1
|
Ogasawara M, Miyashita M, Yamagishi Y, Ota S. Impact of Reduction in Myeloid-derived Suppressor Cells by Wilms' Tumor 1-targeted Dendritic Cell Vaccines on Clinical Outcomes in Acute Leukemia Patients. EJHAEM 2025; 6:e70048. [PMID: 40370631 PMCID: PMC12077754 DOI: 10.1002/jha2.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/29/2025] [Accepted: 04/13/2025] [Indexed: 05/16/2025]
Abstract
Background Myeloid-derived suppressor cells (MDSCs) play a critical role in immunotherapy. Methods We investigated the effects of the Wilms' tumor 1 (WT1) peptide-loaded dendritic cell (DC) vaccination on MDSCs in patients with acute leukemia. Results WT1-DC vaccination reduced MDSCs and enhanced WT1-specific immunity. In complete remission patients, MDSC reduction was accompanied by decreased arginase 1 and indoleamine 2,3-dioxygenase levels and increased interleukin (IL)-12 and interferon-γ levels in plasma. Conversely, patients with disease progression showed increased IL-10 and transforming growth factor-β1. Reduced MDSCs were correlated with WT1-specific immune activation and associated with longer survival. Conclusion These findings indicate that WT1-DC vaccination suppresses MDSCs and improves clinical outcomes. Clinical Trial Registration This study is registered with the University Hospital Medical Information Network (UMIN) in Japan (Registration ID: UMIN000027279).
Collapse
Affiliation(s)
- Masahiro Ogasawara
- Department of HematologySapporo Hokuyu HospitalSapporoJapan
- Institute for Artificial OrganTransplantation and Cell TherapySapporo Hokuyu HospitalSapporoJapan
| | - Mamiko Miyashita
- Institute for Artificial OrganTransplantation and Cell TherapySapporo Hokuyu HospitalSapporoJapan
| | - Yuka Yamagishi
- Cell Processing CenterSapporo Hokuyu HospitalSapporoJapan
| | - Shuichi Ota
- Department of HematologySapporo Hokuyu HospitalSapporoJapan
| |
Collapse
|
2
|
Jamy O, Cicic D. REGAL: galinpepimut-S vs. best available therapy as maintenance therapy for acute myeloid leukemia in second remission. Future Oncol 2025; 21:73-81. [PMID: 39606837 PMCID: PMC11760237 DOI: 10.1080/14796694.2024.2433935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
Patients with relapsed or refractory (r/r) acute myeloid leukemia (AML) have very poor long-term outcomes. Allogeneic stem cell transplantation (allo-SCT) can potentially cure some of these patients who are able to achieve a second or greater remission with salvage chemotherapy. Unfortunately, several barriers exist to transplantation and not all patients with r/r AML are able to proceed to allo-SCT. Therefore, novel therapies to decrease the risk of relapse in these patients are urgently needed. Wilms tumor 1 (WT1) protein has emerged as an encouraging vaccine target in AML due to its overexpression in leukemic blast cells and near absence in normal hematopoietic cells. Maintenance therapy with galinpepimut-S, a multivalent heteroclitic WT1 peptide vaccine, holds promise in early phase trials, in patients with AML by inducing a strong innate immune response against the WT1 antigen, leading to the design of this international, open-label, randomized clinical trial, named REGAL. Clinical trial registration: https://clinicaltrials.gov/study/NCT04229979. The clinical trial identifier is NCT04229979.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- WT1 Proteins/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/therapeutic use
- Remission Induction
- Randomized Controlled Trials as Topic
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/therapeutic use
- Maintenance Chemotherapy/methods
- Female
- Treatment Outcome
- Male
- Adult
- Middle Aged
- Hematopoietic Stem Cell Transplantation
- Salvage Therapy
Collapse
Affiliation(s)
- Omer Jamy
- Division of Hematology/Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dragan Cicic
- Department of Clinical Development, SELLAS Life Sciences Group, Inc., New York, NY, USA
| |
Collapse
|
3
|
Díaz del Moral S, Wagner N, Wagner KD. The Wilms' Tumor Suppressor WT1 in Cardiomyocytes: Implications for Cardiac Homeostasis and Repair. Cells 2024; 13:2078. [PMID: 39768169 PMCID: PMC11674098 DOI: 10.3390/cells13242078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/11/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
The Wilms' tumor suppressor WT1 is essential for the development of the heart, among other organs such as the kidneys and gonads. The Wt1 gene encodes a zinc finger transcription factor that regulates proliferation, cellular differentiation processes, and apoptosis. WT1 is also involved in cardiac homeostasis and repair. In adulthood, WT1-expression levels are lower compared to those observed through development, and WT1 expression is restricted to a few cell types. However, its systemic deletion in adult mice is lethal, demonstrating that its presence is also key for organ maintenance. In response to injury, the epicardium re-activates the expression of WT1, but little is known about the roles it plays in cardiomyocytes, which are the main cell type affected after myocardial infarction. The fact that cardiomyocytes exhibit a low proliferation rate in the adult heart in mammals highlights the need to explore new approaches for cardiac regeneration. The aim of this review is to emphasize the functions carried out by WT1 in cardiomyocytes in cardiac homeostasis and heart regeneration.
Collapse
Affiliation(s)
| | | | - Kay-Dietrich Wagner
- Université Côte d’Azur, CNRS, INSERM, iBV, 06107 Nice, France; (S.D.d.M.); (N.W.)
| |
Collapse
|
4
|
Song Q, Li Y, Wu T, Hu W, Liu Y, Liu A. Feasibility of iodine concentration parameter and extracellular volume fraction derived from dual-energy CT for distinguishing type I and type II epithelial ovarian carcinoma. Abdom Radiol (NY) 2024:10.1007/s00261-024-04752-4. [PMID: 39665991 DOI: 10.1007/s00261-024-04752-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
OBJECTIVES To investigate the feasibility of using the iodine concentration (IC) parameter and extracellular volume (ECV) fraction derived from dual-energy CT for distinguishing between type I and type II epithelial ovarian carcinoma (EOC). METHODS This study retrospectively included 172 patients with EOC preoperatively underwent dual-energy CT scans. Patients were grouped as type I and type II EOC according to postoperatively pathologic results. Normalized IC (NIC, %) values from arterial-phase (AP), venous-phase (VP) and delay-phase (DP) were measured by two observers. ECV fraction (%) was calculated by DP-NIC and hematocrit. Intra-observer correlation coefficient (ICC) was used to assess the agreement between measurements made by two observers. The differences of imaging parameters between the two groups were compared. Logistic regression was used to select independent predictive factors and establish combined parameter. Receiver operating characteristic curve was used to analyze performance of all parameters. RESULTS The ICCs for all parameters exceeded 0.75. All parameters in type II EOC were all significantly higher than those in type I EOC (all P < 0.05). VP-NIC exhibited the highest Area under the curve (AUC) of 0.804, along with 80.39% sensitivity and 71.43% specificity. VP-NIC was identified as the independent factor. The sensitivity and specificity of ECV fraction were 78.43% and 71.43%, respectively. The combined parameter consisting of AP-NIC, VP-NIC, DP-NIC, and ECV fraction yielded an AUC of 0.823, with sensitivity of 76.47% and specificity of 77.14%. The sensitivity of the combined parameter was significantly higher than that of AP-NIC (P = 0.049). CONCLUSION It is valuable for dual-energy CT IC-based parameters and ECV fraction in preoperatively identifying type I and type II EOC. CRITICAL RELEVANCE STATEMENT Dual-energy CT-normalized iodine concentration and extracellular volume fraction achieved satisfactory discriminative efficacy, distinguishing between type I and type II epithelial ovarian carcinoma.
