1
|
Xu C, Chen J, Tan M, Tan Q. The role of macrophage polarization in ovarian cancer: from molecular mechanism to therapeutic potentials. Front Immunol 2025; 16:1543096. [PMID: 40330466 PMCID: PMC12052780 DOI: 10.3389/fimmu.2025.1543096] [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: 12/10/2024] [Accepted: 03/21/2025] [Indexed: 05/08/2025] Open
Abstract
Ovarian cancer (OC) remains the most lethal gynecological malignancy, primarily due to its late-stage diagnosis, frequent recurrence, and resistance to conventional chemotherapy. A critical factor contributing to OC's aggressiveness is the tumor microenvironment (TME), particularly the presence and polarization of tumor-associated macrophages (TAMs). TAMs, often skewed toward an immunosuppressive M2-like phenotype, facilitate tumor growth, angiogenesis, metastasis, and resistance to therapy. This comprehensive review delves into the multifaceted regulation of macrophage polarization in OC, highlighting key molecular pathways such as PTEN loss, Wnt/β-catenin signaling, NF-κB, Myc, STAT3, and JNK, among others. Additionally, it explores the role of chemokines, non-coding RNAs, and various proteins in modulating TAM phenotypes. Emerging evidence underscores the significance of extracellular vesicles (EVs) and ovarian cancer stem cells (CSCs) in promoting M2 polarization, thereby enhancing tumor progression and therapy resistance. The review also identifies critical biomarkers associated with macrophage polarization, including CD163, LILRB1, MUC2, and others, which hold prognostic and therapeutic potential. Therapeutic strategies targeting TAMs are extensively discussed, encompassing oncolytic viruses, engineered EVs, immunotherapies, nanoparticles, targeted therapies, and natural products. These approaches aim to reprogram TAMs from a pro-tumorigenic M2 state to an anti-tumorigenic M1 phenotype, thereby enhancing immune responses and overcoming resistance to treatments such as chemotherapy and immune checkpoint inhibitors. Furthermore, the review addresses the interplay between macrophage polarization and therapy resistance, emphasizing the need for novel interventions to modulate the TME effectively. By synthesizing current knowledge on macrophage polarization in ovarian cancer, this study underscores the potential of targeting TAMs to improve clinical outcomes and personalize treatment strategies for OC patients. Continued research in this domain is essential to develop robust therapeutic frameworks that can mitigate the immunosuppressive TME and enhance the efficacy of existing and novel cancer therapies.
Collapse
Affiliation(s)
| | | | | | - Qingqing Tan
- Department of Gynecology and Obstetrics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| |
Collapse
|
2
|
Wang X, Xing Y, Zhou X, Wang C, Han S, Zhao S. Radiomics Signatures Based on Computed Tomography for Noninvasive Prediction of CXCL10 Expression and Prognosis in Ovarian Cancer. Cancer Rep (Hoboken) 2024; 7:e70030. [PMID: 39443817 PMCID: PMC11499071 DOI: 10.1002/cnr2.70030] [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: 01/09/2024] [Revised: 08/28/2024] [Accepted: 09/17/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) is an aggressive gynecological tumor usually diagnosed with malignant ascites and even observed widespread metastasis or distant spread. AIMS We aimed to develop and identify radiomics models according to computed tomography (CT) for preoperative prediction of CXCL10 expression and prognosis in patients with OC. METHODS Genomic data with CT images and corresponding clinicopathological parameters were extracted from The Cancer Imaging Archive (TCIA) and The Cancer Genome Atlas (TCGA). To analyze the prognosis, we carried out the univariate Cox regression analysis (UCRA), multivariate Cox regression analysis (MCRA), and Kaplan-Meier (KM) analysis. For the data reduction, logistic regression, operator regression, least absolute shrinkage selection, radiomic feature construction, and feature selection were utilized. The predictive performance of the radiomic signatures was assessed using the analyses of the receiver operating characteristic (ROC) curve, decision curve (DCA), and precision-recall (PR) curve. To evaluate the correlation between the radiomic score (Rad-score) and CXCL10 expression, the Wilcoxon rank-sum test was applied. RESULTS Three radiomics models effectively predicted CXCL10 expression levels (AUC = 0.791, 0.748, and 0.718 for the set of training; AUC = 0.761, 0.746, and 0.701 for the set of validation). A higher Rad-score significantly correlated with upregulated CXCL10 expression. CONCLUSION CXCL10 expression can be predicted noninvasively and preoperatively via radiomic signatures based on contrast-enhanced CT images.
Collapse
Affiliation(s)
- Xiaohua Wang
- Department of Gynecology and Obstetrics, Department of GynecologyThe Second Hospital of HeBei Medical University, Affiliated Hospital of Chengde Medical UniversityShijiazhuangChina
| | - Yuanyuan Xing
- Department of Nuclear MedicineAffiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Xuan Zhou
- Department of GynecologyAffiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Chunhui Wang
- Department of GynecologyAffiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Shuyu Han
- Department of GynecologyAffiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Sufen Zhao
- Department of Gynecology and ObstetricsThe Second Hospital of HeBei Medical UniversityShijiazhuangChina
| |
Collapse
|
3
|
Ullah A, Chen Y, Singla RK, Cao D, Shen B. Pro-inflammatory cytokines and CXC chemokines as game-changer in age-associated prostate cancer and ovarian cancer: Insights from preclinical and clinical studies' outcomes. Pharmacol Res 2024; 204:107213. [PMID: 38750677 DOI: 10.1016/j.phrs.2024.107213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/15/2024] [Accepted: 05/09/2024] [Indexed: 06/01/2024]
Abstract
Prostate cancer (PC) and Ovarian cancer (OC) are two of the most common types of cancer that affect the reproductive systems of older men and women. These cancers are associated with a poor quality of life among the aged population. Therefore, finding new and innovative ways to detect, treat, and prevent these cancers in older patients is essential. Finding biomarkers for these malignancies will increase the chance of early detection and effective treatment, subsequently improving the survival rate. Studies have shown that the prevalence and health of some illnesses are linked to an impaired immune system. However, the age-associated changes in the immune system during malignancies such as PC and OC are poorly understood. Recent research has suggested that the excessive production of inflammatory immune mediators, such as interleukin-6 (IL-6), interleukin-8 (IL-8), transforming growth factor (TGF), tumor necrosis factor (TNF), CXC motif chemokine ligand 1 (CXCL1), CXC motif chemokine ligand 12 (CXCL12), and CXC motif chemokine ligand 13 (CXCL13), etc., significantly impact the development of PC and OC in elderly patients. Our review focuses on the latest functional studies of pro-inflammatory cytokines (interleukins) and CXC chemokines, which serve as biomarkers in elderly patients with PC and OC. Thus, we aim to shed light on how these biomarkers affect the development of PC and OC in elderly patients. We also examine the current status and future perspective of cytokines (interleukins) and CXC chemokines-based therapeutic targets in OC and PC treatment for elderly patients.
Collapse
Affiliation(s)
- Amin Ullah
- Department of Abdominal Oncology, Cancer Center of West China Hospital and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yongxiu Chen
- Gynecology Department, Guangdong Women and Children Hospital, No. 521, Xingnan Road, Panyu District, Guangzhou 511442, China
| | - Rajeev K Singla
- Department of Abdominal Oncology, Cancer Center of West China Hospital and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Dan Cao
- Department of Abdominal Oncology, Cancer Center of West China Hospital and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Bairong Shen
- Department of Abdominal Oncology, Cancer Center of West China Hospital and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Stephens AN, Hobbs SJ, Kang SW, Bilandzic M, Rainczuk A, Oehler MK, Jobling TW, Plebanski M, Allman R. A Novel Predictive Multi-Marker Test for the Pre-Surgical Identification of Ovarian Cancer. Cancers (Basel) 2023; 15:5267. [PMID: 37958440 PMCID: PMC10650329 DOI: 10.3390/cancers15215267] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Ovarian cancer remains the most lethal of gynecological malignancies, with the 5-year survival below 50%. Currently there is no simple and effective pre-surgical diagnosis or triage for patients with malignancy, particularly those with early-stage or low-volume tumors. Recently we discovered that CXCL10 can be processed to an inactive form in ovarian cancers and that its measurement has diagnostic significance. In this study we evaluated the addition of processed CXCL10 to a biomarker panel for the discrimination of benign from malignant disease. Multiple biomarkers were measured in retrospectively collected plasma samples (n = 334) from patients diagnosed with benign or malignant disease, and a classifier model was developed using CA125, HE4, Il6 and CXCL10 (active and total). The model provided 95% sensitivity/95% specificity for discrimination of benign from malignant disease. Positive predictive performance exceeded that of "gold standard" scoring systems including CA125, RMI and ROMA% and was independent of menopausal status. In addition, 80% of stage I-II cancers in the cohort were correctly identified using the multi-marker scoring system. Our data suggest the multi-marker panel and associated scoring algorithm provides a useful measurement to assist in pre-surgical diagnosis and triage of patients with suspected ovarian cancer.
Collapse
Affiliation(s)
- Andrew N. Stephens
- Hudson Institute of Medical Research, Clayton 3168, Australia; (S.-W.K.); (M.B.); (A.R.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
- Cleo Diagnostics Ltd., Melbourne 3000, Australia; (S.J.H.); (R.A.)
| | - Simon J. Hobbs
- Cleo Diagnostics Ltd., Melbourne 3000, Australia; (S.J.H.); (R.A.)
| | - Sung-Woon Kang
- Hudson Institute of Medical Research, Clayton 3168, Australia; (S.-W.K.); (M.B.); (A.R.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Maree Bilandzic
- Hudson Institute of Medical Research, Clayton 3168, Australia; (S.-W.K.); (M.B.); (A.R.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Adam Rainczuk
- Hudson Institute of Medical Research, Clayton 3168, Australia; (S.-W.K.); (M.B.); (A.R.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
- Bruker Pty Ltd., Preston 3072, Australia
| | - Martin K. Oehler
- Department of Gynecological Oncology, Royal Adelaide Hospital, Adelaide 5000, Australia;
- Robinson Institute, University of Adelaide, Adelaide 5000, Australia
| | - Tom W. Jobling
- Department of Gynecology Oncology, Monash Medical Centre, Bentleigh East 3165, Australia;
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia;
| | - Richard Allman
- Cleo Diagnostics Ltd., Melbourne 3000, Australia; (S.J.H.); (R.A.)
| |
Collapse
|
5
|
Roy AN, Gupta AM, Banerjee D, Chakrabarti J, Raghavendra PB. Unraveling DPP4 Receptor Interactions with SARS-CoV-2 Variants and MERS-CoV: Insights into Pulmonary Disorders via Immunoinformatics and Molecular Dynamics. Viruses 2023; 15:2056. [PMID: 37896834 PMCID: PMC10612102 DOI: 10.3390/v15102056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 09/23/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Human coronaviruses like MERS CoV are known to utilize dipeptidyl peptidase 4 (DPP4), apart from angiotensin-converting enzyme 2(ACE2) as a potential co-receptor for viral cell entry. DPP4, the ubiquitous membrane-bound aminopeptidase, is closely associated with elevation of disease severity in comorbidities. In SARS-CoV-2, there is inadequate evidence for combination of spike protein variants with DPP4, and underlying adversity in COVID-19. To elucidate this mechanistic basis, we have investigated interaction of spike protein variants with DPP4 through molecular docking and simulation studies. The possible binding interactions between the receptor binding domain (RBD) of different spike variants of SARS-CoV-2 and DPP4 have been compared with interactions observed in the experimentally determined structure of the complex of MERS-CoV with DPP4. Comparative binding affinity confers that Delta-CoV-2: DPP4 shows close proximity with MERS-CoV:DPP4, as depicted from accessible surface area, radius of gyration and number of hydrogen bonding in the interface. Mutations in the delta variant, L452R and T478K directly participate in DPP4 interaction, enhancing DPP4 binding. E484K in alpha and gamma variants of spike protein is also found to interact with DPP4. Hence, DPP4 interaction with spike protein becomes more suitable due to mutation, especially due to L452R, T478K and E484K. Furthermore, perturbation in the nearby residues Y495, Q474 and Y489 is evident due to L452R, T478K and E484K, respectively. Virulent strains of spike protein are more susceptible to DPP4 interaction and are prone to be victimized in patients due to comorbidities. Our results will aid the rational optimization of DPP4 as a potential therapeutic target to manage COVID-19 disease severity.
