1
|
Zou Y, Wan X, Zhou Q, Zhu G, Lin S, Tang Q, Yang X, Wang S. Mechanisms of drug resistance in hepatocellular carcinoma. Biol Proced Online 2025; 27:19. [PMID: 40437363 PMCID: PMC12117952 DOI: 10.1186/s12575-025-00281-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 05/12/2025] [Indexed: 06/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent form of primary liver cancer, associated with high morbidity and mortality worldwide. Despite advancements in diagnostic methods and systemic treatments, including tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs), the development of drug resistance remains a significant challenge in HCC management. Traditional treatments such as surgical resection and transarterial chemoembolization offer limited efficacy, especially in advanced stages. Although novel therapies like lenvatinib, sorafenib, regorafenib, and ICIs have shown promise, their effectiveness is often hindered by primary and acquired resistance, leading to poor long-term survival outcomes. This review focuses on the molecular mechanisms underlying resistance to targeted therapies and immunotherapies in HCC. Key factors contributing to resistance include alterations in the tumor microenvironment (TME), immune evasion, hypoxia, changes in cellular metabolism, and genetic mutations. Additionally, molecular players such as ferroptosis, autophagy, apoptosis, endoplasmic reticulum stress, ABC transporters, and non-coding RNAs(ncRNAs) are discussed as contributors to drug resistance. Understanding these mechanisms is critical for the development of novel therapeutic strategies aimed at overcoming resistance, improving patient outcomes, and ultimately enhancing survival rates in HCC patients.
Collapse
Affiliation(s)
- Yongchun Zou
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Xinliang Wan
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Qichun Zhou
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Gangxing Zhu
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Shanshan Lin
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Qing Tang
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China.
| | - Xiaobing Yang
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China.
| | - Sumei Wang
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China.
| |
Collapse
|
2
|
Nikkilä R, Mäkitie A, Joensuu H, Markkanen S, Elenius K, Monni O, Palotie A, Saarentaus E, Salo T, Bizaki-Vallaskangas A. Novel Genetic Risk Variants Associated with Oral Tongue Squamous Cell Carcinoma. Head Neck Pathol 2025; 19:45. [PMID: 40278994 PMCID: PMC12031715 DOI: 10.1007/s12105-025-01784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 03/28/2025] [Indexed: 04/26/2025]
Abstract
PURPOSE Limited data from genome-wide association studies (GWAS) focusing on oral tongue squamous cell carcinoma (OTSCC) are available. The present study was conducted to explore genetic associations for OTSCC. METHODS A GWAS on 376 cases of OTSCC was conducted using the FinnGen Data Freeze-12 dataset. The case-cohort included 205 males and 171 females. Cases with malignancies involving the base of the tongue or lingual tonsil were excluded from the case-cohort. Individuals with no recorded history of malignancy were used as controls (n = 407,067). A Phenome-wide association study (PheWAS) was performed for the lead variants to assess their co-associations with other cancers. RESULTS GWAS analysis identified three genome-wide significant loci associated with OTSCC (p < 5 × 10-8), located at 5p15.33 (rs27067 near gene LINC01511), 10q24 (rs1007771191 near RPS3AP36), and 20p12.3 (rs1438070080 near PLCB1), respectively. PheWAS showed associations of rs27067 mainly with prostate cancer (OR = 1.06, p = 5.41 × 10-7), and seborrheic keratosis (OR = 1.11, p = 1.51 × 10-11). A co-directional effect with melanoma was also observed (OR = 0.93, p = 6.24 × 10-5). CONCLUSION The GWAS detected two novel genetic associations with OTSCC. Further research is needed to identify the genes at these loci that contribute to the molecular pathogenesis of OTSCC.
Collapse
Affiliation(s)
- Rayan Nikkilä
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer and Research, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Antti Mäkitie
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Heikki Joensuu
- Department of Oncology, HUS Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Saara Markkanen
- Department of Otolaryngology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- The Wellbeing Services County of Pirkanmaa, Tampere, Finland
| | - Klaus Elenius
- Institute of Biomedicine, and MediCity Research Laboratory, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Oncology, Turku University Hospital, Turku, Finland
| | - Outi Monni
- Department of Oncology, HUS Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland and the Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology, and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Elmo Saarentaus
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland and the Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pathology, HUS Helsinki University Hospital, Helsinki, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - Argyro Bizaki-Vallaskangas
- Department of Otolaryngology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- The Wellbeing Services County of Pirkanmaa, Tampere, Finland.
| |
Collapse
|
3
|
Sharafat RH, Saeed A. Ectonucleotidase inhibitors: targeting signaling pathways for therapeutic advancement-an in-depth review. Purinergic Signal 2025; 21:221-265. [PMID: 38958821 PMCID: PMC12061838 DOI: 10.1007/s11302-024-10031-0] [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: 12/12/2023] [Accepted: 06/16/2024] [Indexed: 07/04/2024] Open
Abstract
Ectonucleotidase inhibitors are a family of pharmacological drugs that, by selectively targeting ectonucleotidases, are essential in altering purinergic signaling pathways. The hydrolysis of extracellular nucleotides and nucleosides is carried out by these enzymes, which include ectonucleoside triphosphate diphosphohydrolases (NTPDases) and ecto-5'-nucleotidase (CD73). Ectonucleotidase inhibitors can prevent the conversion of ATP and ADP into adenosine by blocking these enzymes and reduce extracellular adenosine. These molecules are essential for purinergic signaling, which is associated with a variability of physiological and pathological processes. By modifying extracellular nucleotide metabolism and improving purinergic signaling regulation, ectonucleotide pyrophosphatase/phosphodiesterase (ENPP) inhibitors have the potential to improve cancer treatment, inflammatory management, and immune response modulation. Purinergic signaling is affected by CD73 inhibitors because they prevent AMP from being converted to adenosine. These inhibitors are useful in cancer therapy and immunotherapy because they may improve chemotherapy effectiveness and alter immune responses. Purinergic signaling is controlled by NTPDase inhibitors, which specifically target enzymes involved in extracellular nucleotide breakdown. These inhibitors show promise in reducing immunological responses, thrombosis, and inflammation, perhaps assisting in the treatment of cardiovascular and autoimmune illnesses. Alkaline phosphatase (ALP) inhibitors alter the function of enzymes involved in dephosphorylation reactions, which has an impact on a variety of biological processes. By altering the body's phosphate levels, these inhibitors may be used to treat diseases including hyperphosphatemia and certain bone problems. This article provides a guide for researchers and clinicians looking to leverage the remedial capability of ectonucleotidase inhibitors in a variety of illness scenarios by illuminating their processes, advantages, and difficulties.
Collapse
Affiliation(s)
- R Huzaifa Sharafat
- Department of Chemistry, Quaid-I-Azam University, Islamabad, 45321, Pakistan
| | - Aamer Saeed
- Department of Chemistry, Quaid-I-Azam University, Islamabad, 45321, Pakistan.
| |
Collapse
|
4
|
Jaafar HM, Ameen DMH, Mohammad TAM, Jafaar AM. The effects of nanocurcumin on immune-related factors in the ankylosing spondylitis patients: a double-blind, randomized, placebo-controlled clinical trial. Mol Biol Rep 2025; 52:324. [PMID: 40106064 DOI: 10.1007/s11033-025-10397-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND A rheumatic condition characterized by inflammation and increased bone mass at the primary sites of inflammation is called ankylosing spondylitis (AS). The pathophysiology of AS has been linked to Th17 and Regulatory T (Treg) cells, as well as the immunological and miRNA variables that are associated with them. This study looked at how immunological and miRNA variables in AS patients' peripheral blood (PB) were affected by nanocurcumin. METHODS For four months, 30 patients with AS were part of the test group, who got nanocurcumin daily, and 30 patients in the control group, who received a placebo. Using flow cytometry, the frequency of Th17 and Treg was determined. Real-time polymerase chain reaction was used to measure the expression levels of HIF1A, various related microRNAs (miRNAs; miR-18a, miR-206, and miR-30a), and CD39 with CD73 in Patients PBMCs. An enzyme-linked immunosorbent assay was also used to measure the amounts of factors and cytokines secreted in serum. RESULTS Following treatment with nanocurcumin, patients with AS experienced a large rise in Treg cells and a decrease in Th17 cells. The RT-PCR results showed that while miR-18a was dramatically upregulated after nanocurcumin treatment, the expression of miR-206 and miR-30a was significantly downregulated. Additionally, compared to the control group, the nanocurcumin-treated group had lower levels of HIF-1alpha production and higher levels of IGF-1, TGF-β, CD39, and CD73. CONCLUSION According to the findings, Th17 and Treg cell dysregulation, along with other immunological factors and miRNAs in PB, affects the progression of AS. Nanocurcumin therapy has the potential to control these variables, as well as Th17 and Treg cells, and may, therefore, help treat AS and other autoimmune disorders.
