate cancer. This article aimed to provide a more thorough understanding of the diagnosis and treatment of prostate cancer. We summarised and proposed several key issues and directions on applying single-cell multi-omics and spatial transcriptomics to understand the heterogeneity of prostate cancer. Finally, we discussed single-cell omics trends and future directions in prostate cancer.
Collapse
Affiliation(s)
- Xudong Yu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
- Beijing Tumour Minimally Invasive Medical Center of Integrated Traditional Chinese and Western Medicine, Beijing, 101121, China
| | - Ruijia Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Wenfeng Gao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Xuyun Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Yaosheng Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
- Beijing Tumour Minimally Invasive Medical Center of Integrated Traditional Chinese and Western Medicine, Beijing, 101121, China.
| |
Collapse
|
30
|
Zhou Q, Ou Y, Dai X, Chen X, Wu S, Chen W, Hu M, Yang C, Zhang L, Jiang H. Prevalence of tumour-infiltrating CD103 + cells identifies therapeutic-sensitive prostate cancer with poor clinical outcome. Br J Cancer 2023; 128:1466-1477. [PMID: 36759726 PMCID: PMC10070496 DOI: 10.1038/s41416-023-02183-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND The clinical significance and immune correlation of CD103+ cells in prostate cancer (PCa) remain explored. METHODS In total, 1080 patients with PCa underwent radical prostatectomy from three cohorts were enrolled for retrospective analysis. Tumour microarrays were constructed and fresh tumour samples were analysed by flow cytometry. RESULTS High CD103+ cell infiltration correlated with reduced biochemical recurrence (BCR)-free survival in PCa. Adjuvant hormone therapy (HT) prolonged the BCR-free survival for high-risk node-negative diseases with CD103+ cell abundance. CD103+ cell infiltration correlated with less cytotoxic expression and increased infiltration of CD8+ and CD4+ T cells, M1 macrophages and mast cells in PCa. Intratumoral CD8+ T cell was the predominant source of CD103, and the CD103+ subset of CD8+ T cells was featured with high IL-10, PD-1 and CTLA-4 expression. Tumour-infiltrating CD103+ CD8+ T cells exerted anti-tumour function when treated with HT ex vivo. DISCUSSION CD103+ cell infiltration predicted BCR-free survival and response to adjuvant HT in PCa. CD103+ cell infiltration correlated with an enriched but immune-evasive immune landscape. The study supported a model that CD103 expression conferred negative prognostic impact and immunosuppressive function to tumour-infiltrating CD8+ T cells, while the CD103+ CD8+ T cells exhibited a powerful anti-tumour immunity with response to HT.
Collapse
Affiliation(s)
- Quan Zhou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuxi Ou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiyu Dai
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinan Chen
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Siqi Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wensun Chen
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Mengbo Hu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chen Yang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China.
- National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China.
| | - Limin Zhang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China.
- National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China.
| | - Haowen Jiang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China.
- National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China.
- Department of Urology, Jing'an District Central Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
31
|
Li J, Chen L, Billedeau RJ, Stanton TF, Chiang JTP, Lee CC, Li W, Steggerda S, Emberley E, Gross M, Bhupathi D, Che X, Chen J, Dang R, Huang T, Ma Y, MacKinnon A, Makkouk A, Marguier G, Neou S, Sotirovska N, Spurlock S, Zhang J, Zhang W, van Zandt M, Yuan L, Savoy J, Parlati F, Sjogren EB. Discovery of a Series of Potent, Selective, and Orally Bioavailable Nucleoside Inhibitors of CD73 That Demonstrates In Vivo Antitumor Activity. J Med Chem 2023; 66:345-370. [PMID: 36529947 DOI: 10.1021/acs.jmedchem.2c01287] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
CD73 (ecto-5'-nucleotidase) has emerged as an attractive target for cancer immunotherapy of many cancers. CD73 catalyzes the hydrolysis of adenosine monophosphate (AMP) into highly immunosuppressive adenosine that plays a critical role in tumor progression. Herein, we report our efforts in developing orally bioavailable and highly potent small-molecule CD73 inhibitors from the reported hit molecule 2 to lead molecule 20 and then finally to compound 49. Compound 49 was able to reverse AMP-mediated suppression of CD8+ T cells and completely inhibited CD73 activity in serum samples from various cancer patients. In preclinical in vivo studies, orally administered 49 showed a robust dose-dependent pharmacokinetic/pharmacodynamic (PK/PD) relationship that correlated with efficacy. Compound 49 also demonstrated the expected immune-mediated antitumor mechanism of action and was efficacious upon oral administration not only as a single agent but also in combination with either chemotherapeutics or checkpoint inhibitor in the mouse tumor model.
Collapse
Affiliation(s)
- Jim Li
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Lijing Chen
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Roland J Billedeau
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Timothy F Stanton
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - John T P Chiang
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Clarissa C Lee
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Weiqun Li
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Susanne Steggerda
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Ethan Emberley
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Matthew Gross
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Deepthi Bhupathi
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | | | - Jason Chen
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Rosalyn Dang
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Tony Huang
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Yong Ma
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Andrew MacKinnon
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Amani Makkouk
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Gisele Marguier
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Silinda Neou
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Natalija Sotirovska
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Sandra Spurlock
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Jing Zhang
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Winter Zhang
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | | | - Lin Yuan
- NEDP, Branford, Connecticut 06405, United States
| | | | - Francesco Parlati
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Eric B Sjogren
- Calithera Biosciences, 343 Oyster Point Boulevard, South San Francisco, California 94080, United States
| |
Collapse
|
32
|
Catalano M, Shabani S, Venturini J, Ottanelli C, Voltolini L, Roviello G. Lung Cancer Immunotherapy: Beyond Common Immune Checkpoints Inhibitors. Cancers (Basel) 2022; 14:6145. [PMID: 36551630 PMCID: PMC9777293 DOI: 10.3390/cancers14246145] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/27/2022] [Accepted: 11/27/2022] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy is an ever-expanding field in lung cancer treatment research. Over the past two decades, there has been significant progress in identifying immunotherapy targets and creating specific therapeutic agents, leading to a major paradigm shift in lung cancer treatment. However, despite the great success achieved with programmed death protein 1/ligand 1 (PD-1/PD-L1) monoclonal antibodies and with anti-PD-1/PD-L1 plus anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4), only a minority of lung cancer patients respond to treatment, and of these many subsequently experience disease progression. In addition, immune-related adverse events sometimes can be life-threatening, especially when anti-CTLA-4 and anti-PD-1 are used in combination. All of this prompted researchers to identify novel immune checkpoints targets to overcome these limitations. Lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin (Ig) and Immunoreceptor Tyrosine-Based Inhibitory Motif (ITIM) domain (TIGIT), T cell immunoglobulin and mucin-domain containing-3 (TIM-3) are promising molecules now under investigation. This review aims to outline the current role of immunotherapy in lung cancer and to examine efficacy and future applications of the new immune regulating molecules.