Collapse
Affiliation(s)
- Arpan Narayan Roy
- National Institute of Biomedical Genomics, Kalyani 741251, West Bengal, India; (A.N.R.); (D.B.)
| | - Aayatti Mallick Gupta
- Department of Physics of Complex Systems, S. N. Bose National Centre for Basic Sciences, Block-JD, Sector-III, Salt Lake, Kolkata 700106, West Bengal, India; (A.M.G.); (J.C.)
| | - Deboshmita Banerjee
- National Institute of Biomedical Genomics, Kalyani 741251, West Bengal, India; (A.N.R.); (D.B.)
| | - Jaydeb Chakrabarti
- Department of Physics of Complex Systems, S. N. Bose National Centre for Basic Sciences, Block-JD, Sector-III, Salt Lake, Kolkata 700106, West Bengal, India; (A.M.G.); (J.C.)
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block-JD, Sector-III, Salt Lake, Kolkata 700106, West Bengal, India
| | - Pongali B. Raghavendra
- National Institute of Biomedical Genomics, Kalyani 741251, West Bengal, India; (A.N.R.); (D.B.)
| |
Collapse
|
6
|
Matsumoto A, Hiroi M, Mori K, Yamamoto N, Ohmori Y. Differential Anti-Tumor Effects of IFN-Inducible Chemokines CXCL9, CXCL10, and CXCL11 on a Mouse Squamous Cell Carcinoma Cell Line. Med Sci (Basel) 2023; 11:medsci11020031. [PMID: 37218983 DOI: 10.3390/medsci11020031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/11/2023] [Accepted: 04/23/2023] [Indexed: 05/24/2023] Open
Abstract
Chemokines are a group of cytokines involved in the mobilization of leukocytes, which play a role in host defense and a variety of pathological conditions, including cancer. Interferon (IFN)-inducible chemokines C-X-C motif ligand 9 (CXCL), CXCL10, and CXCL11 are anti-tumor chemokines; however, the differential anti-tumor effects of IFN-inducible chemokines are not completely understood. In this study, we investigated the anti-tumor effects of IFN-inducible chemokines by transferring chemokine expression vectors into a mouse squamous cell carcinoma cell line, SCCVII, to generate a cell line stably expressing chemokines and transplanted it into nude mice. The results showed that CXCL9- and CXCL11-expressing cells markedly inhibited tumor growth, whereas CXCL10-expressing cells did not inhibit growth. The NH2-terminal amino acid sequence of mouse CXCL10 contains a cleavage sequence by dipeptidyl peptidase 4 (DPP4), an enzyme that cleaves the peptide chain of chemokines. IHC staining indicated DPP4 expression in the stromal tissue, suggesting CXCL10 inactivation. These results suggest that the anti-tumor effects of IFN-inducible chemokines are affected by the expression of chemokine-cleaving enzymes in tumor tissues.
Collapse
Affiliation(s)
- Ari Matsumoto
- Division of Oral and Maxillofacial Surgery, Department of Diagnostic and Therapeutic Sciences, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado 350-0283, Japan
| | - Miki Hiroi
- Division of Basic Biology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado 350-0283, Japan
| | - Kazumasa Mori
- Division of Oral and Maxillofacial Surgery, Department of Diagnostic and Therapeutic Sciences, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado 350-0283, Japan
| | - Nobuharu Yamamoto
- Division of Oral and Maxillofacial Surgery, Department of Diagnostic and Therapeutic Sciences, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado 350-0283, Japan
| | - Yoshihiro Ohmori
- Division of Microbiology and Immunology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado 350-0283, Japan
| |
Collapse
|
7
|
Michaelides S, Obeck H, Kechur D, Endres S, Kobold S. Migratory Engineering of T Cells for Cancer Therapy. Vaccines (Basel) 2022; 10:1845. [PMID: 36366354 PMCID: PMC9692862 DOI: 10.3390/vaccines10111845] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 10/10/2023] Open
Abstract
Adoptive cell therapy (ACT) and chimeric antigen receptor (CAR) T cell therapy in particular represents an adaptive, yet versatile strategy for cancer treatment. Convincing results in the treatment of hematological malignancies have led to FDA approval for several CAR T cell therapies in defined refractory diseases. In contrast, the treatment of solid tumors with adoptively transferred T cells has not demonstrated convincing efficacy in clinical trials. One of the main reasons for ACT failure in solid tumors is poor trafficking or access of transferred T cells to the tumor site. Tumors employ a variety of mechanisms shielding themselves from immune cell infiltrates, often translating to only fractions of transferred T cells reaching the tumor site. To overcome this bottleneck, extensive efforts are being undertaken at engineering T cells to improve ACT access to solid tumors. In this review, we provide an overview of the immune cell infiltrate in human tumors and the mechanisms tumors employ toward immune exclusion. We will discuss ways in which T cells can be engineered to circumvent these barriers. We give an outlook on ongoing clinical trials targeting immune cell migration to improve ACT and its perspective in solid tumors.
Collapse
Affiliation(s)
- Stefanos Michaelides
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80337 Munich, Germany
| | - Hannah Obeck
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80337 Munich, Germany
| | - Daryna Kechur
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80337 Munich, Germany
| | - Stefan Endres
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80337 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Pettenkoferstrasse 8a, 80336 Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80337 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Pettenkoferstrasse 8a, 80336 Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
| |
Collapse
|
8
|
Wei C, Ma Y, Wang F, Liao Y, Chen Y, Zhao B, Zhao Q, Wang D, Tang D. Igniting Hope for Tumor Immunotherapy: Promoting the "Hot and Cold" Tumor Transition. Clin Med Insights Oncol 2022; 16:11795549221120708. [PMID: 36147198 PMCID: PMC9486259 DOI: 10.1177/11795549221120708] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/05/2022] [Indexed: 12/02/2022] Open
Abstract
The discovery of immune checkpoint inhibitors (ICIs) has ushered a new era for immunotherapy against malignant tumors through the killing effects of cytotoxic T lymphocytes in the tumor microenvironment (TME), resulting in long-lasting tumor suppression and regression. Nevertheless, given that ICIs are highly dependent on T cells in the TME and that most tumors lack T-cell infiltration, promoting the conversion of such immunosuppressive "cold" tumors to "hot" tumors is currently a key challenge in tumor immunotherapy. Herein, we systematically outlined the mechanisms underlying the formation of the immunosuppressive TME in cold tumors, including the role of immunosuppressive cells, impaired antigen presentation, transforming growth factor-β, STAT3 signaling, adenosine, and interferon-γ signaling. Moreover, therapeutic strategies for promoting cold tumors to hot tumors with adequate T-cell infiltration were also discussed. Finally, the prospects of therapeutic tools such as oncolytic viruses, nanoparticles, and photothermal therapy in restoring immune activity in cold tumors were thoroughly reviewed.
Collapse
Affiliation(s)
- Chen Wei
- Clinical Medical College, Yangzhou
University, Yangzhou, China
| | - Yichao Ma
- Clinical Medical College, Yangzhou
University, Yangzhou, China
| | - Fei Wang
- Clinical Medical College, Dalian
Medical University, Dalian, China
| | - Yiqun Liao
- Clinical Medical College, Dalian
Medical University, Dalian, China
| | - Yuji Chen
- Clinical Medical College, Yangzhou
University, Yangzhou, China
| | - Bin Zhao
- Clinical Medical College, Dalian
Medical University, Dalian, China
| | - Qi Zhao
- Clinical Medical College, Yangzhou
University, Yangzhou, China
| | - Daorong Wang
- Department of General Surgery,
Institute of General Surgery, Clinical Medical College, Northern Jiangsu People’s
Hospital, Yangzhou University, Yangzhou, China
| | - Dong Tang
- Department of General Surgery,
Institute of General Surgery, Clinical Medical College, Northern Jiangsu People’s
Hospital, Yangzhou University, Yangzhou, China
| |
Collapse
|
9
|
Cui C, Tian X, Wei L, Wang Y, Wang K, Fu R. New insights into the role of dipeptidyl peptidase 8 and dipeptidyl peptidase 9 and their inhibitors. Front Pharmacol 2022; 13:1002871. [PMID: 36172198 PMCID: PMC9510841 DOI: 10.3389/fphar.2022.1002871] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Dipeptidyl peptidase 8 (DPP8) and 9 (DPP9) are widely expressed in mammals including humans, mainly locate in the cytoplasm. The DPP8 and DPP9 (DPP8/9) belong to serine proteolytic enzymes, they can recognize and cleave N-terminal dipeptides of specific substrates if proline is at the penultimate position. Because the localization of DPP8/9 is different from that of DPP4 and the substrates for DPP8/9 are not yet completely clear, their physiological and pathological roles are still being further explored. In this article, we will review the recent research advances focusing on the expression, regulation, and functions of DPP8/9 in physiology and pathology status. Emerging research results have shown that DPP8/9 is involved in various biological processes such as cell behavior, energy metabolism, and immune regulation, which plays an essential role in maintaining normal development and physiological functions of the body. DPP8/9 is also involved in pathological processes such as tumorigenesis, inflammation, and organ fibrosis. In recent years, related research on immune cell pyroptosis has made DPP8/9 a new potential target for the treatment of hematological diseases. In addition, DPP8/9 inhibitors also have great potential in the treatment of tumors and chronic kidney disease.