Collapse
Affiliation(s)
- Halmat M Jaafar
- College of Pharmacy, Hawler Medical University (HMU), Erbil, Kurdistan Region, Iraq
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | | | - Talar Ahmad Merza Mohammad
- College of Pharmacy, Hawler Medical University (HMU), Erbil, Kurdistan Region, Iraq.
- Pharmacy Department, School of Medicine, Hawler Medical University (HMU), Erbil, Kurdistan Region, Iraq.
| | - Aziz Muzafar Jafaar
- KRG-MOH, Erbil Teaching Hospital (Head of Endocrinology Department), Erbil, Iraq
| |
Collapse
|
5
|
Hosseinkhani N, Alipour S, Ghaffari Jolfayi A, Aghebati-Maleki L, Baghbani E, Alizadeh N, Khaze V, Baradaran B. Docetaxel treatment together with CTLA-4 knockdown enhances reduction of cell viability and amplifies apoptosis stimulation of MCF-7 breast cancer cells. Cytotechnology 2025; 77:19. [PMID: 39676767 PMCID: PMC11638433 DOI: 10.1007/s10616-024-00677-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024] Open
Abstract
Breast cancer is the most frequent cancer in women with a 20% mortality rate. The fate of patients suffering from breast cancer can be influenced by immune cells and tumor cells interaction in the tumor microenvironment (TME). Immune checkpoints such as Cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) are regulators of the immune system and defend normal tissues from immune cell attacks but they can be expressed in breast cancer tissue and facilitate immune evasion of tumoral cells. Based on this, here we studied the role of CTLA-4 silencing by specific siRNA in MCF-7 breast cancer cell line together with Docetaxel treatment which is one of the robust chemotherapy agents to demonstrate the significance of combining chemotherapy with efficient targeted therapy in tumor regression. The MCF-7 breast cancer cell line was transfected with CTLA-4-siRNA through the electroporation method, then received an appropriate dose of Docetaxel determined by MTT assay. Flow cytometry was utilized to investigate the consequence of simultaneous CTLA-4 gene silencing and Docetaxel treatment on the apoptosis and cell cycle of MCF-7 cells. The expression levels of Bax and Bcl-2 were also investigated using quantitative real-time PCR. Compared to control groups, CTLA-4-suppressed and Docetaxel-treated cells became more susceptible to apoptosis and cell cycle arrest at the G2-M phase. The additive effect of CTLA-4 knockdown together with Docetaxel treatment significantly downregulated BCL-2 level and upregulated BAX expression. Our findings support the idea that combining chemotherapy such as Docetaxel with efficient targeted therapy against inhibitory immune checkpoints can be a promising strategy in cancer treatment.
Collapse
Affiliation(s)
- Negar Hosseinkhani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Alipour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghaffari Jolfayi
- Cardiovascular Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Zhu J, Du W, Zeng Y, Liu T, Li J, Wang A, Li Y, Zhang W, Huang JA, Liu Z. CD73 promotes non-small cell lung cancer metastasis by regulating Axl signaling independent of GAS6. Proc Natl Acad Sci U S A 2024; 121:e2404709121. [PMID: 39423241 PMCID: PMC11513981 DOI: 10.1073/pnas.2404709121] [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: 03/06/2024] [Accepted: 09/12/2024] [Indexed: 10/21/2024] Open
Abstract
As catabolic enzyme, CD73 dephosphorylates adenosine monophosphate (AMP) and can also regulate tumor cell proliferation and metastasis. To date, very few studies have explored the role of CD73 in mediating non-small cell lung cancer (NSCLC) metastasis, and the underlying transducing signal has not been elucidated. In the present study, we demonstrated that the CD73/Axl axis could regulate Smad3-induced epithelial-to-mesenchymal transition (EMT) to promote NSCLC metastasis. Mechanically, CD73 can be secreted via the Golgi apparatus transport pathway. Then secreted CD73 may activate AXl by directly bind with site R55 located in Axl extracellular domain independently of GAS6. In addition, we proved that CD73 can stabilize Axl expression via inhibiting CBLB expression. We also identified the distinct function of CD73 activity in adenocarcinoma and squamous cell carcinoma. Our findings indicated a role of CD73 in mediating NSCLC metastasis and propose it as a therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Jianjie Zhu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| | - Wenwen Du
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| | - Yuanyuan Zeng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| | - Ting Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
| | - Jianjun Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| | - Anqi Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| | - Yue Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
| | - Weijie Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
| | - Jian-an Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| | - Zeyi Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou215006, China
| |
Collapse
|
7
|
Gardani CFF, Diz FM, Dondé LB, Rockenbach L, Laufer S, Morrone FB. The potential role of purinergic signaling in cancer therapy: perspectives on anti-CD73 strategies for prostate cancer. Front Immunol 2024; 15:1455469. [PMID: 39355246 PMCID: PMC11442216 DOI: 10.3389/fimmu.2024.1455469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/12/2024] [Indexed: 10/03/2024] Open
Abstract
Purines and pyrimidines are signaling molecules in the tumor microenvironment that affect cancer immunity. The purinergic signaling pathways have been shown to play an important role in the development and progression of cancer. CD39 and CD73 are ectonucleotidases responsible for breaking down ATP or ADP into adenosine, which regulates immunosuppression in various types of cancer. These enzymes have been studied as a potential therapeutic target in immunotherapy, and recent research suggests a correlation between ectonucleotidases and clinical outcomes in cancer.Prostate cancer is the most diagnosed cancer in men, after non-melanoma skin tumors, and is the second leading cause of death in men in the world. Despite having long survival periods, patients often receive excessive or insufficient treatment. Within this complex landscape, the adenosine/CD73 pathway plays a crucial role. Therefore, this review aims to highlight new findings on the potential role of purinergic signaling in cancer treatment and emphasizes the importance of anti-CD73 as a pharmacological strategy for prostate cancer therapy.
Collapse
Affiliation(s)
- Carla Fernanda Furtado Gardani
- Escola de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernando Mendonça Diz
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Centro de Pesquisa Pré-Clínica, Instituto do Cerebro do Rio Grande do Sul (InsCer), Pontíficia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luísa Brandalise Dondé
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Liliana Rockenbach
- Escola de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Stefan Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Fernanda Bueno Morrone
- Escola de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Escola de Ciências da Saúde e da Vida, Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
8
|
Poblano-Pérez LI, Monroy-García A, Fragoso-González G, Mora-García MDL, Castell-Rodríguez A, Mayani H, Álvarez-Pérez MA, Pérez-Tapia SM, Macías-Palacios Z, Vallejo-Castillo L, Montesinos JJ. Mesenchymal Stem/Stromal Cells Derived from Dental Tissues Mediate the Immunoregulation of T Cells through the Purinergic Pathway. Int J Mol Sci 2024; 25:9578. [PMID: 39273524 PMCID: PMC11395442 DOI: 10.3390/ijms25179578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Human dental tissue mesenchymal stem cells (DT-MSCs) constitute an attractive alternative to bone marrow-derived mesenchymal stem cells (BM-MSCs) for potential clinical applications because of their accessibility and anti-inflammatory capacity. We previously demonstrated that DT-MSCs from dental pulp (DP-MSCs), periodontal ligaments (PDL-MSCs), and gingival tissue (G-MSCs) show immunosuppressive effects similar to those of BM, but to date, the DT-MSC-mediated immunoregulation of T lymphocytes through the purinergic pathway remains unknown. In the present study, we compared DP-MSCs, PDL-MSCs, and G-MSCs in terms of CD26, CD39, and CD73 expression; their ability to generate adenosine (ADO) from ATP and AMP; and whether the concentrations of ADO that they generate induce an immunomodulatory effect on T lymphocytes. BM-MSCs were included as the gold standard. Our results show that DT-MSCs present similar characteristics among the different sources analyzed in terms of the properties evaluated; however, interestingly, they express more CD39 than BM-MSCs; therefore, they generate more ADO from ATP. In contrast to those produced by BM-MSCs, the concentrations of ADO produced by DT-MSCs from ATP inhibited the proliferation of CD3+ T cells and promoted the generation of CD4+CD25+FoxP3+CD39+CD73+ Tregs and Th17+CD39+ lymphocytes. Our data suggest that DT-MSCs utilize the adenosinergic pathway as an immunomodulatory mechanism and that this mechanism is more efficient than that of BM-MSCs.