Collapse
Affiliation(s)
- Martina Catalano
- School of Human Health Sciences, University of Florence, 50134 Florence, Italy
| | - Sonia Shabani
- School of Human Health Sciences, University of Florence, 50134 Florence, Italy
| | - Jacopo Venturini
- School of Human Health Sciences, University of Florence, 50134 Florence, Italy
| | - Carlotta Ottanelli
- School of Human Health Sciences, University of Florence, 50134 Florence, Italy
| | - Luca Voltolini
- Thoraco-Pulmonary Surgery Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Giandomenico Roviello
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| |
Collapse
|
33
|
Xue XM, Liu YY, Chen XM, Tao BY, Liu P, Zhou HW, Zhang C, Wang L, Jiang YK, Ding ZW, Shen WD, Zhang J, Yang SM, Wang FY. Pan-cancer analysis identifies NT5E as a novel prognostic biomarker on cancer-associated fibroblasts associated with unique tumor microenvironment. Front Pharmacol 2022; 13:1064032. [PMID: 36569293 PMCID: PMC9768042 DOI: 10.3389/fphar.2022.1064032] [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: 10/07/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Ecto-5'-nucleotidase (NT5E) encodes the cluster of differentiation 73 (CD73), whose overexpression contributes to the formation of immunosuppressive tumor microenvironment and is related to exacerbated prognosis, increased risk of metastasis and resistance to immunotherapy of various tumors. However, the prognostic significance of NT5E in pan-cancer is obscure so far. Methods: We explored the expression level of NT5E in cancers and adjacent tissues and revealed the relationship between the NT5E expression level and clinical outcomes in pan-cancer by utilizing the UCSC Xena database. Then, correlation analyses were performed to evaluate the relationship between NT5E expression and immune infiltration level via EPIC, MCP-counter and CIBERSORT methods, and the enrichment analysis were employed to identify NT5E-interacting molecules and functional pathways. Furthermore, we conducted single-cell analysis to explore the potential role of NT5E on single-cell level based on the CancerSEA database. Meanwhile, gene set enrichment analysis (GSEA) in single-cell level was also conducted in TISCH database and single-cell signature explorer was utilized to evaluate the epithelial-mesenchymal transition (EMT) level in each cell type. Results: The expression level of NT5E was aberrant in almost all cancer types, and was correlated with worse prognosis in several cancers. Notably, NT5E overexpression was related to worse overall survival (OS) in pancreatic adenocarcinoma (PAAD), head and neck squamous cell carcinoma (HNSC), mesothelioma (MESO), stomach adenocarcinoma (STAD), uveal melanoma (UVM) and cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) (p < 0.01). NT5E-related immune microenvironment analysis revealed that NT5E is associated positively with the degree of infiltration of cancer-associated fibroblasts (CAFs) and endothelial cells in most cancers. Enrichment analysis of cellular component (CC) demonstrated the critical part of NT5E played in cell-substrate junction, cell-substrate adherens junction, focal adhesion and external side of plasma membrane. Finally, single-cell analysis of NT5E illuminated that EMT function of CAFs was elevated in basal cell carcinoma (BCC), skin cutaneous melanoma (SKCM), HNSC and PAAD. Conclusion: NT5E could serve as a potential prognostic biomarker for cancers. The potential mechanism may be related to the upregulated EMT function of CAFs, which provides novel inspiration for immunotherapy by targeting CAFs with high NT5E expression.
Collapse
Affiliation(s)
- Xin-miao Xue
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Yu-yang Liu
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Department of Neurosurgery, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Xue-min Chen
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Bing-yan Tao
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Department of Neurosurgery, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Peng Liu
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Han-wen Zhou
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Chi Zhang
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,The Zhantansi Outpatient Department of Central Medical Branch of People’s Liberation Army (PLA) General Hospital Beijing, China
| | - Li Wang
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Yu-ke Jiang
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Zhi-wei Ding
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Wei-dong Shen
- Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Jun Zhang
- Department of Neurosurgery, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China,*Correspondence: Jun Zhang, ; Shi-ming Yang, ; Fang-yuan Wang,
| | - Shi-ming Yang
- Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China,*Correspondence: Jun Zhang, ; Shi-ming Yang, ; Fang-yuan Wang,
| | - Fang-yuan Wang
- Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China,*Correspondence: Jun Zhang, ; Shi-ming Yang, ; Fang-yuan Wang,
| |
Collapse
|
34
|
Hamoud AR, Bach K, Kakrecha O, Henkel N, Wu X, McCullumsmith RE, O’Donovan SM. Adenosine, Schizophrenia and Cancer: Does the Purinergic System Offer a Pathway to Treatment? Int J Mol Sci 2022; 23:ijms231911835. [PMID: 36233136 PMCID: PMC9570456 DOI: 10.3390/ijms231911835] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/23/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
For over a century, a complex relationship between schizophrenia diagnosis and development of many cancers has been observed. Findings from epidemiological studies are mixed, with reports of increased, reduced, or no difference in cancer incidence in schizophrenia patients. However, as risk factors for cancer, including elevated smoking rates and substance abuse, are commonly associated with this patient population, it is surprising that cancer incidence is not higher. Various factors may account for the proposed reduction in cancer incidence rates including pathophysiological changes associated with disease. Perturbations of the adenosine system are hypothesized to contribute to the neurobiology of schizophrenia. Conversely, hyperfunction of the adenosine system is found in the tumor microenvironment in cancer and targeting the adenosine system therapeutically is a promising area of research in this disease. We outline the current biochemical and pharmacological evidence for hypofunction of the adenosine system in schizophrenia, and the role of increased adenosine metabolism in the tumor microenvironment. In the context of the relatively limited literature on this patient population, we discuss whether hypofunction of this system in schizophrenia, may counteract the immunosuppressive role of adenosine in the tumor microenvironment. We also highlight the importance of studies examining the adenosine system in this subset of patients for the potential insight they may offer into these complex disorders.