Collapse
Affiliation(s)
- Chenkai Cui
- Department of Nephrology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xuefei Tian
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Linting Wei
- Department of Nephrology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yinhong Wang
- Department of Nephrology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Kexin Wang
- Department of Nephrology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Rongguo Fu
- Department of Nephrology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Rongguo Fu,
| |
Collapse
|
10
|
Karin N. Chemokines in the Landscape of Cancer Immunotherapy: How They and Their Receptors Can Be Used to Turn Cold Tumors into Hot Ones? Cancers (Basel) 2021; 13:6317. [PMID: 34944943 PMCID: PMC8699256 DOI: 10.3390/cancers13246317] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 02/07/2023] Open
Abstract
Over the last decade, monoclonal antibodies to immune checkpoint inhibitors (ICI), also known as immune checkpoint blockers (ICB), have been the most successful approach for cancer therapy. Starting with mAb to cytotoxic T lymphocyte antigen 4 (CTLA-4) inhibitors in metastatic melanoma and continuing with blockers of the interactions between program cell death 1 (PD-1) and its ligand program cell death ligand 1 (PDL-1) or program cell death ligand 2 (PDL-2), that have been approved for about 20 different indications. Yet for many cancers, ICI shows limited success. Several lines of evidence imply that the limited success in cancer immunotherapy is associated with attempts to treat patients with "cold tumors" that either lack effector T cells, or in which these cells are markedly suppressed by regulatory T cells (Tregs). Chemokines are a well-defined group of proteins that were so named due to their chemotactic properties. The current review focuses on key chemokines that not only attract leukocytes but also shape their biological properties. CXCR3 is a chemokine receptor with 3 ligands. We suggest using Ig-based fusion proteins of two of them: CXL9 and CXCL10, to enhance anti-tumor immunity and perhaps transform cold tumors into hot tumors. Potential differences between CXCL9 and CXCL10 regarding ICI are discussed. We also discuss the possibility of targeting the function or deleting a key subset of Tregs that are CCR8+ by monoclonal antibodies to CCR8. These cells are preferentially abundant in several tumors and are likely to be the key drivers in suppressing anti-cancer immune reactivity.
Collapse
Affiliation(s)
- Nathan Karin
- Department of Immunology, Faculty of Medicine, Technion, P.O. Box 9697, Haifa 31096, Israel
| |
Collapse
|
11
|
Song W, Yin H, Han C, Mao Q, Tang J, Ji Z, Yan X, Wang L, Liu S, Ai C. The role of CXCL10 in prognosis of patients with colon cancer and tumor microenvironment remodeling. Medicine (Baltimore) 2021; 100:e27224. [PMID: 34559115 PMCID: PMC10545341 DOI: 10.1097/md.0000000000027224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUNG Tumor microenvironment (TME) has gradually emerged as an important research topic in the fight against cancer. The immune system is a major contributing factor in TME, and investigations have revealed that tumors are partially infiltrated with numerous immune cell subsets. METHOD We obtained transcriptome RNA-seq data from the the Cancer Genome Atlas databases for 521 patients with colon adenocarcinoma (COAD). ESTIMATE algorithms are then used to estimate the fraction of stromal and immune cells in COAD samples. RESULT A total of 1109 stromal-immune score-related differentially expressed genes were identified and used to generate a high-confidence protein-protein interaction network and univariate COX regression analysis. C-X-C motif chemokine 10 (CXCL10) was identified as the core gene by intersection analysis of data from protein-protein interaction network and univariate COX regression analysis. Then, for CXCL10, we performed gene set enrichment analysis, survival analysis and clinical analysis, and we used CIBERSORT algorithms to estimate the proportion of tumor-infiltrating immune cells in COAD samples. CONCLUSION We discovered that CXCL10 levels could be effective for predicting the prognosis of COAD patients as well as a clue that the status of TME is transitioning from immunological to metabolic activity, which provided additional information for COAD therapies.
Collapse
Affiliation(s)
- Weiwei Song
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University. Beijing, China
| | - Hongli Yin
- German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Chenguang Han
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University. Beijing, China
- College of Nankai University, Tianjin, China
| | - Qiantai Mao
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University. Beijing, China
| | - Jing Tang
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University. Beijing, China
| | - Zhaoshuai Ji
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University. Beijing, China
| | - Xu Yan
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University. Beijing, China
| | - Lan Wang
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University. Beijing, China
| | - Shengnan Liu
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University. Beijing, China
| | - Chao Ai
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University. Beijing, China
| |
Collapse
|
12
|
De Zutter A, Van Damme J, Struyf S. The Role of Post-Translational Modifications of Chemokines by CD26 in Cancer. Cancers (Basel) 2021; 13:cancers13174247. [PMID: 34503058 PMCID: PMC8428238 DOI: 10.3390/cancers13174247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Chemokines are a large family of small chemotactic cytokines that fulfill a central function in cancer. Both tumor-promoting and -impeding roles have been ascribed to chemokines, which they exert in a direct or indirect manner. An important post-translational modification that regulates chemokine activity is the NH2-terminal truncation by peptidases. CD26 is a dipeptidyl peptidase (DPPIV), which typically clips a NH2-terminal dipeptide from the chemokine. With a certain degree of selectivity in terms of chemokine substrate, CD26 only recognizes chemokines with a penultimate proline or alanine. Chemokines can be protected against CD26 recognition by specific amino acid residues within the chemokine structure, by oligomerization or by binding to cellular glycosaminoglycans (GAGs). Upon truncation, the binding affinity for receptors and GAGs is altered, which influences chemokine function. The consequences of CD26-mediated clipping vary, as unchanged, enhanced, and reduced activities are reported. In tumors, CD26 most likely has the most profound effect on CXCL12 and the interferon (IFN)-inducible CXCR3 ligands, which are converted into receptor antagonists upon truncation. Depending on the tumor type, expression of CD26 is upregulated or downregulated and often results in the preferential generation of the chemokine isoform most favorable for tumor progression. Considering the tight relationship between chemokine sequence and chemokine binding specificity, molecules with the appropriate characteristics can be chemically engineered to provide innovative therapeutic strategies in a cancer setting.
Collapse
|
13
|
Kang SW, Rainczuk A, Oehler MK, Jobling TW, Plebanski M, Stephens AN. Active Ratio Test (ART) as a Novel Diagnostic for Ovarian Cancer. Diagnostics (Basel) 2021; 11:diagnostics11061048. [PMID: 34200333 PMCID: PMC8230042 DOI: 10.3390/diagnostics11061048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/04/2021] [Accepted: 06/04/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Despite substantial effort, there remains a lack of biomarker-based, clinically relevant testing for the accurate, non-invasive diagnostic or prognostic profiling of epithelial ovarian cancers (EOC). Our previous work demonstrated that whilst the inflammatory marker C-X-C motif chemokine ligand 10 (CXCL10) has prognostic relevance in ovarian cancer, its use is complicated by the presence of multiple, N-terminally modified variants, mediated by several enzymes including Dipeptidyl Peptidase 4 (DPP4). Methods: In this study, we provide the first evidence for the “Active Ratio Test” (ART) as a novel method to measure biologically relevant CXCL10 proteoforms in clinical samples. Results: In a cohort of 275 patients, ART accurately differentiated patients with malignant EOCs from those with benign gynaecological conditions (AUC 0.8617) and significantly out-performed CA125 alone. Moreover, ART combined with the measurement of CA125 and DPP4 significantly increased prognostic performance (AUC 0.9511; sensitivity 90.0%; specificity 91.7%; Cohen’s d > 1) for EOC detection. Conclusion: Our data demonstrate that ART provides a useful method to accurately discriminate between patients with benign versus malignant EOC, and highlights their relevance to ovarian cancer diagnosis. This marker combination may also be applicable in broader screening applications, to identify or discriminate benign from malignant disease in asymptomatic women.
Collapse
Affiliation(s)
- Sung-Woog Kang
- Hudson Institute of Medical Research, Clayton 3168, Australia; (S.-W.K.); (A.R.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Adam Rainczuk
- Hudson Institute of Medical Research, Clayton 3168, Australia; (S.-W.K.); (A.R.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
- Bruker Pty Ltd., Preston 3072, Australia
| | - Martin K. Oehler
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide 5000, Australia;
- Robinson Institute, University of Adelaide, Adelaide 5000, Australia
| | - Thomas W. Jobling
- Department of Gynaecology Oncology, Monash Medical Centre, Bentleigh East 3165, Australia;
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia;
| | - Andrew N. Stephens
- Hudson Institute of Medical Research, Clayton 3168, Australia; (S.-W.K.); (A.R.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
- Correspondence:
| |
Collapse
|
14
|
Wilson AL, Moffitt LR, Wilson KL, Bilandzic M, Wright MD, Gorrell MD, Oehler MK, Plebanski M, Stephens AN. DPP4 Inhibitor Sitagliptin Enhances Lymphocyte Recruitment and Prolongs Survival in a Syngeneic Ovarian Cancer Mouse Model. Cancers (Basel) 2021; 13:487. [PMID: 33513866 PMCID: PMC7865851 DOI: 10.3390/cancers13030487] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
Immunity plays a key role in epithelial ovarian cancer (EOC) progression with a well-documented correlation between patient survival and high intratumoral CD8+ to T regulatory cell (Treg) ratios. We previously identified dysregulated DPP4 activity in EOCs as a potentially immune-disruptive influence contributing to a reduction in CXCR3-mediated T-cell infiltration in solid tumours. We therefore hypothesized that inhibition of DPP4 activity by sitagliptin, an FDA-approved inhibitor, would improve T-cell infiltration and function in a syngeneic ID8 mouse model of EOC. Daily oral sitagliptin at 50 mg/kg was provided to mice with established primary EOCs. Sitagliptin treatment decreased metastatic tumour burden and significantly increased overall survival and was associated with significant changes to the immune landscape. Sitagliptin increased overall CXCR3-mediated CD8+ T-cell trafficking to the tumour and enhanced the activation and proliferation of CD8+ T-cells in tumour tissue and the peritoneal cavity. Substantial reductions in suppressive cytokines, including CCL2, CCL17, CCL22 and IL-10, were also noted and were associated with reduced CD4+ CD25+ Foxp3+ Treg recruitment in the tumour. Combination therapy with paclitaxel, however, typical of standard-of-care for patients in palliative care, abolished CXCR3-specific T-cell recruitment stimulated by sitagliptin. Our data suggest that sitagliptin may be suitable as an adjunct therapy for patients between chemotherapy cycles as a novel approach to enhance immunity, optimise T-cell-mediated function and improve overall survival.
Collapse
Affiliation(s)
- Amy L. Wilson
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton 3168, Australia; (A.L.W.); (L.R.M.); (M.B.)
- Department of Molecular and Translational Sciences, Monash Health, Clayton 3168, Australia
- Department of Immunology and Pathology, Monash University, Clayton 3800, Australia;
| | - Laura R. Moffitt
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton 3168, Australia; (A.L.W.); (L.R.M.); (M.B.)
- Department of Molecular and Translational Sciences, Monash Health, Clayton 3168, Australia
| | - Kirsty L. Wilson
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia;
| | - Maree Bilandzic
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton 3168, Australia; (A.L.W.); (L.R.M.); (M.B.)
- Department of Molecular and Translational Sciences, Monash Health, Clayton 3168, Australia
| | - Mark D. Wright
- Department of Immunology and Pathology, Monash University, Clayton 3800, Australia;
| | - Mark D. Gorrell
- Centenary Institute, Faculty of Medicine and Health, University of Sydney, Camperdown 2006, Australia;
| | - Martin K. Oehler
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide 5000, Australia;
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia;
| | - Andrew N. Stephens
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton 3168, Australia; (A.L.W.); (L.R.M.); (M.B.)