Collapse
Affiliation(s)
- Qingling Song
- Department of Radiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ye Li
- Department of Radiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tingfan Wu
- United Imaging Research Institute of Innovative Medical Equipment, Shenzhen, China
| | - Wenjun Hu
- Department of Radiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yijun Liu
- Department of Radiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ailian Liu
- Department of Radiology, First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
5
|
Valerie NCK, Sanjiv K, Mortusewicz O, Zhang SM, Alam S, Pires MJ, Stigsdotter H, Rasti A, Langelier MF, Rehling D, Throup A, Purewal-Sidhu O, Desroses M, Onireti J, Wakchaure P, Almlöf I, Boström J, Bevc L, Benzi G, Stenmark P, Pascal JM, Helleday T, Page BDG, Altun M. Coupling cellular drug-target engagement to downstream pharmacology with CeTEAM. Nat Commun 2024; 15:10347. [PMID: 39643609 PMCID: PMC11624193 DOI: 10.1038/s41467-024-54415-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/11/2024] [Indexed: 12/09/2024] Open
Abstract
Cellular target engagement technologies enable quantification of intracellular drug binding; however, simultaneous assessment of drug-associated phenotypes has proven challenging. Here, we present cellular target engagement by accumulation of mutant as a platform that can concomitantly evaluate drug-target interactions and phenotypic responses using conditionally stabilized drug biosensors. We observe that drug-responsive proteotypes are prevalent among reported mutants of known drug targets. Compatible mutants appear to follow structural and biophysical logic that permits intra-protein and paralogous expansion of the biosensor pool. We then apply our method to uncouple target engagement from divergent cellular activities of MutT homolog 1 (MTH1) inhibitors, dissect Nudix hydrolase 15 (NUDT15)-associated thiopurine metabolism with the R139C pharmacogenetic variant, and profile the dynamics of poly(ADP-ribose) polymerase 1/2 (PARP1/2) binding and DNA trapping by PARP inhibitors (PARPi). Further, PARP1-derived biosensors facilitated high-throughput screening for PARP1 binders, as well as multimodal ex vivo analysis and non-invasive tracking of PARPi binding in live animals. This approach can facilitate holistic assessment of drug-target engagement by bridging drug binding events and their biological consequences.
Collapse
Affiliation(s)
- Nicholas C K Valerie
- Science for Life Laboratory, Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, SE-141 52, Sweden.
| | - Kumar Sanjiv
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Oliver Mortusewicz
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Si Min Zhang
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Seher Alam
- Science for Life Laboratory, Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, SE-141 52, Sweden
| | - Maria J Pires
- Science for Life Laboratory, Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, SE-141 52, Sweden
| | - Hannah Stigsdotter
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Azita Rasti
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Marie-France Langelier
- Département de Biochimie and Médecine Moléculaire, Faculté de Médecine, Université de Montréal, Montréal, QC, H3C 3J7, Canada
| | - Daniel Rehling
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, SE-106 91, Sweden
| | - Adam Throup
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Oryn Purewal-Sidhu
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Matthieu Desroses
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Jacob Onireti
- Science for Life Laboratory, Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, SE-141 52, Sweden
| | - Prasad Wakchaure
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Ingrid Almlöf
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Johan Boström
- Science for Life Laboratory, Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, SE-141 52, Sweden
| | - Luka Bevc
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Giorgia Benzi
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Pål Stenmark
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, SE-106 91, Sweden
| | - John M Pascal
- Département de Biochimie and Médecine Moléculaire, Faculté de Médecine, Université de Montréal, Montréal, QC, H3C 3J7, Canada
| | - Thomas Helleday
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
| | - Brent D G Page
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna, SE-171 65, Sweden
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Mikael Altun
- Science for Life Laboratory, Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, SE-141 52, Sweden
| |
Collapse
|
6
|
V U P, T I M, K K M. An integrative analysis to identify pancancer epigenetic biomarkers. Comput Biol Chem 2024; 113:108260. [PMID: 39467487 DOI: 10.1016/j.compbiolchem.2024.108260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/13/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024]
Abstract
Integrating and analyzing the pancancer data collected from different experiments is crucial for gaining insights into the common mechanisms in the molecular level underlying the development and progression of cancers. Epigenetic study of the pancancer data can provide promising results in biomarker discovery. The genes that are epigenetically dysregulated in different cancers are powerful biomarkers for drug-related studies. This paper identifies the genes having altered expression due to aberrant methylation patterns using differential analysis of TCGA pancancer data of 12 different cancers. We identified a comprehensive set of 115 epigenetic biomarker genes out of which 106 genes having pancancer properties. The correlation analysis, gene set enrichment, protein-protein interaction analysis, pancancer characteristics analysis, and diagnostic modeling were performed on these biomarkers to illustrate the power of this signature and found to be important in different molecular operations related to cancer. An accuracy of 97.56% was obtained on TCGA pancancer gene expression dataset for predicting the binary class tumor or normal. The source code and dataset of this work are available at https://github.com/panchamisuneeth/EpiPanCan.git.
Collapse
Affiliation(s)
- Panchami V U
- Adi Shankara Institute of Engineering and Technology, Ernakulam, 683574, Kerala, India; Government Engineering College Thrissur, 680009, Kerala, India; APJ Abdul Kalam Technological University, 695016, Kerala, India.
| | - Manish T I
- SCMS School of Engineering and Technology, Ernakulam, 683576, Kerala, India; APJ Abdul Kalam Technological University, 695016, Kerala, India
| | - Manesh K K
- Government Engineering College Thrissur, 680009, Kerala, India; APJ Abdul Kalam Technological University, 695016, Kerala, India
| |
Collapse
|
7
|
Wagner KD, Safwan-Zaiter H, Wagner N. A Dual Role of the Senescence Marker P16Ink4a in Liver Endothelial Cell Function. Cells 2024; 13:1929. [PMID: 39682678 PMCID: PMC11640762 DOI: 10.3390/cells13231929] [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: 10/23/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
P16Ink4a is a well-established marker of senescence. Although P16Ink4a is expressed in endothelial cells, little is known about its function in these cells. Using isolated liver endothelial cells with silencing or overexpression of P16Ink4a, we show here that dependent on P16Ink4a levels, different pathways and functions are affected. High levels of P16Ink4a reduce proliferation and induce senescence, while low levels have the opposite effects. Only high P16Ink4a expression reduces in vitro angiogenesis. Expression profiling reveals an inflammatory phenotype upon silencing of P16Ink4a, while P16Ink4a overexpression is associated with a profile associated with DNA damage, repair and senescence. Low levels of P16Ink4a induce reactive oxygen species (ROS) generation and increase endothelial cell leakage. Collectively, P16Ink4a represents an "antagonistic pleiotropy" gene, which is, on the one hand, required to prevent ROS generation and endothelial damage and, on the other hand, inhibits angiogenesis through induction of senescence at high levels.