Collapse
Affiliation(s)
- Abdul-Rizaq Hamoud
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Karen Bach
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Ojal Kakrecha
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Nicholas Henkel
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Xiaojun Wu
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Robert E. McCullumsmith
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
- Neurosciences Institute, ProMedica, Toledo, OH 43606, USA
| | - Sinead M. O’Donovan
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
- Correspondence:
| |
Collapse
|
35
|
Casadó‐Llombart S, Ajami T, Consuegra‐Fernández M, Carreras E, Aranda F, Armiger N, Alcaraz A, Mengual L, Lozano F. Gene variation impact on prostate cancer progression: Lymphocyte modulator, activation, and cell adhesion gene variant contribution. Prostate 2022; 82:1331-1337. [PMID: 35767366 PMCID: PMC9542726 DOI: 10.1002/pros.24407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/29/2022] [Accepted: 06/01/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND The view of prostate cancer (PCa) progression as a result of the interaction of epithelial cancer cells with the host's immune system is supported by the presence of tumor infiltrating lymphocytes (TILs). TILs fate and interaction with the tumor microenvironment is mediated by accessory molecules such as CD5 and CD6, two signal-transducing coreceptors involved in fine-tuning of T cell responses. While the nature of the CD5 ligand is still controversial, CD6 binds CD166/ALCAM, a cell adhesion molecule involved in progression and dissemination of epithelial cancers, including PCa. The purpose of the present study was to determine the role of CD5, CD6, and CD166/ALCAM gene variants in PCa. METHODS Functionally relevant CD5 (rs2241002 and rs2229177), CD6 (rs17824933, rs11230563, and rs12360861) and CD166/ALCAM (rs6437585, rs579565, rs1044243, and rs35271455) single nucleotide polymorphisms (SNPs) were genotyped in germline DNA samples from 376 PCa patients. Their association with PCa prognostic factors, namely biochemical recurrence (BCR) and International Society of Urological Pathology (ISUP) grade was analyzed by generalized linear models and survival analyses. RESULT Proportional hazards regression showed that the minor CD6 rs12360861AA and CD166/ALCAM rs579565AA genotypes were associated with earlier BCR, with hazard ratios of 2.65 (95% CI: 1.39-5.05, p = 0.003) and 1.86, (95% CI: 1.02-3.39, p = 0.043), respectively. Individually, none of the analyzed SNPs was significantly associated with ISUP grade, but haplotype analyses revealed association of the CD5 rs2241002C -rs2229177T haplotype with ISUP grade ≥2, with odds ratio of 1.52 (95% CI: 1.05-2.21, p = 0.026). CONCLUSION The results show the impact on PCa aggressiveness and recurrence brought about by gene variants involved in modulation of lymphocyte activation (CD5, CD6) and immune-epithelial cell adhesion (CD166/ALCAM) in PCa aggressiveness and recurrence, thus supporting a role for host immune response in PCa pathophysiology.
Collapse
Affiliation(s)
- Sergi Casadó‐Llombart
- Immunoreceptors del Sistema Innat i AdaptatiuInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Tarek Ajami
- Laboratori i Servei d'UrologiaHospital Clínic de BarcelonaBarcelonaSpain
| | - Marta Consuegra‐Fernández
- Immunoreceptors del Sistema Innat i AdaptatiuInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Esther Carreras
- Immunoreceptors del Sistema Innat i AdaptatiuInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Fernando Aranda
- Immunoreceptors del Sistema Innat i AdaptatiuInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Noelia Armiger
- Immunoreceptors del Sistema Innat i AdaptatiuInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Antonio Alcaraz
- Laboratori i Servei d'UrologiaHospital Clínic de BarcelonaBarcelonaSpain
- Genètica i tumors urològicsInstitut d'Investigacions Biomèdiques August Pi i Sunyer, IDIBAPSBarcelonaSpain
| | - Lourdes Mengual
- Laboratori i Servei d'UrologiaHospital Clínic de BarcelonaBarcelonaSpain
- Genètica i tumors urològicsInstitut d'Investigacions Biomèdiques August Pi i Sunyer, IDIBAPSBarcelonaSpain
- Departament de Biomedicina, Facultat de Medicina i Ciències de la SalutUniversitat de Barcelona (UB)BarcelonaSpain
| | - Francisco Lozano
- Immunoreceptors del Sistema Innat i AdaptatiuInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
- Genètica i tumors urològicsInstitut d'Investigacions Biomèdiques August Pi i Sunyer, IDIBAPSBarcelonaSpain
- Departament de Biomedicina, Facultat de Medicina i Ciències de la SalutUniversitat de Barcelona (UB)BarcelonaSpain
- Servei d'Immunologia, Centre de Diagnòstic BiomèdicHospital Clínic de BarcelonaBarcelonaSpain
| |
Collapse
|
36
|
Gao Z, Wang L, Song Z, Ren M, Yang Y, Li J, Shen K, Li Y, Ding Y, Yang Y, Zhou Y, Wei C, Gu J. Intratumoral CD73: An immune checkpoint shaping an inhibitory tumor microenvironment and implicating poor prognosis in Chinese melanoma cohorts. Front Immunol 2022; 13:954039. [PMID: 36131912 PMCID: PMC9483101 DOI: 10.3389/fimmu.2022.954039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAs a novel immune checkpoint, CD73 has been reported to play prominent roles in several malignancies. However, the significance of CD73 in melanoma remains ambiguous. This study sought to reveal the impact of CD73 on the tumor microenvironment (TME) and patients’ prognosis, and to investigate whether CD73 could be a therapeutic target in Chinese melanomas, which were dominated by acral and mucosal subtypes.MethodsTwo independent Chinese cohorts of 194 patients with melanoma were enrolled. CD73 and PD-L1 expression as well as CD8+ and CD56+ cell infiltrations were evaluated by immunohistochemistry in 194 resected melanoma samples. Clinical outcomes of patients were assessed utilizing the Kaplan-Meier plotter and Cox proportional hazard analysis. RNA-seq data was obtained from TCGA database. Gene set functional annotations were performed based on GO, KEGG and GSEA analysis. CIBERSORT, ssGSEA and TIMER were used to explore the association between CD73 and immune infiltration. These findings were validated by establishing tumor xenograft model, and functions of tumor-infiltrating immune cells were examined by flow cytometry and immunofluorescence.ResultsHigh CD73 expression showed poorer clinical outcomes and was identified as an independent prognostic indicator for survival in two cohorts. Expression of CD73 was more prevalent than PD-L1 in Chinese melanoma cohorts (54.6% vs 23.2%). Co-expression of both immune checkpoints was infrequent (12.9%) in melanoma, and 54.4% of PD-L1 negative cases showed elevated expression of CD73. CD73high tumors showed a microenvironment with fewer CD8+ T cells and CD56+ NK cells infiltration, which displayed a dysfunctional phenotype. With the treatment of CD73 inhibitor APCP, the amount of CD8+ T cells and CD56+ NK cells infiltrated in tumors was elevated and the immunosuppressive effect of CD73 was eliminated.ConclusionsHigh CD73 expression was associated with an inhibitory TME and adverse clinical outcomes of melanoma. In comparison to PD-L1, CD73 was more prevalent and possessed more definite prognostic significance. Therefore, it may serve as a prognostic indicator and immunotherapeutic target next to PD-L1 in melanoma for Chinese population.
