1
|
Farhangnia P, Khorramdelazad H, Nickho H, Delbandi AA. Current and future immunotherapeutic approaches in pancreatic cancer treatment. J Hematol Oncol 2024; 17:40. [PMID: 38835055 DOI: 10.1186/s13045-024-01561-6] [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: 04/09/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
Pancreatic cancer is a major cause of cancer-related death, but despondently, the outlook and prognosis for this resistant type of tumor have remained grim for a long time. Currently, it is extremely challenging to prevent or detect it early enough for effective treatment because patients rarely exhibit symptoms and there are no reliable indicators for detection. Most patients have advanced or spreading cancer that is difficult to treat, and treatments like chemotherapy and radiotherapy can only slightly prolong their life by a few months. Immunotherapy has revolutionized the treatment of pancreatic cancer, yet its effectiveness is limited by the tumor's immunosuppressive and hard-to-reach microenvironment. First, this article explains the immunosuppressive microenvironment of pancreatic cancer and highlights a wide range of immunotherapy options, including therapies involving oncolytic viruses, modified T cells (T-cell receptor [TCR]-engineered and chimeric antigen receptor [CAR] T-cell therapy), CAR natural killer cell therapy, cytokine-induced killer cells, immune checkpoint inhibitors, immunomodulators, cancer vaccines, and strategies targeting myeloid cells in the context of contemporary knowledge and future trends. Lastly, it discusses the main challenges ahead of pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hamid Nickho
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Cao Z, Guan M, Cheng C, Wang F, Jing Y, Zhang K, Jiao J, Ruan L, Chen Z. KIF20B and MET, hub genes of DIAPHs, predict poor prognosis and promote pancreatic cancer progression. Pathol Res Pract 2024; 254:155046. [PMID: 38266456 DOI: 10.1016/j.prp.2023.155046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND The DIAPHs (DIAPH1, DIAPH2, and DIAPH3) are members of the diaphanous subfamily of the formin family. KIF20B and MET, hub genes of DIAPHs, play crucial roles in cytoskeletal remodeling, cell migration, and adhesion. However, their combined prognostic and treatment value in pancreatic adenocarcinoma (PC) warrants further investigation. METHODS Multiomics analysis tools were used to comprehensively assess the genomic expression and prognostic value of KIF20B and MET in PC. Immune cell infiltration, functional enrichment, single-cell RNA-seq (scRNA) analysis, potential therapeutic drugs, and nomograms were established and analyzed. CCK-8 levels, transwell assay, Co-IP assay, mass spectrometry, and western blotting were performed to assess the role of KIF20B and MET as modulators of β-catenin and Lactate Dehydrogenase A (LDHA) in vitro. Xenograft tumor models were used to evaluate the anti-tumor effects in vivo. RESULTS DIAPHs, KIF20B, and MET were overexpressed and functioned as poor prognostic markers of PC. Immunoinfiltration analysis revealed that pDC and NK cells were enriched with low expression levels of KIF20B and MET, whereas Th2 cells were enriched with high expression levels of these two genes. The copy number variations (CNVs) in KIF20B and MET were positively correlated with B cell and CD4 + T cell infiltration. Immunological checkpoints NT5E and CD44 were positively correlated with KIF20B and MET expression. Moreover, the nomogram constructed based on KIF20B and MET demonstrated predictive value for overall survival. scRNA-Seq analysis indicated that KIF20B and MET were enriched in endothelial, malignant, B, T, and CD8 + T cells, which correlated with glycolysis and the epithelial-mesenchymal transition (EMT). The interactions of KIF20B and MET with β-catenin and LDHA were verified by Co-IP assay and mass spectrometry. Knockdown of KIF20B and MET downregulates β-catenin and LDHA in vitro. Furthermore, dual knockdown of KIF20B and MET exhibited a synergistic suppressive effect on PC progression in vitro and in vivo. CONCLUSION DIAPHs, KIF20B, and MET are promising candidates for the prognosis and treatment of PC. More importantly, downregulation of KIF20B and MET inhibited pancreatic cancer progression by regulating LDHA and EMT.
Collapse
Affiliation(s)
- Zhangqi Cao
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Mingwei Guan
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chienshan Cheng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fengjiao Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yanhua Jing
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ke Zhang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Juying Jiao
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Linjie Ruan
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhen Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
3
|
Chen Q, Yin H, He J, Xie Y, Wang W, Xu H, Zhang L, Shi C, Yu J, Wu W, Liu L, Pu N, Lou W. Tumor Microenvironment Responsive CD8 + T Cells and Myeloid-Derived Suppressor Cells to Trigger CD73 Inhibitor AB680-Based Synergistic Therapy for Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302498. [PMID: 37867243 PMCID: PMC10667825 DOI: 10.1002/advs.202302498] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/20/2023] [Indexed: 10/24/2023]
Abstract
CD73 plays a critical role in the pathogenesis and immune escape in pancreatic ductal adenocarcinoma (PDAC). AB680, an exceptionally potent and selective inhibitor of CD73, is administered in an early clinical trial, in conjunction with gemcitabine and anti-PD-1 therapy, for the treatment of PDAC. Nevertheless, the specific therapeutic efficacy and immunoregulation within the microenvironment of AB680 monotherapy in PDAC have yet to be fully elucidated. In this study, AB680 exhibits a significant effect in augmenting the infiltration of responsive CD8+ T cells and prolongs the survival in both subcutaneous and orthotopic murine PDAC models. In parallel, it also facilitates chemotaxis of myeloid-derived suppressor cells (MDSCs) by tumor-derived CXCL5 in an AMP-dependent manner, which may potentially contribute to enhanced immunosuppression. The concurrent administration of AB680 and PD-1 blockade, rather than gemcitabine, synergistically restrain tumor growth. Notably, gemcitabine weakened the efficacy of AB680, which is dependent on CD8+ T cells. Finally, the supplementation of a CXCR2 inhibitor is validated to further enhance the therapeutic efficacy when combined with AB680 plus PD-1 inhibitor. These findings systematically demonstrate the efficacy and immunoregulatory mechanism of AB680, providing a novel, efficient, and promising immunotherapeutic combination strategy for PDAC.
Collapse
Affiliation(s)
- Qiangda Chen
- Department of Pancreatic SurgeryCancer CenterDepartment of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Hanlin Yin
- Department of Pancreatic SurgeryCancer CenterDepartment of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Junyi He
- Department of Pancreatic SurgeryCancer CenterDepartment of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Yuqi Xie
- Department of Pancreatic SurgeryCancer CenterDepartment of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Wenquan Wang
- Department of Pancreatic SurgeryCancer CenterDepartment of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Huaxiang Xu
- Department of Pancreatic SurgeryCancer CenterDepartment of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Lei Zhang
- Department of Pancreatic SurgeryCancer CenterDepartment of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Chenye Shi
- Department of Pancreatic SurgeryCancer CenterDepartment of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Jun Yu
- Departments of Medicine and OncologyJohns Hopkins University School of MedicineBaltimoreMD21287USA
| | - Wenchuan Wu
- Department of Pancreatic SurgeryCancer CenterDepartment of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Liang Liu
- Department of Pancreatic SurgeryCancer CenterDepartment of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Ning Pu
- Department of Pancreatic SurgeryCancer CenterDepartment of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Wenhui Lou
- Department of Pancreatic SurgeryCancer CenterDepartment of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| |
Collapse
|
4
|
Yu W, Li M, Xia J. Identification of immunogenic cell death‑related prognostic signatures in pancreatic cancer. Oncol Lett 2023; 26:473. [PMID: 37809045 PMCID: PMC10551861 DOI: 10.3892/ol.2023.14061] [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: 12/12/2022] [Accepted: 08/22/2023] [Indexed: 10/10/2023] Open
Abstract
Targeting immunogenic cell death (ICD) may enable the response of pancreatic cancer to immune checkpoint inhibitors (ICIs). The aim of the present study was to elucidate the role of ICD-related genes in pancreatic cancer. Utilizing the k-means method, consensus clustering was employed to effectively group patients with pancreatic cancer. Subsequently, a set of differentially expressed genes was identified between the two subtypes related to ICD, facilitating the execution of a comprehensive enrichment analysis. Furthermore, the construction of an ICD-related prognostic signature (IRPS) was accomplished through LASSO Cox regression, thereby enabling the assessment of responses to both chemotherapy and immunotherapy. In addition, the biological functionality of 5'-nucleotidase ecto (NT5E) was elucidated through experimental investigations. Patients characterized as the ICD high subtype experienced a comparatively shorter overall survival. This subtype exhibited a noteworthy correlation with HLA families and immune checkpoint molecules, underscoring its immunological significance. Subsequently, patients with elevated IRPS risk scores displayed resistance towards immunotherapy interventions. Of note, synergistic downregulation of NT5E in combination with Gemcitabine was observed to significantly induce tumor cell apoptosis, emphasizing its potential therapeutic value. Leveraging ICD-related genes, a novel classification system was meticulously devised to comprehensively evaluate both the clinical outcomes and therapeutic responses of patients diagnosed with pancreatic cancer.
Collapse
Affiliation(s)
- Wenjing Yu
- Department of Laboratory Medicine, The 13th People's Hospital of Chongqing, Chongqing 400053, P.R. China
| | - Mei Li
- Department of Laboratory Medicine, The 13th People's Hospital of Chongqing, Chongqing 400053, P.R. China
| | - Jing Xia
- Department of Laboratory Medicine, The 13th People's Hospital of Chongqing, Chongqing 400053, P.R. China
| |
Collapse
|
5
|
Chen X, Li Y, Zhou Z, Zhang Y, Chang L, Gao X, Li Q, Luo H, Westover KD, Zhu J, Wei X. Dynamic ultrasound molecular-targeted imaging of senescence in evaluation of lapatinib resistance in HER2-positive breast cancer. Cancer Med 2023; 12:19904-19920. [PMID: 37792675 PMCID: PMC10587953 DOI: 10.1002/cam4.6607] [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: 03/24/2023] [Revised: 07/21/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Prolonged treatment of HER2+ breast cancer with lapatinib (LAP) causes cellular senescence and acquired drug resistance, which often associating with poor prognosis for patients. We aim to explore the correlation between cellular senescence and LAP resistance in HER2+ breast cancer, screen for molecular marker of reversible senescence, and construct targeted nanobubbles for ultrasound molecular imaging to dynamically evaluate LAP resistance. METHODS AND RESULTS In this study, we established a new cellular model of reversible cellular senescence using LAP and HER2+ breast cancer cells and found that reversible senescence contributed to LAP resistance in HER2+ breast cancer. Then, we identified ecto-5'-nucleotidase (NT5E) as a marker of reversible senescence in HER2+ breast cancer. Based on this, we constructed NT5E-targeted nanobubbles (NT5E-FITC-NBs) as a new molecular imaging modality which could both target reversible senescent cells and be used for ultrasound imaging. NT5E-FITC-NBs showed excellent physical and imaging characteristics. As an ultrasound contrast agent, NT5E-FITC-NBs could accurately identify reversible senescent cells both in vitro and in vivo. CONCLUSIONS Our data demonstrate that cellular senescence-based ultrasound-targeted imaging can identify reversible senescence and evaluate LAP resistance effectively in HER2+ breast cancer cells, which has the potential to improve cancer treatment outcomes by altering therapeutic strategies ahead of aggressive recurrences.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Department of Diagnostic and Therapeutic UltrasonographyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjinChina
- Department of UltrasoundTianjin HospitalTianjinChina
| | - Ying Li
- Breast Cancer CenterTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Zhiwei Zhou
- Department of Radiation Oncology and BiochemistryUniversity of Texas Southwestern Medical CenterTexasDallasUSA
| | - Yanqiu Zhang
- Department of Diagnostic and Therapeutic UltrasonographyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Luchen Chang
- Department of Diagnostic and Therapeutic UltrasonographyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Xiujun Gao
- School of Biomedical Engineering and Technology, Tianjin Medical UniversityTianjinChina
| | - Qing Li
- Cancer CenterDaping Hospital, Third Military Medical UniversityChongqingChina
| | - Hao Luo
- Cancer CenterDaping Hospital, Third Military Medical UniversityChongqingChina
| | - Kenneth D. Westover
- Department of Radiation Oncology and BiochemistryUniversity of Texas Southwestern Medical CenterTexasDallasUSA
| | - Jialin Zhu
- Department of Diagnostic and Therapeutic UltrasonographyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Xi Wei
- Department of Diagnostic and Therapeutic UltrasonographyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjinChina
| |
Collapse
|
6
|
Guo X, Yu S, Ren X, Li L. Immune checkpoints represent a promising breakthrough in targeted therapy and prognosis of myelodysplastic syndrome. Heliyon 2023; 9:e19222. [PMID: 37810157 PMCID: PMC10558320 DOI: 10.1016/j.heliyon.2023.e19222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 06/27/2023] [Accepted: 08/16/2023] [Indexed: 10/10/2023] Open
Abstract
Myelodysplastic syndrome (MDS) is a hematological malignancy of undetermined etiology, possibly linked to chromosomal structural alterations, genetic mutations, presentation and carcinogenicity of variant antigens on cell surface, and the generation of pro-inflammatory microenvironment in the bone marrow. Current drugs are unable to cure this disease, and therefore, decreasing the survival and proliferation of malignant cells to delay disease progression and extend the survival time of patients becomes the primary approach to management. In recent years, the immune system has received increasing attention for its potential role in the occurrence and development of MDS, leading to the emergence of immunoregulation as a viable treatment option. The current review provides a brief overview of pathogenesis of MDS and current treatment principles. In the meantime, the significance of immune proteins in treatment and prognosis of MDS is also discussed.