Collapse
Affiliation(s)
| | | | - Nicole Wagner
- CNRS, INSERM, iBV, Université Côte d’Azur, 06107 Nice, France;
| |
Collapse
|
8
|
Lorusso B, Nogara A, Fioretzaki R, Corradini E, Bove R, Roti G, Gherli A, Montanaro A, Monica G, Cavazzini F, Bonomini S, Graiani G, Silini EM, Gnetti L, Pilato FP, Cerasoli G, Quaini F, Lagrasta CAM. CD26 Is Differentially Expressed throughout the Life Cycle of Infantile Hemangiomas and Characterizes the Proliferative Phase. Int J Mol Sci 2024; 25:9760. [PMID: 39337249 PMCID: PMC11432178 DOI: 10.3390/ijms25189760] [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: 07/09/2024] [Revised: 08/30/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
Infantile hemangiomas (IHs) are benign vascular neoplasms of childhood (prevalence 5-10%) due to the abnormal proliferation of endothelial cells. IHs are characterized by a peculiar natural life cycle enclosing three phases: proliferative (≤12 months), involuting (≥13 months), and involuted (up to 4-7 years). The mechanisms underlying this neoplastic disease still remain uncovered. Twenty-seven IH tissue specimens (15 proliferative and 12 involuting) were subjected to hematoxylin and eosin staining and a panel of diagnostic markers by immunohistochemistry. WT1, nestin, CD133, and CD26 were also analyzed. Moreover, CD31pos/CD26pos proliferative hemangioma-derived endothelial cells (Hem-ECs) were freshly isolated, exposed to vildagliptin (a DPP-IV/CD26 inhibitor), and tested for cell survival and proliferation by MTT assay, FACS analysis, and Western blot assay. All IHs displayed positive CD31, GLUT1, WT1, and nestin immunostaining but were negative for D2-40. Increased endothelial cell proliferation in IH samples was documented by ki67 labeling. All endothelia of proliferative IHs were positive for CD26 (100%), while only 10 expressed CD133 (66.6%). Surprisingly, seven involuting IH samples (58.3%) exhibited coexisting proliferative and involuting aspects in the same hemangiomatous lesion. Importantly, proliferative areas were characterized by CD26 immunolabeling, at variance from involuting sites that were always CD26 negative. Finally, in vitro DPP-IV pharmacological inhibition by vildagliptin significantly reduced Hem-ECs proliferation through the modulation of ki67 and induced cell cycle arrest associated with the upregulation of p21 protein expression. Taken together, our findings suggest that CD26 might represent a reliable biomarker to detect proliferative sites and unveil non-regressive IHs after a 12-month life cycle.
Collapse
Affiliation(s)
- Bruno Lorusso
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (B.L.); (A.N.); (R.F.); (E.C.); (R.B.); (G.R.); (A.G.); (A.M.); (G.M.); (F.C.); (F.Q.)
| | - Antonella Nogara
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (B.L.); (A.N.); (R.F.); (E.C.); (R.B.); (G.R.); (A.G.); (A.M.); (G.M.); (F.C.); (F.Q.)
| | - Rodanthi Fioretzaki
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (B.L.); (A.N.); (R.F.); (E.C.); (R.B.); (G.R.); (A.G.); (A.M.); (G.M.); (F.C.); (F.Q.)
- Department of Medical Oncology, Metaxa Cancer Hospital of Piraeus, 185 37 Piraeus, Greece
| | - Emilia Corradini
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (B.L.); (A.N.); (R.F.); (E.C.); (R.B.); (G.R.); (A.G.); (A.M.); (G.M.); (F.C.); (F.Q.)
| | - Roberta Bove
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (B.L.); (A.N.); (R.F.); (E.C.); (R.B.); (G.R.); (A.G.); (A.M.); (G.M.); (F.C.); (F.Q.)
| | - Giovanni Roti
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (B.L.); (A.N.); (R.F.); (E.C.); (R.B.); (G.R.); (A.G.); (A.M.); (G.M.); (F.C.); (F.Q.)
- Translational Hematology and Chemogenomics (THEC), University of Parma, 43126 Parma, Italy
- Hematology and BMT Unit, University Hospital of Parma, 43126 Parma, Italy;
| | - Andrea Gherli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (B.L.); (A.N.); (R.F.); (E.C.); (R.B.); (G.R.); (A.G.); (A.M.); (G.M.); (F.C.); (F.Q.)
- Translational Hematology and Chemogenomics (THEC), University of Parma, 43126 Parma, Italy
| | - Anna Montanaro
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (B.L.); (A.N.); (R.F.); (E.C.); (R.B.); (G.R.); (A.G.); (A.M.); (G.M.); (F.C.); (F.Q.)
- Translational Hematology and Chemogenomics (THEC), University of Parma, 43126 Parma, Italy
| | - Gregorio Monica
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (B.L.); (A.N.); (R.F.); (E.C.); (R.B.); (G.R.); (A.G.); (A.M.); (G.M.); (F.C.); (F.Q.)
- Translational Hematology and Chemogenomics (THEC), University of Parma, 43126 Parma, Italy
| | - Filippo Cavazzini
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (B.L.); (A.N.); (R.F.); (E.C.); (R.B.); (G.R.); (A.G.); (A.M.); (G.M.); (F.C.); (F.Q.)
- Translational Hematology and Chemogenomics (THEC), University of Parma, 43126 Parma, Italy
| | - Sabrina Bonomini
- Hematology and BMT Unit, University Hospital of Parma, 43126 Parma, Italy;
| | - Gallia Graiani
- Center of Dental Medicine, University of Parma, 43126 Parma, Italy;
| | - Enrico Maria Silini
- Pathology Section, University Hospital of Parma, 43126 Parma, Italy; (E.M.S.); (L.G.); (F.P.P.)
| | - Letizia Gnetti
- Pathology Section, University Hospital of Parma, 43126 Parma, Italy; (E.M.S.); (L.G.); (F.P.P.)
| | - Francesco Paolo Pilato
- Pathology Section, University Hospital of Parma, 43126 Parma, Italy; (E.M.S.); (L.G.); (F.P.P.)
| | - Giuseppe Cerasoli
- Pediatric Surgery, Ospedale dei Bambini of Parma, University Hospital of Parma, 43126 Parma, Italy;
| | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (B.L.); (A.N.); (R.F.); (E.C.); (R.B.); (G.R.); (A.G.); (A.M.); (G.M.); (F.C.); (F.Q.)
| | - Costanza Anna Maria Lagrasta
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (B.L.); (A.N.); (R.F.); (E.C.); (R.B.); (G.R.); (A.G.); (A.M.); (G.M.); (F.C.); (F.Q.)
| |
Collapse
|
9
|
Gugulothu KN, Anvesh Sai P, Suraparaju S, Karuturi SP, Pendli G, Kamma RB, Nimmagadda K, Modepalli A, Mamilla M, Vashist S. WT1 Cancer Vaccine in Advanced Pancreatic Cancer: A Systematic Review. Cureus 2024; 16:e56934. [PMID: 38665761 PMCID: PMC11043900 DOI: 10.7759/cureus.56934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Advanced pancreatic cancer is one of the prominent contributors to cancer-related mortality globally. Chemotherapy, especially gemcitabine, is generally used for the treatment of advanced pancreatic cancer. Despite the treatment, the fatality rate for advanced pancreatic cancer is alarmingly high. Thus, the dire need for better treatment alternatives has drawn focus to cancer vaccinations. The Wilms tumor gene (WT1), typically associated with Wilms tumor, is found to be excessively expressed in some cancers, such as pancreatic cancer. This characteristic feature is harvested to develop cancer vaccines against WT1. This review aims to systematically summarize the clinical trials investigating the efficacy and safety of WT1 vaccines in patients with advanced pancreatic cancer. An extensive literature search was conducted on databases Medline, Web of Science, ScienceDirect, and Google Scholar using the keywords "Advanced pancreatic cancer," "Cancer vaccines," "WT1 vaccines," and "Pulsed DC vaccines," and the results were exclusively studied to construct this review. WT1 vaccines work by introducing peptides from the WT1 protein to trigger an immune response involving cytotoxic T lymphocytes via antigen-presenting cells. Upon activation, these lymphocytes induce apoptosis in cancer cells by specifically targeting those with increased WT1 levels. WT1 vaccinations, which are usually given in addition to chemotherapy, have demonstrated clinically positive results and minimal side effects. However, there are several challenges to their widespread use, such as the immunosuppressive nature of tumors and heterogeneity in expression. Despite these limitations, the risk-benefit profile of cancer vaccines is encouraging, especially for the WT1 vaccine in the treatment of advanced pancreatic cancer. Considering the fledgling status of their development, large multicentric, variables-matched, extensive analysis across diverse demographics is considered essential.