- Department of Molecular and Translational Sciences, Monash Health, Clayton 3168, Australia
| |
Collapse
|
15
|
Moffitt LR, Bilandzic M, Wilson AL, Chen Y, Gorrell MD, Oehler MK, Plebanski M, Stephens AN. Hypoxia Regulates DPP4 Expression, Proteolytic Inactivation, and Shedding from Ovarian Cancer Cells. Int J Mol Sci 2020; 21:8110. [PMID: 33143089 PMCID: PMC7672561 DOI: 10.3390/ijms21218110] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 10/28/2020] [Indexed: 01/03/2023] Open
Abstract
The treatment of ovarian cancer has not significantly changed in decades and it remains one of the most lethal malignancies in women. The serine protease dipeptidyl peptidase 4 (DPP4) plays key roles in metabolism and immunity, and its expression has been associated with either pro- or anti-tumour effects in multiple tumour types. In this study, we provide the first evidence that DPP4 expression and enzyme activity are uncoupled under hypoxic conditions in ovarian cancer cells. Whilst we identified strong up-regulation of DPP4 mRNA expression under hypoxic growth, the specific activity of secreted DPP4 was paradoxically decreased. Further investigation revealed matrix metalloproteinases (MMP)-dependent inactivation and proteolytic shedding of DPP4 from the cell surface, mediated by at least MMP10 and MMP13. This is the first report of uncoupled DPP4 expression and activity in ovarian cancer cells, and suggests a previously unrecognized, cell- and tissue-type-dependent mechanism for the regulation of DPP4 in solid tumours. Further studies are necessary to identify the functional consequences of DPP4 processing and its potential prognostic or therapeutic value.
Collapse
Affiliation(s)
- Laura R. Moffitt
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Maree Bilandzic
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Amy L. Wilson
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Yiqian Chen
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Mark D. Gorrell
- Centenary Institute, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Martin K. Oehler
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia;
- Robinson Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia;
| | - Andrew N. Stephens
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia; (L.R.M.); (M.B.); (A.L.W.); (Y.C.)
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| |
Collapse
|
16
|
Bassendine MF, Bridge SH, McCaughan GW, Gorrell MD. COVID-19 and comorbidities: A role for dipeptidyl peptidase 4 (DPP4) in disease severity? J Diabetes 2020; 12:649-658. [PMID: 32394639 DOI: 10.1111/1753-0407.13052] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is caused by a novel betacoronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), similar to SARS-CoV and Middle East respiratory syndrome (MERS-CoV), which cause acute respiratory distress syndrome and case fatalities. COVID-19 disease severity is worse in older obese patients with comorbidities such as diabetes, hypertension, cardiovascular disease, and chronic lung disease. Cell binding and entry of betacoronaviruses is via their surface spike glycoprotein; SARS-CoV binds to the metalloprotease angiotensin-converting enzyme 2 (ACE2), MERS-CoV utilizes dipeptidyl peptidase 4 (DPP4), and recent modeling of the structure of SARS-CoV-2 spike glycoprotein predicts that it can interact with human DPP4 in addition to ACE2. DPP4 is a ubiquitous membrane-bound aminopeptidase that circulates in plasma; it is multifunctional with roles in nutrition, metabolism, and immune and endocrine systems. DPP4 activity differentially regulates glucose homeostasis and inflammation via its enzymatic activity and nonenzymatic immunomodulatory effects. The importance of DPP4 for the medical community has been highlighted by the approval of DPP4 inhibitors, or gliptins, for the treatment of type 2 diabetes mellitus. This review discusses the dysregulation of DPP4 in COVID-19 comorbid conditions; DPP4 activity is higher in older individuals and increased plasma DPP4 is a predictor of the onset of metabolic syndrome. DPP4 upregulation may be a determinant of COVID-19 disease severity, which creates interest regarding the use of gliptins in management of COVID-19. Also, knowledge of the chemistry and biology of DPP4 could be utilized to develop novel therapies to block viral entry of some betacoronaviruses, potentially including SARS-CoV-2.
Collapse
Affiliation(s)
- Margaret F Bassendine
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Simon H Bridge
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Geoffrey W McCaughan
- Centenary Institute and The University of Sydney Faculty of Medicine and Health, Sydney, Australia
| | - Mark D Gorrell
- Centenary Institute and The University of Sydney Faculty of Medicine and Health, Sydney, Australia
| |
Collapse
|
17
|
Karin N. CXCR3 Ligands in Cancer and Autoimmunity, Chemoattraction of Effector T Cells, and Beyond. Front Immunol 2020; 11:976. [PMID: 32547545 PMCID: PMC7274023 DOI: 10.3389/fimmu.2020.00976] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022] Open
Abstract
CXCR3 is a chemokine receptor with three ligands; CXCL9, CXCL10, and CXCL11. CXCL11 binds CXCR3 with a higher affinity than the other ligands leading to receptor internalization. Long ago we reported that one of these chemokines, CXCL10, not only attracts CXCR3+ CD4+ and CD8+ effector T cells to sites of inflammation, but also direct their polarization into highly potent effector T cells. Later we showed that CXCL11 directs the linage development of T-regulatory-1 cells (Tr1). We also observed that CXCL11 and CXCL10 induce different signaling cascades via CXCR3. Collectively this suggests that CXCR3 ligands differentially regulate the biological function of T cells via biased signaling. It is generally accepted that tumor cells evolved to express several chemokine receptors and secrete their ligands. Vast majority of these chemokines support tumor growth by different mechanisms that are discussed. We suggest that CXCL10 and possibly CXCL9 differ from other chemokines by their ability to restrain tumor growth and enhance anti-tumor immunity. Along with this an accumulating number of studies showed in various human cancers a clear association between poor prognosis and low expression of CXCL10 at tumor sites, and vice versa. Finally, we discuss the possibility that CXCL9 and CXCL10 may differ in their biological function via biased signaling and its possible relevance to cancer immunotherapy. The current mini review focuses on exploring the role of CXCR3 ligands in directing the biological properties of CD4+ and CD8+ T cells in the context of cancer and autoimmunity. We believe that the combined role of these chemokines in attracting T cells and also directing their biological properties makes them key drivers of immune function.
Collapse
Affiliation(s)
- Nathan Karin
- Department of Immunology, Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
18
|
Bronger H, Magdolen V, Goettig P, Dreyer T. Proteolytic chemokine cleavage as a regulator of lymphocytic infiltration in solid tumors. Cancer Metastasis Rev 2020; 38:417-430. [PMID: 31482487 PMCID: PMC6890590 DOI: 10.1007/s10555-019-09807-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the past decade, immune-based therapies such as monoclonal antibodies against tumor epitopes or immune checkpoint inhibitors have become an integral part of contemporary cancer treatment in many entities. However, a fundamental prerequisite for the success of such therapies is a sufficient trafficking of tumor-infiltrating lymphocytes into the tumor microenvironment. This infiltration is facilitated by chemokines, a group of about 50 small proteins capable of chemotactically guiding leukocytes. Proteolytic inactivation of chemokines leading to an impaired infiltration of immune effector cells appears to be an efficient immune escape mechanism of solid cancers. The CXCR3 and CX3CR1 chemokine receptor ligands CXCL9-11 and CX3CL1, respectively, are mainly responsible for the tumor-suppressive lymphocytic infiltration into the tumor micromilieu. Their structure explains the biochemical basis of their proteolytic cleavage, while in vivo data from mouse models and patient samples shed light on the corresponding processes in cancer. The emerging roles of proteases, e.g., matrix metalloproteinases, cathepsins, and dipeptidyl peptidase 4, in chemokine inactivation define new resistance mechanisms against immunotherapies and identify attractive new targets to enhance immune intervention in cancer.
Collapse
Affiliation(s)
- Holger Bronger
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Straße 22, D-81675, Munich, Germany.
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Straße 22, D-81675, Munich, Germany
| | - Peter Goettig
- Division of Structural Biology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Tobias Dreyer
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Straße 22, D-81675, Munich, Germany
| |
Collapse
|
19
|
Metabolism of Estrogens: Turnover Differs Between Platinum-Sensitive and -Resistant High-Grade Serous Ovarian Cancer Cells. Cancers (Basel) 2020; 12:cancers12020279. [PMID: 31979221 PMCID: PMC7072378 DOI: 10.3390/cancers12020279] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/09/2020] [Accepted: 01/21/2020] [Indexed: 02/04/2023] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is currently treated with cytoreductive surgery and platinum-based chemotherapy. The majority of patients show a primary response; however, many rapidly develop drug resistance. Antiestrogens have been studied as low toxic treatment options for HGSOC, with higher response rates in platinum-sensitive cases. Mechanisms for this difference in response remain unknown. Therefore, the present study investigated the impact of platinum resistance on steroid metabolism in six established HGSOC cell lines sensitive and resistant against carboplatin using a high-resolution mass spectrometry assay to simultaneously quantify the ten main steroids of the estrogenic metabolic pathway. An up to 60-fold higher formation of steroid hormones and their sulfated or glucuronidated metabolites was observed in carboplatin-sensitive cells, which was reversible by treatment with interleukin-6 (IL-6). Conversely, treatment of carboplatin-resistant cells expressing high levels of endogenous IL-6 with the monoclonal anti-IL-6R antibody tocilizumab changed their status to “platinum-sensitive”, exhibiting a decreased IC50 value for carboplatin, decreased growth, and significantly higher estrogen metabolism. Analysis of these metabolic differences could help to detect platinum resistance in HGSOC patients earlier, thereby allowing more efficient interventions.
Collapse
|
20
|
Eckert EC, Nace RA, Tonne JM, Evgin L, Vile RG, Russell SJ. Generation of a Tumor-Specific Chemokine Gradient Using Oncolytic Vesicular Stomatitis Virus Encoding CXCL9. MOLECULAR THERAPY-ONCOLYTICS 2019; 16:63-74. [PMID: 31930167 PMCID: PMC6951834 DOI: 10.1016/j.omto.2019.12.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/07/2019] [Indexed: 02/08/2023]
Abstract
Genetically modified vesicular stomatitis virus (VSV) is an attractive agent for cancer treatment due to rapid intratumoral replication and observed clinical responses. Although VSV selectively kills malignant cells and can boost antitumor immunity, limited induction of intratumoral immune infiltration remains a barrier to efficacy in some cancer models. Here we engineered the oncolytic VSV platform to encode the T cell chemokine CXCL9, which is known to mediate the recruitment of activated CD8+ cytotoxic T cells and CD4+ T helper cells, and demonstrates conserved protein function between mice and humans. Chemotactic activity of the virally encoded chemokine was confirmed in vitro. Intratumoral concentration of CXCL9 was shown to increase after VSV therapy in three different cancer models, but to a much greater degree after VSV-CXCL9 therapy as compared with VSV control viruses. Despite a steep chemokine gradient from the tumor to the bloodstream, tumor trafficking of adoptively transferred and endogenous T cells was not measurably increased following VSV-CXCL9 therapy. Our results indicate that oncolytic VSV infection promotes release of CXCL9 in the tumor microenvironment, but further boosting of the functional chemokine gradient through virus engineering has little incremental impact on intratumoral immune cell infiltration in mouse and human tumor models.