Collapse
Affiliation(s)
- Luis Ignacio Poblano-Pérez
- Mesenchymal Stem Cell Laboratory, Oncology Research Unit, Oncology Hospital, Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Alberto Monroy-García
- Immunology and Cancer Laboratory, Oncology Research Unit, Oncology Hospital, Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Gladis Fragoso-González
- Institute of Biomedical Research, Department of Immunology, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - María de Lourdes Mora-García
- Immunobiology Laboratory, Cell Differentiation and Cancer Unit, Facultad de Estudios Superiores-Zaragoza, Universidad Nacional Autónoma de México, Mexico City 09230, Mexico
| | - Andrés Castell-Rodríguez
- Department of Cellular and Tissue Biology, Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Héctor Mayani
- Hematopoietic Stem Cell Laboratory, Oncology Research Unit, Oncology Hospital, Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Marco Antonio Álvarez-Pérez
- Tissue Bioengineering Laboratory, Postgraduate Studies, Research Division, Faculty of Dentistry, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Sonia Mayra Pérez-Tapia
- Research and Development in Biotherapeutic Unit (UDIBI), National School of Biological Sciences, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- National Laboratory for Specialized Services of Investigation, Development and Innovation (I+D+i) for Pharma Chemicals and Biotechnological Products (LANSEIDI-FarBiotec-CONACyT), Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Department of Immunology, National School of Biological Sciences, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Zaira Macías-Palacios
- Research and Development in Biotherapeutic Unit (UDIBI), National School of Biological Sciences, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- National Laboratory for Specialized Services of Investigation, Development and Innovation (I+D+i) for Pharma Chemicals and Biotechnological Products (LANSEIDI-FarBiotec-CONACyT), Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Luis Vallejo-Castillo
- Research and Development in Biotherapeutic Unit (UDIBI), National School of Biological Sciences, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- National Laboratory for Specialized Services of Investigation, Development and Innovation (I+D+i) for Pharma Chemicals and Biotechnological Products (LANSEIDI-FarBiotec-CONACyT), Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Juan José Montesinos
- Mesenchymal Stem Cell Laboratory, Oncology Research Unit, Oncology Hospital, Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| |
Collapse
|
9
|
Mirza MR, Tandaric L, Henriksen JR, Mäenpää J, Christensen RD, Waldstrøm M, Lindemann K, Roed H, Auranen A, Akslen LA, Thomsen LCV, Lindberg SN, Madsen K, Bjørge L. NSGO-OV-UMB1/ENGOT-OV30: A phase II study of durvalumab in combination with the anti-CD73 monoclonal antibody Oleclumab in patients with relapsed ovarian cancer. Gynecol Oncol 2024; 188:103-110. [PMID: 38943691 DOI: 10.1016/j.ygyno.2024.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
OBJECTIVES In patients with epithelial ovarian cancer (EOC), the clinical efficacy of monotherapy with immune checkpoint inhibitors (ICIs) against PD-1/PD-L1 is modest. To enhance response rates to these immunotherapeutic agents and broaden the indications for their use, new approaches involving combinational therapy are needed. The immune regulator CD73 is a potential target, as it promotes tumor escape by producing immunosuppressive extracellular adenosine in the tumor microenvironment. Here, we present the results from the NSGO-OV-UMB1/ENGOT-OV-30 trial evaluating the activity of combining the anti-CD73 antibody oleclumab with the anti-PD-L1 checkpoint inhibitor durvalumab in patients with recurrent EOC. METHODS In this phase II open-label non-randomized study, patients with CD73-positive relapsed EOC were intravenously administered oleclumab (3000 mg, Q2W) and durvalumab (1500 mg, Q4W). The primary endpoint was disease control rate (DCR) at 16 weeks. The expression of PD-L1 and CD8 was assessed by immunohistochemistry of archival tumors. RESULTS This trial included 25 patients with a median age of 66 years (47-77 years). Twenty-two patients were evaluable for treatment activity analysis. The DCR was 27%, the median progression-free survival was 2.7 months (95% CI: 2.2-4.2) and the median overall survival was 8.4 months (95% CI: 5.0-13.4). Infiltration of CD8+ cells and PD-L1 expression on tumor cells were observed in partially overlapping sets of 74% of the tumor samples. Neither CD8- nor PD-L1-positivity were significantly associated with better DCR. CONCLUSIONS Combined treatment with oleclumab and durvalumab was safe and demonstrated limited anti-tumor activity in patients with recurrent EOC.
Collapse
MESH Headings
- Humans
- Female
- Middle Aged
- 5'-Nucleotidase/antagonists & inhibitors
- 5'-Nucleotidase/immunology
- Aged
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/pharmacology
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/pathology
- Carcinoma, Ovarian Epithelial/drug therapy
- Carcinoma, Ovarian Epithelial/immunology
- Carcinoma, Ovarian Epithelial/pathology
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/pathology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- GPI-Linked Proteins/immunology
- GPI-Linked Proteins/antagonists & inhibitors
- Adult
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/pharmacology
Collapse
Affiliation(s)
- M R Mirza
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Nordic Society of Gynaecological Oncology Clinical Trial Unit (NSGO-CTU), Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
| | - L Tandaric
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Norway; Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - J R Henriksen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - J Mäenpää
- Department of Obstetrics and Gynecology and Tays Cancer Centre, Tampere University Hospital and Tampere University, Finland
| | - R D Christensen
- Research Unit of General Practice, University of Southern Denmark, Institute of Public Health, Odense, Denmark
| | - M Waldstrøm
- Department of Pathology, Hvidovre Hospital, Hvidovre, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - K Lindemann
- Department of Gynecological Oncology, Oslo University Hospital, Division of Cancer Medicine, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - H Roed
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - A Auranen
- Department of Obstetrics and Gynecology and Tays Cancer Centre, Tampere University Hospital and Tampere University, Finland
| | - L A Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Section for Pathology, University of Bergen, Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - L C V Thomsen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Norway; Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - S N Lindberg
- Nordic Society of Gynaecological Oncology Clinical Trial Unit (NSGO-CTU), Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - K Madsen
- Nordic Society of Gynaecological Oncology Clinical Trial Unit (NSGO-CTU), Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - L Bjørge
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Norway; Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
10
|
Shukla S, Dalai P, Agrawal-Rajput R. Metabolic crosstalk: Extracellular ATP and the tumor microenvironment in cancer progression and therapy. Cell Signal 2024; 121:111281. [PMID: 38945420 DOI: 10.1016/j.cellsig.2024.111281] [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: 04/28/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Adenosine 5'-triphosphate (ATP) is a vital element in energy information. It plays a critical role in transmitting signals inside the body, which is necessary for controlling the life activities of all cells, including tumor cells [1]. Its significance extends from intracellular signaling pathways to tumor regression. Purinergic signaling, a form of extracellular paracrine signaling, relies on purine nucleotides. Extracellular ectonucleotidases convert these purine nucleotides to their respective di and mono-phosphate nucleoside forms, contributing significantly to immune biology, cancer biology, and inflammation studies. ATP functions as a mighty damage-linked molecular pattern when released outside the cell, accumulating in inflammatory areas. In the tumor microenvironment (TME), purinergic receptors such as ATP-gated ion channels P2X1-5 and G protein-coupled receptors (GPCR) (P2Y) interact with ATP and other nucleotides, influencing diverse immune cell activities. CD39 and CD73-mediated extracellular ATP degradation contributes to immunosuppression by diminishing ATP-dependent activation and generating adenosine (ADO), potentially hindering antitumor immunity and promoting tumor development. Unraveling the complexities of extracellular ATP (e-ATP) and ADO effects on the TME poses challenges in identifying optimal treatment targets, yet ongoing investigations aim to devise strategies combating e-ATP/ADO-induced immunosuppression, ultimately enhancing anti-tumor immunity. This review explores e-ATP metabolism, its purinergic signaling, and therapeutic strategies targeting associated receptors and enzymes.
Collapse
Affiliation(s)
- Sourav Shukla
- Immunology Lab, Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar 382426, Gujarat, India
| | - Parameswar Dalai
- Immunology Lab, Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar 382426, Gujarat, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar 382426, Gujarat, India.
| |
Collapse
|
11
|
Kaplinsky N, Williams K, Watkins D, Adams M, Stanbery L, Nemunaitis J. Regulatory role of CD39 and CD73 in tumor immunity. Future Oncol 2024; 20:1367-1380. [PMID: 38652041 PMCID: PMC11321403 DOI: 10.2217/fon-2023-0871] [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: 10/12/2023] [Accepted: 03/07/2024] [Indexed: 04/25/2024] Open
Abstract
CD39 is the rate-limiting enzyme for the molecular signal cascade leading to the generation of ADP and adenosine monophosphate (AMP). In conjunction with CD73, CD39 converts adenosine triphosphate (ATP) to ADP and AMP, which leads to the accumulation of immunosuppressive adenosine in the tumor microenvironment. This review focuses on the role of CD39 and CD73 in immune response and malignant progression, including the expression of CD39 within the tumor microenvironment and its relationship to immune effector cells, and its role in antigen presentation. The role of CD39- and CD73-targeting therapeutics and cancer-directed clinical trials investigating CD39 modulation are also explored.