Collapse
Affiliation(s)
| | | | - Sonika Suraparaju
- Internal Medicine, Sri Padmavathi Medical College for Women, Tirupati, IND
| | | | - Ganesh Pendli
- Internal Medicine, PES Institute of Medical Sciences and Research, Kuppam, IND
| | - Ravi Babu Kamma
- Internal Medicine, Sri Venkata Sai (SVS) Medical College, Mahabubnagar, IND
| | | | - Alekhya Modepalli
- Internal Medicine, Sri Padmavathi Medical College for Women, Tirupati, IND
| | - Mahesh Mamilla
- Internal Medicine, Sri Venkateswara Medical College, Tirupati, IND
| | | |
Collapse
|
10
|
Lee JU, Kim SH, Lee SH, Ji MJ, Jin JA, So HJ, Song ML, Lee HK, Kang TW. Combinational Pulsing of TAAs Enforces Dendritic Cell-Based Immunotherapy through T-Cell Proliferation and Interferon-γ Secretion in LLC1 Mouse Model. Cancers (Basel) 2024; 16:409. [PMID: 38254898 PMCID: PMC10814594 DOI: 10.3390/cancers16020409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
NSCLC, the most common type of lung cancer, is often diagnosed late due to minimal early symptoms. Its high risk of recurrence or metastasis post-chemotherapy makes DC-based immunotherapy a promising strategy, offering targeted cancer destruction, low side effects, memory formation, and overcoming the immune evasive ability of cancers. However, the limited response to DCs pulsed with single antigens remains a significant challenge. To overcome this, we enhanced DC antigen presentation by pulsing with TAAs. Our study focused on enhancing DC-mediated immune response specificity and intensity by combinatorial pulsing of TAAs, selected for their prevalence in NSCLC. We selected four types of TAAs expressed in NSCLC and pulsed DCs with the optimal combination. Next, we administered TAAs-pulsed DCs into the LLC1 mouse model to evaluate their anti-tumor efficacy. Our results showed that TAAs-pulsed DCs significantly reduced tumor size and promoted apoptosis in tumor tissue. Moreover, TAAs-pulsed DCs significantly increased total T cells in the spleen compared to the unpulsed DCs. Additionally, in vitro stimulation of splenocytes from the TAAs-pulsed DCs showed notable T-cell proliferation and increased IFN-γ secretion. Our findings demonstrate the potential of multiple TAA pulsing to enhance the antigen-presenting capacity of DCs, thereby strengthening the immune response against tumors.
Collapse
Affiliation(s)
- Jae-Ung Lee
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| | - Sang-Heon Kim
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| | - Sung-Hoon Lee
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| | - Min-Jae Ji
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| | - Jeong-Ah Jin
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| | - Hyung-Joon So
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| | | | - Hong-Ki Lee
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
- EHLCell Clinic, Seoul 06029, Republic of Korea
| | - Tae-Wook Kang
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| |
Collapse
|
11
|
Morales AE, Gumenick R, Genovese CM, Jang YY, Ouedraogo A, Ibáñez de Garayo M, Pannellini T, Patel S, Bott ME, Alvarez J, Mun SS, Totonchy J, Gautam A, Delgado de la Mora J, Chang S, Wirth D, Horenstein M, Dao T, Scheinberg DA, Rubinstein PG, Semeere A, Martin J, Godfrey CC, Moser CB, Matining RM, Campbell TB, Borok MZ, Krown SE, Cesarman E. Wilms' tumor 1 (WT1) antigen is overexpressed in Kaposi Sarcoma and is regulated by KSHV vFLIP. PLoS Pathog 2024; 20:e1011881. [PMID: 38190392 PMCID: PMC10898863 DOI: 10.1371/journal.ppat.1011881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 02/27/2024] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
In people living with HIV, Kaposi Sarcoma (KS), a vascular neoplasm caused by KS herpesvirus (KSHV/HHV-8), remains one of the most common malignancies worldwide. Individuals living with HIV, receiving otherwise effective antiretroviral therapy, may present with extensive disease requiring chemotherapy. Hence, new therapeutic approaches are needed. The Wilms' tumor 1 (WT1) protein is overexpressed and associated with poor prognosis in several hematologic and solid malignancies and has shown promise as an immunotherapeutic target. We found that WT1 was overexpressed in >90% of a total 333 KS biopsies, as determined by immunohistochemistry and image analysis. Our largest cohort from ACTG, consisting of 294 cases was further analyzed demonstrating higher WT1 expression was associated with more advanced histopathologic subtypes. There was a positive correlation between the proportion of infected cells within KS tissues, assessed by expression of the KSHV-encoded latency-associated nuclear antigen (LANA), and WT1 positivity. Areas with high WT1 expression showed sparse T-cell infiltrates, consistent with an immune evasive tumor microenvironment. We show that major oncogenic isoforms of WT1 are overexpressed in primary KS tissue and observed WT1 upregulation upon de novo infection of endothelial cells with KSHV. KSHV latent viral FLICE-inhibitory protein (vFLIP) upregulated total and major isoforms of WT1, but upregulation was not seen after expression of mutant vFLIP that is unable to bind IKKƴ and induce NFκB. siRNA targeting of WT1 in latent KSHV infection resulted in decreased total cell number and pAKT, BCL2 and LANA protein expression. Finally, we show that ESK-1, a T cell receptor-like monoclonal antibody that recognizes WT1 peptides presented on MHC HLA-A0201, demonstrates increased binding to endothelial cells after KSHV infection or induction of vFLIP expression. We propose that oncogenic isoforms of WT1 are upregulated by KSHV to promote tumorigenesis and immunotherapy directed against WT1 may be an approach for KS treatment.