Collapse
Affiliation(s)
- Elizabeth C Eckert
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA.,Clinical and Translational Science Track, Mayo Graduate School of Biomedical Science, Mayo Clinic, Rochester, MN 55905, USA
| | - Rebecca A Nace
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jason M Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Laura Evgin
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard G Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Stephen J Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
21
|
Bilandzic M, Rainczuk A, Green E, Fairweather N, Jobling TW, Plebanski M, Stephens AN. Keratin-14 (KRT14) Positive Leader Cells Mediate Mesothelial Clearance and Invasion by Ovarian Cancer Cells. Cancers (Basel) 2019; 11:cancers11091228. [PMID: 31443478 PMCID: PMC6769856 DOI: 10.3390/cancers11091228] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/12/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Epithelial ovarian cancer metastasis is driven by spheroids, which are heterogeneous cancer cell aggregates released from the primary tumour mass that passively disseminate throughout the peritoneal cavity to promote tumour spread, disease recurrence, and acquired chemoresistance. Despite their clinical importance, the molecular events that control spheroid attachment and invasion into underlying healthy tissues remain poorly understood. We examined a novel in vitro invasion model using imaging mass spectrometry to establish a “snapshot” of the spheroid/mesothelial interface. Amongst numerous adhesion-related proteins, we identified a sub-population of highly motile, invasive cells that expressed the basal epithelial marker KRT14 as an absolute determinant of invasive potential. The loss of KRT14 completely abrogated the invasive capacity, but had no impact on cell viability or proliferation, suggesting an invasion-specific role. Our data demonstrate KRT14 cells as an ovarian cancer “leader cell” phenotype underlying tumor invasion, and suggest their importance as a clinically relevant target in directed anti-tumour therapies.
Collapse
Affiliation(s)
- Maree Bilandzic
- Hudson Institute of Medical Research, Clayton 3168, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Adam Rainczuk
- Hudson Institute of Medical Research, Clayton 3168, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
- Bruker Biosciences Pty Ltd., Preston 3078, Australia
| | - Emma Green
- Hudson Institute of Medical Research, Clayton 3168, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Nicole Fairweather
- Hudson Institute of Medical Research, Clayton 3168, Australia
- Department of Gynaecology Oncology Monash Health, Monash Medical Centre, Moorabbin 3189, Australia
| | - Thomas W Jobling
- Department of Gynaecology Oncology Monash Health, Monash Medical Centre, Moorabbin 3189, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Andrew N Stephens
- Hudson Institute of Medical Research, Clayton 3168, Australia.
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia.
| |
Collapse
|
22
|
Josahkian JA, Saggioro FP, Vidotto T, Ventura HT, Candido Dos Reis FJ, de Sousa CB, Tiezzi DG, de Andrade JM, Koti M, Squire JA. Increased STAT1 Expression in High Grade Serous Ovarian Cancer Is Associated With a Better Outcome. Int J Gynecol Cancer 2018; 28:459-465. [PMID: 29303938 DOI: 10.1097/igc.0000000000001193] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Recently it has been demonstrated that constitutively activated signal transducer and activator of transcription 1 (STAT1) gene expression may act as a biomarker of ovarian cancer chemotherapy response. In this study, our objective was to validate the use of STAT1 immunohistochemistry as a prognostic biomarker for disease outcome using a cohort derived from Latin America. METHODS We evaluated a cohort of Brazilian high-grade serous ovarian cancer, comprising 65 patients with outcome data covering more than 5 years to determine the prognostic and predictive value of STAT1 expression levels. High-grade serous ovarian cancer tumors were used to construct a tissue microarray. Exploratory analyses were conducted on clinical, histopathological, and STAT1 expression data that included descriptive statistics and Pearson correlative analyses. Survival curves for disease-free survival and overall survival were obtained by the Kaplan-Meier method, and the significance of homogeneity between the classes was assessed by log-rank statistics (Mantel-Cox). RESULTS High expression of STAT1 in tumors was significantly associated with improved disease-free survival (P = 0.0256) and overall survival (P = 0.0193). Proportional hazards regression analysis showed STAT1 expression had an independent effect on both disease-free survival (P = 0.0358) and overall survival (P = 0.0469). CONCLUSIONS These findings from a Brazilian cohort of patients with ovarian cancer reinforce the association of high STAT1 expression with better response to chemotherapy, providing additional validation of this protein as both a prognostic and predictive biomarker. Collectively, these results together with other recently published studies increase the feasibility of using the STAT1 pathway for the development of novel immunomodulator drugs that could enhance response to treatment.
Collapse
|
23
|
García-Hernández MDLL, Uribe-Uribe NO, Espinosa-González R, Kast WM, Khader SA, Rangel-Moreno J. A Unique Cellular and Molecular Microenvironment Is Present in Tertiary Lymphoid Organs of Patients with Spontaneous Prostate Cancer Regression. Front Immunol 2017; 8:563. [PMID: 28567040 PMCID: PMC5434117 DOI: 10.3389/fimmu.2017.00563] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 04/27/2017] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Multiple solid cancers contain tertiary lymphoid organs (TLO). However, it is unclear whether they promote tumor rejection, facilitate tumor evasion, or simply whether they are a byproduct of chronic inflammation. We hypothesize that although chronic inflammation induces TLO formation, the tumor milieu can modulate TLO organization and functions in prostate cancer. Therefore, our study seeks to elucidate the cellular and molecular signatures in unique prostatectomy specimens from evanescent carcinoma patients to identify markers of cancer regression, which could be harnessed to modulate local immunosuppression or potentially enhance TLO function. METHODS We used multicolor immunofluorescence to stain prostate tissues, collected at different stages of cancer progression (prostatic intraepithelial neoplasia, intermediate and advanced cancer) or from patients with evanescent prostate carcinoma. Tissues were stained with antibodies specific for pro-inflammatory molecules (cyclooxygenase 2, CXCL10, IL17), tumor-infiltrating immune cells (mature DC-LAMP+ dendritic cells, CD3+ T cells, CD3+Foxp3+ regulatory T cells (Treg), T bet+ Th1 cells, granzyme B+ cytotoxic cells), and stromal cell populations (lymphatic vessels, tumor neovessels, high endothelial venules (HEV), stromal cells), which promote prostate tumor growth or are critical components of tumor-associated TLO. RESULTS Generally, inflammatory cells are located at the margins of tumors. Unexpectedly, we found TLO within prostate tumors from patients at different stages of cancer and in unique samples from patients with spontaneous cancer remission. In evanescent prostate carcinomas, accumulation of Treg was compromised, while Tbet+ T cells and CD8 T cells were abundant in tumor-associated TLO. In addition, we found a global decrease in tumor neovascularization and the coverage by cells positive for cyclooxygenase 2 (COX2). Finally, consistent with tumor regression, prostate stem cell antigen was considerably reduced in TLO and tumor areas from evanescent carcinoma patients. CONCLUSION Collectively, our results suggest that COX2 and Treg are attractive therapeutic targets that can be harnessed to enhance TLO-driven tumor immunity against prostate cancer. Specially, the presence of HEV and lymphatics indicate that TLO can be used as a platform for delivery of cell-based and/or COX2 blocking therapies to improve control of tumor growth in prostate cancer.
Collapse
Affiliation(s)
| | - Norma Ofelia Uribe-Uribe
- Department of Anatomy and Anatomical Pathology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Ricardo Espinosa-González
- Department of Anatomy and Anatomical Pathology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - W. Martin Kast
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Department of Urology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, USA
| | - Shabaana A. Khader
- Department of Molecular Microbiology, Washington University in Saint Louis, St. Louis, MO, USA
| | | |
Collapse
|
24
|
Wang XM, Holz LE, Chowdhury S, Cordoba SP, Evans KA, Gall MG, Vieira de Ribeiro AJ, Zheng YZ, Levy MT, Yu DM, Yao TW, Polak N, Jolly CJ, Bertolino P, McCaughan GW, Gorrell MD. The pro-fibrotic role of dipeptidyl peptidase 4 in carbon tetrachloride-induced experimental liver injury. Immunol Cell Biol 2017; 95:443-453. [PMID: 27899813 DOI: 10.1038/icb.2016.116] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 11/04/2016] [Accepted: 11/24/2016] [Indexed: 12/19/2022]
Abstract
Liver fibrosis is a progressive pathological process involving inflammation and extracellular matrix deposition. Dipeptidyl peptidase 4 (DPP4), also known as CD26, is a cell surface glycoprotein and serine protease. DPP4 binds to fibronectin, can inactivate specific chemokines, incretin hormone and neuropeptides, and influences cell adhesion and migration. Such properties suggest a pro-fibrotic role for this peptidase but this hypothesis needs in vivo examination. Experimental liver injury was induced with carbon tetrachloride (CCl4) in DPP4 gene knockout (gko) mice. DPP4 gko had less liver fibrosis and inflammation and fewer B cell clusters than wild type mice in the fibrosis model. DPP4 inhibitor-treated mice also developed less liver fibrosis. DNA microarray and PCR showed that many immunoglobulin (Ig) genes and some metabolism-associated transcripts were differentially expressed in the gko strain compared with wild type. CCl4-treated DPP4 gko livers had more IgM+ and IgG+ intrahepatic lymphocytes, and fewer CD4+, IgD+ and CD21+ intrahepatic lymphocytes. These data suggest that DPP4 is pro-fibrotic in CCl4-induced liver fibrosis and that the mechanisms of DPP4 pro-fibrotic action include energy metabolism, B cells, NK cells and CD4+ cells.
Collapse
Affiliation(s)
- Xin M Wang
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- A.W. Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Lauren E Holz
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Sumaiya Chowdhury
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Shaun P Cordoba
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Kathryn A Evans
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Margaret G Gall
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Yuan Zhou Zheng
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Miriam T Levy
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- A.W. Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Denise Mt Yu
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Tsun-Wen Yao
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Natasa Polak
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Christopher J Jolly
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Patrick Bertolino
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Geoffrey W McCaughan
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- A.W. Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Mark D Gorrell
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- A.W. Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| |
Collapse
|
25
|
Henderson JM, Zhang HE, Polak N, Gorrell MD. Hepatocellular carcinoma: Mouse models and the potential roles of proteases. Cancer Lett 2017; 387:106-113. [PMID: 27045475 DOI: 10.1016/j.canlet.2016.03.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 03/24/2016] [Accepted: 03/24/2016] [Indexed: 02/07/2023]
Abstract
Primary liver cancer is the second most common cause of mortality from cancer. The most common models of hepatocellular carcinoma, which use a chemical and/or metabolic insult, xenograft, or genetic manipulation, are discussed in this review. In the tumour microenvironment lymphocytes, fibroblasts, endothelial cells and antigen presenting cells are important determinants of cell fate. These cells make a range of proteases that modify the biological activity of other proteins, particularly extracellular matrix proteins that alter cell migration of tumour cells, fibroblasts and leucocytes, and chemokines that alter leucocyte migration. The DPP4 family of post-proline peptidase enzymes modifies cell movement and the activities of many bioactive molecules including growth factors and chemokines.