Collapse
Affiliation(s)
- Xinyu Guo
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, Heping District 154 Anshan Road, Tianjin, China
| | - Shunjie Yu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, Heping District 154 Anshan Road, Tianjin, China
| | - Xiaotong Ren
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, Heping District 154 Anshan Road, Tianjin, China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, Heping District 154 Anshan Road, Tianjin, China
| |
Collapse
|
7
|
Posta M, Győrffy B. Analysis of a large cohort of pancreatic cancer transcriptomic profiles to reveal the strongest prognostic factors. Clin Transl Sci 2023; 16:1479-1491. [PMID: 37260110 PMCID: PMC10432876 DOI: 10.1111/cts.13563] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/04/2023] [Accepted: 05/11/2023] [Indexed: 06/02/2023] Open
Abstract
Pancreatic adenocarcinoma remains a leading cause of cancer-related deaths. In order to develop appropriate therapeutic and prognostic tools, a comprehensive mapping of the tumor's molecular abnormalities is essential. Here, our aim was to integrate available transcriptomic data to uncover genes whose elevated expression is simultaneously linked to cancer pathogenesis and inferior survival. A comprehensive search was performed in GEO to identify clinical studies with transcriptome-level gene expression data of pancreatic carcinoma with overall survival data and normal pancreatic tissues. After quantile normalization, the entire database was used to identify genes with altered expression. Cox proportional hazard regression was employed to uncover genes most strongly correlated with survival with a Bonferroni corrected p < 0.01. Perturbed biological processes and molecular pathways were identified to enable the understanding of underlying processes. A total of 16 available datasets were combined. The aggregated database comprised data of 1640 samples for 20,443 genes. When comparing with normal pancreatic tissues, a total of 2612 upregulated and 1977 downregulated genes were uncovered in pancreatic carcinoma. Among these, we found 24 genes with higher expression which significantly correlated with overall survival length also. The most significant genes were ANXA8, FAM83A, KRT6A, MET, MUC16, NT5E, and SLC2A1. These genes remained significant after a multivariate analysis also including grade and stage. Here, we assembled a large-scale database of pancreatic carcinoma samples and used this cohort to identify carcinoma-specific genes linked to altered survival outcomes. As our analysis focused on genes with higher expression, these could serve as future therapy targets.
Collapse
Affiliation(s)
- Máté Posta
- Károly Rácz Doctoral School of Clinical MedicineSemmelweis UniversityBudapestHungary
- Oncology Biomarker Research Group, Institute of EnzymologyResearch Centre for Natural SciencesBudapestHungary
- Systems Biology of Reproduction Research Group, Institute of EnzymologyResearch Centre for Natural SciencesBudapestHungary
| | - Balázs Győrffy
- Department of Bioinformatics and Department of PediatricsSemmelweis UniversityBudapestHungary
| |
Collapse
|
8
|
Effer B, Perez I, Ulloa D, Mayer C, Muñoz F, Bustos D, Rojas C, Manterola C, Vergara-Gómez L, Dappolonnio C, Weber H, Leal P. Therapeutic Targets of Monoclonal Antibodies Used in the Treatment of Cancer: Current and Emerging. Biomedicines 2023; 11:2086. [PMID: 37509725 PMCID: PMC10377242 DOI: 10.3390/biomedicines11072086] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer is one of the leading global causes of death and disease, and treatment options are constantly evolving. In this sense, the use of monoclonal antibodies (mAbs) in immunotherapy has been considered a fundamental aspect of modern cancer therapy. In order to avoid collateral damage, it is indispensable to identify specific molecular targets or biomarkers of therapy and/or diagnosis (theragnostic) when designing an appropriate immunotherapeutic regimen for any type of cancer. Furthermore, it is important to understand the currently employed mAbs in immunotherapy and their mechanisms of action in combating cancer. To achieve this, a comprehensive understanding of the biology of cancer cell antigens, domains, and functions is necessary, including both those presently utilized and those emerging as potential targets for the design of new mAbs in cancer treatment. This review aims to provide a description of the therapeutic targets utilized in cancer immunotherapy over the past 5 years, as well as emerging targets that hold promise as potential therapeutic options in the application of mAbs for immunotherapy. Additionally, the review explores the mechanisms of actin of the currently employed mAbs in immunotherapy.
Collapse
Affiliation(s)
- Brian Effer
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Isabela Perez
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Daniel Ulloa
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Carolyn Mayer
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Francisca Muñoz
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Diego Bustos
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Claudio Rojas
- Programa de Doctorado en Ciencias Médicas, Universidad de la Frontera, Temuco 4811230, Chile
- Centro de Estudios Morfológicos y Quirúrgicos de La, Universidad de La Frontera, Temuco 4811230, Chile
| | - Carlos Manterola
- Programa de Doctorado en Ciencias Médicas, Universidad de la Frontera, Temuco 4811230, Chile
- Centro de Estudios Morfológicos y Quirúrgicos de La, Universidad de La Frontera, Temuco 4811230, Chile
| | - Luis Vergara-Gómez
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Camila Dappolonnio
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Helga Weber
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Pamela Leal
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
- Department of Agricultural Sciences and Natural Resources, Faculty of Agricultural and Forestry Science, Universidad de La Frontera, Temuco 4810296, Chile
| |
Collapse
|
9
|
Petruk N, Siddiqui A, Tadayon S, Määttä J, Mattila PK, Jukkola A, Sandholm J, Selander KS. CD73 regulates zoledronate-induced lymphocyte infiltration in triple-negative breast cancer tumors and lung metastases. Front Immunol 2023; 14:1179022. [PMID: 37533856 PMCID: PMC10390692 DOI: 10.3389/fimmu.2023.1179022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/23/2023] [Indexed: 08/04/2023] Open
Abstract
Introduction Bisphosphonates (BPs) are bone-protecting osteoclast inhibitors, typically used in the treatment of osteoporosis and skeletal complications of malignancies. When given in the adjuvant setting, these drugs may also prevent relapses and prolong overall survival in early breast cancer (EBC), specifically among postmenopausal patients. Because of these findings, adjuvant nitrogen-containing BPs (N-BPs), such as zoledronate (ZOL), are now the standard of care for high-risk EBC patients, but there are no benefit-associated biomarkers, and the efficacy remains low. BPs have been demonstrated to possess anti-tumor activities, but the mechanisms by which they provide the beneficial effects in EBC are not known. Methods We used stably transfected 4T1 breast cancer cells together with suppression of CD73 (sh-CD73) or control cells (sh-NT). We compared ZOL effects on tumor growth and infiltrating lymphocytes (TILs) into tumors and lung metastases using two mouse models. B cell depletion was performed using anti-CD20 antibody. Results Sh-CD73 4T1 cells were significantly more sensitive to the growth inhibitory effects of n-BPs in vitro. However, while ZOL-induced growth inhibition was similar between the tumor groups in vivo, ZOL enhanced B and T lymphocyte infiltration into the orthotopic tumors with down-regulated CD73. A similar trend was detected in lung metastases. ZOL-induced tumor growth inhibition was found to be augmented with B cell depletion in sh-NT tumors, but not in sh-CD73 tumors. As an internal control, ZOL effects on bone were similar in mice bearing both tumor groups. Discussion Taken together, these results indicate that ZOL modifies TILs in breast cancer, both in primary tumors and metastases. Our results further demonstrate that B cells may counteract the growth inhibitory effects of ZOL. However, all ZOL-induced TIL effects may be influenced by immunomodulatory characteristics of the tumor.
Collapse
Affiliation(s)
- Nataliia Petruk
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Arafat Siddiqui
- Institute of Biomedicine, University of Turku, Turku, Finland
- Western Cancer Centre FICAN West, Turku, Finland
| | - Sina Tadayon
- MediCity Research Laboratory, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Jorma Määttä
- Institute of Biomedicine, University of Turku, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Pieta K. Mattila
- Institute of Biomedicine, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Arja Jukkola
- Department of Oncology, Tampere University Hospital, Tays Cancer Center, Tampere, Finland
| | - Jouko Sandholm
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Katri S. Selander
- Department of Oncology and Radiation Therapy, Oulu University Hospital, Oulu, Finland
- Cancer Research and Translational Medicine Research Unit, University of Oulu, Oulu, Finland
| |
Collapse
|
10
|
Zhang M, Dai X, Xiang Y, Xie L, Sun M, Shi J. Advances in CD73 inhibitors for immunotherapy: Antibodies, synthetic small molecule compounds, and natural compounds. Eur J Med Chem 2023; 258:115546. [PMID: 37302340 DOI: 10.1016/j.ejmech.2023.115546] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/20/2023] [Accepted: 06/04/2023] [Indexed: 06/13/2023]
Abstract
Tumors, a disease with a high mortality rate worldwide, have become a serious threat to human health. Exonucleotide-5'-nucleotidase (CD73) is an emerging target for tumor therapy. Its inhibition can significantly reduce adenosine levels in the tumor microenvironment. It has a better therapeutic effect on adenosine-induced immunosuppression. In the immune response, extracellular ATP exerts immune efficacy by activating T cells. However, dead tumor cells release excess ATP, overexpress CD39 and CD73 on the cell membrane and catabolize this ATP to adenosine. This leads to further immunosuppression. There are a number of inhibitors of CD73 currently under investigation. These include antibodies, synthetic small molecule inhibitors and a number of natural compounds with prominent roles in the anti-tumor field. However, only a small proportion of the CD73 inhibitors studied to date have successfully reached the clinical stage. Therefore, effective and safe inhibition of CD73 in oncology therapy still holds great therapeutic potential. This review summarizes the currently reported CD73 inhibitors, describes their inhibitory effects and pharmacological mechanisms, and provides a brief review of them. It aims to provide more information for further research and development of CD73 inhibitors.
Collapse
Affiliation(s)
- Mingxue Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Xiaoqin Dai
- Department of Traditional Chinese Medicine, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan Province, China
| | - Yu Xiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Linshen Xie
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
| | - Minghan Sun
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
11
|
Xu S, Wang C, Yang L, Wu J, Li M, Xiao P, Xu Z, Xu Y, Wang K. Targeting immune checkpoints on tumor-associated macrophages in tumor immunotherapy. Front Immunol 2023; 14:1199631. [PMID: 37313405 PMCID: PMC10258331 DOI: 10.3389/fimmu.2023.1199631] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/16/2023] [Indexed: 06/15/2023] Open
Abstract
Unprecedented breakthroughs have been made in cancer immunotherapy in recent years. Particularly immune checkpoint inhibitors have fostered hope for patients with cancer. However, immunotherapy still exhibits certain limitations, such as a low response rate, limited efficacy in certain populations, and adverse events in certain tumors. Therefore, exploring strategies that can improve clinical response rates in patients is crucial. Tumor-associated macrophages (TAMs) are the predominant immune cells that infiltrate the tumor microenvironment and express a variety of immune checkpoints that impact immune functions. Mounting evidence indicates that immune checkpoints in TAMs are closely associated with the prognosis of patients with tumors receiving immunotherapy. This review centers on the regulatory mechanisms governing immune checkpoint expression in macrophages and strategies aimed at improving immune checkpoint therapies. Our review provides insights into potential therapeutic targets to improve the efficacy of immune checkpoint blockade and key clues to developing novel tumor immunotherapies.