Collapse
Affiliation(s)
- Zixu Gao
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lu Wang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhengqing Song
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ming Ren
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Yang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianrui Li
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kangjie Shen
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yinlam Li
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiteng Ding
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanwen Yang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuhong Zhou
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuanyuan Wei
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Jianying Gu, ; Chuanyuan Wei,
| | - Jianying Gu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Jianying Gu, ; Chuanyuan Wei,
| |
Collapse
|
37
|
Zhou Q, Yang C, Mou Z, Wu S, Dai X, Chen X, Ou Y, Zhang L, Sha J, Jiang H. Identification and validation of a poor clinical outcome subtype of primary prostate cancer with Midkine abundance. Cancer Sci 2022; 113:3698-3709. [PMID: 36018546 PMCID: PMC9633304 DOI: 10.1111/cas.15546] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 11/30/2022] Open
Abstract
Recent studies identified Midkine (MDK) as playing a key role in immune regulation. In this study, we aimed to discover the clinical significance and translational relevance in prostate cancer (PCa). We retrospectively analyzed 759 PCa patients who underwent radical prostatectomy from Huashan Hospital, Fudan University (training cohort, n = 369) and Chinese Prostate Cancer Consortium (validation cohort, n = 390). A total of 325 PCa patients from The Cancer Genome Atlas (TCGA) database (external cohort) were analyzed for exploration. Immune landscape and antitumor immunity were assessed through immunohistochemistry and flow cytometry. Patient‐derived explant culture system was applied for evaluating the targeting potential of MDK. We found that intratumoral MDK expression correlated with PCa progression, which indicated an unfavorable biochemical recurrence (BCR)‐free survival for postoperative PCa patients. Addition of MDK expression to the postoperative risk assessment tool CAPRA‐S could improve its prognostic value. Tumors with MDK abundance characterized the tumor‐infiltrating CD8+ T cells with less cytotoxicity production and increased immune checkpoint expression, which were accompanied by enriched immunosuppressive contexture. Moreover, MDK inhibition could reactivate CD8+ T cell antitumor immunity. MDK mRNA expression negatively correlated with androgen receptor activity signature and positively associated with radiotherapy‐related signature. In conclusion, intratumoral MDK expression could serve as an independent prognosticator for BCR in postoperative PCa patients. MDK expression impaired the antitumor function of CD8+ T cells through orchestrating an immunoevasive microenvironment, which could be reversed by MDK inhibition. Moreover, tumors with MDK enrichment possessed potential sensitivity to postoperative radiotherapy while resistance to adjuvant hormonal therapy of PCa. MDK could be considered as a potential therapeutic target for PCa.
Collapse
Affiliation(s)
- Quan Zhou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chen Yang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
| | - Zezhong Mou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Siqi Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiyu Dai
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinan Chen
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuxi Ou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Limin Zhang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianjun Sha
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haowen Jiang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Han Z, Feng Y, Deng Y, Tang Z, Cai S, Zhuo Y, Liang Y, Ye J, Cai Z, Yang S, Liang Y, Hon CT, Chen J, Zhong W. Integrated analysis reveals prognostic value and progression-related role of AMIGO2 in prostate cancer. Transl Androl Urol 2022; 11:914-928. [PMID: 35958903 PMCID: PMC9360515 DOI: 10.21037/tau-21-1148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 06/26/2022] [Indexed: 11/28/2022] Open
Abstract
Background Even though emerging studies supplied evidence that Adhesion Molecule with Ig Like Domain family 2 (AMIGO2) plays a critical role in numerous cancers, comprehensive analysis of the prognostic value and significant role of AMIGO2 in prostate cancer (PCa) have not been described. Methods Differentially expressed analysis, survival analysis and univariate cox regression analysis were first performed to explore the diagnostic and prognostic role of AMIGO2 in various cancers, especially in PCa. Tissue microarray were used to examined the association between AMGIO2 and clinical features. Multivariate cox regression analysis, concordance index, nomogram construction, the receiver operator characteristic curve and calibration curves were further used to discover the effects of AMIGO2 on recurrence-free survival (RFS) and clinicopathological characteristics, including age, Gleason score (GS) and tumor stage. Genetic and Epigenetic Alterations analysis were further conducted to explore the potential effect of AMIGO2 in PCa and examined by biological function analysis and in vitro experiments. Results AMIGO2 was associated with poor RFS (P<0.05) and differentially expressed (P<0.05) in multiple cancer type, especially in PCa. Besides, decreasing the expression of AMIGO2 inhibited PCa cell proliferation and colony formation in vitro. In addition, AMIGO2 was a reliable prognostic marker providing additional information (C-index: 0.7) that supplement the currently used prognosis evaluation system, e.g., T stage (C-index: 0.62) and GS (C-index: 0.65). A novel nomogram was established based on AMIGO2, tumor stage and GS with accuracies (areas under curve) of 0.70, 0.78 and 0.82 for predicting 3-, 5- and 7-year RFS, respectively. Bioinformatic analysis and in vitro examination also suggested that AMIGO2 might involve in the progression of PCa tumors inducing epithelial mesenchymal transition (EMT). Conclusions We identified AMIGO2 as a pan-cancer gene that could not only be a prognostic biomarker in various cancers, especially in PCa, but may functionally promoting PCa progression via EMT and mediating docetaxel resistance, suggesting AMIGO2 as a potential target for future treatment of PCa.