Collapse
Affiliation(s)
- Ayana E. Morales
- Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Ruby Gumenick
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Caitlyn M. Genovese
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Yun Yeong Jang
- Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Ariene Ouedraogo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Maite Ibáñez de Garayo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Tania Pannellini
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Sanjay Patel
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Matthew E. Bott
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Julio Alvarez
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Sung Soo Mun
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Jennifer Totonchy
- School of Pharmacy, Chapman University, Irvine, California, United States of America
| | - Archana Gautam
- Department of Allergy and Immunology, Icahn School of Medicine, New York, New York, United States of America
| | - Jesus Delgado de la Mora
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Stephanie Chang
- Cornell University, Ithaca, New York, United States of America
| | - Dagmar Wirth
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research Braunschweig, Germany
| | - Marcelo Horenstein
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Tao Dao
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - David A. Scheinberg
- Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Paul G. Rubinstein
- Section of Hematology/Oncology, John H. Stroger Jr Hospital of Cook County (Cook County Hospital), Ruth M. Rothstein Core Center, University of Illinois, Chicago, Illinois, United States of America
| | - Aggrey Semeere
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Jeffrey Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
| | - Catherine C. Godfrey
- Office of the Global AIDS Coordinator, Department of State, Washington, DC, United States of America
| | - Carlee B. Moser
- Center for Biostatistics in AIDS Research, Harvard T H Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Roy M. Matining
- Center for Biostatistics in AIDS Research, Harvard T H Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Thomas B. Campbell
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Margaret Z. Borok
- Department of Internal Medicine, University of Zimbabwe, Harare, Zimbabwe
| | - Susan E. Krown
- Memorial Sloan Kettering Cancer Center (emerita), New York, New York, United States of America
| | - Ethel Cesarman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, United States of America
| |
Collapse
|
12
|
Guo PC, Zuo J, Huang KK, Lai GY, Zhang X, An J, Li JX, Li L, Wu L, Lin YT, Wang DY, Xu JS, Hao SJ, Wang Y, Li RH, Ma W, Song YM, Liu C, Liu CY, Dai Z, Xu Y, Sharma AD, Ott M, Ou-Yang Q, Huo F, Fan R, Li YY, Hou JL, Volpe G, Liu LQ, Esteban MA, Lai YW. Cell atlas of CCl 4-induced progressive liver fibrosis reveals stage-specific responses. Zool Res 2023; 44:451-466. [PMID: 36994536 PMCID: PMC10236302 DOI: 10.24272/j.issn.2095-8137.2023.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/11/2023] [Indexed: 03/12/2023] Open
Abstract
Chronic liver injury leads to progressive liver fibrosis and ultimately cirrhosis, a major cause of morbidity and mortality worldwide. However, there are currently no effective anti-fibrotic therapies available, especially for late-stage patients, which is partly attributed to the major knowledge gap regarding liver cell heterogeneity and cell-specific responses in different fibrosis stages. To reveal the multicellular networks regulating mammalian liver fibrosis from mild to severe phenotypes, we generated a single-nucleus transcriptomic atlas encompassing 49 919 nuclei corresponding to all main liver cell types at different stages of murine carbon tetrachloride (CCl 4)-induced progressive liver fibrosis. Integrative analysis distinguished the sequential responses to injury of hepatocytes, hepatic stellate cells and endothelial cells. Moreover, we reconstructed cell-cell interactions and gene regulatory networks implicated in these processes. These integrative analyses uncovered previously overlooked aspects of hepatocyte proliferation exhaustion and disrupted pericentral metabolic functions, dysfunction for clearance by apoptosis of activated hepatic stellate cells, accumulation of pro-fibrotic signals, and the switch from an anti-angiogenic to a pro-angiogenic program during CCl 4-induced progressive liver fibrosis. Our dataset thus constitutes a useful resource for understanding the molecular basis of progressive liver fibrosis using a relevant animal model.
Collapse
Affiliation(s)
- Peng-Cheng Guo
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
| | - Jing Zuo
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
| | - Ke-Ke Huang
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510799, China
| | - Guang-Yao Lai
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health and Guangzhou Medical University, Guangzhou, Guangdong 510530, China
| | - Xiao Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
| | - Juan An
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jin-Xiu Li
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Li
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Liang Wu
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Yi-Ting Lin
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Dong-Ye Wang
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Jiang-Shan Xu
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
| | - Shi-Jie Hao
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Wang
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
| | - Rong-Hai Li
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
| | - Wen Ma
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
| | - Yu-Mo Song
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
| | - Chang Liu
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
| | - Chuan-Yu Liu
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
| | - Zhen Dai
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Yan Xu
- Biotherapy Centre, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Amar Deep Sharma
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover 30625, Germany
| | - Michael Ott
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover 30625, Germany
| | - Qing Ou-Yang
- Department of Hepatobiliary Surgery and Liver Transplant Center, General Hospital of Southern Theater Command, Guangzhou, Guangdong 510010, China
| | - Feng Huo
- Department of Hepatobiliary Surgery and Liver Transplant Center, General Hospital of Southern Theater Command, Guangzhou, Guangdong 510010, China
| | - Rong Fan
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, Guangdong 510515, China
| | - Yong-Yin Li
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, Guangdong 510515, China
| | - Jin-Lin Hou
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, Guangdong 510515, China
| | - Giacomo Volpe
- Hematology and Cell Therapy Unit, IRCCS-Istituto Tumori 'Giovanni Paolo II', Bari 70124, Italy
| | - Long-Qi Liu
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
| | - Miguel A Esteban
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510799, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health and Guangzhou Medical University, Guangzhou, Guangdong 510530, China
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany. E-mail:
| | - Yi-Wei Lai
- BGI-Hangzhou, Hangzhou, Zhejiang 310012, China
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China. E-mail:
| |
Collapse
|
13
|
Low JJW, Sulaiman SA, Johdi NA, Abu N. Immunomodulatory effects of extracellular vesicles in glioblastoma. Front Cell Dev Biol 2022; 10:996805. [PMID: 36467419 PMCID: PMC9708723 DOI: 10.3389/fcell.2022.996805] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2024] Open
Abstract
Glioblastoma (GB) is a type of brain cancer that can be considered aggressive. Glioblastoma treatment has significant challenges due to the immune privilege site of the brain and the presentation of an immunosuppressive tumor microenvironment. Extracellular vesicles (EVs) are cell-secreted nanosized vesicles that engage in intercellular communication via delivery of cargo that may cause downstream effects such as tumor progression and recipient cell modulation. Although the roles of extracellular vesicles in cancer progression are well documented, their immunomodulatory effects are less defined. Herein, we focus on glioblastoma and explain the immunomodulatory effects of extracellular vesicles secreted by both tumor and immune cells in detail. The tumor to immune cells, immune cells to the tumor, and intra-immune cells extracellular vesicles crosstalks are involved in various immunomodulatory effects. This includes the promotion of immunosuppressive phenotypes, apoptosis, and inactivation of immune cell subtypes, which affects the central nervous system and peripheral immune system response, aiding in its survival and progression in the brain.
Collapse
Affiliation(s)
| | | | | | - Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), UKM Medical Centre, Universiti Kebangsaan Malaysia, Jalan Yaa’cob Latiff, Bandar Tun Razak, Kuala Lumpur, Malaysia
| |
Collapse
|
14
|
Safwan-Zaiter H, Wagner N, Wagner KD. P16INK4A-More Than a Senescence Marker. Life (Basel) 2022; 12:1332. [PMID: 36143369 PMCID: PMC9501954 DOI: 10.3390/life12091332] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Aging is a biological feature that is characterized by gradual degeneration of function in cells, tissues, organs, or an intact organism due to the accumulation of environmental factors and stresses with time. Several factors have been attributed to aging such as oxidative stress and augmented production or exposure to reactive oxygen species, inflammatory cytokines production, telomere shortening, DNA damage, and, importantly, the deposit of senescent cells. These are irreversibly mitotically inactive, yet metabolically active cells. The reason underlying their senescence lies within the extrinsic and the intrinsic arms. The extrinsic arm is mainly characterized by the expression and the secretory profile known as the senescence-associated secretory phenotype (SASP). The intrinsic arm results from the impact of several genes meant to regulate the cell cycle, such as tumor suppressor genes. P16INK4A is a tumor suppressor and cell cycle regulator that has been linked to aging and senescence. Extensive research has revealed that p16 expression is significantly increased in senescent cells, as well as during natural aging or age-related pathologies. Based on this fact, p16 is considered as a specific biomarker for detecting senescent cells and aging. Other studies have found that p16 is not only a senescence marker, but also a protein with many functions outside of senescence and aging. In this paper, we discuss and shed light on several studies that show the different functions of p16 and provide insights in its role in several biological processes besides senescence and aging.