Collapse
Affiliation(s)
- Shumin Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Chenyang Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Lingge Yang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Jiaji Wu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Mengshu Li
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Peng Xiao
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhiyong Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Yun Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| |
Collapse
|
12
|
Strickland LN, Faraoni EY, Ruan W, Yuan X, Eltzschig HK, Bailey-Lundberg JM. The resurgence of the Adora2b receptor as an immunotherapeutic target in pancreatic cancer. Front Immunol 2023; 14:1163585. [PMID: 37187740 PMCID: PMC10175829 DOI: 10.3389/fimmu.2023.1163585] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense desmoplastic stroma that impedes drug delivery, reduces parenchymal blood flow, and suppresses the anti-tumor immune response. The extracellular matrix and abundance of stromal cells result in severe hypoxia within the tumor microenvironment (TME), and emerging publications evaluating PDAC tumorigenesis have shown the adenosine signaling pathway promotes an immunosuppressive TME and contributes to the overall low survival rate. Hypoxia increases many elements of the adenosine signaling pathway, resulting in higher adenosine levels in the TME, further contributing to immune suppression. Extracellular adenosine signals through 4 adenosine receptors (Adora1, Adora2a, Adora2b, Adora3). Of the 4 receptors, Adora2b has the lowest affinity for adenosine and thus, has important consequences when stimulated by adenosine binding in the hypoxic TME. We and others have shown that Adora2b is present in normal pancreas tissue, and in injured or diseased pancreatic tissue, Adora2b levels are significantly elevated. The Adora2b receptor is present on many immune cells, including macrophages, dendritic cells, natural killer cells, natural killer T cells, γδ T cells, B cells, T cells, CD4+ T cells, and CD8+ T cells. In these immune cell types, adenosine signaling through Adora2b can reduce the adaptive anti-tumor response, augmenting immune suppression, or may contribute to transformation and changes in fibrosis, perineural invasion, or the vasculature by binding the Adora2b receptor on neoplastic epithelial cells, cancer-associated fibroblasts, blood vessels, lymphatic vessels, and nerves. In this review, we discuss the mechanistic consequences of Adora2b activation on cell types in the tumor microenvironment. As the cell-autonomous role of adenosine signaling through Adora2b has not been comprehensively studied in pancreatic cancer cells, we will also discuss published data from other malignancies to infer emerging therapeutic considerations for targeting the Adora2b adenosine receptor to reduce the proliferative, invasive, and metastatic potential of PDAC cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Jennifer M. Bailey-Lundberg
- Department of Anesthesiology, Critical Care, and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
13
|
Bertoni APS, Valandro C, Brasil RÁ, Zeiser FA, Wink MR, Furlanetto TW, da Costa CA. NT5E DNA methylation in papillary thyroid cancer: Novel opportunities for precision oncology. Mol Cell Endocrinol 2023; 570:111915. [PMID: 37059175 DOI: 10.1016/j.mce.2023.111915] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/01/2023] [Accepted: 03/23/2023] [Indexed: 04/16/2023]
Abstract
The ectoenzyme CD73, encoded by the NT5E gene, has emerged as a potential prognostic and therapeutic marker for papillary thyroid carcinoma (PTC), which has increased in incidence in recent decades. Here, from The Cancer Genome Atlas Thyroid Cancer (TCGA-THCA) database, we extracted and combined clinical features, levels of NT5E mRNA, and DNA methylation of PTC samples and performed multivariate and random forest analyses to evaluate the prognostic relevance and the potential of discriminating between adjacent non-malignant and thyroid tumor samples. As a result, we revealed that lower levels of methylation at the cg23172664 site were independently associated with BRAF-like phenotype (p = 0.002), age over 55 years (p = 0.012), presence of capsule invasion (p = 0.007) and presence of positive lymph node metastasis (LNM) (p = 0.04). The methylation levels of cg27297263 and cg23172664 sites showed significant and inversely correlations with levels of NT5E mRNA expression (r = -0.528 and r = -0.660, respectively), and their combination was able to discriminate between adjacent non-malignant and tumor samples with a precision of 96%-97% and 84%-85%, respectively. These data suggest that combining cg23172664 and cg27297263 sites may bring new insights to reveal new subsets of patients with papillary thyroid carcinoma.
Collapse
Affiliation(s)
- Ana Paula Santin Bertoni
- Software Innovation Laboratory - SOFTWARELAB, Applied Computing Graduate Program, Universidade do Vale do Rio dos Sinos (UNISINOS), São Leopoldo, Brazil; Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre-UFCSPA, Porto Alegre, RS, Brazil
| | - Cleiton Valandro
- Software Innovation Laboratory - SOFTWARELAB, Applied Computing Graduate Program, Universidade do Vale do Rio dos Sinos (UNISINOS), São Leopoldo, Brazil
| | - Rafael Ávila Brasil
- Software Innovation Laboratory - SOFTWARELAB, Applied Computing Graduate Program, Universidade do Vale do Rio dos Sinos (UNISINOS), São Leopoldo, Brazil
| | - Felipe André Zeiser
- Software Innovation Laboratory - SOFTWARELAB, Applied Computing Graduate Program, Universidade do Vale do Rio dos Sinos (UNISINOS), São Leopoldo, Brazil
| | - Márcia Rosângela Wink
- Software Innovation Laboratory - SOFTWARELAB, Applied Computing Graduate Program, Universidade do Vale do Rio dos Sinos (UNISINOS), São Leopoldo, Brazil
| | - Tania Weber Furlanetto
- Programa de Pós-Graduação em Medicina: Ciências Médicas, UFRGS, Porto Alegre, RS, Brazil
| | - Cristiano André da Costa
- Software Innovation Laboratory - SOFTWARELAB, Applied Computing Graduate Program, Universidade do Vale do Rio dos Sinos (UNISINOS), São Leopoldo, Brazil.
| |
Collapse
|
14
|
Ma L, Feng Y, Zhou Z. A close look at current γδ T-cell immunotherapy. Front Immunol 2023; 14:1140623. [PMID: 37063836 PMCID: PMC10102511 DOI: 10.3389/fimmu.2023.1140623] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/08/2023] [Indexed: 04/03/2023] Open
Abstract
Owing to their antitumor and major histocompatibility complex (MHC)-independent capacities, γδ T cells have gained popularity in adoptive T-cell immunotherapy in recent years. However, many unknowns still exist regarding γδ T cells, and few clinical data have been collected. Therefore, this review aims to describe all the main features of the applications of γδ T cells and provide a systematic view of current γδ T-cell immunotherapy. Specifically, this review will focus on how γδ T cells performed in treating cancers in clinics, on the γδ T-cell clinical trials that have been conducted to date, and the role of γδ T cells in the pharmaceutical industry.
Collapse
Affiliation(s)
- Ling Ma
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Research and Development Department, Beijing Dingchengtaiyuan (DCTY) Biotech Co., Ltd., Beijing, China
| | - Yanmin Feng
- Research and Development Department, Beijing Dingchengtaiyuan (DCTY) Biotech Co., Ltd., Beijing, China
| | - Zishan Zhou
- Research and Development Department, Beijing Dingchengtaiyuan (DCTY) Biotech Co., Ltd., Beijing, China
- *Correspondence: Zishan Zhou,
| |
Collapse
|
15
|
Sun BY, Yang ZF, Wang ZT, Liu G, Zhou C, Zhou J, Fan J, Gan W, Yi Y, Qiu SJ. Integrative analyses identify CD73 as a prognostic biomarker and immunotherapeutic target in intrahepatic cholangiocarcinoma. World J Surg Oncol 2023; 21:90. [PMID: 36899373 PMCID: PMC9999525 DOI: 10.1186/s12957-023-02970-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND CD73 promotes progression in several malignancies and is considered as a novel immune checkpoint. However, the function of CD73 in intrahepatic cholangiocarcinoma (ICC) remains uncertain. In this study, we aim to investigate the role of CD73 in ICC. METHODS Multi-omics data of 262 ICC patients from the FU-iCCA cohort were analyzed. Two single-cell datasets were downloaded to examine the expression of CD73 at baseline and in response to immunotherapy. Functional experiments were performed to explore the biological functions of CD73 in ICC. The expression of CD73 and HHLA2 and infiltrations of CD8 + , Foxp3 + , CD68 + , and CD163 + immune cells were evaluated by immunohistochemistry in 259 resected ICC samples from Zhongshan Hospital. The prognostic value of CD73 was assessed by Cox regression analysis. RESULTS CD73 correlated with poor prognosis in two ICC cohorts. Single-cell atlas of ICC indicated high expression of CD73 on malignant cells. TP53 and KRAS gene mutations were more frequent in patients with high CD73 expression. CD73 promoted ICC proliferation, migration, invasion, and epithelial-mesenchymal transition. High CD73 expression was associated with a higher ratio of Foxp3 + /CD8 + tumor-infiltrating lymphocytes (TILs) and CD163 + /CD68 + tumor-associated macrophages (TAMs). A positive correlation between CD73 and CD44 was observed, and patients with high CD73 expression showed elevated expression of HHLA2. CD73 expression in malignant cells was significantly upregulated in response to immunotherapy. CONCLUSIONS High expression of CD73 is associated with poor prognosis and a suppressive tumor immune microenvironment in ICC. CD73 could potentially be a novel biomarker for prognosis and immunotherapy in ICC.
Collapse
Affiliation(s)
- Bao-Ye Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Zhang-Fu Yang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Zhu-Tao Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Gao Liu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Cheng Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Wei Gan
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
| | - Yong Yi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
| | - Shuang-Jian Qiu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
16
|
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 2023:10.1007/s00262-023-03416-4. [PMID: 36881132 DOI: 10.1007/s00262-023-03416-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
17
|
Xia C, Yin S, To KKW, Fu L. CD39/CD73/A2AR pathway and cancer immunotherapy. Mol Cancer 2023; 22:44. [PMID: 36859386 PMCID: PMC9979453 DOI: 10.1186/s12943-023-01733-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/26/2023] [Indexed: 03/03/2023] Open
Abstract
Cancer development is closely associated with immunosuppressive tumor microenvironment (TME) that attenuates antitumor immune responses and promotes tumor cell immunologic escape. The sequential conversion of extracellular ATP into adenosine by two important cell-surface ectonucleosidases CD39 and CD73 play critical roles in reshaping an immunosuppressive TME. The accumulated extracellular adenosine mediates its regulatory functions by binding to one of four adenosine receptors (A1R, A2AR, A2BR and A3R). The A2AR elicits its profound immunosuppressive function via regulating cAMP signaling. The increasing evidence suggests that CD39, CD73 and A2AR could be used as novel therapeutic targets for manipulating the antitumor immunity. In recent years, monoclonal antibodies or small molecule inhibitors targeting the CD39/CD73/A2AR pathway have been investigated in clinical trials as single agents or in combination with anti-PD-1/PD-L1 therapies. In this review, we provide an updated summary about the pathophysiological function of the adenosinergic pathway in cancer development, metastasis and drug resistance. The targeting of one or more components of the adenosinergic pathway for cancer therapy and circumvention of immunotherapy resistance are also discussed. Emerging biomarkers that may be used to guide the selection of CD39/CD73/A2AR-targeting treatment strategies for individual cancer patients is also deliberated.
Collapse
Affiliation(s)
- Chenglai Xia
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, 528000, China. .,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 515150, China.
| | - Shuanghong Yin
- grid.284723.80000 0000 8877 7471Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, 528000 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China
| | - Kenneth K. W. To
- grid.10784.3a0000 0004 1937 0482School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
18
|
Liu W, Yu X, Yuan Y, Feng Y, Wu C, Huang C, Xie P, Li S, Li X, Wang Z, Qi L, Chen Y, Shi L, Li MJ, Huang Z, Tang B, Chang A, Hao J. CD73, a Promising Therapeutic Target of Diclofenac, Promotes Metastasis of Pancreatic Cancer through a Nucleotidase Independent Mechanism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206335. [PMID: 36563135 PMCID: PMC9951332 DOI: 10.1002/advs.202206335] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/04/2022] [Indexed: 06/17/2023]
Abstract
CD73, a cell surface-bound nucleotidase, facilitates extracellular adenosine formation by hydrolyzing 5'-AMP to adenosine. Several studies have shown that CD73 plays an essential role in immune escape, cell proliferation and tumor angiogenesis, making it an attractive target for cancer therapies. However, there are limited clinical benefits associated with the mainstream enzymatic inhibitors of CD73, suggesting that the mechanism underlying the role of CD73 in tumor progression is more complex than anticipated, and further investigation is necessary. In this study, CD73 is found to overexpress in the cytoplasm of pancreatic ductal adenocarcinoma (PDAC) cells and promotes metastasis in a nucleotidase-independent manner, which cannot be restrained by the CD73 monoclonal antibodies or small-molecule enzymatic inhibitors. Furthermore, CD73 promotes the metastasis of PDAC by binding to the E3 ligase TRIM21, competing with the Snail for its binding site. Additionally, a CD73 transcriptional inhibitor, diclofenac, a non-steroidal anti-inflammatory drug, is more effective than the CD73 blocking antibody for the treatment of PDAC metastasis. Diclofenac also enhances the therapeutic efficacy of gemcitabine in the spontaneous KPC (LSL-KrasG12D/+ , LSL-Trp53R172H/+ , and Pdx-1-Cre) pancreatic cancer model. Therefore, diclofenac may be an effective anti-CD73 therapy, when used alone or in combination with gemcitabine-based chemotherapy regimen, for metastatic PDAC.