Collapse
Affiliation(s)
| | - Kada Williams
- University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Dean Watkins
- University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Molly Adams
- University of Toledo College of Medicine, Toledo, OH 43614, USA
| | | | | |
Collapse
|
12
|
Wang H, Wei Y, Wang N. Purinergic pathways and their clinical use in the treatment of acute myeloid leukemia. Purinergic Signal 2024:10.1007/s11302-024-09997-8. [PMID: 38446337 DOI: 10.1007/s11302-024-09997-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Despite the use of various therapies such as hematopoietic stem cell transplantation and chimeric antigen receptor T cell therapy (CAR-T), the prognosis of patients with acute myeloid leukemia (AML) is still generally poor. However, immunotherapy is currently a hot topic in the treatment of hematological tumors. Extracellular adenosine triphosphate (ATP) can be converted to adenosine diphosphate (ADP) via CD39, and ADP can be converted to adenosine via CD73, which can bind to P1 and P2 receptors to exert immunomodulatory effects. Research on the mechanism of the purinergic signaling pathway can provide a new direction for the treatment of AML, and inhibitors of this signaling pathway have been discovered by several researchers and gradually applied in the clinic. In this paper, the mechanism of the purinergic signaling pathway and its clinical application are described, revealing a new target for the treatment of AML and subsequent improvement in patient prognosis.
Collapse
Affiliation(s)
- Huijuan Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yujie Wei
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Na Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
13
|
Zhang C, Wang K, Wang H. Adenosine in cancer immunotherapy: Taking off on a new plane. Biochim Biophys Acta Rev Cancer 2023; 1878:189005. [PMID: 37913941 DOI: 10.1016/j.bbcan.2023.189005] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023]
Abstract
As a new pillar of cancer therapy, tumor immunotherapy has brought irreplaceable durable responses in tumors. Considering its low response rate, additional immune regulatory mechanisms will be critical for the development of next-generation immune therapeutics. As a key regulatory mechanism, adenosine (ADO) protects tissues from excessive immune responses, but as a metabolite highly concentrated in tumor microenvironments, extracellular adenosine acts on adenosine receptors (mainly A2A receptors) expressed on MDSCs, Tregs, NK cells, effector T cells, DCs, and macrophages to promote tumor cell escape from immune surveillance by inhibiting the immune response. Amounting preclinical studies have demonstrated the adenosine pathway as a novel checkpoint for immunotherapy. Large number of adenosine pathway targeting clinical trials are now underway, including antibodies against CD39 and CD73 as well as A2A receptor inhibitors. There has been evidence of antitumor efficacy of these inhibitors in early clinical trials among a variety of tumors such as breast cancer, prostate cancer, non-small cell lung cancer, etc. As more clinical trial results are published, the combination of blockade of this pathway with immune checkpoint inhibitors, targeted drugs, traditional chemotherapy medications, radiotherapy and endocrine therapy will provide cancer patients with better clinical outcomes. We would elaborate on the role of CD39-CD73-A2AR pathway in the contribution of tumor microenvironment and the targeting of the adenosinergic pathway for cancer therapy in the review.
Collapse
Affiliation(s)
- Chenyue Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Kai Wang
- Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
14
|
Bakhtiyari M, Liaghat M, Aziziyan F, Shapourian H, Yahyazadeh S, Alipour M, Shahveh S, Maleki-Sheikhabadi F, Halimi H, Forghaniesfidvajani R, Zalpoor H, Nabi-Afjadi M, Pornour M. The role of bone marrow microenvironment (BMM) cells in acute myeloid leukemia (AML) progression: immune checkpoints, metabolic checkpoints, and signaling pathways. Cell Commun Signal 2023; 21:252. [PMID: 37735675 PMCID: PMC10512514 DOI: 10.1186/s12964-023-01282-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/17/2023] [Indexed: 09/23/2023] Open
Abstract
Acute myeloid leukemia (AML) comprises a multifarious and heterogeneous array of illnesses characterized by the anomalous proliferation of myeloid cells in the bone marrow microenvironment (BMM). The BMM plays a pivotal role in promoting AML progression, angiogenesis, and metastasis. The immune checkpoints (ICs) and metabolic processes are the key players in this process. In this review, we delineate the metabolic and immune checkpoint characteristics of the AML BMM, with a focus on the roles of BMM cells e.g. tumor-associated macrophages, natural killer cells, dendritic cells, metabolic profiles and related signaling pathways. We also discuss the signaling pathways stimulated in AML cells by BMM factors that lead to AML progression. We then delve into the roles of immune checkpoints in AML angiogenesis, metastasis, and cell proliferation, including co-stimulatory and inhibitory ICs. Lastly, we discuss the potential therapeutic approaches and future directions for AML treatment, emphasizing the potential of targeting metabolic and immune checkpoints in AML BMM as prognostic and therapeutic targets. In conclusion, the modulation of these processes through the use of directed drugs opens up new promising avenues in combating AML. Thereby, a comprehensive elucidation of the significance of these AML BMM cells' metabolic and immune checkpoints and signaling pathways on leukemic cells can be undertaken in the future investigations. Additionally, these checkpoints and cells should be considered plausible multi-targeted therapies for AML in combination with other conventional treatments in AML. Video Abstract.
Collapse
Affiliation(s)
- Maryam Bakhtiyari
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mahsa Liaghat
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Fatemeh Aziziyan
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hooriyeh Shapourian
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sheida Yahyazadeh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maedeh Alipour
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Shaghayegh Shahveh
- American Association of Naturopath Physician (AANP), Washington, DC, USA
| | - Fahimeh Maleki-Sheikhabadi
- Department of Hematology and Blood Banking, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Halimi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razieh Forghaniesfidvajani
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Majid Pornour
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA.
| |
Collapse
|
15
|
Mougola Bissiengou P, Montcho Comlan JG, Atsame Ebang G, Sylla Niang M, Djoba Siawaya JF. Prostate malignant tumor and benign prostatic hyperplasia microenvironments in black African men: Limited infiltration of CD8+ T lymphocytes, NK-cells, and high frequency of CD73+ stromal cells. Cancer Rep (Hoboken) 2023; 6 Suppl 1:e1817. [PMID: 37092584 PMCID: PMC10440842 DOI: 10.1002/cnr2.1817] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Anti-cancerous immunology has yet to be investigated in the African black population, despite being the dawn of precision medicine. AIM Here we investigated the tumor microenvironment of prostate cancer and benign prostatic hyperplasia (BPH) in black Africans. METHODS Through immunohistochemistry analysis of prostate cancer and BPH patients' biopsies, we investigated the expression and distribution of CD73, CCD8 T-lymphocytes, and natural killer cells. In addition, we looked at tumor-infiltrating features CD8 T-lymphocytes and natural killer cells. RESULTS We show for the first time in black Africans a high expression of CD73 in epithelial-stromal cells and virtually no infiltration of CD8 T lymphocytes and natural killer cells in the tumoral area. In addition, CD73 was seven (7) times more likely to be expressed in prostate cancer stromal tissues than in benign prostatic hyperplasia tissues (odds ratio = 7.2; χ2 = 21; p < .0001). In addition, PSA concentration was significantly higher in prostate cancer patients than in BPH patients (p < .001). Also, the PSA-based ROC. analysis showed an area under the curve of 0.87 (p < .0001). CONCLUSION CD73 expression is more likely expressed in prostate cancer stromal tissues than in benign prostatic hyperplasia tissues. The features of prostate cancer in Black Africans suggest CD73 expression as a possible target for immunotherapy in this population.
Collapse
Affiliation(s)
- Pélagie Mougola Bissiengou
- Service d'Immunologie, Département des Sciences Fondamentales, Faculté de MédecineUniversité des Sciences de la SantéLibrevilleGabon
- Service d'Immunologie, Département des Sciences Biologiques et Pharmaceutiques Appliquées, Faculté de Médecine, de Pharmacie et d'OdontostomatologieUniversité Cheikh Anta DiopDakarSenegal
| | - Jérôme Gaston Montcho Comlan
- Service d'Immunologie, Département des Sciences Biologiques et Pharmaceutiques Appliquées, Faculté de Médecine, de Pharmacie et d'OdontostomatologieUniversité Cheikh Anta DiopDakarSenegal
| | - Gabrielle Atsame Ebang
- Unité d'anatomie‐Cytologie‐PathologieCentre Hospitalier Universitaire de LibrevilleLibrevilleGabon
| | - Maguette Sylla Niang
- Service d'Immunologie, Département des Sciences Biologiques et Pharmaceutiques Appliquées, Faculté de Médecine, de Pharmacie et d'OdontostomatologieUniversité Cheikh Anta DiopDakarSenegal
| | - Joel Fleury Djoba Siawaya
- Service LaboratoireCentre Hospitalier Universitaire Mère‐Enfant Fondation Jeanne EBORILibrevilleGabon
| |
Collapse
|
16
|
Liu Z, Quan L, Ma F, Yang M, Jiang X, Chen X. Determination of adenosine by CRISPR-Cas12a system based on duplexed aptamer and molecular beacon reporter linked to gold nanoparticles. Mikrochim Acta 2023; 190:173. [PMID: 37020072 PMCID: PMC10075494 DOI: 10.1007/s00604-023-05748-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/13/2023] [Indexed: 04/07/2023]
Abstract
Adenosine as a potential tumor marker is of great value for clinical disease diagnosis. Since the CRISPR-cas12a system is only capable of recognizing nucleic acid targets we expanded the CRISPR-cas12a system to determine small molecules by designing a duplexed aptamer (DA) converting g-RNA recognition of adenosine to recognition of aptamer complementary DNA strands (ACD). To further improve the sensitivity of determination, we designed a molecule beacon (MB)/gold nanoparticle (AuNP)-based reporter, which has higher sensitivity than traditional ssDNA reporter. In addition, the AuNP-based reporter enables more efficient and fast determination. The determination of adenosine under 488-nm excitation can be realized within 7 min, which is more than 4 times faster than traditional ssDNA reporter. The linear determination range of the assay to adenosine was 0.5-100 μM with the determination limit of 15.67 nM. The assay was applied to recovery determination of adenosine in serum samples with satisfactory results. The recoveries were between 91 and 106% and the RSD values of different concertation were below 4.8%. This sensitive, highly selective, and stable sensing system is expected to play a role in the clinical determination of adenosine and other biomolecules.