Collapse
Affiliation(s)
- Zhaodong Han
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuanfa Feng
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.,Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yulin Deng
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.,Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Zhenfeng Tang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.,Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Shanghua Cai
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yangjia Zhuo
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yingke Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jianheng Ye
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhouda Cai
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shuai Yang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yuxiang Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chi Tin Hon
- Macau Institute of Systems Engineering, Macau University of Science and Technology, Avenida Wai Long, Macau, China
| | - Jiahong Chen
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, China
| | - Weide Zhong
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.,Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.,Macau Institute of Systems Engineering, Macau University of Science and Technology, Avenida Wai Long, Macau, China
| |
Collapse
|
39
|
Iser IC, Vedovatto S, Oliveira FD, Beckenkamp LR, Lenz G, Wink MR. The crossroads of adenosinergic pathway and epithelial-mesenchymal plasticity in cancer. Semin Cancer Biol 2022; 86:202-213. [PMID: 35779713 DOI: 10.1016/j.semcancer.2022.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 10/31/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a key mechanism related to tumor progression, invasion, metastasis, resistance to therapy and poor prognosis in several types of cancer. However, targeting EMT or partial-EMT, as well as the molecules involved in this process, has remained a challenge. Recently, the CD73 enzyme, which hydrolyzes AMP to produce adenosine (ADO), has been linked to the EMT process. This relationship is not only due to the production of the immunosuppressant ADO but also to its role as a receptor for extracellular matrix proteins, being involved in cell adhesion and migration. This article reviews the crosstalk between the adenosinergic pathway and the EMT program and the impact of this interrelation on cancer development and progression. An in silico analysis of RNAseq datasets showed that several tumor types have a significant correlation between an EMT score and NT5E (CD73) and ENTPD1 (CD39) expressions, with the strongest correlations in prostate adenocarcinoma. Furthermore, it is evident that the cooperation between EMT and adenosinergic pathway in tumor progression is context and tumor-dependent. The increased knowledge about this topic will help broaden the view to explore new treatments and therapies for different types of cancer.
Collapse
Affiliation(s)
- Isabele Cristiana Iser
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Samlai Vedovatto
- Department of Biophysics and Center of Biotechnology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fernanda Dittrich Oliveira
- Department of Biophysics and Center of Biotechnology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Liziane Raquel Beckenkamp
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Guido Lenz
- Department of Biophysics and Center of Biotechnology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Márcia Rosângela Wink
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil.
| |
Collapse
|
40
|
Trigos AS, Pasam A, Banks P, Wallace R, Guo C, Keam S, Thorne H, Mitchell C, Lade S, Clouston D, Hakansson A, Liu Y, Blyth B, Murphy D, Lawrentschuk N, Bolton D, Moon D, Darcy P, Haupt Y, Williams SG, Castro E, Olmos D, Goode D, Neeson P, Sandhu S. Tumor immune microenvironment of primary prostate cancer with and without germline mutations in homologous recombination repair genes. J Immunother Cancer 2022; 10:jitc-2021-003744. [PMID: 35764368 PMCID: PMC9240881 DOI: 10.1136/jitc-2021-003744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Aberrations in homologous recombination repair (HRR) genes are emerging as important biomarkers for personalized treatment in prostate cancer (PCa). HRR deficiency (HRD) could affect the tumor immune microenvironment (TIME), potentially contributing to differential responses to poly ADP-ribose polymerase (PARP) inhibitors and immune checkpoint inhibitors. Spatial distribution of immune cells in a range of cancers identifies novel disease subtypes and is related to prognosis. In this study we aimed to determine the differences in the TIME of PCa with and without germline (g) HRR mutations. METHODS We performed gene expression analysis, multiplex immunohistochemistry of T and B cells and quantitative spatial analysis of PCa samples from 36 patients with gHRD and 26 patients with sporadic PCa. Samples were archival tumor tissue from radical prostatectomies with the exception of one biopsy. Results were validated in several independent cohorts. RESULTS Although the composition of the T cell and B cells was similar in the tumor areas of gHRD-mutated and sporadic tumors, the spatial profiles differed between these cohorts. We describe two T-cell spatial profiles across primary PCa, a clustered immune spatial (CIS) profile characterized by dense clusters of CD4+ T cells closely interacting with PD-L1+ cells, and a free immune spatial (FIS) profile of CD8+ cells in close proximity to tumor cells. gHRD tumors had a more T-cell inflamed microenvironment than sporadic tumors. The CIS profile was mainly observed in sporadic tumors, whereas a FIS profile was enriched in gHRD tumors. A FIS profile was associated with lower Gleason scores, smaller tumors and longer time to biochemical recurrence and metastasis. CONCLUSIONS gHRD-mutated tumors have a distinct immune microenvironment compared with sporadic tumors. Spatial profiling of T-cells provides additional information beyond T-cell density and is associated with time to biochemical recurrence, time to metastasis, tumor size and Gleason scores.
Collapse
Affiliation(s)
- Anna Sofia Trigos
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Anupama Pasam
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Patricia Banks
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Roslyn Wallace
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Christina Guo
- Institute of Cancer Research Sutton, Sutton, Surrey, UK,Royal Marsden Hospital Sutton, Sutton, London, UK
| | - Simon Keam
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Heather Thorne
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - kConFab
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Catherine Mitchell
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Stephen Lade
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | | | - Yang Liu
- Veracyte Inc, South San Francisco, California, USA
| | - Benjamin Blyth
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Declan Murphy
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia,Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Nathan Lawrentschuk
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Daniel Moon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Phil Darcy
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Ygal Haupt
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Scott G Williams
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Elena Castro
- Instituto de Investigacion Biomedica de Malaga, Malaga, Spain
| | - David Olmos
- Instituto de Investigacion Biomedica de Malaga, Malaga, Spain,Medical Oncology Department, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - David Goode
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul Neeson
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Shahneen Sandhu
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
41
|
CD73/Adenosine Pathway Involvement in the Interaction of Non-Small Cell Lung Cancer Stem Cells and Bone Cells in the Pre-Metastatic Niche. Int J Mol Sci 2022; 23:ijms23095126. [PMID: 35563517 PMCID: PMC9104817 DOI: 10.3390/ijms23095126] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 11/17/2022] Open
Abstract
Adenosinergic signaling is an important regulator of tissue homeostasis and extracellular accumulation of adenosine (Ado) and is associated with different pathologies, such as cancer. In non-small-cell lung cancer (NSCLC), a subset of CD133/CXCR4+ cancer stem cell (CSCs) has been demonstrated to initiate bone metastases. Here we investigated how NSCLC CSCs interact with osteoclasts (OCs) and osteoblasts (OBs) by modulating Ado production and OC activity. We proved that CSC-spheres, generated in vitro from NSCLC cell lines, express CD38, PC-1, and CD73, enzymes of the non-canonical adenosinergic pathway, produce high level of Ado, and down-regulate A1R and A3R inhibitory receptors, while expressing A2AR and A2BR. To address the Ado role and modulation of the in-bone pre-metastatic niche, we performed co-cultures of CSC-spheres with OCs and OBs cells. Firstly, we verified that active OCs do not activate non-canonical the adenosinergic pathway, conversely to OBs. OCs co-cultured with CSC-spheres increase Ado production that is related to the OC resorption activity and contributes to T-cell suppression. Finally, we proved the efficacy of anti-CD73 agents in blocking NSCLC cell migration. Overall, we assessed the importance of adenosinergic signaling in the interaction between CSCs and OCs at the pre-metastatic niche, with therapeutic implications related to Ado production.