Collapse
Affiliation(s)
- Weishuai Liu
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Xiaozhou Yu
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Yudong Yuan
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Yixing Feng
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Chao Wu
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Chongbiao Huang
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Peng Xie
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Shengnan Li
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Xiaofeng Li
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Ziyang Wang
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Lisha Qi
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Yanan Chen
- School of MedicineNankai UniversityTianjin300071China
| | - Lei Shi
- Tianjin Medical UniversityTianjin300070China
| | | | - Zhiyong Huang
- Tianjin Institute of Industrial Biotechnology Chinese Academy of SciencesTianjin300308China
| | - Bo Tang
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Antao Chang
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Jihui Hao
- Key Laboratory of Cancer Prevention and TherapyNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjin300060China
| |
Collapse
|
19
|
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: 2.0] [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
|
20
|
Fu Z, Chen S, Zhu Y, Zhang D, Xie P, Jiao Q, Chi J, Xu S, Xue Y, Lu X, Song X, Cristofanilli M, Gradishar WJ, Kalinsky K, Yin Y, Zhang B, Wan Y. Proteolytic regulation of CD73 by TRIM21 orchestrates tumor immunogenicity. SCIENCE ADVANCES 2023; 9:eadd6626. [PMID: 36608132 PMCID: PMC9821867 DOI: 10.1126/sciadv.add6626] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 12/06/2022] [Indexed: 05/24/2023]
Abstract
Despite the rapid utilization of immunotherapy, emerging challenges to the current immune checkpoint blockade need to be resolved. Here, we report that elevation of CD73 levels due to its aberrant turnover is correlated with poor prognosis in immune-cold triple-negative breast cancers (TNBCs). We have identified TRIM21 as an E3 ligase that governs CD73 destruction. Disruption of TRIM21 stabilizes CD73 that in turn enhances CD73-catalyzed production of adenosine, resulting in the suppression of CD8+ T cell function. Replacement of lysine 133, 208, 262, and 321 residues by arginine on CD73 attenuated CD73 ubiquitylation and degradation. Diminishing of CD73 ubiquitylation remarkably promotes tumor growth and impedes antitumor immunity. In addition, a TRIM21high/CD73low signature in a subgroup of human breast malignancies was associated with a favorable immune profile. Collectively, our findings uncover a mechanism that governs CD73 proteolysis and point to a new therapeutic strategy by modulating CD73 ubiquitylation.
Collapse
Affiliation(s)
- Ziyi Fu
- Department of Obstetrics and Gynecology, Department of Pharmacology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Siqi Chen
- Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yueming Zhu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, USA
| | - Donghong Zhang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ping Xie
- Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Qiao Jiao
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Junlong Chi
- Department of Obstetrics and Gynecology, Department of Pharmacology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Shipeng Xu
- Department of Obstetrics and Gynecology, Department of Pharmacology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yifan Xue
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xinghua Lu
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xinxin Song
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - William J. Gradishar
- Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kevin Kalinsky
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bin Zhang
- Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yong Wan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
21
|
Jacoberger-Foissac C, Cousineau I, Bareche Y, Allard D, Chrobak P, Allard B, Pommey S, Messaoudi N, McNicoll Y, Soucy G, Koseoglu S, Masia R, Lake AC, Seo H, Eeles CB, Rohatgi N, Robson SC, Turcotte S, Haibe-Kains B, Stagg J. CD73 Inhibits cGAS-STING and Cooperates with CD39 to Promote Pancreatic Cancer. Cancer Immunol Res 2023; 11:56-71. [PMID: 36409930 PMCID: PMC9812927 DOI: 10.1158/2326-6066.cir-22-0260] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/07/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022]
Abstract
The ectonucleotidases CD39 and CD73 catalyze extracellular ATP to immunosuppressive adenosine, and as such, represent potential cancer targets. We investigated biological impacts of CD39 and CD73 in pancreatic ductal adenocarcinoma (PDAC) by studying clinical samples and experimental mouse tumors. Stromal CD39 and tumoral CD73 expression significantly associated with worse survival in human PDAC samples and abolished the favorable prognostic impact associated with the presence of tumor-infiltrating CD8+ T cells. In mouse transplanted KPC tumors, both CD39 and CD73 on myeloid cells, as well as CD73 on tumor cells, promoted polarization of infiltrating myeloid cells towards an M2-like phenotype, which enhanced tumor growth. CD39 on tumor-specific CD8+ T cells and pancreatic stellate cells also suppressed IFNγ production by T cells. Although therapeutic inhibition of CD39 or CD73 alone significantly delayed tumor growth in vivo, targeting of both ectonucleotidases exhibited markedly superior antitumor activity. CD73 expression on human and mouse PDAC tumor cells also protected against DNA damage induced by gemcitabine and irradiation. Accordingly, large-scale pharmacogenomic analyses of human PDAC cell lines revealed significant associations between CD73 expression and gemcitabine chemoresistance. Strikingly, increased DNA damage in CD73-deficient tumor cells associated with activation of the cGAS-STING pathway. Moreover, cGAS expression in mouse KPC tumor cells was required for antitumor activity of the CD73 inhibitor AB680 in vivo. Our study, thus, illuminates molecular mechanisms whereby CD73 and CD39 seemingly cooperate to promote PDAC progression.
Collapse
Affiliation(s)
- Célia Jacoberger-Foissac
- Faculty of Pharmacy, University of Montreal., Cancer Axis, Centre de Recherche Du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada., Institut du Cancer de Montréal
| | - Isabelle Cousineau
- Cancer Axis, Centre de Recherche Du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada., Institut du Cancer de Montréal
| | - Yacine Bareche
- Faculty of Pharmacy, University of Montreal., Cancer Axis, Centre de Recherche Du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada., Institut du Cancer de Montréal
| | - David Allard
- Faculty of Pharmacy, University of Montreal., Cancer Axis, Centre de Recherche Du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada., Institut du Cancer de Montréal
| | - Pavel Chrobak
- Cancer Axis, Centre de Recherche Du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada., Institut du Cancer de Montréal
| | - Bertrand Allard
- Cancer Axis, Centre de Recherche Du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada., Institut du Cancer de Montréal
| | - Sandra Pommey
- Cancer Axis, Centre de Recherche Du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada., Institut du Cancer de Montréal
| | - Nouredin Messaoudi
- Department of Surgery, University of Antwerp, Antwerp, Belgium., Department of Surgery, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel and Europe Hospitals, Brussels, Belgium
| | - Yannic McNicoll
- Surgery Department, Hôpital Jean-Talon, CIUSSS NIM, Montreal, Quebec, Canada
| | - Geneviève Soucy
- Pathology Service, Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | | | - Ricard Masia
- Surface Oncology, Inc. Cambridge, Massachusetts, USA
| | | | - Heewon Seo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Christopher B. Eeles
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Neha Rohatgi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Simon C. Robson
- Center for Inflammation Research, Gastroenterology, Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Simon Turcotte
- Cancer Axis, Centre de Recherche Du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada., Institut du Cancer de Montréal., Hepatopancreatobiliary Surgery & Liver Transplantation Service, Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada., Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada, Department of Computer Science, University of Toronto, Toronto, Ontario, Canada, Ontario Institute for Cancer Research, Toronto, Ontario, Canada, Vector Institute for Artificial Intelligence, Toronto, Ontario, Canada
| | - John Stagg
- Faculty of Pharmacy, University of Montreal., Cancer Axis, Centre de Recherche Du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada., Institut du Cancer de Montréal.,Correspondence: 900 St-Denis Street, Montréal, QC, Canada, H2X 0A9; ; Tel: 514-890-8000 ex:25170
| |
Collapse
|
22
|
O'Brien BJ, Faraoni EY, Strickland LN, Ma Z, Mota V, Mota S, Chen X, Mills T, Eltzschig HK, DelGiorno KE, Bailey‐Lundberg JM. CD73-generated extracellular adenosine promotes resolution of neutrophil-mediated tissue injury and restrains metaplasia in pancreatitis. FASEB J 2023; 37:e22684. [PMID: 36468677 PMCID: PMC9753971 DOI: 10.1096/fj.202201537r] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/07/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
Pancreatitis is currently the leading cause of gastrointestinal hospitalizations in the US. This condition occurs in response to abdominal injury, gallstones, chronic alcohol consumption or, less frequently, the cause remains idiopathic. CD73 is a cell surface ecto-5'-nucleotidase that generates extracellular adenosine, which can contribute to resolution of inflammation by binding adenosine receptors on infiltrating immune cells. We hypothesized genetic deletion of CD73 would result in more severe pancreatitis due to decreased generation of extracellular adenosine. CD73 knockout (CD73-/- ) and C57BL/6 (wild type, WT) mice were used to evaluate the progression and response of caerulein-induced acute and chronic pancreatitis. In response to caerulein-mediated chronic or acute pancreatitis, WT mice display resolution of pancreatitis at earlier timepoints than CD73-/- mice. Using immunohistochemistry and analysis of single-cell RNA-seq (scRNA-seq) data, we determined CD73 localization in chronic pancreatitis is primarily observed in mucin/ductal cell populations and immune cells. In murine pancreata challenged with caerulein to induce acute pancreatitis, we compared CD73-/- to WT mice and observed a significant infiltration of Ly6G+, MPO+, and Granzyme B+ cells in CD73-/- compared to WT pancreata and we quantified a significant increase in acinar-to-ductal metaplasia demonstrating sustained metaplasia and inflammation in CD73-/- mice. Using neutrophil depletion in CD73-/- mice, we show neutrophil depletion significantly reduces metaplasia defined by CK19+ cells per field and significantly reduces acute pancreatitis. These data identify CD73 enhancers as a potential therapeutic strategy for patients with acute and chronic pancreatitis as adenosine generation and activation of adenosine receptors is critical to resolve persistent inflammation in the pancreas.
Collapse
Affiliation(s)
- Baylee J. O'Brien
- Center for Perioperative Medicine, Department of Anesthesiology, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Erika Y. Faraoni
- Center for Perioperative Medicine, Department of Anesthesiology, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Lincoln N. Strickland
- Center for Perioperative Medicine, Department of Anesthesiology, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Zhibo Ma
- Gene Expression LaboratoryThe Salk Institute for Biological SciencesSan DiegoCaliforniaUSA
| | - Victoria Mota
- Center for Perioperative Medicine, Department of Anesthesiology, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Samantha Mota
- Center for Perioperative Medicine, Department of Anesthesiology, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
- The Graduate School of Biomedical SciencesThe University of Texas MD Anderson Cancer Center and The University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Xuebo Chen
- Center for Perioperative Medicine, Department of Anesthesiology, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Tingting Mills
- Department of Biochemistry, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Holger K. Eltzschig
- Center for Perioperative Medicine, Department of Anesthesiology, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Kathleen E. DelGiorno
- Department of Cell and Developmental BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Jennifer M. Bailey‐Lundberg
- Center for Perioperative Medicine, Department of Anesthesiology, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
- The Graduate School of Biomedical SciencesThe University of Texas MD Anderson Cancer Center and The University of Texas Health Science Center at HoustonHoustonTexasUSA
| |
Collapse
|
23
|
Faraoni EY, Strickland LN, O’Brien BJ, Barraza JF, Thosani NC, Wray CJ, Mills TW, Bailey-Lundberg JM. Radiofrequency ablation in combination with CD73 inhibitor AB680 reduces tumor growth and enhances anti-tumor immunity in a syngeneic model of pancreatic ductal adenocarcinoma. Front Oncol 2022; 12:995027. [PMID: 36147911 PMCID: PMC9486545 DOI: 10.3389/fonc.2022.995027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/19/2022] [Indexed: 11/30/2022] Open
Abstract
Pancreatic ductal adenocarcinoma presents a 5-year overall survival rate of 11%, placing an imperative need for the discovery and application of innovative treatments. Radiofrequency ablation represents a promising therapy for PDA, as studies show it induces coagulative necrosis and a host adaptive immune response. In this work we evaluated the effects of RFA treatment in vivo by establishing a syngeneic mouse model of PDA and performing tumor ablation in one flank. Our studies revealed RFA acutely impaired PDA tumor growth; however, such effects were not sustained one week after treatment. Adenosine (ADO) pathway represents a strong immunosuppressive mechanism that was shown to play a role in PDA progression and preliminary data from ongoing clinical studies suggest ADO pathway inhibition may improve therapeutic outcomes. Thus, to investigate whether ADO generation may be involved in tumor growth relapse after RFA, we evaluated adenosine-monophosphate (AMP), ADO and inosine (INO) levels by HPLC and found they were acutely increased after treatment. Thus, we evaluated an in vivo CD73 inhibition in combination with RFA to study ADO pathway implication in RFA response. Results showed combination therapy of RFA and a CD73 small molecule inhibitor (AB680) in vivo promoted sustained tumor growth impairment up to 10 days after treatment as evidenced by increased necrosis and anti-tumor immunity, suggesting RFA in combination with CD73 inhibitors may improve PDA patient response.