Collapse
Affiliation(s)
- Zhenhua Liu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | | | - Fanghui Ma
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Minghui Yang
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410083, China.
| | - Xinyu Jiang
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China.
| | - Xiang Chen
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, 410083, China.
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410000, China.
| |
Collapse
|
17
|
Zhulai GA, Shibaev MI. Relationship between the Gene Expression of Adenosine Kinase Isoforms and the Expression of CD39 and CD73 Ectonucleotidases in Colorectal Cancer. Acta Naturae 2023; 15:42-49. [PMID: 37538807 PMCID: PMC10395772 DOI: 10.32607/actanaturae.11871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/03/2023] [Indexed: 08/05/2023] Open
Abstract
Tumor cells have the capacity to create an adenosine-rich immunosuppressive environment, which can interfere with antitumor immunotherapy. Approaches are currently being developed with a view to suppressing the production of adenosine or its signals. Such approaches include the use of antibodies to inhibit CD39, CD73, and adenosine-receptor antagonists. However, the abundance of enzymatic pathways that control the ATP-adenosine balance, as well as the still poorly understood intracellular adenosine regulation, makes the hoped-for success unlikely. In the present study, the enzyme adenosine kinase (ADK) needed to convert adenosine to adenosine monophosphate, thereby regulating its levels, was investigated. To do so, peripheral blood samples from patients with colorectal cancer (CRC) (n = 31) were collected with blood samples from healthy donors (n = 17) used as controls. ADK gene expression levels and those of its long (ADK-L) and short (ADK-S) isoforms were measured. The relationship between the levels of ADK gene expression and that of CD39, CD73, and A2aR genes was analyzed. It turned out that in the group of CRC patients (stages III-IV), the level of ADK-L mRNA was lower (p < 0.0011) when compared to that of the control. For the first time, an average correlation was found between the level of expression of CD39 and ADK-S (r = -0.468 at p = 0.043) and between CD73 and ADK-L (r = 0.518 at p = 0.0232) in CRC patients. Flow cytometry was used to assess the content of CD39/CD73-expressing CD8+, CD4+ and Treg lymphocytes, as well as their relationship with the level of ADK gene expression in CRC patients. But no significant correlations were found.
Collapse
Affiliation(s)
- G A Zhulai
- Institute of Biology, Karelian Research Centre, Russian Academy of Sciences, Petrozavodsk, 185910 Russian Federation
| | - M I Shibaev
- Endoscopic Department, Baranov Republican Hospital, Petrozavodsk, 185910 Russian Federation
| |
Collapse
|
18
|
Amaro A, Reggiani F, Fenoglio D, Gangemi R, Tosi A, Parodi A, Banelli B, Rigo V, Mastracci L, Grillo F, Cereghetti A, Tastanova A, Ghosh A, Sallustio F, Emionite L, Daga A, Altosole T, Filaci G, Rosato A, Levesque M, Maio M, Pfeffer U, Croce M. Guadecitabine increases response to combined anti-CTLA-4 and anti-PD-1 treatment in mouse melanoma in vivo by controlling T-cells, myeloid derived suppressor and NK cells. J Exp Clin Cancer Res 2023; 42:67. [PMID: 36934257 PMCID: PMC10024396 DOI: 10.1186/s13046-023-02628-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/21/2023] [Indexed: 03/20/2023] Open
Abstract
BACKGROUND The combination of Programmed Cell Death 1 (PD-1) and Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) blockade has dramatically improved the overall survival rate for malignant melanoma. Immune checkpoint blockers (ICBs) limit the tumor's immune escape yet only for approximately a third of all tumors and, in most cases, for a limited amount of time. Several approaches to overcome resistance to ICBs are being investigated among which the addition of epigenetic drugs that are expected to act on both immune and tumor cells. Guadecitabine, a dinucleotide prodrug of a decitabine linked via phosphodiester bond to a guanosine, showed promising results in the phase-1 clinical trial, NIBIT-M4 (NCT02608437). METHODS We used the syngeneic B16F10 murine melanoma model to study the effects of immune checkpoint blocking antibodies against CTLA-4 and PD-1 in combination, with and without the addition of Guadecitabine. We comprehensively characterized the tumor's and the host's responses under different treatments by flow cytometry, multiplex immunofluorescence and methylation analysis. RESULTS In combination with ICBs, Guadecitabine significantly reduced subcutaneous tumor growth as well as metastases formation compared to ICBs and Guadecitabine treatment. In particular, Guadecitabine greatly enhanced the efficacy of combined ICBs by increasing effector memory CD8+ T cells, inducing effector NK cells in the spleen and reducing tumor infiltrating regulatory T cells and myeloid derived suppressor cells (MDSC), in the tumor microenvironment (TME). Guadecitabine in association with ICBs increased serum levels of IFN-γ and IFN-γ-induced chemokines with anti-angiogenic activity. Guadecitabine led to a general DNA-demethylation, in particular of sites of intermediate methylation levels. CONCLUSIONS These results indicate Guadecitabine as a promising epigenetic drug to be added to ICBs therapy.
Collapse
Affiliation(s)
- Adriana Amaro
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| | - Francesco Reggiani
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| | - Daniela Fenoglio
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Rosaria Gangemi
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| | - Anna Tosi
- Immunology and Molecular Oncology Diagnostics, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Alessia Parodi
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| | - Barbara Banelli
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| | - Valentina Rigo
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| | - Luca Mastracci
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| | - Federica Grillo
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| | - Alessandra Cereghetti
- Department of Dermatology, University of Zurich, University Hospital of Zurich, Zurich, Switzerland
| | - Aizhan Tastanova
- Department of Dermatology, University of Zurich, University Hospital of Zurich, Zurich, Switzerland
| | - Adhideb Ghosh
- Functional Genomics Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Fabio Sallustio
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Laura Emionite
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| | - Antonio Daga
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| | - Tiziana Altosole
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| | - Gilberto Filaci
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Diagnostics, Istituto Oncologico Veneto IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Mitchell Levesque
- Department of Dermatology, University of Zurich, University Hospital of Zurich, Zurich, Switzerland
| | | | - Ulrich Pfeffer
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy.
| | - Michela Croce
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| |
Collapse
|
19
|
Rahimifard K, Shahbazi M, Oliaei F, Akbari R, Tarighi M, Mohammadnia-Afrouzi M. Increased frequency of CD39 +CD73 + regulatory T cells and Deltex-1 gene expression level in kidney transplant recipients with excellent long-term graft function. Transpl Immunol 2023; 78:101823. [PMID: 36921728 DOI: 10.1016/j.trim.2023.101823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/27/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND The ability of regulatory T cells (Tregs) to limit inflammatory responses has been demonstrated. However, different subpopulations of this cell have varying abilities to suppress alloreactive immune responses. The primary goal of this study was to assess the frequency of CD4+FOXP3+CD39+CD73+ Tregs and Deltex-1 gene expression on long-term renal transplant function. METHODS A total of 49 subjects were divided into 3 groups: (i) the excellent long-term graft function (ELTGF) group, (ii) the chronic graft dysfunction (CGD) group, and (iii) the healthy control (HC) group. Following sample collection, peripheral blood mononuclear cells (PBMCs) were isolated, and the Deltex-1 gene expression level and the frequency of CD4+FOXP3+CD39+CD73+ Tregs were evaluated. RESULTS The ELTGF group had more CD4+FOXP3+ Tregs than the CGD group, but the difference was not statistically significant (P = 0.07). However, the frequency of CD4+FOXP3+CD39+CD73+ Tregs and the ratio of these cells to total CD4+ lymphocytes significantly increased in the ELTGF group than in the CGD group (P = 0.04 and P = 0.02 respectively). In addition, the expression level of the Deltex-1 gene was significantly lower in the CGD group than in the other 2 groups (P = 0.01 and P = 0.04 respectively). CONCLUSIONS Given the increased frequency of CD4+FOXP3+CD39+CD73+ Tregs and the expression level of the Deltex-1 gene in the ELTGF group, it appears that these factors probably improved function and long-term survival of the transplanted organ through the suppression of alloreactive responses and reduction of inflammation. In other words, one of the immunological mechanisms involved in the CGD group may be a deficiency in Tregs.