Collapse
|
42
|
Su Y, Xu B, Shen Q, Lei Z, Zhang W, Hu T. LIMK2 Is a Novel Prognostic Biomarker and Correlates With Tumor Immune Cell Infiltration in Lung Squamous Cell Carcinoma. Front Immunol 2022; 13:788375. [PMID: 35273591 PMCID: PMC8902256 DOI: 10.3389/fimmu.2022.788375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Previous research found that LIM domain kinase 2 (LIMK2) expression correlated with a poor prognosis in many cancers. However, its role in lung squamous cell carcinoma (LUSC) has not yet been clarified. Our study aimed to clarify the role of LIMK2 in LUSC prognosis prediction and explore the relationship between LIMK2 and immune infiltration in LUSC. In this study, we first analyzed the expression level and prognostic value of LIMK2 across cancers. Subsequently, we explored the association of LIMK2 expression with immune infiltrating cells and immune checkpoints. our study found that LIMK2 was highly expressed and positively associated with the overall survival of LUSC. Moreover, our study further indicated that LIMK2 expression was significantly negatively correlated with immune cell infiltration and immune checkpoints in LUSC. Finally, we confirmed upstream regulatory noncoding RNAs (ncRNAs) of LIMK2, and the PVT1 and DHRS4-AS1/miR-423-5p/LIMK2 regulatory axes were successfully constructed in LUSC. Put together, LIMK2 is a novel prognostic biomarker and correlates with tumor immune cell infiltration in LUSC, and the expression of LIMK2 is regulated by the PVT1 and DHRS4-AS1/miR-423-5p axes.
Collapse
Affiliation(s)
- Yongcheng Su
- Cancer Research Center, Xiamen University School of Medicine, Xiamen, China
| | - Beibei Xu
- Department of General Surgery, The First Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qianwen Shen
- Cancer Research Center, Xiamen University School of Medicine, Xiamen, China
| | - Ziyu Lei
- Cancer Research Center, Xiamen University School of Medicine, Xiamen, China
| | - Wenqing Zhang
- Cancer Research Center, Xiamen University School of Medicine, Xiamen, China
| | - Tianhui Hu
- Cancer Research Center, Xiamen University School of Medicine, Xiamen, China.,Shenzhen Research Institute of Xiamen University, Shenzhen, China
| |
Collapse
|
43
|
Sakellakis M, Flores L, Ramachandran S. Patterns of indolence in prostate cancer (Review). Exp Ther Med 2022; 23:351. [PMID: 35493432 PMCID: PMC9019743 DOI: 10.3892/etm.2022.11278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/11/2022] [Indexed: 11/20/2022] Open
Abstract
Although prostate cancer is a major cause of cancer-related mortality worldwide, most patients will have a relatively indolent clinical course. Contrary to most other types of cancer, even the diagnosis of locally advanced or metastatic disease is not always lethal. The present review aimed to summarize what is known regarding the underlying mechanisms related to the indolent course of subsets of prostate cancer, at various stages. The data suggested that no specific gene alteration by itself was responsible for carcinogenesis or disease aggressiveness. However, pathway analysis identified genetic aberrations in multiple critical pathways that tend to accumulate over the course of the disease. The progression from indolence into aggressive disease is associated with a complex interplay in which genetic and epigenetic factors are involved. The effect of the immune tumor microenvironment is also very important. Emerging evidence has suggested that the upregulation of pathways related to cellular aging and senescence can identify patients with indolent disease. In addition, a number of tumors enter a long-lasting quiescent state. Further research will determine whether halting tumor evolution is a feasible option, and whether the life of patients can be markedly prolonged by inducing tumor senescence or long-term dormancy.
Collapse
Affiliation(s)
- Minas Sakellakis
- Fourth Oncology Department and Comprehensive Clinical Trials Center, Metropolitan Hospital, 18547 Athens, Greece
| | - Laura Flores
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX 77025, USA
| | - Sumankalai Ramachandran
- Department of Genitourinary Oncology, MD Anderson Cancer Center, University of Texas, Houston, TX 77025, USA
| |
Collapse
|
44
|
High Keratin-7 Expression in Benign Peri-Tumoral Prostatic Glands Is Predictive of Bone Metastasis Onset and Prostate Cancer-Specific Mortality. Cancers (Basel) 2022; 14:cancers14071623. [PMID: 35406395 PMCID: PMC8997075 DOI: 10.3390/cancers14071623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 12/10/2022] Open
Abstract
BACKGROUND New predictive biomarkers are needed to accurately predict metastasis-free survival (MFS) and cancer-specific survival (CSS) in localized prostate cancer (PC). Keratin-7 (KRT7) overexpression has been associated with poor prognosis in several cancers and is described as a novel prostate progenitor marker in the mouse prostate. METHODS KRT7 expression was evaluated in prostatic cell lines and in human tissue by immunohistochemistry (IHC, on advanced PC, n = 91) and immunofluorescence (IF, on localized PC, n = 285). The KRT7 mean fluorescence intensity (MFI) was quantified in different compartments by digital analysis and correlated to clinical endpoints in the localized PC cohort. RESULTS KRT7 is expressed in prostatic cell lines and found in the basal and supra-basal compartment from healthy prostatic glands and benign peri-tumoral glands from localized PC. The KRT7 staining is lost in luminal cells from localized tumors and found as an aberrant sporadic staining (2.2%) in advanced PC. In the localized PC cohort, high KRT7 MFI above the 80th percentile in the basal compartment was significantly and independently correlated with MFS and CSS, and with hypertrophic basal cell phenotype. CONCLUSION High KRT7 expression in benign glands is an independent biomarker of MFS and CSS, and its expression is lost in tumoral cells. These results require further validation on larger cohorts.