Collapse
Affiliation(s)
- Erika Y. Faraoni
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Lincoln N. Strickland
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Baylee J. O’Brien
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Joseph F. Barraza
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Nirav C. Thosani
- Center for Interventional Gastroenterology at UTHealth (iGUT), McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Curtis J. Wray
- Department of Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Tingting W. Mills
- Department of Biochemistry, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jennifer M. Bailey-Lundberg
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Center for Interventional Gastroenterology at UTHealth (iGUT), McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Anesthesiology, Center for Perioperative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
24
|
Liu Q, Zhao H, Guo Y, Zhang K, Shang F, Liu T. Bioinformatics-Based Analysis: Noncoding RNA-Mediated COL10A1 Is Associated with Poor Prognosis and Immune Cell Infiltration in Pancreatic Cancer. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7904982. [PMID: 36105715 PMCID: PMC9467764 DOI: 10.1155/2022/7904982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022]
Abstract
Background Collagen type X alpha 1 (COL10A1) is a structural component of the extracellular matrix that is aberrantly expressed in a variety of cancer tissues. However, its role in pancreatic cancer progression is not well understood. Methods The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and Gene Expression Profiling Interaction Analysis (GEPIA) data were employed to explore the expression of COL10A1 in normal and tumor tissues and its prognostic value in pancreatic adenocarcinoma. The clinical data of pancreatic cancer in TCGA were used to explore the relationship between COL10A1 and clinical features. Genes coexpressed with COL10A1 were explored using multiple databases and analyzed for functional enrichment. In addition, the lncRNA/miRNA/COL10A1 axis that may be involved in COL10A1 regulation in pancreatic cancer was explored by constructing a competitive endogenous RNA (ceRNA) regulatory axis. Finally, COL10A1 was analyzed for correlation with immune cell infiltration and various immune checkpoint molecules in pancreatic cancer. Results It was found that the expression of COL10A1 was significantly increased in pancreatic cancer tissues. High expression of COL10A1 was related to the clinicopathological characteristics and the worse prognosis of pancreatic cancer patients. The TUG1/miR-144-3p/COL10A1 axis was identified as the most likely upstream noncoding RNA pathway for COL10A1 in pancreatic cancer. Besides, in pancreatic adenocarcinoma, the expression level of COL10A1 showed a significant positive correlation with tumor immune cell infiltration, biomarkers of immune cells, and expression of immune checkpoint molecules. Conclusion COL10A1 is an early diagnostic marker, and its high expression correlates with immune infiltration in pancreatic cancer. The TUG1/miR-144-3p/COL10A1 axis was identified as the most likely upstream noncoding RNA pathway for COL10A1 in pancreatic cancer.
Collapse
Affiliation(s)
- Qi Liu
- Department of Colorectal Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| | - Hongyu Zhao
- Department of Gastroenterology and Center of Digestive Endoscopy, the Second Hospital of Jilin University, Changchun 130041, China
| | - Yu Guo
- Department of Colorectal Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| | - Kai Zhang
- Department of Colorectal Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| | - Fengjia Shang
- Department of Colorectal Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| | - Tongjun Liu
- Department of Colorectal Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| |
Collapse
|
25
|
Gössling GCL, Zhen DB, Pillarisetty VG, Chiorean EG. Combination immunotherapy for pancreatic cancer: challenges and future considerations. Expert Rev Clin Immunol 2022; 18:1173-1186. [PMID: 36045547 DOI: 10.1080/1744666x.2022.2120471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION : Immune checkpoint inhibitors (ICI) have not yielded significant efficacy in pancreatic ductal adenocarcinoma (PDA), despite the role of the innate and adaptive immune systems on progression and survival. However, recently identified pathways have identified new targets and generated promising clinical investigations into promoting an effective immune-mediated antitumor response in PDA. AREAS COVERED : We review biological mechanisms associated with immunotherapy resistance and outline strategies for therapeutic combinations with established and novel therapies in PDA. EXPERT OPINION : Pancreatic cancers rarely benefits from treatment with ICI due to an immunosuppressive tumor microenvironment (TME). New understandings of factors associated with the suppressive TME, include low and poor quality neoantigens, constrained effector T cells infiltration, and the presence of a dense, suppressive myeloid cell population. These findings have been translated into new clinical investigations evaluating novel therapies in combination with ICI and/or standard systemic chemotherapy and radiotherapy. The epithelial, immune, and stromal compartments are intricately related in PDA, and the framework for successful targeting of this disease requires a comprehensive and personalized approach.
Collapse
Affiliation(s)
| | - David B Zhen
- University of Washington School of Medicine, Seattle, WA, USA.,Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Venu G Pillarisetty
- University of Washington School of Medicine, Seattle, WA, USA.,Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - E Gabriela Chiorean
- University of Washington School of Medicine, Seattle, WA, USA.,Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
26
|
Chen XM, Liu YY, Tao BY, Xue XM, Zhang XX, Wang LL, Zhong H, Zhang J, Yang SM, Jiang QQ. NT5E upregulation in head and neck squamous cell carcinoma: A novel biomarker on cancer-associated fibroblasts for predicting immunosuppressive tumor microenvironment. Front Immunol 2022; 13:975847. [PMID: 36091055 PMCID: PMC9458906 DOI: 10.3389/fimmu.2022.975847] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/03/2022] [Indexed: 11/22/2022] Open
Abstract
Despite tremendous progress made in the diagnosis and managements, head and neck squamous cell carcinoma (HNSC) remains a global medical dilemma with dismal clinical prognosis and high mortality. Gene NT5E encodes the ecto-5’-nucleotidase (CD73), which facilitates the formation of immunosuppressive tumor microenvironment (TME) permissive for tumor progression in various malignancies. Nevertheless, the cell subsets NT5E expressed on and the potential function of NT5E in the TME of HNSC remain virgin lands in HNSC. In this study, we comprehensively performed integrated prognostic analysis and elucidated that NT5E was an independent prognostic indicator for HNSC, for which a high NT5E level predicted poor overall survival (OS), disease-specific survival (DSS) and progression-free interval (PFI) in HNSC patients (p<0.05). Enrichment analyses revealed the close correlation between NT5E and ECM remodeling, and the latent function of NT5E may involve in epithelial-to-mesenchymal transition (EMT) and metastasis during HNSC progression. HNSC-related immune infiltration analysis and single-cell type analysis demonstrated that NT5E expression was significantly positively associated with cancer-associated fibroblasts (CAFs) in HNSC (p<0.01). NT5E-related TME analysis revealed that NT5E-high group are characterized by low neoantigen loads (NAL, p<0.001) and tumor mutation burden (TMB, p<0.01), indicating high-NT5E-expression HNSC patients may be recalcitrant to immunotherapy. In-situ multicolor immunofluorescence staining was later conducted and the results further verified our findings. Taken together, NT5E could be a novel biomarker in HNSC. Predominantly expressed on CAFs, the upregulation of NT5E might predict an immunosuppressive TME for HNSC patients who may benefit little from immunotherapy. Targeting CAFs with high NT5E expression might be a novel therapeutic strategy for HNSC patients.
Collapse
Affiliation(s)
- 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
| | - 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
| | - 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
| | - 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
| | - Xin-xin Zhang
- 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
| | - Lin-lin 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
| | - Hui Zhong
- 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
- Southern Medical University, Guangzhou, China
| | - Jun Zhang
- Department of Neurosurgery, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Qing-qing Jiang, ; Shi-ming Yang, ; Jun Zhang,
| | - 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: Qing-qing Jiang, ; Shi-ming Yang, ; Jun Zhang,
| | - Qing-qing Jiang
- 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: Qing-qing Jiang, ; Shi-ming Yang, ; Jun Zhang,
| |
Collapse
|
27
|
ALCAM: A Novel Surface Marker on EpCAMlow Circulating Tumor Cells. Biomedicines 2022; 10:biomedicines10081983. [PMID: 36009530 PMCID: PMC9405826 DOI: 10.3390/biomedicines10081983] [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: 06/25/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Current strategies in circulating tumor cell (CTC) isolation in pancreatic cancer heavily rely on the EpCAM and cytokeratin cell status. EpCAM is generally not considered a good marker given its transitory change during Epithelial to Mesenchymal Transition (EMT) or reverse EMT. There is a need to identify other surface markers to capture the complete repertoire of PDAC CTCs. The primary objective of the study is to characterize alternate surface biomarkers to EpCAM on CTCs that express low or negligible levels of surface EpCAM in pancreatic cancer patients. Methods: Flow cytometry and surface mass spectrometry were used to identify proteins expressed on the surface of PDAC CTCs in culture. CTCs were grown under conditions of attachment and in co-culture with naïve neutrophils. Putative biomarkers were then validated in GEMMs and patient samples. Results: Surface proteomic profiling of CTCs identified several novel protein biomarkers. ALCAM was identified as a novel robust marker in GEMM models and in patient samples. Conclusions: We identified several novel surface biomarkers on CTCs expressed under differing conditions of culture. ALCAM was validated and identified as a novel alternate surface marker on EpCAMlow CTCs.
Collapse
|
28
|
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: 4.5] [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
|
29
|
Tang HD, Wang Y, Xie P, Tan SY, Li HF, Shen H, Zhang Z, Lei ZQ, Zhou JH. The Crosstalk Between Immune Infiltration, Circulating Tumor Cells, and Metastasis in Pancreatic Cancer: Identification of HMGB3 From a Multiple Omics Analysis. Front Genet 2022; 13:892177. [PMID: 35754798 PMCID: PMC9213737 DOI: 10.3389/fgene.2022.892177] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/19/2022] [Indexed: 12/27/2022] Open
Abstract
Metastasis is the major cause of death in patients with pancreatic ductal adenocarcinoma (PDAC), and circulating tumor cells (CTCs) play an important role in the development of metastasis. However, few studies have uncovered the metastasis mechanism of PDAC based on CTCs. In this study, the existing bulk RNA-sequencing (bulk RNA-seq) and single-cell sequencing (scRNA-seq) data for CTCs in pancreatic cancer were obtained from the Gene Expression Omnibus (GEO) database. Analysis of tumor-infiltrating immune cells (TIICs) by CIBERSORT showed that the CTCs enriched from the peripheral blood of metastatic PDAC were found to contain a high proportion of T cell regulators (Tregs) and macrophages, while the proportion of dendritic cells (DCs) was lower than that enriched from localized PDAC. Through weighted gene co-expression network analysis (WGCNA) and the result of scRNA-seq, we identified the hub module (265 genes) and 87 marker genes, respectively, which were highly associated with metastasis. The results of functional enrichment analysis indicated that the two gene sets mentioned above are mainly involved in cell adhesion and cytoskeleton and epithelial–mesenchymal transition (EMT). Finally, we found that HMGB3 was the hub gene according to the Venn diagram. The expression of HMGB3 in PDAC was significantly higher than that in normal tissues (protein and mRNA levels). HMGB3 expression was significantly positively correlated with both EMT-related molecules and CTC cluster–related markers. Furthermore, it was also found that HMGB3 mutations were favorably related to tumor-associated immune cells through the TIMER2.0 online tool. We further demonstrated that PDAC patients with higher HMGB3 expression had significantly worse overall survival (OS) in multiple datasets. In summary, our study suggests that HMGB3 is a hub gene associated with EMT in CTCs, the formation of CTC clusters, and infiltration patterns of immune cells favorable for tumor progression and metastasis to distant organs.