Collapse
Affiliation(s)
- Kimiya Rahimifard
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mehdi Shahbazi
- Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Farshid Oliaei
- Kidney Transplantation Center, Shahid Beheshti Hospital, Babol University of Medical Sciences, Babol, Iran
| | - Roghayeh Akbari
- Kidney Transplantation Center, Shahid Beheshti Hospital, Babol University of Medical Sciences, Babol, Iran
| | - Mona Tarighi
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mousa Mohammadnia-Afrouzi
- Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
20
|
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: 132] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [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
|
21
|
Ahangar NK, Khalaj-Kondori M, Alizadeh N, Mokhtarzadeh A, Baghbanzadeh A, Shadbad MA, Dolatkhah K, Baradaran B. Silencing tumor-intrinsic HHLA2 potentiates the anti-tumoral effect of paclitaxel on MG63 cells: Another side of immune checkpoint. Gene 2023; 855:147086. [PMID: 36535461 DOI: 10.1016/j.gene.2022.147086] [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: 04/14/2022] [Revised: 11/17/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Osteosarcoma is common type of bone cancer; however, the prognosis of patients with metastatic osteosarcoma is poor. As a new inhibitory immune checkpoint molecule, HHLA2 is upregulated in osteosarcoma. Herein, we studied the significance of tumor-intrinsic HHLA2 in MG-63 growth. Also, we examined the influence of combined therapy of HHLA2 knockdown with paclitaxel on the apoptosis, cell cycle, migration, and stemness of MG-63 cells. METHODS The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay was performed to study the half-maximal inhibitory concentration (IC50) of paclitaxel and the cytotoxicity of HHLA2-small interfering RNA (siRNA) on MG-63 cells. The apoptosis and cell cycle were analyzed using flow cytometry. The wound-healing and colony formation assays were conducted to investigate the effect of paclitaxel and HHLA2 knockdown on the migration and stemness of MG-63 cells, respectively. QRT-PCR was used to determine the Bax, caspase-3, and Bcl-2 mRNA expression levels. RESULTS HHLA2 silencing has enhanced the chemosensitivity of MG-63 cells to paclitaxel. Besides, HHLA2 knockdown has increased the paclitaxel-induced cytotoxic effect on MG-63 cells. In terms of stimulating apoptosis, decreasing clonogenicity, halting the cell cycle at the sub G1 phase, and inhibiting migration, tumor-intrinsic HHLA2 silencing has increased these anti-tumor effects of paclitaxel on MG-63 cells. Besides, HHLA2 knockdown has potentiated paclitaxel-mediated Bcl-2 downregulation and paclitaxel-mediated caspase-3 and Bax upregulation in MG-63 cells. CONCLUSION Tumor-intrinsic HHLA2 knockdown increases the anti-tumoral effect of paclitaxel on MG-63 cells and enhances the chemosensitivity of MG-63 cells to paclitaxel.
Collapse
Affiliation(s)
- Noora Karim Ahangar
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 5166616471, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 5166616471, Iran.
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Iran
| | | | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Iran.
| |
Collapse
|
22
|
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: 2.0] [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
|
23
|
Bai Y, Zhang X, Zheng J, Liu Z, Yang Z, Zhang X. Overcoming high level adenosine-mediated immunosuppression by DZD2269, a potent and selective A2aR antagonist. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:302. [PMID: 36229853 PMCID: PMC9563815 DOI: 10.1186/s13046-022-02511-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/03/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Adenosine is a potent immunosuppressant whose levels in the tumor microenvironment (TME) are often much higher than those in normal tissues. Binding of adenosine to its receptor A2aR activates a cascade of genes and leads to immunosuppression. In addition, immune checkpoint blockage markedly increases A2aR expression in T cells, which could dampen their anti-tumor response. Several A2aR antagonists are under clinical development, but with limited clinical benefit reported so far. These A2aR antagonists showed much diminished activity at high adenosine levels found in TME, which may explain their clinical underperformance. We report the discovery and early clinical development of DZD2269, a novel A2aR antagonist which can fully block A2aR mediated immunosuppression commonly found in TME. Adenosine stimulates phosphorylation of cyclic AMP response element binding protein (CREB) in T cells and inhibits anti-tumor cytokine secretion in PBMCs in a dose-dependent manner. DZD2269 was able to reverse the immunosuppression induced by high concentrations of adenosine, as demonstrated by inhibiting CREB phosphorylation in T cells, restoring Th1 cytokine secretion in PBMCs, and stimulating dendritic cells (DCs) maturation. As a single agent, DZD2269 showed anti-tumor growth in multiple syngeneic mouse tumor models, and more profound anti-tumor effects were observed when DZD2269 was in combination with immune checkpoint inhibitors, radiotherapy, or chemotherapy. A good PK/PD relationship was observed in these animal models. In the phase 1 clinical study, downregulation of pCREB was detected in human T cells, consistent with preclinical prediction. Our data support further clinical development of DZD2269 in patients with cancer. METHODS The selectivity of DZD2269 for adenosine receptors was tested in engineered cell lines, and its efficacy in blocking A2aR signaling and reversing adenosine-mediated immunosuppression was assessed in human T cells and peripheral blood mononuclear cells (PBMCs). The anti-tumor effects of DZD2269 were evaluated in multiple syngeneic mouse models as a single agent as well as in combination with chemotherapy, radiotherapy, or immune checkpoint inhibitors. A phase 1 study in healthy volunteers (NCT04932005) has been initiated to assess safety, pharmacokinetics (PK) and pharmacodynamics (PD) of DZD2269. RESULTS Adenosine stimulates phosphorylation of cyclic AMP response element binding protein (CREB) in T cells and inhibits anti-tumor cytokine secretion in PBMCs in a dose-dependent manner. DZD2269 was able to reverse the immunosuppression induced by high concentrations of adenosine, as demonstrated by inhibiting CREB phosphorylation in T cells, restoring Th1 cytokine secretion in PBMCs, and stimulating dendritic cells (DCs) maturation. As a single agent, DZD2269 showed anti-tumor growth in multiple syngeneic mouse tumor models, and more profound anti-tumor effects were observed when DZD2269 was in combination with immune checkpoint inhibitors, radiotherapy, or chemotherapy. A good PK/PD relationship was observed in these animal models. In the phase 1 clinical study, downregulation of pCREB was detected in human T cells, consistent with preclinical prediction. CONCLUSION DZD2269 is a novel A2aR antagonist which can fully block A2aR mediated immunosuppression commonly found in TME. Clinical development of DZD2269 in patients with cancer is warranted (NCT04634344).
Collapse
Affiliation(s)
- Yu Bai
- grid.11135.370000 0001 2256 9319Biomed-X Center, Academy for Advanced Interdisciplinary Studies, Peking University, 100871 Beijing, China ,Dizal Pharmaceuticals, 199 Liangjing Rd, Zhangjiang Hi-Tech Park, Pudong District, 201203 Shanghai, China
| | - Xin Zhang
- Dizal Pharmaceuticals, 199 Liangjing Rd, Zhangjiang Hi-Tech Park, Pudong District, 201203 Shanghai, China
| | - Jie Zheng
- Dizal Pharmaceuticals, 199 Liangjing Rd, Zhangjiang Hi-Tech Park, Pudong District, 201203 Shanghai, China
| | - Ziyi Liu
- Dizal Pharmaceuticals, 199 Liangjing Rd, Zhangjiang Hi-Tech Park, Pudong District, 201203 Shanghai, China
| | - Zhenfan Yang
- Dizal Pharmaceuticals, 199 Liangjing Rd, Zhangjiang Hi-Tech Park, Pudong District, 201203 Shanghai, China
| | - Xiaolin Zhang
- grid.11135.370000 0001 2256 9319Biomed-X Center, Academy for Advanced Interdisciplinary Studies, Peking University, 100871 Beijing, China ,Dizal Pharmaceuticals, 199 Liangjing Rd, Zhangjiang Hi-Tech Park, Pudong District, 201203 Shanghai, China
| |
Collapse
|
24
|
Liu Z, Liu X, Shen H, Xu X, Zhao X, Fu R. Adenosinergic axis and immune checkpoint combination therapy in tumor: A new perspective for immunotherapy strategy. Front Immunol 2022; 13:978377. [PMID: 36159861 PMCID: PMC9493240 DOI: 10.3389/fimmu.2022.978377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/23/2022] [Indexed: 11/21/2022] Open
Abstract
There are two figures and one table in this review, the review consists of 5823 words, without the description of figures and table, but including references. Tumor cells escape anti-tumor immune responses in various ways, including functionally shaping the microenvironment through the secretion of various chemokines and, cytokines. Adenosine is a powerful immunosuppressive metabolite, that is frequently elevated in the extracellular tumor microenvironment (TME). Thus, it has recently been proposed as a novel antitumor immunoassay for targeting adenosine- generating enzymes, such as CD39, CD73, and adenosine receptors. In recent years, the discovery of the immune checkpoints, such as programmed cell death 1(PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA-4), has also greatly changed treatment methods and ideas for malignant tumors. Malignant tumor immunotherapy has been developed from point-to-point therapy targeting immune checkpoints, combining different points of different pathways to create a therapy based on the macroscopic immune regulatory system network. This article reviews the theoretical basis of the adenosine energy axis and immune checkpoint combined therapy for malignant tumors and the latest advances in malignant tumors.