Collapse
Affiliation(s)
| | - Nicole Wagner
- CNRS, INSERM, iBV, Université Côte d’Azur, 06107 Nice, France
| | | |
Collapse
|
15
|
Wagner N, Wagner KD. Peroxisome Proliferator-Activated Receptors and the Hallmarks of Cancer. Cells 2022; 11:cells11152432. [PMID: 35954274 PMCID: PMC9368267 DOI: 10.3390/cells11152432] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/11/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) function as nuclear transcription factors upon the binding of physiological or pharmacological ligands and heterodimerization with retinoic X receptors. Physiological ligands include fatty acids and fatty-acid-derived compounds with low specificity for the different PPAR subtypes (alpha, beta/delta, and gamma). For each of the PPAR subtypes, specific pharmacological agonists and antagonists, as well as pan-agonists, are available. In agreement with their natural ligands, PPARs are mainly focused on as targets for the treatment of metabolic syndrome and its associated complications. Nevertheless, many publications are available that implicate PPARs in malignancies. In several instances, they are controversial for very similar models. Thus, to better predict the potential use of PPAR modulators for personalized medicine in therapies against malignancies, it seems necessary and timely to review the three PPARs in relation to the didactic concept of cancer hallmark capabilities. We previously described the functions of PPAR beta/delta with respect to the cancer hallmarks and reviewed the implications of all PPARs in angiogenesis. Thus, the current review updates our knowledge on PPAR beta and the hallmarks of cancer and extends the concept to PPAR alpha and PPAR gamma.
Collapse
Affiliation(s)
- Nicole Wagner
- Correspondence: (N.W.); (K.-D.W.); Tel.: +33-489-153-713 (K.-D.W.)
| | | |
Collapse
|
16
|
Wagner KD, Wagner N. The Senescence Markers p16INK4A, p14ARF/p19ARF, and p21 in Organ Development and Homeostasis. Cells 2022; 11:cells11121966. [PMID: 35741095 PMCID: PMC9221567 DOI: 10.3390/cells11121966] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023] Open
Abstract
It is widely accepted that senescent cells accumulate with aging. They are characterized by replicative arrest and the release of a myriad of factors commonly called the senescence-associated secretory phenotype. Despite the replicative cell cycle arrest, these cells are metabolically active and functional. The release of SASP factors is mostly thought to cause tissue dysfunction and to induce senescence in surrounding cells. As major markers for aging and senescence, p16INK4, p14ARF/p19ARF, and p21 are established. Importantly, senescence is also implicated in development, cancer, and tissue homeostasis. While many markers of senescence have been identified, none are able to unambiguously identify all senescent cells. However, increased levels of the cyclin-dependent kinase inhibitors p16INK4A and p21 are often used to identify cells with senescence-associated phenotypes. We review here the knowledge of senescence, p16INK4A, p14ARF/p19ARF, and p21 in embryonic and postnatal development and potential functions in pathophysiology and homeostasis. The establishment of senolytic therapies with the ultimate goal to improve healthy aging requires care and detailed knowledge about the involvement of senescence and senescence-associated proteins in developmental processes and homeostatic mechanism. The review contributes to these topics, summarizes open questions, and provides some directions for future research.
Collapse
|
17
|
van Amerongen RA, Hagedoorn RS, Remst DFG, Assendelft DC, van der Steen DM, Wouters AK, van de Meent M, Kester MGD, de Ru AH, Griffioen M, van Veelen PA, Falkenburg JHF, Heemskerk MHM. WT1-specific TCRs directed against newly identified peptides install antitumor reactivity against acute myeloid leukemia and ovarian carcinoma. J Immunother Cancer 2022; 10:jitc-2021-004409. [PMID: 35728869 PMCID: PMC9214430 DOI: 10.1136/jitc-2021-004409] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2022] [Indexed: 12/24/2022] Open
Abstract
Background Transcription factor Wilms’ tumor gene 1 (WT1) is an ideal tumor target based on its expression in a wide range of tumors, low-level expression in normal tissues and promoting role in cancer progression. In clinical trials, WT1 is targeted using peptide-based or dendritic cell-based vaccines and T-cell receptor (TCR)-based therapies. Antitumor reactivities were reported, but T-cell reactivity is hampered by self-tolerance to WT1 and limited number of WT1 peptides, which were thus far selected based on HLA peptide binding algorithms. Methods In this study, we have overcome both limitations by searching in the allogeneic T-cell repertoire of healthy donors for high-avidity WT1-specific T cells, specific for WT1 peptides derived from the HLA class I associated ligandome of primary leukemia and ovarian carcinoma samples. Results Using broad panels of malignant cells and healthy cell subsets, T-cell clones were selected that demonstrated potent and specific anti-WT1 T-cell reactivity against five of the eight newly identified WT1 peptides. Notably, T-cell clones for WT1 peptides previously used in clinical trials lacked reactivity against tumor cells, suggesting limited processing and presentation of these peptides. The TCR sequences of four T-cell clones were analyzed and TCR gene transfer into CD8+ T cells installed antitumor reactivity against WT1-expressing solid tumor cell lines, primary acute myeloid leukemia (AML) blasts, and ovarian carcinoma patient samples. Conclusions Our approach resulted in a set of naturally expressed WT1 peptides and four TCRs that are promising candidates for TCR gene transfer strategies in patients with WT1-expressing tumors, including AML and ovarian carcinoma.
Collapse
Affiliation(s)
- Rosa A van Amerongen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Renate S Hagedoorn
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dennis F G Remst
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Danique C Assendelft
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk M van der Steen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne K Wouters
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marian van de Meent
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michel G D Kester
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arnoud H de Ru
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Mirjam H M Heemskerk
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
18
|
Evaluating Established Roles, Future Perspectives and Methodological Heterogeneity for Wilms’ Tumor 1 (WT1) Antigen Detection in Adult Renal Cell Carcinoma, Using a Novel N-Terminus Targeted Antibody (Clone WT49). Biomedicines 2022; 10:biomedicines10040912. [PMID: 35453662 PMCID: PMC9026801 DOI: 10.3390/biomedicines10040912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/23/2022] [Accepted: 04/11/2022] [Indexed: 11/20/2022] Open
Abstract
Renal cell carcinoma (RCC) is arguably the deadliest form of genitourinary malignancy and is nowadays viewed as a heterogeneous series of cancers, with the same origin but fundamentally different metabolisms and clinical behaviors. Immunohistochemistry (IHC) is increasingly necessary for RCC subtyping and definitive diagnosis. WT1 is a complex gene involved in carcinogenesis. To address reporting heterogeneity and WT1 IHC standardization, we used a recent N-terminus targeted monoclonal antibody (clone WT49) to evaluate WT1 protein expression in 56 adult RCC (aRCC) cases. This is the largest WT1 IHC investigation focusing exclusively on aRCCs and the first report on clone WT49 staining in aRCCs. We found seven (12.5%) positive cases, all clear cell RCCs, showing exclusively nuclear staining for WT1. We did not disregard cytoplasmic staining in any of the negative cases. Extratumoral fibroblasts, connecting tubules and intratumoral endothelial cells showed the same exclusively nuclear WT1 staining pattern. We reviewed WT1 expression patterns in aRCCs and the possible explanatory underlying metabolomics. For now, WT1 protein expression in aRCCs is insufficiently investigated, with significant discrepancies in the little data reported. Emerging WT1-targeted RCC immunotherapy will require adequate case selection and sustained efforts to standardize the quantification of tumor-associated antigens for aRCC and its many subtypes.