Collapse
Affiliation(s)
- Hao-Dong Tang
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Yang Wang
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
| | - Peng Xie
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Si-Yuan Tan
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Hai-Feng Li
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Hao Shen
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Zheng Zhang
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
| | - Zheng-Qing Lei
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
| | - Jia-Hua Zhou
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China.,Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
| |
Collapse
|
30
|
Liu Z, Wu X, Wang Q, Li Z, Liu X, Sheng X, Zhu H, Zhang M, Xu J, Feng X, Wu B, Lv X. CD73-Adenosine A 1R Axis Regulates the Activation and Apoptosis of Hepatic Stellate Cells Through the PLC-IP 3-Ca 2+/DAG-PKC Signaling Pathway. Front Pharmacol 2022; 13:922885. [PMID: 35784730 PMCID: PMC9245432 DOI: 10.3389/fphar.2022.922885] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022] Open
Abstract
Alcohol-related liver fibrosis (ALF) is a form of alcohol-related liver disease (ALD) that generally occurs in response to heavy long-term drinking. Ecto-5'-nucleotidase (NT5E), also known as CD73, is a cytomembrane protein linked to the cell membrane via a GPI anchor that regulates the conversion of extracellular ATP to adenosine. Adenosine and its receptors are important regulators of the cellular response. Previous studies showed that CD73 and adenosine A1 receptor (A1R) were important in alcohol-related liver disease, however the exact mechanism is unclear. The aim of this study was to elucidate the role and mechanism of the CD73-A1R axis in both a murine model of alcohol and carbon tetrachloride (CCl4) induced ALF and in an in vitro model of fibrosis induced by acetaldehyde. The degree of liver injury was determined by measuring serum AST and ALT levels, H & E staining, and Masson's trichrome staining. The expression levels of fibrosis indicators and PLC-IP3-Ca2+/DAG-PKC signaling pathway were detected by quantitative real-time PCR, western blotting, ELISA, and calcium assay. Hepatic stellate cell (HSC) apoptosis was detected using the Annexin V-FITC/PI cell apoptosis detection kit. Knockdown of CD73 significantly attenuated the accumulation of α-SMA and COL1a1 damaged the histological architecture of the mouse liver induced by alcohol and CCl4. In vitro, CD73 inhibition attenuated acetaldehyde-induced fibrosis and downregulated A1R expression in HSC-T6 cells. Inhibition of CD73/A1R downregulated the expression of the PLC-IP3-Ca2+/DAG-PKC signaling pathway. In addition, silencing of CD73/A1R promoted apoptosis in HSC-T6 cells. In conclusion, the CD73-A1R axis can regulate the activation and apoptosis of HSCs through the PLC-IP3-Ca2+/DAG-PKC signaling pathway.
Collapse
Affiliation(s)
- Zhenni Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xue Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Qi Wang
- Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Zixuan Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xueqi Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiaodong Sheng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Hong Zhu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Mengda Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Junrui Xu
- General Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaowen Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Baoming Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiongwen Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, China
| |
Collapse
|
31
|
Huang WL, Wu SF, Huang X, Zhou S. Integrated Analysis of ECT2 and COL17A1 as Potential Biomarkers for Pancreatic Cancer. DISEASE MARKERS 2022; 2022:9453549. [PMID: 35722628 PMCID: PMC9200569 DOI: 10.1155/2022/9453549] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/25/2022] [Indexed: 11/30/2022]
Abstract
Background Pancreatic cancer (PC) is a malignant tumor of the digestive tract. It presents with atypical clinical symptoms and lacks specific diagnostic indicators. This study is aimed at exploring the potential biomarkers of PC. Methods TCGA database pancreatic cancer dataset was normalized and used to identify differentially expressed genes (DEGs). Survival, independent prognostic, and clinical correlation analyses were performed on DEGs to screen for key genes. DNA methylation, mutation, and copy number variation (CNV) analyses were used to analyze genetic variants in key genes. GSEA was performed to explore the functional enrichment of the key genes. Based on the expression of key genes, construction of a competing endogenous RNA (ceRNA) network, analysis of the tumor microenvironment (TME), and prediction of chemotherapeutic drug sensitivity were performed. Furthermore, the GEO database was used to validate the reliability of key genes. Results Two key genes (ECT2 and COL17A1) were identified, which were highly expressed in PC. The mRNA expression of ECT2 and COL17A1 was associated with DNA methylation and CNV. The cell cycle, proteasome, and pathways in cancer were enriched in the high-COL17A1 and ECT2 groups. The TME results showed that immune scores were decreased in the high-ECT2 group. CeRNA network results showed that there were eleven miRNAs were involved in the regulation of ECT2 and COL17A1. Moreover, pRRophetic analysis showed that 20 chemotherapeutic drugs were associated with ECT2 and COL17A1 expression. Conclusions Collectively, ECT2 and COL17A1 may be potential biomarkers for PC, providing a new direction for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Wen-liang Huang
- MRI Room, The Second Affiliated Hospital of Luohe Medical College, Luohe, China
| | - Shu-fen Wu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Luohe Medical College, Luohe, China
| | - Xiao Huang
- Department of Clinical Laboratory, Zhengzhou University First Affiliated Hospital, Zhengzhou, China
| | - Shan Zhou
- MRI Room, The Second Affiliated Hospital of Luohe Medical College, Luohe, China
| |
Collapse
|
32
|
Yin C, Alqahtani A, Noel MS. The Next Frontier in Pancreatic Cancer: Targeting the Tumor Immune Milieu and Molecular Pathways. Cancers (Basel) 2022; 14:2619. [PMID: 35681599 PMCID: PMC9179513 DOI: 10.3390/cancers14112619] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/13/2022] [Accepted: 05/19/2022] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with abysmal prognosis. It is currently the third most common cause of cancer-related mortality, despite being the 11th most common cancer. Chemotherapy is standard of care in all stages of pancreatic cancer, yet survival, particularly in the advanced stages, often remains under one year. We are turning to immunotherapies and targeted therapies in PDAC in order to directly attack the core features that make PDAC notoriously resistant to chemotherapy. While the initial studies of these agents in PDAC have generally been disappointing, we find optimism in recent preclinical and early clinical research. We find that despite the immunosuppressive effects of the PDAC tumor microenvironment, new strategies, such as combining immune checkpoint inhibitors with vaccine therapy or chemokine receptor antagonists, help elicit strong immune responses. We also expand on principles of DNA homologous recombination repair and highlight opportunities to use agents, such as PARP inhibitors, that exploit deficiencies in DNA repair pathways. Lastly, we describe advances in direct targeting of driver mutations and metabolic pathways and highlight some technological achievements such as novel KRAS inhibitors.
Collapse
Affiliation(s)
| | | | - Marcus S. Noel
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA; (C.Y.); (A.A.)
| |
Collapse
|
33
|
The Effects of CD73 on Gastrointestinal Cancer Progression and Treatment. JOURNAL OF ONCOLOGY 2022; 2022:4330329. [PMID: 35620732 PMCID: PMC9130010 DOI: 10.1155/2022/4330329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/04/2022] [Indexed: 11/18/2022]
Abstract
Gastrointestinal (GI) cancer is a common and deadly malignant tumor. CD73, a cell-surface protein, acts as a switch of the adenosine-related signaling pathway that can cause significant immunosuppression. Recent evidence has emerged that CD73 is a promising immunotherapy target for regaining immune cell function and restraining tumorigenesis, and a growing stream of research indicates that combining immunotherapy with other therapies can effectively improve the prognosis and survival of GI cancer patients. Several immune checkpoint inhibitors have been approved for use in GI cancer recently; however, they have demonstrated limited efficacy. Solving the problem of immunosuppression in GI cancer is the key to developing an effective therapeutic option and the modulation of CD73 expression may provide an answer. In this review, we discuss current research on CD73 in gastric, liver, pancreatic, and colorectal cancer to evaluate its therapeutic potential as an immunotherapy target in GI cancers.
Collapse
|
34
|
Wang S, Wang R, Gao F, Huang J, Zhao X, Li D. Pan-cancer analysis of the DNA methylation patterns of long non-coding RNA. Genomics 2022; 114:110377. [PMID: 35513292 DOI: 10.1016/j.ygeno.2022.110377] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/23/2022] [Accepted: 04/27/2022] [Indexed: 11/04/2022]
Abstract
Long non-coding RNA (lncRNA) regulated by abnormal DNA methylation (ADM-lncRNA) emerges as a biomarker for cancer diagnosis and treatment. This study comprehensively described the methylation patterns of lncRNA in pan-cancer using the cancer data set in The Cancer Genome Atlas (TCGA). Based on the cancer heterogeneity of ADM-lncRNA in pan-cancer, we constructed a co-expression network of pan-cancer ADM-lncRNA (pADM-lncRNA) in 10 cancers, highlighting the combined action mode of abnormal DNA methylation, and indicating the internal connection among different cancers. Functional analysis revealed the pan-carcinogenic pathway of pADM-lncRNA and suggested potential factors for cancer heterogeneity and tumor immune microenvironment changes. Survival analysis showed the potential of pADM-lncRNA-mRNA co-expression pair as cancer biomarkers. Revealing the action mode of lncRNA and DNA methylation in cancer may help understand the key molecular mechanisms of cell carcinogenesis.
Collapse
Affiliation(s)
- Shijia Wang
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing 100069, China
| | - Rendong Wang
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing 100069, China
| | - Fang Gao
- Health Management Center, Binzhou People's Hospital, Shandong Province, China
| | - Jun Huang
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing 100069, China
| | - Xiaoxiao Zhao
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing 100069, China
| | - Dongguo Li
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
35
|
Head and Neck Squamous Cell Carcinoma: NT5E Could Be a Prognostic Biomarker. Appl Bionics Biomech 2022; 2022:3051907. [PMID: 35510041 PMCID: PMC9061055 DOI: 10.1155/2022/3051907] [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: 02/17/2022] [Accepted: 04/08/2022] [Indexed: 11/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a type of tumour with a relatively poor prognosis. In recent years, immune checkpoint inhibitors, such as CTLA-4 and PD-1/PDL-1 inhibitors, have improved the treatment status of advanced tumours. However, the emergence of drug resistance has brought difficulties to clinical treatment, and new immune checkpoint research is imminent. The hypoxia-adenosine pathway, in which CD73 encoded by the NT5E gene is a key enzyme for adenosine production, has been identified as an immune checkpoint of great potential. Therefore, NT5E may play an important role in HNSCC. We performed a detailed bioinformatics analysis of NT5E in HNSCC, and the results showed that the overexpression of NT5E in HNSCC was associated with poor prognosis. Our further investigation of the coexpression pattern of HNSCC could provide a reference for drug resistance and immunotherapy studies.
Collapse
|
36
|
Kolbe K, Wittner M, Hartjen P, Hüfner AD, Degen O, Ackermann C, Cords L, Stellbrink HJ, Haag F, Schulze zur Wiesch J. Inversed Ratio of CD39/CD73 Expression on γδ T Cells in HIV Versus Healthy Controls Correlates With Immune Activation and Disease Progression. Front Immunol 2022; 13:867167. [PMID: 35529864 PMCID: PMC9074873 DOI: 10.3389/fimmu.2022.867167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/15/2022] [Indexed: 12/16/2022] Open
Abstract
Background γδ T cells are unconventional T cells that have been demonstrated to be crucial for the pathogenesis and potentially for the cure of HIV-1 infection. The ectonucleotidase CD39 is part of the purinergic pathway that regulates immune responses by degradation of pro-inflammatory ATP in concert with CD73. Few studies on the expression of the ectoenzymes CD73 and CD39 on human γδ T cells in HIV have been performed to date. Methods PBMC of n=86 HIV-1-infected patients were compared to PBMC of n=26 healthy individuals using 16-color flow cytometry determining the surface expression of CD39 and CD73 on Vδ1 and Vδ2 T cells in association with differentiation (CD45RA, CD28, CD27), activation and exhaustion (TIGIT, PD-1, CD38, and HLA-DR), and assessing the intracellular production of pro- and anti-inflammatory cytokines (IL-2, TGF-ß, TNF-α, Granzyme B, IL-10, IFN-γ) after in vitro stimulation with PMA/ionomycin. Results CD39 and CD73 expression on γδ T cells were inversed in HIV infection which correlated with HIV disease progression and immune activation. CD39, but not CD73 expression on γδ T cells of ART-treated patients returned to levels comparable with those of healthy individuals. Only a small subset (<1%) of γδ T cells co-expressed CD39 and CD73 in healthy or HIV-infected individuals. There were significantly more exhausted and terminally differentiated CD39+ Vδ1 T cells regardless of the disease status. Functionally, IL-10 was only detectable in CD39+ γδ T cells after in vitro stimulation in all groups studied. Viremic HIV-infected patients showed the highest levels of IL-10 production. The highest percentage of IL-10+ cells was found in the small CD39/CD73 co-expressing γδ T-cell population, both in healthy and HIV-infected individuals. Also, CD39+ Vδ2 T cells produced IL-10 more frequently than their CD39+ Vδ1 counterparts in all individuals regardless of the HIV status. Conclusions Our results point towards a potential immunomodulatory role of CD39+ and CD73+ γδ T cells in the pathogenesis of chronic HIV infection that needs further investigation.