Collapse
|
25
|
Guo S, Han F, Zhu W. CD39 - A bright target for cancer immunotherapy. Biomed Pharmacother 2022; 151:113066. [PMID: 35550530 DOI: 10.1016/j.biopha.2022.113066] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 11/27/2022] Open
Abstract
The ATP-adenosine pathway functions as a key modulator of innate and adaptive immunity within the tumor microenvironment, and cancer immune evasion largely involves the generation of high amounts of immunosuppressive extracellular adenosine (eADO). Consequently, inhibition of eADO-generating enzymes and/or eADO receptors can effectively restore the antitumor immunity of multiple immune cells. With several clinical strategies currently being explored to modulating the eADO pathway in patients with cancer, recent clinical data with antagonists targeting CD73 and A2A receptor have demonstrated a promising therapeutic potential in cancer. Recent findings reveal that the ectonucleotidase CD39, the limiting enzyme been viewed as "immunological switch", converts ATP-driven pro-inflammatory milieu to an anti-inflammatory state mediated by adenosine. Owing to its superior feature of CD39 antagonism that rely not only on preventing the accumulation of adenosine but also on the stabilization of extracellular ATP to restore antitumor immunity, several inhibitors and clinical trials based on CD39 are being evaluated. Consequently, there is currently a focus on understanding the role of CD39 in governing immunity and how therapeutic strategies targeting this pathway alter the antitumor potential. We herein review the impact of CD39 on tumor microenvironment with a focus on treatment preference. Additionally, we also discuss the implication for rational combination therapies, molecular regulation, as well as potential limitations.
Collapse
Affiliation(s)
- Shuwei Guo
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Fengfeng Han
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wei Zhu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
26
|
Adenosine-Metabolizing Enzymes, Adenosine Kinase and Adenosine Deaminase, in Cancer. Biomolecules 2022; 12:biom12030418. [PMID: 35327609 PMCID: PMC8946555 DOI: 10.3390/biom12030418] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/05/2022] [Accepted: 03/06/2022] [Indexed: 12/17/2022] Open
Abstract
The immunosuppressive effect of adenosine in the microenvironment of a tumor is well established. Presently, researchers are developing approaches in immune therapy that target inhibition of adenosine or its signaling such as CD39 or CD73 inhibiting antibodies or adenosine A2A receptor antagonists. However, numerous enzymatic pathways that control ATP-adenosine balance, as well as understudied intracellular adenosine regulation, can prevent successful immunotherapy. This review contains the latest data on two adenosine-lowering enzymes: adenosine kinase (ADK) and adenosine deaminase (ADA). ADK deletes adenosine by its phosphorylation into 5′-adenosine monophosphate. Recent studies have revealed an association between a long nuclear ADK isoform and an increase in global DNA methylation, which explains epigenetic receptor-independent role of adenosine. ADA regulates the level of adenosine by converting it to inosine. The changes in the activity of ADA are detected in patients with various cancer types. The article focuses on the biological significance of these enzymes and their roles in the development of cancer. Perspectives of future studies on these enzymes in therapy for cancer are discussed.
Collapse
|
27
|
Baghbani E, Noorolyai S, Rahmani S, Shanehbandi D, Shadbad MA, Aghebati-Maleki L, Mokhtarzadeh A, Brunetti O, Fasano R, Silvestris N, Baradaran B. Silencing tumor-intrinsic CD73 enhances the chemosensitivity of NSCLC and potentiates the anti-tumoral effects of cisplatin: An in vitro study. Biomed Pharmacother 2021; 145:112370. [PMID: 34862113 DOI: 10.1016/j.biopha.2021.112370] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022] Open
Abstract
AIMS Besides suppressing anti-tumoral immune responses, tumor-intrinsic inhibitory immune checkpoints have been implicated in tumor development. Herein, we aimed to investigate the significance of tumor-intrinsic CD73, as an inhibitory immune checkpoint, in non-small cell lung cancer (NSCLC) development and propose a novel therapeutic approach. MAIN METHODS We investigated the cell viability, chemosensitivity, apoptosis, migration, and the cell cycle of A-549 and NCI-H1299 following treatment with cisplatin and CD73-small interfering RNA (siRNA) transfection. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay was used to study the viability of studied groups and chemosensitivity of tumoral cells. Flow cytometry and 4',6-diamidino-2-phenylindole (DAPI) staining were used to investigate the apoptosis of NSCLC cells. Flow cytometry and the wound-healing assay were used to investigate the cell cycle and migration of NSCLC cells, respectively. The mRNA expression levels of c-Myc, caspase 3, ROCK, and MMP-9 were investigated to study the underlying molecular mechanism. KEY FINDINGS CD73-siRNA transfection has significantly decreased the cell viability and enhanced the chemosensitivity of A-549 and NCI-H1299 cells to cisplatin. CD73-siRNA has considerably stimulated apoptosis, arrested the cell cycle, inhibited tumor migration, downregulated the mRNA expression of c-Myc, MMP-9, and ROCK, and upregulated caspase 3 expression in NSCLC cells. Besides, combined cisplatin therapy with CD73-siRNA transfection has potentiated the aforementioned anti-tumoral effects of cisplatin on NSCLC cells. SIGNIFICANCE Besides suppressing anti-tumoral immune responses, tumor-intrinsic CD73 can facilitate NSCLC development, and the combined cisplatin therapy with CD73-siRNA transfection can substantially enhance the chemosensitivity of NSCLC to cisplatin and potentiates cisplatin-induced anti-tumoral effects on NSCLC.
Collapse
Affiliation(s)
- Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Noorolyai
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shima Rahmani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Oronzo Brunetti
- Medical Oncology Unit-IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy
| | - Rossella Fasano
- Medical Oncology Unit-IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit-IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy; Department of Biomedical Sciences and Human Oncology DIMO-University of Bari, Bari, Italy.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
28
|
Kang BW, Chau I. Emerging agents for metastatic pancreatic cancer: spotlight on early phase clinical trials. Expert Opin Investig Drugs 2021; 30:1089-1107. [PMID: 34727804 DOI: 10.1080/13543784.2021.1995354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Despite the recent development of new chemotherapeutic regimens and combination strategies, metastatic pancreatic cancer (mPC) still shows only a modest response to conventional cytotoxic agents. However, several novel therapeutic agents targeting the unique features of mPC are showing promise in clinical trials. AREA COVERED This article reviews the current state of development of new agents targeting various systems and molecular pathways. We searched PubMed and clinicaltrials.gov in September 2021 with a special focus on ongoing early phase clinical trials to identify the promising therapeutic strategies for mPC. EXPERT OPINION Extensive tumor heterogeneity, complex tumor microenvironment, genetic alterations of the oncogenic signaling pathways, metabolic dysregulation, and a low immunogenicity are hurdles for current treatment approaches. Ongoing research efforts strive to overcome these hurdles and are showing some promising early results.
Collapse
Affiliation(s)
- Byung Woog Kang
- Department of Oncology/Hematology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Kyungpook National University, Daegu, Republic of Korea
| | - Ian Chau
- Department of Medicine, Royal Marsden Hospital, London, Surrey, UK
| |
Collapse
|
29
|
Shadbad MA, Asadzadeh Z, Derakhshani A, Hosseinkhani N, Mokhtarzadeh A, Baghbanzadeh A, Hajiasgharzadeh K, Brunetti O, Argentiero A, Racanelli V, Silvestris N, Baradaran B. A scoping review on the potentiality of PD-L1-inhibiting microRNAs in treating colorectal cancer: Toward single-cell sequencing-guided biocompatible-based delivery. Biomed Pharmacother 2021; 143:112213. [PMID: 34560556 DOI: 10.1016/j.biopha.2021.112213] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 12/15/2022] Open
Abstract
Tumoral programmed cell death ligand 1 (PD-L1) has been implicated in the immune evasion and development of colorectal cancer. Although monoclonal immune checkpoint inhibitors can exclusively improve the prognosis of patients with microsatellite instability-high (MSI-H) and tumor mutational burden-high (TMB-H) colorectal cancer, specific tumor-suppressive microRNAs (miRs) can regulate multiple oncogenic pathways and inhibit the de novo expression of oncoproteins, like PD-L1, both in microsatellite stable (MSS) and MSI-H colorectal cancer cells. This scoping review aimed to discuss the currently available evidence regarding the therapeutic potentiality of PD-L1-inhibiting miRs for colorectal cancer. For this purpose, the Web of Science, Scopus, and PubMed databases were systematically searched to obtain peer-reviewed studies published before 17 March 2021. We have found that miR-191-5p, miR-382-3p, miR-148a-3p, miR-93-5p, miR-200a-3p, miR-200c-3p, miR-138-5p, miR-140-3p, and miR-15b-5p can inhibit tumoral PD-L1 in colorectal cancer cells. Besides inhibiting PD-L1, miR-140-3p, miR-382-3p, miR-148a-3p, miR-93-5p, miR-200a-3p, miR-200c-3p, miR-138-5p, and miR-15b-5p can substantially reduce tumor migration, inhibit tumor development, stimulate anti-tumoral immune responses, decrease tumor viability, and enhance the chemosensitivity of colorectal cancer cells regardless of the microsatellite state. Concerning the specific, effective, and safe delivery of these miRs, the single-cell sequencing-guided biocompatible-based delivery of these miRs can increase the specificity of miR delivery, decrease the toxicity of traditional nanoparticles, transform the immunosuppressive tumor microenvironment into the proinflammatory one, suppress tumor development, decrease tumor migration, and enhance the chemosensitivity of tumoral cells regardless of the microsatellite state.