Collapse
Affiliation(s)
- James M Henderson
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales 2006 Australia
| | - Hui Emma Zhang
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales 2006 Australia
| | - Natasa Polak
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales 2006 Australia
| | - Mark D Gorrell
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales 2006 Australia.
| |
Collapse
|
26
|
Au KK, Le Page C, Ren R, Meunier L, Clément I, Tyrishkin K, Peterson N, Kendall-Dupont J, Childs T, Francis JA, Graham CH, Craig AW, Squire JA, Mes-Masson AM, Koti M. STAT1-associated intratumoural T H1 immunity predicts chemotherapy resistance in high-grade serous ovarian cancer. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2016. [PMID: 27917296 DOI: 10.1002/cjp2.55] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
High-grade serous ovarian carcinoma (HGSC) accounts for 70% of all epithelial ovarian cancers but clinical management is challenged by a lack of accurate prognostic and predictive biomarkers of chemotherapy response. This study evaluated the role of Signal Transducer and Activator of Transcription 1 (STAT1) as an independent prognostic and predictive biomarker and its correlation with intratumoural CD8+ T cells in a second independent biomarker validation study. Tumour STAT1 expression and intratumoural CD8+ T cell infiltration were assessed by immunohistochemistry as a multicentre validation study conducted on 734 chemotherapy-naïve HGSCs. NanoString-based profiling was performed to correlate expression of STAT1 target genes CXCL9, CXCL10 and CXCL11 with CD8A transcript expression in 143 primary tumours. Multiplexed cytokine analysis of pre-treatment plasma from resistant and sensitive patients was performed to assess systemic levels of STAT1-induced cytokines. STAT1 was validated as a prognostic and predictive biomarker in both univariate and multivariate models and its expression correlated significantly with intra-epithelial CD8+ T cell infiltration in HGSC. STAT1 levels increased the prognostic and predictive value of intratumoural CD8+ T cells, confirming their synergistic role as biomarkers in HGSC. In addition, expression of STAT1 target genes (CXCL9, CXCL10 and CXCL11) correlated significantly with levels of, and CD8A transcripts from intratumoural CD8+ T cells within the resistant and sensitive tumours. Our findings provide compelling evidence that high levels of STAT1, STAT1-induced chemokines and CD8+ T cells correlate with improved chemotherapy response in HGSC. These results identify STAT1 and its target genes as novel biomarkers of chemosensitivity in HGSC. These findings provide new translational opportunities for patient stratification for immunotherapies based on emerging biomarkers of inflammation in HGSC. An improved understanding of the role of interferon-inducible genes will be foundational for developing immunomodulatory therapies in ovarian cancer.
Collapse
Affiliation(s)
- Katrina K Au
- Department of Biomedical and Molecular Sciences Queen's University Kingston Ontario K7L 3N6 Canada
| | - Cécile Le Page
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal Montreal Quebec H2X 0A9 Canada
| | - Runhan Ren
- Department of Biomedical and Molecular Sciences Queen's University Kingston Ontario K7L 3N6 Canada
| | - Liliane Meunier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal Montreal Quebec H2X 0A9 Canada
| | - Isabelle Clément
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal Montreal Quebec H2X 0A9 Canada
| | - Kathrin Tyrishkin
- Department of Pathology and Molecular Medicine Kingston General Hospital Kingston Ontario K7L 3N6 Canada
| | - Nichole Peterson
- Department of Obstetrics and Gynecology Kingston General Hospital Kingston Ontario K7L 3N6 Canada
| | - Jennifer Kendall-Dupont
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal Montreal Quebec H2X 0A9 Canada
| | - Timothy Childs
- Department of Pathology and Molecular Medicine Kingston General Hospital Kingston Ontario K7L 3N6 Canada
| | - Julie-Ann Francis
- Department of Obstetrics and Gynecology Kingston General Hospital Kingston Ontario K7L 3N6 Canada
| | - Charles H Graham
- Department of Biomedical and Molecular Sciences Queen's University Kingston Ontario K7L 3N6 Canada
| | - Andrew W Craig
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioK7L 3N6Canada; Cancer Biology and Genetics DivisionQueen's Cancer Research Institute, Queen's UniversityOntarioK7L 3N6Canada
| | - Jeremy A Squire
- Departments of Genetics and Pathology Faculdade de Medicina de Ribeirão Preto - USP Av. Bandeirantes Ribeirão Preto São Paulo 3900 Brazil
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de MontréalMontrealQuebecH2X 0A9Canada; Department of MedicineUniversité de MontréalMontrealQuebecH3C 3J7Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioK7L 3N6Canada; Department of Obstetrics and GynecologyKingston General HospitalKingstonOntarioK7L 3N6Canada; Cancer Biology and Genetics DivisionQueen's Cancer Research Institute, Queen's UniversityOntarioK7L 3N6Canada
| |
Collapse
|
27
|
Au KK, Le Page C, Ren R, Meunier L, Clément I, Tyrishkin K, Peterson N, Kendall-Dupont J, Childs T, Francis JA, Graham CH, Craig AW, Squire JA, Mes-Masson AM, Koti M. STAT1-associated intratumoural T H1 immunity predicts chemotherapy resistance in high-grade serous ovarian cancer. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2016. [PMID: 27917296 DOI: 10.1002/cjp2.55]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
High-grade serous ovarian carcinoma (HGSC) accounts for 70% of all epithelial ovarian cancers but clinical management is challenged by a lack of accurate prognostic and predictive biomarkers of chemotherapy response. This study evaluated the role of Signal Transducer and Activator of Transcription 1 (STAT1) as an independent prognostic and predictive biomarker and its correlation with intratumoural CD8+ T cells in a second independent biomarker validation study. Tumour STAT1 expression and intratumoural CD8+ T cell infiltration were assessed by immunohistochemistry as a multicentre validation study conducted on 734 chemotherapy-naïve HGSCs. NanoString-based profiling was performed to correlate expression of STAT1 target genes CXCL9, CXCL10 and CXCL11 with CD8A transcript expression in 143 primary tumours. Multiplexed cytokine analysis of pre-treatment plasma from resistant and sensitive patients was performed to assess systemic levels of STAT1-induced cytokines. STAT1 was validated as a prognostic and predictive biomarker in both univariate and multivariate models and its expression correlated significantly with intra-epithelial CD8+ T cell infiltration in HGSC. STAT1 levels increased the prognostic and predictive value of intratumoural CD8+ T cells, confirming their synergistic role as biomarkers in HGSC. In addition, expression of STAT1 target genes (CXCL9, CXCL10 and CXCL11) correlated significantly with levels of, and CD8A transcripts from intratumoural CD8+ T cells within the resistant and sensitive tumours. Our findings provide compelling evidence that high levels of STAT1, STAT1-induced chemokines and CD8+ T cells correlate with improved chemotherapy response in HGSC. These results identify STAT1 and its target genes as novel biomarkers of chemosensitivity in HGSC. These findings provide new translational opportunities for patient stratification for immunotherapies based on emerging biomarkers of inflammation in HGSC. An improved understanding of the role of interferon-inducible genes will be foundational for developing immunomodulatory therapies in ovarian cancer.
Collapse
Affiliation(s)
- Katrina K Au
- Department of Biomedical and Molecular Sciences Queen's University Kingston Ontario K7L 3N6 Canada
| | - Cécile Le Page
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal Montreal Quebec H2X 0A9 Canada
| | - Runhan Ren
- Department of Biomedical and Molecular Sciences Queen's University Kingston Ontario K7L 3N6 Canada
| | - Liliane Meunier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal Montreal Quebec H2X 0A9 Canada
| | - Isabelle Clément
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal Montreal Quebec H2X 0A9 Canada
| | - Kathrin Tyrishkin
- Department of Pathology and Molecular Medicine Kingston General Hospital Kingston Ontario K7L 3N6 Canada
| | - Nichole Peterson
- Department of Obstetrics and Gynecology Kingston General Hospital Kingston Ontario K7L 3N6 Canada
| | - Jennifer Kendall-Dupont
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal Montreal Quebec H2X 0A9 Canada
| | - Timothy Childs
- Department of Pathology and Molecular Medicine Kingston General Hospital Kingston Ontario K7L 3N6 Canada
| | - Julie-Ann Francis
- Department of Obstetrics and Gynecology Kingston General Hospital Kingston Ontario K7L 3N6 Canada
| | - Charles H Graham
- Department of Biomedical and Molecular Sciences Queen's University Kingston Ontario K7L 3N6 Canada
| | - Andrew W Craig
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioK7L 3N6Canada; Cancer Biology and Genetics DivisionQueen's Cancer Research Institute, Queen's UniversityOntarioK7L 3N6Canada
| | - Jeremy A Squire
- Departments of Genetics and Pathology Faculdade de Medicina de Ribeirão Preto - USP Av. Bandeirantes Ribeirão Preto São Paulo 3900 Brazil
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de MontréalMontrealQuebecH2X 0A9Canada; Department of MedicineUniversité de MontréalMontrealQuebecH3C 3J7Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioK7L 3N6Canada; Department of Obstetrics and GynecologyKingston General HospitalKingstonOntarioK7L 3N6Canada; Cancer Biology and Genetics DivisionQueen's Cancer Research Institute, Queen's UniversityOntarioK7L 3N6Canada
| |
Collapse
|
28
|
Au KK, Le Page C, Ren R, Meunier L, Clément I, Tyrishkin K, Peterson N, Kendall-Dupont J, Childs T, Francis JA, Graham CH, Craig AW, Squire JA, Mes-Masson AM, Koti M. STAT1-associated intratumoural T H1 immunity predicts chemotherapy resistance in high-grade serous ovarian cancer. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2016; 2:259-270. [PMID: 27917296 PMCID: PMC5129574 DOI: 10.1002/cjp2.55] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/24/2016] [Accepted: 08/06/2016] [Indexed: 12/15/2022]
Abstract
High‐grade serous ovarian carcinoma (HGSC) accounts for 70% of all epithelial ovarian cancers but clinical management is challenged by a lack of accurate prognostic and predictive biomarkers of chemotherapy response. This study evaluated the role of Signal Transducer and Activator of Transcription 1 (STAT1) as an independent prognostic and predictive biomarker and its correlation with intratumoural CD8+ T cells in a second independent biomarker validation study. Tumour STAT1 expression and intratumoural CD8+ T cell infiltration were assessed by immunohistochemistry as a multicentre validation study conducted on 734 chemotherapy‐naïve HGSCs. NanoString‐based profiling was performed to correlate expression of STAT1 target genes CXCL9, CXCL10 and CXCL11 with CD8A transcript expression in 143 primary tumours. Multiplexed cytokine analysis of pre‐treatment plasma from resistant and sensitive patients was performed to assess systemic levels of STAT1‐induced cytokines. STAT1 was validated as a prognostic and predictive biomarker in both univariate and multivariate models and its expression correlated significantly with intra‐epithelial CD8+ T cell infiltration in HGSC. STAT1 levels increased the prognostic and predictive value of intratumoural CD8+ T cells, confirming their synergistic role as biomarkers in HGSC. In addition, expression of STAT1 target genes (CXCL9, CXCL10 and CXCL11) correlated significantly with levels of, and CD8A transcripts from intratumoural CD8+ T cells within the resistant and sensitive tumours. Our findings provide compelling evidence that high levels of STAT1, STAT1‐induced chemokines and CD8+ T cells correlate with improved chemotherapy response in HGSC. These results identify STAT1 and its target genes as novel biomarkers of chemosensitivity in HGSC. These findings provide new translational opportunities for patient stratification for immunotherapies based on emerging biomarkers of inflammation in HGSC. An improved understanding of the role of interferon‐inducible genes will be foundational for developing immunomodulatory therapies in ovarian cancer.