Collapse
|
19
|
Ueda Y, Usuki K, Fujita J, Matsumura I, Aotsuka N, Sekiguchi N, Nakazato T, Iwasaki H, Takahara‐Matsubara M, Sugimoto S, Goto M, Naoe T, Kizaki M, Miyazaki Y, Aakashi K. Phase 1/2 study evaluating the safety and efficacy of DSP-7888 dosing emulsion in myelodysplastic syndromes. Cancer Sci 2022; 113:1377-1392. [PMID: 34932235 PMCID: PMC8990724 DOI: 10.1111/cas.15245] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/11/2021] [Accepted: 12/06/2021] [Indexed: 11/27/2022] Open
Abstract
DSP-7888 is an immunotherapeutic cancer vaccine derived from the Wilms' tumor gene 1 (WT1) protein. This phase 1/2 open-label study evaluated the safety and efficacy of DSP-7888 dosing emulsion in patients with myelodysplastic syndromes (MDS). DSP-7888 was administered intradermally (3.5 or 10.5 mg) every 2 weeks for 6 months and then every 2-4 weeks until lack of benefit. Twelve patients were treated in phase 1 (3.5 mg, n = 6; 10.5 mg, n = 6), with no dose-limiting toxicities reported. Thus, the 10.5 mg dose was selected as the recommended phase 2 dose, and 35 patients were treated in phase 2. Forty-seven patients received ≥1 dose of the study drug and comprised the safety analysis set. The most common adverse drug reaction (ADR) was injection site reactions (ISR; 91.5%). Grade 3 ISR were common (58.8%) in phase 1 but occurred less frequently in 2 (22.9%) following implementation of risk minimization strategies. Other common ADR were pyrexia (10.6%) and febrile neutropenia (8.5%). In the efficacy analysis set, comprising patients with higher-risk MDS after azacitidine failure in phases 1 and 2 (n = 42), the disease control rate was 19.0%, and the median overall survival (OS) was 8.6 (90% confidence interval [CI], 6.8-10.3) months. Median OS was 10.0 (90% CI, 7.6-11.4) months in patients with a WT1-specific immune response (IR; n = 33) versus 4.1 (90% CI, 2.3-8.1) months in those without a WT1-specific IR (n = 9; P = .0034). The acceptable safety and clinical activity findings observed support the continued development of DSP-7888 dosing emulsion.
Collapse
Affiliation(s)
- Yasunori Ueda
- Department of Hematology/OncologyKurashiki Central HospitalOkayamaJapan
| | - Kensuke Usuki
- Department of HematologyNTT Medical Center TokyoTokyoJapan
| | - Jiro Fujita
- Department of Hematology and OncologyOsaka University HospitalOsakaJapan
| | - Itaru Matsumura
- Department of Hematology and RheumatologyKindai University HospitalOsakaJapan
| | - Nobuyuki Aotsuka
- Department of Hematology OncologyJapanese Red Cross Narita HospitalChibaJapan
| | - Naohiro Sekiguchi
- Department of HematologyNational Hospital Organization Disaster Medical CenterTokyoJapan
| | - Tomonori Nakazato
- Department of HematologyYokohama Municipal Citizen’s HospitalKanagawaJapan
| | - Hiromi Iwasaki
- Department of HematologyNational Hospital Organization Kyushu Medical CenterFukuokaJapan
| | | | | | | | - Tomoki Naoe
- National Hospital Organization Nagoya Medical CenterAichiJapan
| | | | - Yasushi Miyazaki
- Department of HematologyAtomic Bomb Disease and Hibakusha Medicine UnitAtomic Bomb Disease InstituteNagasaki UniversityNagasakiJapan
| | - Koichi Aakashi
- Department of Medicine and Biosystemic Science Faculty of MedicineKyushu UniversityFukuokaJapan
| |
Collapse
|
20
|
Transcriptional Regulation of Cardiac Development and Disease. Int J Mol Sci 2022; 23:ijms23062945. [PMID: 35328365 PMCID: PMC8953235 DOI: 10.3390/ijms23062945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/07/2022] [Indexed: 02/01/2023] Open
|
21
|
Li X, Zhong J, Deng X, Guo X, Lu Y, Lin J, Huang X, Wang C. Targeting Myeloid-Derived Suppressor Cells to Enhance the Antitumor Efficacy of Immune Checkpoint Blockade Therapy. Front Immunol 2022; 12:754196. [PMID: 35003065 PMCID: PMC8727744 DOI: 10.3389/fimmu.2021.754196] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/30/2021] [Indexed: 12/11/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that are activated under pathological conditions, such as cancer, or mature myeloid cells that are converted immune-suppressive cells via tumor-derived exosomes, and potently support the tumor processes at different levels. Currently, multiple studies have demonstrated that MDSCs induce immune checkpoint blockade (ICB) therapy resistance through their contribution to the immunosuppressive network in the tumor microenvironment. In addition, non-immunosuppressive mechanisms of MDSCs such as promotion of angiogenesis and induction of cancer stem cells also exert a powerful role in tumor progression. Thus, MDSCs are potential therapeutic targets to enhance the antitumor efficacy of ICB therapy in cases of multiple cancers. This review focuses on the tumor-promoting mechanism of MDSCs and provides an overview of current strategies that target MDSCs with the objective of enhancing the antitumor efficacy of ICB therapy.
Collapse
Affiliation(s)
- Xueyan Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China
| | - Jiahui Zhong
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xue Deng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xuan Guo
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Yantong Lu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Juze Lin
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China
| | - Xuhui Huang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China
| | - Changjun Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, China
| |
Collapse
|
22
|
Dynamic Spatiotemporal Expression Pattern of the Senescence-Associated Factor p16Ink4a in Development and Aging. Cells 2022; 11:cells11030541. [PMID: 35159350 PMCID: PMC8833900 DOI: 10.3390/cells11030541] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 02/01/2023] Open
Abstract
A plethora of factors have been attributed to underly aging, including oxidative stress, telomere shortening and cellular senescence. Several studies have shown a significant role of the cyclin-dependent kinase inhibitor p16ink4a in senescence and aging. However, its expression in development has been less well documented. Therefore, to further clarify a potential role of p16 in development and aging, we conducted a developmental expression study of p16, as well as of p19ARF and p21, and investigated their expression on the RNA level in brain, heart, liver, and kidney of mice at embryonic, postnatal, adult, and old ages. P16 expression was further assessed on the protein level by immunohistochemistry. Expression of p16 was highly dynamic in all organs in embryonic and postnatal stages and increased dramatically in old mice. Expression of p19 and p21 was less variable and increased to a moderate extent at old age. In addition, we observed a predominant expression of p16 mRNA and protein in liver endothelial cells versus non-endothelial cells of old mice, which suggests a functional role specifically in liver endothelium of old subjects. Thus, p16 dynamic spatiotemporal expression might implicate p16 in developmental and physiological processes in addition to its well-known function in the build-up of senescence.