Collapse
Affiliation(s)
- Mahdi Abdoli Shadbad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Iran
| | - Afshin Derakhshani
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | | | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Iran
| | | | - Oronzo Brunetti
- Istituto Tumori BariGiovanni Paolo II, Istituto Nazionale dei Tumori (IRCCS), Bari, Italy
| | - Antonella Argentiero
- Istituto Tumori BariGiovanni Paolo II, Istituto Nazionale dei Tumori (IRCCS), Bari, Italy
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Nicola Silvestris
- Istituto Tumori BariGiovanni Paolo II, Istituto Nazionale dei Tumori (IRCCS), Bari, Italy; Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, Bari, Italy.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Iran.
| |
Collapse
|
30
|
Shadbad MA, Asadzadeh Z, Hosseinkhani N, Derakhshani A, Alizadeh N, Brunetti O, Silvestris N, Baradaran B. A Systematic Review of the Tumor-Infiltrating CD8 + T-Cells/PD-L1 Axis in High-Grade Glial Tumors: Toward Personalized Immuno-Oncology. Front Immunol 2021; 12:734956. [PMID: 34603316 PMCID: PMC8486082 DOI: 10.3389/fimmu.2021.734956] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/02/2021] [Indexed: 12/11/2022] Open
Abstract
Based on preclinical findings, programmed death-ligand 1 (PD-L1) can substantially attenuate CD8+ T-cell-mediated anti-tumoral immune responses. However, clinical studies have reported controversial results regarding the significance of the tumor-infiltrating CD8+ T-cells/PD-L1 axis on the clinical picture and the response rate of patients with high-grade glial tumors to anti-cancer therapies. Herein, we conducted a systematic review according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statements to clarify the clinical significance of the tumor-infiltrating CD8+ T-cells/PD-L1 axis and elucidate the impact of this axis on the response rate of affected patients to anti-cancer therapies. Indeed, a better understanding of the impact of this axis on the response rate of affected patients to anti-cancer therapies can provide valuable insights to address the futile response rate of immune checkpoint inhibitors in patients with high-grade glial tumors. For this purpose, we systematically searched Scopus, Web of Science, Embase, and PubMed to obtain peer-reviewed studies published before 1 January 2021. We have observed that PD-L1 overexpression can be associated with the inferior prognosis of glioblastoma patients who have not been exposed to chemo-radiotherapy. Besides, exposure to anti-cancer therapies, e.g., chemo-radiotherapy, can up-regulate inhibitory immune checkpoint molecules in tumor-infiltrating CD8+ T-cells. Therefore, unlike unexposed patients, increased tumor-infiltrating CD8+ T-cells in anti-cancer therapy-exposed tumoral tissues can be associated with the inferior prognosis of affected patients. Because various inhibitory immune checkpoints can regulate anti-tumoral immune responses, the single-cell sequencing of the cells residing in the tumor microenvironment can provide valuable insights into the expression patterns of inhibitory immune checkpoints in the tumor micromovement. Thus, administrating immune checkpoint inhibitors based on the data from the single-cell sequencing of these cells can increase patients’ response rates, decrease the risk of immune-related adverse events development, prevent immune-resistance development, and reduce the risk of tumor recurrence.
Collapse
Affiliation(s)
- Mahdi Abdoli Shadbad
- Research Center for Evidence-Based Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negar Hosseinkhani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Derakhshani
- Laboratory of Experimental Pharmacology, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Oronzo Brunetti
- Medical Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy.,Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
31
|
Vautrot V, Bentayeb H, Causse S, Garrido C, Gobbo J. Tumor-Derived Exosomes: Hidden Players in PD-1/PD-L1 Resistance. Cancers (Basel) 2021; 13:cancers13184537. [PMID: 34572764 PMCID: PMC8467727 DOI: 10.3390/cancers13184537] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Immunotherapies such as anti-PD-1/PD-L1 have garnered increasing importance in cancer therapy, leading to substantial improvements in patient care and survival. However, a certain proportion of patients present tumors that resist these treatments. Exosomes, small vesicles secreted by almost every cell, including tumor cells, have proven to be key actors in this resistance. In this review, we describe the involvement of immune checkpoints and immune modulators in tumor-derived exosomes (TEXs) in the context of cancer. We will focus on the most promising proteins under scrutiny for use in combination with PD-1 blockade therapy in a clinical setting: PD-L1, CTLA-4, TIM-3, CD73/39, LAG-3, and TIGIT. Finally, we will discuss how they can change the game in immunotherapy, notably through their role in immunoresistance and how they can guide therapeutic decisions, as well as the current obstacles in the field. Abstract Recently, immunotherapy has garnered increasing importance in cancer therapy, leading to substantial improvements in patient care and survival. By blocking the immune checkpoints—protein regulators of the immune system—immunotherapy prevents immune tolerance toward tumors and reactivates the immune system, prompting it to fight cancer cell growth and diffusion. A widespread strategy for this is the blockade of the interaction between PD-L1 and PD-1. However, while patients generally respond well to immunotherapy, a certain proportion of patients present tumors that resist these treatments. This portion can be very high in some cancers and hinders cancer curability. For this reason, current efforts are focusing on combining PD-1/PD-L1 immunotherapy with the targeting of other immune checkpoints to counter resistance and achieve better results. Exosomes, small vesicles secreted by almost any cell, including tumor cells, have proven to be key actors in this resistance. The exosomes released by tumor cells spread the immune-suppressive properties of the tumor throughout the tumor microenvironment and participate in establishing metastatic niches. In this review, we will describe immune checkpoints and immune modulators whose presence in tumor-derived exosomes (TEXs) has been established. We will focus on the most promising proteins under scrutiny for use in combination with PD-1 blockade therapy in a clinical setting, such as PD-L1, CTLA-4, TIM-3, CD73/39, LAG-3, and TIGIT. We will explore the immunosuppressive impact of these exosomal proteins on a variety of immune cells. Finally, we will discuss how they can change the game in immunotherapy and guide therapeutic decisions, as well as the current limits of this approach. Depending on the viewpoint, these exosomal proteins may either provide key missing information on tumor growth and resistance mechanisms or they may be the next big challenge to overcome in improving cancer treatment.
Collapse
Affiliation(s)
- Valentin Vautrot
- Research Center UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC, INSERM, F-21000 Dijon, France; (V.V.); (H.B.); (S.C.); (C.G.)
- Unité de Formation et de Recherches Sciences de la Santé, University of Bourgogne Franche-Comté, F-21000 Dijon, France
- Centre Georges-François Leclerc, F-21079 Dijon, France
| | - Hafidha Bentayeb
- Research Center UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC, INSERM, F-21000 Dijon, France; (V.V.); (H.B.); (S.C.); (C.G.)
- Unité de Formation et de Recherches Sciences de la Santé, University of Bourgogne Franche-Comté, F-21000 Dijon, France
- Centre Georges-François Leclerc, F-21079 Dijon, France
| | - Sébastien Causse
- Research Center UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC, INSERM, F-21000 Dijon, France; (V.V.); (H.B.); (S.C.); (C.G.)
- Unité de Formation et de Recherches Sciences de la Santé, University of Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Carmen Garrido
- Research Center UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC, INSERM, F-21000 Dijon, France; (V.V.); (H.B.); (S.C.); (C.G.)
- Unité de Formation et de Recherches Sciences de la Santé, University of Bourgogne Franche-Comté, F-21000 Dijon, France
- Centre Georges-François Leclerc, F-21079 Dijon, France
| | - Jessica Gobbo
- Research Center UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC, INSERM, F-21000 Dijon, France; (V.V.); (H.B.); (S.C.); (C.G.)
- Unité de Formation et de Recherches Sciences de la Santé, University of Bourgogne Franche-Comté, F-21000 Dijon, France
- Centre Georges-François Leclerc, F-21079 Dijon, France
- Centre Georges-François Leclerc, Early Phase Unit INCa CLIP², Department of Oncology, F-21079 Dijon, France
- Clinical Investigation Center CIC1432, Module Plurithématique, INSERM, F-21079 Dijon, France
- Correspondence:
| |
Collapse
|