Collapse
Affiliation(s)
- Katrina K Au
- Department of Biomedical and Molecular Sciences Queen's University Kingston Ontario K7L 3N6 Canada
| | - Cécile Le Page
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal Montreal Quebec H2X 0A9 Canada
| | - Runhan Ren
- Department of Biomedical and Molecular Sciences Queen's University Kingston Ontario K7L 3N6 Canada
| | - Liliane Meunier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal Montreal Quebec H2X 0A9 Canada
| | - Isabelle Clément
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal Montreal Quebec H2X 0A9 Canada
| | - Kathrin Tyrishkin
- Department of Pathology and Molecular Medicine Kingston General Hospital Kingston Ontario K7L 3N6 Canada
| | - Nichole Peterson
- Department of Obstetrics and Gynecology Kingston General Hospital Kingston Ontario K7L 3N6 Canada
| | - Jennifer Kendall-Dupont
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal Montreal Quebec H2X 0A9 Canada
| | - Timothy Childs
- Department of Pathology and Molecular Medicine Kingston General Hospital Kingston Ontario K7L 3N6 Canada
| | - Julie-Ann Francis
- Department of Obstetrics and Gynecology Kingston General Hospital Kingston Ontario K7L 3N6 Canada
| | - Charles H Graham
- Department of Biomedical and Molecular Sciences Queen's University Kingston Ontario K7L 3N6 Canada
| | - Andrew W Craig
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioK7L 3N6Canada; Cancer Biology and Genetics DivisionQueen's Cancer Research Institute, Queen's UniversityOntarioK7L 3N6Canada
| | - Jeremy A Squire
- Departments of Genetics and Pathology Faculdade de Medicina de Ribeirão Preto - USP Av. Bandeirantes Ribeirão Preto São Paulo 3900 Brazil
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de MontréalMontrealQuebecH2X 0A9Canada; Department of MedicineUniversité de MontréalMontrealQuebecH3C 3J7Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioK7L 3N6Canada; Department of Obstetrics and GynecologyKingston General HospitalKingstonOntarioK7L 3N6Canada; Cancer Biology and Genetics DivisionQueen's Cancer Research Institute, Queen's UniversityOntarioK7L 3N6Canada
| |
Collapse
|
29
|
Intratumoral interferon-gamma increases chemokine production but fails to increase T cell infiltration of human melanoma metastases. Cancer Immunol Immunother 2016; 65:1189-99. [PMID: 27522581 DOI: 10.1007/s00262-016-1881-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/11/2016] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Optimal approaches to induce T cell infiltration of tumors are not known. Chemokines CXCL9, CXCL10, and CXCL11 support effector T cell recruitment and may be induced by IFN. This study tests the hypothesis that intratumoral administration of IFNγ will induce CXCL9-11 and will induce T cell recruitment and anti-tumor immune signatures in melanoma metastases. PATIENTS AND METHODS Nine eligible patients were immunized with a vaccine comprised of 12 class I MHC-restricted melanoma peptides and received IFNγ intratumorally. Effects on the tumor microenvironment were evaluated in sequential tumor biopsies. Adverse events (AEs) were recorded. T cell responses to vaccination were assessed in PBMC by IFNγ ELISPOT assay. Tumor biopsies were evaluated for immune cell infiltration, chemokine protein expression, and gene expression. RESULTS Vaccination and intratumoral administration of IFNγ were well tolerated. Circulating T cell responses to vaccine were detected in six of nine patients. IFNγ increased production of chemokines CXCL10, CXCL11, and CCL5 in patient tumors. Neither vaccination alone, nor the addition of IFNγ promoted immune cell infiltration or induced anti-tumor immune gene signatures. CONCLUSION The melanoma vaccine induced circulating T cell responses, but it failed to infiltrate metastases, thus highlighting the need for combination strategies to support T cell infiltration. A single intratumoral injection of IFNγ induced T cell-attracting chemokines; however, it also induced secondary immune regulation that may paradoxically limit immune infiltration and effector functions. Alternate dosing strategies or additional combinatorial treatments may be needed to promote trafficking and retention of tumor-reactive T cells in melanoma metastases.
Collapse
|
30
|
CXCL9 and CXCL10 predict survival and are regulated by cyclooxygenase inhibition in advanced serous ovarian cancer. Br J Cancer 2016; 115:553-63. [PMID: 27490802 PMCID: PMC4997538 DOI: 10.1038/bjc.2016.172] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/18/2016] [Accepted: 05/11/2016] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Tumour-infiltrating lymphocytes (TILs) are associated with improved survival in several epithelial cancers. The two chemokines CXCL9 and CXCL10 facilitate chemotactic recruitment of TILs, and their intratumoral accumulation is a conceivable way to improve TIL-dependent immune intervention in cancer. However, the prognostic impact of CXCL9 and CXCL10 in high-grade serous ovarian cancer (HGSC) is largely unknown. METHODS One hundred and eighty four cases of HGSC were immunohistochemically analyzed for CXCL9, CXCL10. TILs were assessed using CD3, CD56 and FOXP3 staining. Chemokine regulation was investigated using the ovarian cancer cell lines OV-MZ-6 and SKOV-3. RESULTS High expression of CXCL9 and CXCL10 was associated with an approximately doubled overall survival (n=70, CXCL9: HR 0.41; P=0.006; CXCL10: HR 0.46; P=0.010) which was confirmed in an independent validation set (n=114; CXCL9: HR 0.60; P=0.019; CXCL10: HR 0.52; P=0.005). Expression of CXCR3 ligands significantly correlated with TILs. In human ovarian cancer cell lines the cyclooxygenase (COX) metabolite Prostaglandin E2 was identified as negative regulator of chemokine secretion, whereas COX inhibition by indomethacin significantly upregulated CXCL9 and CXCL10. In contrast, celecoxib, the only COX inhibitor prospectively evaluated for therapy of ovarian cancer, suppressed NF-κB activation and inhibited chemokine release. CONCLUSION Our results support the notion that CXCL9 and CXCL10 exert tumour-suppressive function by TIL recruitment in human ovarian cancer. COX inhibition by indomethacin, not by celecoxib, may be a promising approach to concomitantly improve immunotherapies.
Collapse
|
31
|
Ovarian cancer and the immune system - The role of targeted therapies. Gynecol Oncol 2016; 142:349-56. [PMID: 27174875 DOI: 10.1016/j.ygyno.2016.05.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/03/2016] [Accepted: 05/07/2016] [Indexed: 01/21/2023]
Abstract
The majority of patients with epithelial ovarian cancer are diagnosed with advanced disease. While many of these patients will respond initially to chemotherapy, the majority will relapse and die of their disease. Targeted therapies that block or activate specific intracellular signaling pathways have been disappointing. In the past 15years, the role of the immune system in ovarian cancer has been investigated. Patients with a more robust immune response, as documented by the presence of lymphocytes infiltrating within their tumor, have increased survival and better response to chemotherapy. In addition, a strong immunosuppressive environment often accompanies ovarian cancer. Recent research has identified potential therapies that leverage the immune system to identify and destroy tumor cells that previously evaded immunosurveillance mechanisms. In this review, we discuss the role of the immune system in ovarian cancer and focus on specific pathways and molecules that show a potential for targeted therapy. We also review the ongoing clinical trials using targeted immunotherapy in ovarian cancer. The role of targeted immunotherapy in patients with ovarian cancer represents a field of growing research and clinical importance.
Collapse
|
32
|
Sato Y, Motoyama S, Nanjo H, Wakita A, Yoshino K, Sasaki T, Nagaki Y, Liu J, Imai K, Saito H, Minamiya Y. CXCL10 Expression Status is Prognostic in Patients with Advanced Thoracic Esophageal Squamous Cell Carcinoma. Ann Surg Oncol 2015; 23:936-42. [PMID: 26464192 DOI: 10.1245/s10434-015-4909-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND CXCL10, a member of the CXC chemokine family, is known to mediate chemotaxis, apoptosis, angiogenesis, and cell growth. It is also reportedly involved in tumor development and can affect prognosis in several cancers. However, the precise relationship between CXCL10 and the prognosis of patients with esophageal squamous cell carcinoma (ESCC) is not fully understood. METHODS We used ESCC tissue microarrays containing samples from 177 patients to test whether the CXCL10 expression status, determined using immunohistochemical analysis, is predictive of prognosis. We also tested whether CXCL10 expression status could serve as a clinically useful marker for evaluating the need for adjuvant chemotherapy after surgery. RESULTS We found that high CXCL10 expression in clinical samples was an independent prognostic factor and was predictive of a favorable 5-year overall survival and disease-specific survival (p = 0.0102 and 0.0332, respectively). Additionally, no significant difference was detected between patients in the CXCL10-high group treated with surgery alone and those treated with surgery followed by adjuvant chemotherapy. In the CXCL10-low group, on the other hand, patients treated with surgery followed by adjuvant chemotherapy had better 5-year overall survival than those treated with surgery alone. CONCLUSIONS High CXCL10 expression is an independent prognostic factor and has the potential to serve as a clinically useful marker of the need for adjuvant chemotherapy after surgery in patients with advanced thoracic ESCC.
Collapse
Affiliation(s)
- Yusuke Sato
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan.
| | - Satoru Motoyama
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroshi Nanjo
- Department of Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Akiyuki Wakita
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Kei Yoshino
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Tomohiko Sasaki
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Yushi Nagaki
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Jiajia Liu
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Kazuhiro Imai
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Hajime Saito
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Yoshihiro Minamiya
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
33
|
Zhang H, Maqsudi S, Rainczuk A, Duffield N, Lawrence J, Keane FM, Justa-Schuch D, Geiss-Friedlander R, Gorrell MD, Stephens AN. Identification of novel dipeptidyl peptidase 9 substrates by two-dimensional differential in-gel electrophoresis. FEBS J 2015; 282:3737-3757. [PMID: 26175140 DOI: 10.1111/febs.13371] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/22/2015] [Accepted: 07/07/2015] [Indexed: 12/26/2022]
Abstract
Dipeptidyl peptidase 9 (DPP9) is a member of the S9B/DPPIV (DPP4) serine protease family, which cleaves N-terminal dipeptides at an Xaa-Pro consensus motif. Cytoplasmic DPP9 has roles in epidermal growth factor signalling and in antigen processing, whilst the role of the recently discovered nuclear form of DPP9 is unknown. Mice lacking DPP9 proteolytic activity die as neonates. We applied a modified 2D differential in-gel electrophoresis approach to identify novel DPP9 substrates, using mouse embryonic fibroblasts lacking endogenous DPP9 activity. A total of 111 potential new DPP9 substrates were identified, with nine proteins/peptides confirmed as DPP9 substrates by MALDI-TOF or immunoblotting. Moreover, we also identified the dipeptide Val-Ala as a consensus site for DPP9 cleavage that was not recognized by DPP8, suggesting different in vivo roles for these closely related enzymes. The relative kinetics for the cleavage of these nine candidate substrates by DPP9, DPP8 and DPP4 were determined. This is the first identification of DPP9 substrates from cells lacking endogenous DPP9 activity. These data greatly expand the potential roles of DPP9 and suggest different in vivo roles for DPP9 and DPP8.