Collapse
Affiliation(s)
- Katharina Kolbe
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg Lübeck Borstel Riems, Hamburg, Germany
| | - Melanie Wittner
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg Lübeck Borstel Riems, Hamburg, Germany
- *Correspondence: Melanie Wittner,
| | - Philip Hartjen
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anja-Dorothee Hüfner
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Infectious Diseases Clinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Olaf Degen
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Infectious Diseases Clinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christin Ackermann
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leon Cords
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Schulze zur Wiesch
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg Lübeck Borstel Riems, Hamburg, Germany
| |
Collapse
|
37
|
Pu N, Chen Q, Yin H, Zhang J, Zhao G, Habib JR, Chen J, Yu J, Lou W, Wu W. Identification of an Immune-Related BAT Signature for Predicting Adjuvant Chemotherapy Response and Overall Survival in Patients with Resected Ductal Adenocarcinoma of the Pancreas. J Gastrointest Surg 2022; 26:869-886. [PMID: 35059985 DOI: 10.1007/s11605-021-05232-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 12/08/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND Adjuvant chemotherapy (ACT) is widely accepted in patients with pancreatic ductal adenocarcinoma (PDAC) after surgery; however, effective models for predicting ACT response are scarce. Thus, the objective of this study was to develop a novel signature for predicting its response and overall survival (OS) in resected PDAC patients. METHODS A total of 50 PDAC patients with the transcriptome expression profiles, information about chemotherapy, and relevant clinical data were retrieved from the Cancer Genome Atlas (TCGA), and twenty-nine patients with tissue specimens and clinical data from our hospital were included as a validation. A novel gene signature was developed using bioinformatic differentially expressed genes (DEGs) analysis, Lasso-penalized Cox regression, and multivariate Cox regression studies. RESULTS Between chemotherapy-resistant and chemotherapy-sensitive cohorts, 569 DEGs were identified, with 490 upregulated and 79 downregulated genes mainly specialized in the regulation of peptide/protein/hormone secretion, calcium ion homeostasis, and T cell activation regulation in biological processes. After Lasso-penalized Cox and multivariate Cox regression analysis, BAT (BCHE, ADH1A, and TNS4) signature was established to predict ACT response and OS. Moreover, BAT signature was verified as an independent risk factor for ACT response (p = 0.042) and OS (median OS: 17.5 months vs. 34.8 months, p = 0.040) and significantly associated with immune infiltrations (p < 0.05). Then, this signature was further validated as the independent risk factor for recurrence-free survival (RFS) in PDAC patients receiving postoperative ACT (median RFS: 9.0 months vs. not reached, p = 0.014), and tumor-infiltrating CD4+ and CD8+ T cells were further validated to be significantly decreased in tissues with higher BAT signature scores (p = 0.015 and 0.021, respectively). CONCLUSION The BAT signature is a novel formulated and independent risk factor for predicting ACT response and long-term survival in patients with resected PDAC. This signature could comprehensively reflect local immune-related response, tumor purity, potential biological behavior, and chemo drug susceptibility.
Collapse
Affiliation(s)
- Ning Pu
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Qiangda Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hanlin Yin
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jicheng Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guochao Zhao
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Joseph R Habib
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jie Chen
- Department of Cardiothoracic Surgery, Naval Medical Center of PLA, Shanghai, 200052, China
| | - Jun Yu
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Wenhui Lou
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenchuan Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
38
|
A Novel Anti-CD73 Antibody That Selectively Inhibits Membrane CD73 Shows Antitumor Activity and Induces Tumor Immune Escape. Biomedicines 2022; 10:biomedicines10040825. [PMID: 35453575 PMCID: PMC9031174 DOI: 10.3390/biomedicines10040825] [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: 02/28/2022] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023] Open
Abstract
CD73 catalyzes the conversion of ATP to adenosine, which is involved in various physiological and pathological processes, including tumor immune escape. Because CD73 expression and activity are particularly high on cancer cells and contribute to the immunosuppressive properties of the tumor environment, it is considered an attractive target molecule for specific cancer therapies. In line, several studies demonstrated that CD73 inhibition has a significant antitumor effect. However, complete blocking of CD73 activity can evoke autoimmune phenomena and adverse side effects. We developed a CD73-specific antibody, 22E6, that specifically inhibits the enzymatic activity of membrane-tethered CD73 present in high concentrations on cancer cells and cancer cell-derived extracellular vesicles but has no inhibitory effect on soluble CD73. Inhibition of CD73 on tumor cells with 22E6 resulted in multiple effects on tumor cells in vitro, including increased apoptosis and interference with chemoresistance. Intriguingly, in a xenograft mouse model of acute lymphocytic leukemia (ALL), 22E6 treatment resulted in an initial tumor growth delay in some animals, followed by a complete loss of CD73 expression on ALL cells in all 22E6 treated animals, indicating tumor immune escape. Taken together, 22E6 shows great potential for cancer therapy, favorably in combination with other drugs.
Collapse
|
39
|
Faraoni EY, Ju C, Robson SC, Eltzschig HK, Bailey-Lundberg JM. Purinergic and Adenosinergic Signaling in Pancreatobiliary Diseases. Front Physiol 2022; 13:849258. [PMID: 35360246 PMCID: PMC8964054 DOI: 10.3389/fphys.2022.849258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Adenosine 5'-triphosphate (ATP), other nucleotides, and the nucleoside analogue, adenosine, all have the capacity to modulate cellular signaling pathways. The cellular processes linked to extracellular purinergic signaling are crucial in the initiation, evolution, and resolution of inflammation. Injured or dying cells in the pancreatobiliary tract secrete or release ATP, which results in sustained purinergic signaling mediated through ATP type-2 purinergic receptors (P2R). This process can result in chronic inflammation, fibrosis, and tumor development. In contrast, signaling via the extracellular nucleoside derivative adenosine via type-1 purinergic receptors (P1R) is largely anti-inflammatory, promoting healing. Failure to resolve inflammation, as in the context of primary sclerosing cholangitis or chronic pancreatitis, is a risk factor for parenchymal and end-organ scarring with the associated risk of pancreatobiliary malignancies. Emerging immunotherapeutic strategies suggest that targeting purinergic and adenosinergic signaling can impact the growth and invasive properties of cancer cells, potentiate anti-tumor immunity, and also block angiogenesis. In this review, we dissect out implications of disordered purinergic responses in scar formation, end-organ injury, and in tumor development. We conclude by addressing promising opportunities for modulation of purinergic/adenosinergic signaling in the prevention and treatment of pancreatobiliary diseases, inclusive of cancer.
Collapse
Affiliation(s)
- Erika Y. Faraoni
- Department of Anesthesiology, Center for Perioperative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Cynthia Ju
- Department of Anesthesiology, Center for Perioperative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Simon C. Robson
- Departments of Internal Medicine and Anesthesiology, Center for Inflammation Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Holger K. Eltzschig
- Department of Anesthesiology, Center for Perioperative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jennifer M. Bailey-Lundberg
- Department of Anesthesiology, Center for Perioperative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
40
|
Zhou Y, Fan Y, Mao Y, Lou M, Liu X, Yuan K, Tong J. NCAPG is a prognostic biomarker of immune infiltration in non-small-cell lung cancer. Biomark Med 2022; 16:523-535. [PMID: 35199566 DOI: 10.2217/bmm-2021-1090] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Purpose: Non-small-cell lung cancer (NSCLC) is the leading cause of cancer-related deaths. The protein NCAPG plays a significant role in tumor development. Patients & methods: We set up a tissue microarray (containing 140 NSCLC and ten normal lung tissues) and performed immunohistochemistry to assess NCAPG expression in the tissues of 140 patients. The prognostic value of NCAPG in NSCLC was assessed using the univariate and multivariate Cox proportional hazards regression models and Kaplan-Meier plots. We analyzed the association between NCAPG and immune infiltration in NSCLC. Results: Multifactorial analysis and Kaplan-Meier plots revealed that upregulation of NCAPG expression was an independent factor in the prognosis of NSCLC. Data from CIBERSORT showed a negative correlation between NCAPG and the expression of memory CD4+ T cells, CD8+ T cells, dendritic cells, macrophages, mast cells and natural killer cells (p < 0.001). Gene set enrichment analysis revealed that cell cycle, adhesion and proliferation were significantly enriched in samples with a high NCAPG expression. Conclusion: NCAPG is a novel biomarker of prognosis and is associated with immune cell infiltration in the tumor microenvironment. Thus it may be a potential target in NSCLC treatment.
Collapse
Affiliation(s)
- Yong Zhou
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China.,Heart & Lung Disease Laboratory, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Yongfei Fan
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Yifeng Mao
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Ming Lou
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Xiaoshuang Liu
- Nanjing Jinling Hospital: East Region Military Command General Hospital, Nanjing, Jiangsu, 210002, China
| | - Kai Yuan
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China.,Heart & Lung Disease Laboratory, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Jichun Tong
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| |
Collapse
|
41
|
Computational investigation of adenosine 5′-(α,β-methylene)-diphosphate (AMPCP) derivatives as ecto-5′-nucleotidase (CD73) inhibitors by using 3D-QSAR, molecular docking, and molecular dynamics simulations. Struct Chem 2022. [DOI: 10.1007/s11224-021-01863-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
42
|
Tian Y, Zhou Y, Liu J, Yi L, Gao Z, Yuan K, Tong J. Correlation of SIDT1 with Poor Prognosis and Immune Infiltration in Patients with Non-Small Cell Lung Cancer. Int J Gen Med 2022; 15:803-816. [PMID: 35125883 PMCID: PMC8807869 DOI: 10.2147/ijgm.s347171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/05/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Yubin Tian
- School of Medical, Dalian Medical University, Dalian, People’s Republic of China
- The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Yong Zhou
- The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Junhui Liu
- School of Medical, Dalian Medical University, Dalian, People’s Republic of China
- The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Lei Yi
- The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Zhaojia Gao
- The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
- Heart and Lung Disease Laboratory, The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Kai Yuan
- School of Medical, Dalian Medical University, Dalian, People’s Republic of China
- Heart and Lung Disease Laboratory, The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
- Correspondence: Kai Yuan; Jichun Tong, Email ;
| | - Jichun Tong
- The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
- Heart and Lung Disease Laboratory, The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| |
Collapse
|
43
|
Gao ZW, Liu C, Yang L, Chen HC, Yang LF, Zhang HZ, Dong K. CD73 Severed as a Potential Prognostic Marker and Promote Lung Cancer Cells Migration via Enhancing EMT Progression. Front Genet 2021; 12:728200. [PMID: 34868205 PMCID: PMC8635862 DOI: 10.3389/fgene.2021.728200] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 10/25/2021] [Indexed: 12/25/2022] Open
Abstract
To investigate the expression levels and prognostic value of CD73 in lung cancer. And moreover, to identify the effect and potential mechanism of CD73 on lung cancer cells proliferation and migration. CD73 expression levels in lung cancer were analyzed base on GEPIA2 and GEO database. GEPIA2 and Kaplan-Meier Plotter (KM Plotter) was used to analyzed the correlation between CD73 expression and prognosis. GEO dataset were analyzed via GEO2R. CD73 overexpression cell model was construction via recombinant lentivirus transfection into A549 and NCI-H520 cells. CCK8 assay were used to investigate cells proliferation. Migration and invasion ability were evaluated by scratch and transwell methods. Base on GEPIA2, GSE32683, GSE116959 and GSE37745 dataset, we found that CD73 expression were significant higher in tumor tissues of lung adenocarcinoma (LUAD) compared with that in non-tumor normal tissues and in lung squamous cell carcinoma (LUSC), while there were no significant difference of CD73 expression between LUSC and normal control tissues. Interestingly, a high CD73 level predict poor overall survival (OS) of LUSC. However, GEPIA2 and KM plotter showed the opposite conclusion of prognostic value of CD73 in LUAD. By using cell experiments, we found that CD73 overexpression promoted proliferation and migration of LUAD A549 cells. However, there was no significant effect of CD73 overexpression on LUSC NCI-H520 cells. Furthermore, CD73 overexpression facilitates epithelial to mesenchymal transition (EMT) progression of A549 cells. In conclusion, our results indicated that CD73 expression were increased in LUAD and might be an poor prognostic marker for LUSC patients. CD73 play an important role in LUAD cells proliferation and migration. These data allowed to support CD73 as a therapeutic target for LUAD.