Collapse
|
45
|
Challenges of the Immunotherapy: Perspectives and Limitations of the Immune Checkpoint Inhibitor Treatment. Int J Mol Sci 2022; 23:ijms23052847. [PMID: 35269988 PMCID: PMC8910928 DOI: 10.3390/ijms23052847] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 02/04/2023] Open
Abstract
Immunotherapy is a quickly developing type of treatment and the future of therapy in oncology. This paper is a review of recent findings in the field of immunotherapy with an emphasis on immune checkpoint inhibitors. The challenges that immunotherapy might face in near future, such as primary and acquired resistance and the irAEs, are described in this article, as well as the perspectives such as identification of environmental modifiers of immunity and development of anti-cancer vaccines and combined therapies. There are multiple factors that may be responsible for immunoresistance, such as genomic factors, factors related to the immune system cells or to the cancer microenvironment, factors emerging from the host cells, as well as other factors such as advanced age, biological sex, diet, many hormones, existing comorbidities, and the gut microbiome.
Collapse
|
46
|
Mamdani H, Matosevic S, Khalid AB, Durm G, Jalal SI. Immunotherapy in Lung Cancer: Current Landscape and Future Directions. Front Immunol 2022; 13:823618. [PMID: 35222404 PMCID: PMC8864096 DOI: 10.3389/fimmu.2022.823618] [Citation(s) in RCA: 178] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/20/2022] [Indexed: 12/11/2022] Open
Abstract
Over the past decade, lung cancer treatment has undergone a major paradigm shift. A greater understanding of lung cancer biology has led to the development of many effective targeted therapies as well as of immunotherapy. Immune checkpoint inhibitors (ICIs) have shown tremendous benefit in the treatment of non-small cell lung cancer (NSCLC) and are now being used as first-line therapies in metastatic disease, consolidation therapy following chemoradiation in unresectable locally advanced disease, and adjuvant therapy following surgical resection and chemotherapy in resectable disease. Despite these benefits, predicting who will respond to ICIs has proven to be difficult and there remains a need to discover new predictive immunotherapy biomarkers. Furthermore, resistance to ICIs in lung cancer is frequent either because of a lack of response or disease progression after an initial response. The utility of ICIs in the treatment of small cell lung cancer (SCLC) remains limited to first-line treatment of extensive stage disease in combination with chemotherapy with modest impact on overall survival. It is thus important to explore and exploit additional targets to reap the full benefits of immunotherapy in the treatment of lung cancer. Here, we will summarize the current state of immunotherapy in lung cancer, discuss novel targets, and explore the intersection between DNA repair defects and immunotherapy.
Collapse
Affiliation(s)
- Hirva Mamdani
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Sandro Matosevic
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, United States
| | - Ahmed Bilal Khalid
- Department of Internal Medicine, Indiana University, Indianapolis, IN, United States
| | - Gregory Durm
- Department of Internal Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Shadia I. Jalal
- Department of Internal Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
47
|
Immunogenic cell death and its therapeutic or prognostic potential in high-grade glioma. Genes Immun 2022; 23:1-11. [PMID: 35046546 PMCID: PMC8866117 DOI: 10.1038/s41435-021-00161-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/14/2021] [Accepted: 12/30/2021] [Indexed: 12/22/2022]
Abstract
Immunogenic cell death (ICD) has emerged as a key component of therapy-induced anti-tumor immunity. Over the past few years, ICD was found to play a pivotal role in a wide variety of novel and existing treatment modalities. The clinical application of these techniques in cancer treatment is still in its infancy. Glioblastoma (GBM) is the most lethal primary brain tumor with a dismal prognosis despite maximal therapy. The development of new therapies in this aggressive type of tumors remains highly challenging partially due to the cold tumor immune environment. GBM could therefore benefit from ICD-based therapies stimulating the anti-tumor immune response. In what follows, we will describe the mechanisms behind ICD and the ICD-based (pre)clinical advances in anticancer therapies focusing on GBM.
Collapse
|
48
|
Mao C, Ding Y, Xu N. A Double-Edged Sword Role of Cytokines in Prostate Cancer Immunotherapy. Front Oncol 2021; 11:688489. [PMID: 34868907 PMCID: PMC8635015 DOI: 10.3389/fonc.2021.688489] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 10/25/2021] [Indexed: 01/02/2023] Open
Abstract
Prostate cancer (PC) is one of the most common malignancies among men and is the second leading cause of cancer death. PC immunotherapy has taken relatively successful steps in recent years, and these treatments are still being developed and tested. Evidence suggests that immunotherapy using cytokines as essential mediators in the immune system may help treat cancer. It has been shown that cytokines play an important role in anti-tumor defense. On the other hand, other cytokines can also favor the tumor and suppress anti-tumor responses. Moreover, the dose of cytokine in cancer cytokine-based immunotherapy, as well as the side effects of high doses, can also affect the outcomes of treatment. Cytokines can also be determinative in the outcome of other immunotherapy methods used in PC. In this review, the role of cytokines in the pathogenesis of cancer and their impacts on the main types of immunotherapies in the treatment of PC are discussed.
Collapse
Affiliation(s)
- Chenyu Mao
- Department of Medical Oncology Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yongfeng Ding
- Department of Medical Oncology Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Nong Xu
- Department of Medical Oncology Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
49
|
Liu G, Zhang Q, Liu G, Li D, Zhang L, Gu Z, Tian H, Zhang Y, Tian X. Disruption of adenosine 2A receptor improves the anti-tumor function of anti-mesothelin CAR T cells both in vitro and in vivo. Exp Cell Res 2021; 409:112886. [PMID: 34673000 DOI: 10.1016/j.yexcr.2021.112886] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/11/2021] [Accepted: 10/14/2021] [Indexed: 12/30/2022]
Abstract
Chimeric antigen receptor (CAR) T cells have been successfully used for the treatment of hematological malignancies including acute and chronic lymphoblastic leukemia. However, results of CAR T cell projects in solid tumors have been less impressive to date, partly because of immunosuppressive tumor microenvironment (TME). It is widely known that high adenosine production is an important factor causing tumor-induced immunosuppression in TME, and adenosine mediates the suppression of anti-tumor T cell responses via binding and signaling through adenosine 2a receptor (A2aR). Previous studies have shown that adenosine generated by cancer cells significantly inhibits T cell anti-tumor activity through binding and then activating adenosine 2A receptors (A2aRs) of T cells. Based on the previous work, in our study, we evaluated whether A2aR disruption by shRNA could enhance the anti-tumor function of anti-mesothelin (MSLN) CAR T cells both in vitro and in vivo. For this goal above, we used MSLN-positive human ovarian serous carcinoma cells (SKOV3) and human colon cancer cells (HCT116) as target cancer cells while MSLN-negative human ovarian cancer cells (ES2) as non-target cancer cells. We observed that targeting cell-intrinsic A2aR through shRNA overexpression caused significant A2aR disruption in CAR T cells and profoundly increased CAR T cell efficacy in both CAR T cell cytokine production and cytotoxicity towards MSLN-positive cancer cells in vitro. More importantly, in SKOV3 xenograft mouse models, anti-MSLN CAR-T cells significantly reduced the tumor burden compared with non-transduced T cells, and the anti-tumor activity of A2aR-disrupted anti-MSLN CAR-T cells was stronger than that of wild-type anti-MSLN CAR-T cells. Altogether, our study showed enhanced anti-tumor efficacy caused by shRNA-mediated A2aR disruption in anti-MSLN CAR T cells both in vitro and in vivo, which proved that shRNA-mediated modification of gene expression might be an excellent strategy for improving CAR T cell function in immunosuppressive tumor microenvironment (TME) and could potentially improve the outcome of treatment in clinical trials.