Collapse
Affiliation(s)
- Hui Zhang
- Molecular Hepatology, Liver Injury and Cancer Group, Centenary Institute, Sydney Medical School, University of Sydney, Australia
| | - Sadiqa Maqsudi
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Adam Rainczuk
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Nadine Duffield
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Josie Lawrence
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Fiona M Keane
- Molecular Hepatology, Liver Injury and Cancer Group, Centenary Institute, Sydney Medical School, University of Sydney, Australia
| | - Daniela Justa-Schuch
- Department of Molecular Biology, Faculty of Medicine, Georg-August-University of Goettingen, Germany
| | - Ruth Geiss-Friedlander
- Department of Molecular Biology, Faculty of Medicine, Georg-August-University of Goettingen, Germany
| | - Mark D Gorrell
- Molecular Hepatology, Liver Injury and Cancer Group, Centenary Institute, Sydney Medical School, University of Sydney, Australia
| | - Andrew N Stephens
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
- Epworth Research Institute, Epworth HealthCare, Richmond, Victoria, Australia
| |
Collapse
|
34
|
Waumans Y, Baerts L, Kehoe K, Lambeir AM, De Meester I. The Dipeptidyl Peptidase Family, Prolyl Oligopeptidase, and Prolyl Carboxypeptidase in the Immune System and Inflammatory Disease, Including Atherosclerosis. Front Immunol 2015; 6:387. [PMID: 26300881 PMCID: PMC4528296 DOI: 10.3389/fimmu.2015.00387] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/13/2015] [Indexed: 12/19/2022] Open
Abstract
Research from over the past 20 years has implicated dipeptidyl peptidase (DPP) IV and its family members in many processes and different pathologies of the immune system. Most research has been focused on either DPPIV or just a few of its family members. It is, however, essential to consider the entire DPP family when discussing any one of its members. There is a substantial overlap between family members in their substrate specificity, inhibitors, and functions. In this review, we provide a comprehensive discussion on the role of prolyl-specific peptidases DPPIV, FAP, DPP8, DPP9, dipeptidyl peptidase II, prolyl carboxypeptidase, and prolyl oligopeptidase in the immune system and its diseases. We highlight possible therapeutic targets for the prevention and treatment of atherosclerosis, a condition that lies at the frontier between inflammation and cardiovascular disease.
Collapse
Affiliation(s)
- Yannick Waumans
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp , Antwerp , Belgium
| | - Lesley Baerts
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp , Antwerp , Belgium
| | - Kaat Kehoe
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp , Antwerp , Belgium
| | - Anne-Marie Lambeir
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp , Antwerp , Belgium
| | - Ingrid De Meester
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp , Antwerp , Belgium
| |
Collapse
|
35
|
Kreuzinger C, Gamperl M, Wolf A, Heinze G, Geroldinger A, Lambrechts D, Boeckx B, Smeets D, Horvat R, Aust S, Hamilton G, Zeillinger R, Cacsire Castillo-Tong D. Molecular characterization of 7 new established cell lines from high grade serous ovarian cancer. Cancer Lett 2015; 362:218-28. [DOI: 10.1016/j.canlet.2015.03.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 03/23/2015] [Accepted: 03/31/2015] [Indexed: 12/27/2022]
|
36
|
Zsiros E, Duttagupta P, Dangaj D, Li H, Frank R, Garrabrant T, Hagemann IS, Levine BL, June CH, Zhang L, Wang E, Marincola FM, Bedognetti D, Powell DJ, Tanyi J, Feldman MD, Kandalaft LE, Coukos G. The Ovarian Cancer Chemokine Landscape Is Conducive to Homing of Vaccine-Primed and CD3/CD28-Costimulated T Cells Prepared for Adoptive Therapy. Clin Cancer Res 2015; 21:2840-50. [PMID: 25712684 DOI: 10.1158/1078-0432.ccr-14-2777] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 02/06/2015] [Indexed: 12/18/2022]
Abstract
PURPOSE Chemokines are implicated in T-cell trafficking. We mapped the chemokine landscape in advanced stage ovarian cancer and characterized the expression of cognate receptors in autologous dendritic cell (DC)-vaccine primed T cells in the context of cell-based immunotherapy. EXPERIMENTAL DESIGN The expression of all known human chemokines in patients with primary ovarian cancer was analyzed on two independent microarray datasets and validated on tissue microarray. Peripheral blood T cells from five HLA-A2 patients with recurrent ovarian cancer, who previously received autologous tumor DC vaccine, underwent CD3/CD28 costimulation and expansion ex vivo. Tumor-specific T cells were identified by HER2/neu pentamer staining and were evaluated for the expression and functionality of chemokine receptors important for homing to ovarian cancer. RESULTS The chemokine landscape of ovarian cancer is heterogeneous with high expression of known lymphocyte-recruiting chemokines (CCL2, CCL4, and CCL5) in tumors with intraepithelial T cells, whereas CXCL10, CXCL12, and CXCL16 are expressed quasi-universally, including in tumors lacking tumor-infiltrating T cells. DC-vaccine primed T cells were found to express the cognate receptors for the above chemokines. Ex vivo CD3/CD28 costimulation and expansion of vaccine-primed Tcells upregulated CXCR3 and CXCR4, and enhanced their migration toward universally expressed chemokines in ovarian cancer. CONCLUSIONS DC-primed tumor-specific T cells are armed with the appropriate receptors to migrate toward universal ovarian cancer chemokines, and these receptors are further upregulated by ex vivo CD3/CD28 costimulation, which render T cells more fit for migrating toward these chemokines. Clin Cancer Res; 21(12); 2840-50. ©2015 AACR.
Collapse
Affiliation(s)
- Emese Zsiros
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania. Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, New York. Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, New York
| | - Priyanka Duttagupta
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Denarda Dangaj
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania. Ludwig Cancer Research Center, University of Lausanne, Lausanne, Switzerland
| | - Hongzhe Li
- Department of Epidemiology & Biostatistics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Renee Frank
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Thomas Garrabrant
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ian S Hagemann
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bruce L Levine
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Carl H June
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lin Zhang
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ena Wang
- National Institutes of Health, Bethesda, Maryland. Sidra Medical and Research Centre, Doha, Qatar
| | - Francesco M Marincola
- National Institutes of Health, Bethesda, Maryland. Sidra Medical and Research Centre, Doha, Qatar
| | - Davide Bedognetti
- National Institutes of Health, Bethesda, Maryland. Sidra Medical and Research Centre, Doha, Qatar
| | - Daniel J Powell
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Janos Tanyi
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael D Feldman
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lana E Kandalaft
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania. Ludwig Cancer Research Center, University of Lausanne, Lausanne, Switzerland. Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania. Ludwig Cancer Research Center, University of Lausanne, Lausanne, Switzerland. Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
37
|
Zhang H, Chen Y, Wadham C, McCaughan GW, Keane FM, Gorrell MD. Dipeptidyl peptidase 9 subcellular localization and a role in cell adhesion involving focal adhesion kinase and paxillin. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1853:470-480. [PMID: 25486458 DOI: 10.1016/j.bbamcr.2014.11.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 11/24/2014] [Accepted: 11/26/2014] [Indexed: 12/12/2022]
Abstract
Dipeptidyl peptidase 9 (DPP9) is a ubiquitously expressed member of the DPP4 gene and protease family. Deciphering the biological functions of DPP9 and its roles in pathogenesis has implicated DPP9 in tumor biology, the immune response, apoptosis, intracellular epidermal growth factor-dependent signaling and cell adhesion and migration. We investigated the intracellular distribution of DPP9 chimeric fluorescent proteins and consequent functions of DPP9. We showed that while some DPP9 is associated with mitochondria, the strongest co-localization was with microtubules. Under steady state conditions, DPP9 was not seen at the plasma membrane, but upon stimulation with either phorbol 12-myristate 13-acetate or epidermal growth factor, some DPP9 re-distributed towards the ruffling membrane. DPP9 was seen at the leading edge of the migrating cell and co-localized with the focal adhesion proteins, integrin-β1 and talin. DPP9 gene silencing and treatment with a DPP8/DPP9 specific inhibitor both reduced cell adhesion and migration. Expression of integrin-β1 and talin was decreased in DPP9-deficient and DPP9-enzyme-inactive cells. There was a concomitant decrease in the phosphorylation of focal adhesion kinase and paxillin, indicating that DPP9 knockdown or enzyme inhibition suppressed the associated adhesion signaling pathway, causing impaired cell movement. These novel findings provide mechanistic insights into the regulatory role of DPP9 in cell movement, and may thus implicate DPP9 in tissue and tumor growth and metastasis.
Collapse
Affiliation(s)
- Hui Zhang
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Yiqian Chen
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Carol Wadham
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Geoffrey W McCaughan
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Fiona M Keane
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Mark D Gorrell
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
38
|
Lamon S, Zacharewicz E, Stephens AN, Russell AP. EPO-receptor is present in mouse C2C12 and human primary skeletal muscle cells but EPO does not influence myogenesis. Physiol Rep 2014; 2:e00256. [PMID: 24760510 PMCID: PMC4002236 DOI: 10.1002/phy2.256] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The role and regulation of the pleiotropic cytokine erythropoietin (EPO) in skeletal muscle are controversial. EPO exerts its effects by binding its specific receptor (EPO‐R), which activates intracellular signaling and gene transcription in response to internal and external stress signals. EPO is suggested to play a direct role in myogenesis via the EPO‐R, but several studies have questioned the effect of EPO treatment in muscle in vitro and in vivo. The lack of certainty surrounding the use of nonspecific EPO‐R antibodies contributes to the ambiguity of the field. Our study demonstrates that the EPO‐R gene and protein are expressed at each stage of mouse C2C12 and human skeletal muscle cell proliferation and differentiation and validates a specific antibody for the detection of the EPO‐R protein. However, in our experimental conditions, EPO treatment had no effect on mouse C2C12 and human muscle cell proliferation, differentiation, protein synthesis or EPO‐R expression. While an increase in Akt and MAPK phosphorylation was observed, we demonstrate that this effect resulted from the stress caused by changing medium and not from EPO treatment. We therefore suggest that skeletal muscle EPO‐R might be present in a nonfunctional form, or too lowly expressed to play a role in muscle cell function. The EPO‐R is expressed at the gene and protein level in mouse and human myoblasts and myotubes. However, EPO treatment does not seem to activate the EPO‐R and its downstream signaling pathways in skeletal muscle cells, questioning its functionality.
Collapse
Affiliation(s)
- Séverine Lamon
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Victoria, Australia
| | | | | | | |
Collapse
|