Collapse
Affiliation(s)
- Zhao-Wei Gao
- Department of Clinical Laboratory, The Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| | - Chong Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| | - Lan Yang
- Department of Clinical Laboratory, The Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| | - Hao-Chuan Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| | - Long-Fei Yang
- Department of Clinical Laboratory, The Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| | - Hui-Zhong Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| | - Ke Dong
- Department of Clinical Laboratory, The Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
44
|
Chen Q, Pu N, Yin H, Zhang J, Zhao G, Lou W, Wu W. A metabolism-relevant signature as a predictor for prognosis and therapeutic response in pancreatic cancer. Exp Biol Med (Maywood) 2021; 247:120-130. [PMID: 34632851 DOI: 10.1177/15353702211049220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Although several altered metabolic genes have been identified to be involved in the tumorigenesis and advance of pancreatic cancer (PC), their prognostic values remained unclear. The purpose of this study was to explore new targets and establish a metabolic signature to predict prognosis and chemotherapy response for optimal individualized treatment. The expression data of PC patients from two independent cohorts and metabolism-related genes from KEGG were utilized and analyzed for the establishment of the signature via lasso regression. Then, the differentially expressed candidate genes were further confirmed via online data mining platform and qRT-PCR of clinical specimens. Then, the analyses of gene set enrichment, mutation, and chemotherapeutic response were performed via R package. As results showed, 109 differentially expressed metabolic genes were screened out in PC. Then a metabolism-related five-gene signature comprising B3GNT3, BCAT1, KYNU, LDHA, and TYMS was constructed and showed excellent ability for predicting survival. A novel nomogram coordinating the metabolic signature and other independent prognostic parameters was developed and showed better predictive power in predicting survival. In addition, this metabolic signature was significantly involved in the activation of multiple oncological pathways and regulation of the tumor immune microenvironment. The patients with high risk scores had higher tumor mutation burdens and were prone to be more sensitive to chemotherapy. In summary, our work identified a new metabolic signature and established a superior prognostic nomogram which may supply more indications to explore novel strategies for diagnosis and treatment.
Collapse
Affiliation(s)
- Qiangda Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ning Pu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hanlin Yin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jicheng Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guochao Zhao
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenhui Lou
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenchuan Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
45
|
Nykänen N, Mäkelä R, Arjonen A, Härmä V, Lewandowski L, Snowden E, Blaesius R, Jantunen I, Kuopio T, Kononen J, Rantala JK. Ex Vivo Drug Screening Informed Targeted Therapy for Metastatic Parotid Squamous Cell Carcinoma. Front Oncol 2021; 11:735820. [PMID: 34604070 PMCID: PMC8481915 DOI: 10.3389/fonc.2021.735820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
The purpose of ex vivo drug screening in the context of precision oncology is to serve as a functional diagnostic method for therapy efficacy modeling directly on patient-derived tumor cells. Here, we report a case study using integrated multiomics ex vivo drug screening approach to assess therapy efficacy in a rare metastatic squamous cell carcinoma of the parotid gland. Tumor cells isolated from lymph node metastasis and distal subcutaneous metastasis were used for imaging-based single-cell resolution drug screening and reverse-phase protein array-based drug screening assays to inform the treatment strategy after standard therapeutic options had been exhausted. The drug targets discovered on the basis of the ex vivo measured drug efficacy were validated with histopathology, genomic profiling, and in vitro cell biology methods, and targeted treatments with durable clinical responses were achieved. These results demonstrate the use of serial ex vivo drug screening to inform adjuvant therapy options prior to and during treatment and highlight HER2 as a potential therapy target also in metastatic squamous cell carcinoma of the salivary glands.
Collapse
Affiliation(s)
| | | | | | - Ville Härmä
- Misvik Biology Oy, Turku, Finland.,Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | | | - Eileen Snowden
- Genomic Sciences, BD Technologies, Research Triangle Park, Durham, NC, United States
| | - Rainer Blaesius
- Genomic Sciences, BD Technologies, Research Triangle Park, Durham, NC, United States
| | - Ismo Jantunen
- Central Finland Health Care District, Jyväskylä, Finland
| | - Teijo Kuopio
- Central Finland Health Care District, Jyväskylä, Finland.,Department of Biological and Environmental Science, Jyväskylä, Finland
| | | | - Juha K Rantala
- Misvik Biology Oy, Turku, Finland.,Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
46
|
Chen Q, Yin H, Pu N, Zhang J, Zhao G, Wenhui L, Wu W. Chemokine C-C motif ligand 21 synergized with programmed death-ligand 1 blockade restrains tumor growth. Cancer Sci 2021; 112:4457-4469. [PMID: 34402138 PMCID: PMC8586683 DOI: 10.1111/cas.15110] [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: 04/16/2021] [Revised: 08/09/2021] [Accepted: 08/12/2021] [Indexed: 12/27/2022] Open
Abstract
Programmed death‐ligand 1 (PD‐L1) blockade has revolutionized the prognosis of several cancers, but shows a weak effect on pancreatic cancer (PC) due to poor effective immune infiltration. Chemokine C‐C motif ligand 21 (CCL21), a chemokine promoting T cell immunity by recruiting and colocalizing dendritic cells (DCs) and T cells, serves as a potential antitumor agent in many cancers. However, its antitumor response and mechanism combined with PD‐L1 blockade in PC remain unclear. In our study, we found CCL21 played an important role in leukocyte chemotaxis, inflammatory response, and positive regulation of PI3K‐AKT signaling in PC using Metascape and gene set enrichment analysis. The CCL21 level was verified to be positively correlated with infiltration of CD8+ T cells by the CIBERSORT algorithm, but no significant difference in survival was observed in either The Cancer Genome Atlas or the International Cancer Genome Consortium cohort when stratified by CCL21 expression. Additionally, we found the growth rate of allograft tumors was reduced and T cell infiltration was increased, but tumor PD‐L1 abundance elevated simultaneously in the CCL21‐overexpressed tumors. Then, CCL21 was further verified to increase tumor PD‐L1 level through the AKT‐glycogen synthase kinase‐3β axis in human PC cells, which partly impaired the antitumor T cell immunity. Finally, the combination of CCL21 and PD‐L1 blockade showed superior synergistic tumor suppression in vitro and in vivo. Together, our findings suggested that CCL21 in combination with PD‐L1 blockade might be an efficient and promising option for the treatment of PC.
Collapse
Affiliation(s)
- Qiangda Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hanlin Yin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ning Pu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jicheng Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guochao Zhao
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lou Wenhui
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenchuan Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
47
|
CD73 Promotes Tumor Progression in Patients with Esophageal Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13163982. [PMID: 34439161 PMCID: PMC8393769 DOI: 10.3390/cancers13163982] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary The immune system plays important roles in antitumor activities. However, increasing evidence shows that tumor cells develop several mechanisms to escape the immune attack, resulting in immunosuppression. One of the most important immunosuppressive pathways is the CD73-adenosinergic pathway. In addition, this pathway participates in the development of cancer, including tumor cell proliferation, angiogenesis, and anti-inflammation mechanisms. Moreover, CD73 can mediate the invasion and metastasis of tumor cells via the regulation of cell interactions with the extracellular matrix components. Therefore, overcoming immunosuppression to restore the antitumor functions of T cells may be explored as a potential treatment strategy. Overexpression of CD73 promotes the malignant properties of cancers and is associated with specific clinical characteristics and worse prognosis in many types of cancers. The current study is the first to investigate the role of CD73 in determining the clinical outcomes of patients with esophageal squamous cell carcinoma. Abstract Cluster of differentiation (CD)-73 plays pivotal roles in the regulation of immune reactions via the production of extracellular adenosine, and the overexpression of CD73 is associated with worse outcomes in several types of cancers. Here, we identified 167 esophageal squamous cell carcinoma (ESCC) patients who underwent esophagectomy, including 64 and 103 patients with high and low expression levels of CD73, respectively. Univariate and multivariate analyses showed high expression of CD73 was an independent prognostic factor for worse disease-free survival and overall survival. In addition, we selected another cohort consisting of 38 ESCC patients receiving nivolumab or pembrolizumab and found that treatment response and survival benefit to immunotherapy were strongly correlated with the expression levels of CD73/programmed death ligand 1. Moreover, the transwell assay revealed knockdown of CD73 in two ESCC cell lines, TE1 and KYSE30, exhibited significantly reduced abilities of cell invasion and migration. CD73 silencing also showed that the protein expression levels of CD73, vimentin, and snail were downregulated, while those of E-cadherin were upregulated in Western blotting. The findings of our study indicate CD73 may be an independent prognostic factor for ESCC patients who underwent esophagectomy. Furthermore, it may be associated with the patient responses to immunotherapy.
Collapse
|
48
|
CD39 Regulation and Functions in T Cells. Int J Mol Sci 2021; 22:ijms22158068. [PMID: 34360833 PMCID: PMC8348030 DOI: 10.3390/ijms22158068] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/15/2022] Open
Abstract
CD39 is an enzyme which is responsible, together with CD73, for a cascade converting adenosine triphosphate into adenosine diphosphate and cyclic adenosine monophosphate, ultimately leading to the release of an immunosuppressive form of adenosine in the tumor microenvironment. Here, we first review the environmental and genetic factors shaping CD39 expression. Second, we report CD39 functions in the T cell compartment, highlighting its role in regulatory T cells, conventional CD4+ T cells and CD8+ T cells. Finally, we compile a list of studies, from preclinical models to clinical trials, which have made essential contributions to the discovery of novel combinatorial approaches in the treatment of cancer.
Collapse
|
49
|
Kepp O, Bezu L, Yamazaki T, Di Virgilio F, Smyth MJ, Kroemer G, Galluzzi L. ATP and cancer immunosurveillance. EMBO J 2021; 40:e108130. [PMID: 34121201 DOI: 10.15252/embj.2021108130] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/24/2021] [Accepted: 04/05/2021] [Indexed: 12/14/2022] Open
Abstract
While intracellular adenosine triphosphate (ATP) occupies a key position in the bioenergetic metabolism of all the cellular compartments that form the tumor microenvironment (TME), extracellular ATP operates as a potent signal transducer. The net effects of purinergic signaling on the biology of the TME depend not only on the specific receptors and cell types involved, but also on the activation status of cis- and trans-regulatory circuitries. As an additional layer of complexity, extracellular ATP is rapidly catabolized by ectonucleotidases, culminating in the accumulation of metabolites that mediate distinct biological effects. Here, we discuss the molecular and cellular mechanisms through which ATP and its degradation products influence cancer immunosurveillance, with a focus on therapeutically targetable circuitries.
Collapse
Affiliation(s)
- Oliver Kepp
- Equipe labellisée par la Ligue contre le cancer, Centre de Recherche des Cordeliers, INSERM U1138, Sorbonne Université, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Lucillia Bezu
- Equipe labellisée par la Ligue contre le cancer, Centre de Recherche des Cordeliers, INSERM U1138, Sorbonne Université, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | | | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Qld, Australia
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Centre de Recherche des Cordeliers, INSERM U1138, Sorbonne Université, Université de Paris, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.,Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.,Université de Paris, Paris, France
| |
Collapse
|
50
|
Boes DM, Godoy-Hernandez A, McMillan DGG. Peripheral Membrane Proteins: Promising Therapeutic Targets across Domains of Life. MEMBRANES 2021; 11:346. [PMID: 34066904 PMCID: PMC8151925 DOI: 10.3390/membranes11050346] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/28/2021] [Accepted: 05/05/2021] [Indexed: 11/16/2022]
Abstract
Membrane proteins can be classified into two main categories-integral and peripheral membrane proteins-depending on the nature of their membrane interaction. Peripheral membrane proteins are highly unique amphipathic proteins that interact with the membrane indirectly, using electrostatic or hydrophobic interactions, or directly, using hydrophobic tails or GPI-anchors. The nature of this interaction not only influences the location of the protein in the cell, but also the function. In addition to their unique relationship with the cell membrane, peripheral membrane proteins often play a key role in the development of human diseases such as African sleeping sickness, cancer, and atherosclerosis. This review will discuss the membrane interaction and role of periplasmic nitrate reductase, CymA, cytochrome c, alkaline phosphatase, ecto-5'-nucleotidase, acetylcholinesterase, alternative oxidase, type-II NADH dehydrogenase, and dihydroorotate dehydrogenase in certain diseases. The study of these proteins will give new insights into their function and structure, and may ultimately lead to ground-breaking advances in the treatment of severe diseases.
Collapse
Affiliation(s)
- Deborah M. Boes
- Department of Biotechnology, Delft University of Technology, Van der Maasweg 9, NL-2629 HZ Delft, The Netherlands; (D.M.B.); (A.G.-H.)
| | - Albert Godoy-Hernandez
- Department of Biotechnology, Delft University of Technology, Van der Maasweg 9, NL-2629 HZ Delft, The Netherlands; (D.M.B.); (A.G.-H.)
| | - Duncan G. G. McMillan
- Department of Biotechnology, Delft University of Technology, Van der Maasweg 9, NL-2629 HZ Delft, The Netherlands; (D.M.B.); (A.G.-H.)
- School of Fundamental Sciences, Massey University, Palmerston North, Private Bag 11 222, New Zealand
| |
Collapse
|