Collapse
Affiliation(s)
- Guodi Liu
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231, China
| | - Qian Zhang
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231, China
| | - Guoping Liu
- Department of General Surgery, Changhai Hospital, Shanghai, 200433, China
| | - Dehua Li
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231, China
| | - Linsong Zhang
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231, China
| | - Zhangjie Gu
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231, China
| | - Huixin Tian
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231, China
| | - Yong Zhang
- Department of Pathology, Tumor Hospital of China Medical University and Liao Ning Cancer Hospital and Institute, Shenyang, 110042, China.
| | - Xiaoli Tian
- Shanghai Yihao Biological Technology Co., Ltd, Shanghai, 200231, China.
| |
Collapse
|
50
|
Magagna I, Gourdin N, Kieffer Y, Licaj M, Mhaidly R, Andre P, Morel A, Vincent-Salomon A, Paturel C, Mechta-Grigoriou F. CD73-Mediated Immunosuppression Is Linked to a Specific Fibroblast Population That Paves the Way for New Therapy in Breast Cancer. Cancers (Basel) 2021; 13:cancers13235878. [PMID: 34884993 PMCID: PMC8657241 DOI: 10.3390/cancers13235878] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/14/2021] [Accepted: 11/19/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAF) are heterogeneous with multiple functions in breast cancer. Recently, we identified a specific CAF subpopulation (referred to as CAF-S1), which promotes immunosuppression and immunotherapy resistance. METHODS AND RESULTS Here, by studying a large collection of human samples, we highlight the key function of CD73/NT5E in CAF-S1-mediated immunosuppression in breast cancer. We first reveal that CD73 protein level specifically accumulates in CAF-S1 in breast cancer patients. Interestingly, infiltration of regulatory T lymphocytes (Tregs) is significantly correlated with CD73 expression in stroma but not in epithelium, indicating that CD73 contributes to immunosuppression when expressed in CAF-S1 and not in tumor cells. By performing functional assays based on relevant systems using primary CAF-S1 isolated from patients, we demonstrate that CAF-S1 increase the content in both PD-1+ and CTLA-4+ Tregs. Importantly, the use of a blocking anti-CD73 antibody on CAF-S1 reduces CAF-S1-mediated immunosuppression by preventing expression of these immune checkpoints on Tregs. CONCLUSIONS Our data support the potential clinical benefit of using both anti-CD73 and immune-checkpoint inhibitors in breast cancer patients for inhibiting CAF-S1-mediated immunosuppression and enhancing anti-tumor immune response.
Collapse
Affiliation(s)
- Ilaria Magagna
- Equipe labellisée Ligue Nationale Contre le Cancer, Stress and Cancer Laboratory, Institut Curie, PSL Research University, 26, rue d’Ulm, 75005 Paris, France; (I.M.); (Y.K.); (M.L.); (R.M.)
- Inserm, U830, 75005 Paris, France
- Innate Pharma, 117 Avenue de Luminy BP 30191, 13276 Marseille, France; (N.G.); (P.A.); (A.M.); (C.P.)
| | - Nicolas Gourdin
- Innate Pharma, 117 Avenue de Luminy BP 30191, 13276 Marseille, France; (N.G.); (P.A.); (A.M.); (C.P.)
| | - Yann Kieffer
- Equipe labellisée Ligue Nationale Contre le Cancer, Stress and Cancer Laboratory, Institut Curie, PSL Research University, 26, rue d’Ulm, 75005 Paris, France; (I.M.); (Y.K.); (M.L.); (R.M.)
- Inserm, U830, 75005 Paris, France
| | - Monika Licaj
- Equipe labellisée Ligue Nationale Contre le Cancer, Stress and Cancer Laboratory, Institut Curie, PSL Research University, 26, rue d’Ulm, 75005 Paris, France; (I.M.); (Y.K.); (M.L.); (R.M.)
- Inserm, U830, 75005 Paris, France
| | - Rana Mhaidly
- Equipe labellisée Ligue Nationale Contre le Cancer, Stress and Cancer Laboratory, Institut Curie, PSL Research University, 26, rue d’Ulm, 75005 Paris, France; (I.M.); (Y.K.); (M.L.); (R.M.)
- Inserm, U830, 75005 Paris, France
| | - Pascale Andre
- Innate Pharma, 117 Avenue de Luminy BP 30191, 13276 Marseille, France; (N.G.); (P.A.); (A.M.); (C.P.)
| | - Ariane Morel
- Innate Pharma, 117 Avenue de Luminy BP 30191, 13276 Marseille, France; (N.G.); (P.A.); (A.M.); (C.P.)
| | - Anne Vincent-Salomon
- Hospital Group, Department of Diagnostic and Theranostic Medicine, Institut Curie, 75005 Paris, France;
| | - Carine Paturel
- Innate Pharma, 117 Avenue de Luminy BP 30191, 13276 Marseille, France; (N.G.); (P.A.); (A.M.); (C.P.)
| | - Fatima Mechta-Grigoriou
- Equipe labellisée Ligue Nationale Contre le Cancer, Stress and Cancer Laboratory, Institut Curie, PSL Research University, 26, rue d’Ulm, 75005 Paris, France; (I.M.); (Y.K.); (M.L.); (R.M.)
- Inserm, U830, 75005 Paris, France
- Correspondence: ; Tel.: +33-(0)1-56-24-66-53; Fax: +33-(0)1-56-24-66-50
| |
Collapse
|