Collapse
|
23
|
Oommen S, Cantero Peral S, Qureshi MY, Holst KA, Burkhart HM, Hathcock MA, Kremers WK, Brandt EB, Larsen BT, Dearani JA, Edwards BS, Maleszewski JJ, Nelson TJ. Autologous Umbilical Cord Blood-Derived Mononuclear Cell Therapy Promotes Cardiac Proliferation and Adaptation in a Porcine Model of Right Ventricle Pressure Overload. Cell Transplant 2022; 31:9636897221120434. [PMID: 36086821 PMCID: PMC9465577 DOI: 10.1177/09636897221120434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 07/19/2022] [Accepted: 07/31/2022] [Indexed: 11/29/2022] Open
Abstract
Congenital heart diseases, including single ventricle circulations, are clinically challenging due to chronic pressure overload and the inability of the myocardium to compensate for lifelong physiological demands. To determine the clinical relevance of autologous umbilical cord blood-derived mononuclear cells (UCB-MNCs) as a therapy to augment cardiac adaptation following surgical management of congenital heart disease, a validated model system of right ventricular pressure overload due to pulmonary artery banding (PAB) in juvenile pigs has been employed. PAB in a juvenile porcine model and intramyocardial delivery of UCB-MNCs was evaluated in three distinct 12-week studies utilizing serial cardiac imaging and end-of-study pathology evaluations. PAB reproducibly induced pressure overload leading to chronic right ventricular remodeling including significant myocardial fibrosis and elevation of heart failure biomarkers. High-dose UCB-MNCs (3 million/kg) delivered into the right ventricular myocardium did not cause any detectable safety issues in the context of arrhythmias or abnormal cardiac physiology. In addition, this high-dose treatment compared with placebo controls demonstrated that UCB-MNCs promoted a significant increase in Ki-67-positive cardiomyocytes coupled with an increase in the number of CD31+ endothelium. Furthermore, the incorporation of BrdU-labeled cells within the myocardium confirmed the biological potency of the high-dose UCB-MNC treatment. Finally, the cell-based treatment augmented the physiological adaptation compared with controls with a trend toward increased right ventricular mass within the 12 weeks of the follow-up period. Despite these adaptations, functional changes as measured by echocardiography and magnetic resonance imaging did not demonstrate differences between cohorts in this surgical model system. Therefore, this randomized, double-blinded, placebo-controlled pre-clinical trial establishes the safety of UCB-MNCs delivered via intramyocardial injections in a dysfunctional right ventricle and validates the induction of cardiac proliferation and angiogenesis as transient paracrine mechanisms that may be important to optimize long-term outcomes for surgically repaired congenital heart diseases.
Collapse
Affiliation(s)
- Saji Oommen
- Division of Cardiovascular Diseases,
Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Susana Cantero Peral
- Division of Cardiovascular Diseases,
Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Kimberly A. Holst
- Department of Cardiovascular Surgery,
Mayo Clinic, Rochester, MN, USA
| | - Harold M. Burkhart
- Pediatric Cardiothoracic Surgery, The
University of Oklahoma, Oklahoma City, OK, USA
| | | | - Walter K. Kremers
- Biomedical Statistics and Informatics,
Mayo Clinic, Rochester, MN, USA
| | - Emma B. Brandt
- Division of Cardiovascular Diseases,
Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Joseph A. Dearani
- Department of Cardiovascular Surgery,
Mayo Clinic, Rochester, MN, USA
| | | | | | - Timothy J. Nelson
- Division of Cardiovascular Diseases,
Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
24
|
Cytosolic Self-DNA—A Potential Source of Chronic Inflammation in Aging. Cells 2021; 10:cells10123544. [PMID: 34944052 PMCID: PMC8700131 DOI: 10.3390/cells10123544] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/28/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022] Open
Abstract
Aging is the consequence of a lifelong accumulation of stochastic damage to tissues and cellular components. Advancing age closely associates with elevated markers of innate immunity and low-grade chronic inflammation, probably reflecting steady increasing incidents of cellular and tissue damage over the life course. The DNA sensing cGAS-STING signaling pathway is activated by misplaced cytosolic self-DNA, which then initiates the innate immune responses. Here, we hypothesize that the stochastic release of various forms of DNA from the nucleus and mitochondria, e.g., because of DNA damage, altered nucleus integrity, and mitochondrial damage, can result in chronic activation of inflammatory responses that characterize the aging process. This cytosolic self-DNA-innate immunity axis may perturb tissue homeostasis and function that characterizes human aging and age-associated pathology. Proper techniques and experimental models are available to investigate this axis to develop therapeutic interventions.
Collapse
|
25
|
Schmidt V, Sieckmann T, Kirschner KM, Scholz H. WT1 regulates HOXB9 gene expression in a bidirectional way. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2021; 1864:194764. [PMID: 34508900 DOI: 10.1016/j.bbagrm.2021.194764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/09/2021] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
The homeoboxB9 (HOXB9) gene is necessary for specification of the anterior-posterior body axis during embryonic development and expressed in various types of cancer. Here we show that the Wilms tumor transcription factor WT1 regulates the HOXB9 gene in a bidirectional manner. Silencing of WT1 activates HOXB9 in Wt1 expressing renal cell adenocarcinoma-derived 786-0 cells, mesonephric M15 cells and ex vivo cultured murine embryonic kidneys. In contrast, HOXB9 expression in U2OS osteosarcoma and human embryonic kidney (HEK) 293 cells, which lack endogenous WT1, is enhanced by overexpression of WT1. Consistently, Hoxb9 promoter activity is stimulated by WT1 in transiently transfected U2OS and HEK293 cells, but inhibited in M15 cells with CRISPR/Cas9-mediated Wt1 deletion. Electrophoretic mobility shift assay and chromatin immunoprecipitation demonstrate binding of WT1 to the HOXB9 promoter in WT1-overexpressing U2OS cells and M15 cells. BASP1, a transcriptional co-repressor of WT1, is associated with the HOXB9 promoter in the chromatin of these cell lines. Co-transfection of U2OS and HEK293 cells with BASP1 plus WT1 prevents the stimulatory effect of WT1 on the HOXB9 promoter. Our findings identify HOXB9 as a novel downstream target gene of WT1. Depending on the endogenous expression of WT1, forced changes in WT1 can either stimulate or repress HOXB9, and the inhibitory effect of WT1 on transcription of HOXB9 involves BASP1. Consistent with inhibition of Hoxb9 expression by WT1, both transcripts are distributed in an almost non-overlapping pattern in embryonic mouse kidneys. Regulation of HOXB9 expression by WT1 might become relevant during kidney development and cancer progression.
Collapse
Affiliation(s)
- Valentin Schmidt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Vegetative Physiologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Tobias Sieckmann
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Vegetative Physiologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Karin M Kirschner
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Vegetative Physiologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Holger Scholz
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Vegetative Physiologie, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
26
|
Every Beat You Take-The Wilms' Tumor Suppressor WT1 and the Heart. Int J Mol Sci 2021; 22:ijms22147675. [PMID: 34299295 PMCID: PMC8306835 DOI: 10.3390/ijms22147675] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/06/2021] [Accepted: 07/16/2021] [Indexed: 12/23/2022] Open
Abstract
Nearly three decades ago, the Wilms’ tumor suppressor Wt1 was identified as a crucial regulator of heart development. Wt1 is a zinc finger transcription factor with multiple biological functions, implicated in the development of several organ systems, among them cardiovascular structures. This review summarizes the results from many research groups which allowed to establish a relevant function for Wt1 in cardiac development and disease. During development, Wt1 is involved in fundamental processes as the formation of the epicardium, epicardial epithelial-mesenchymal transition, coronary vessel development, valve formation, organization of the cardiac autonomous nervous system, and formation of the cardiac ventricles. Wt1 is further implicated in cardiac disease and repair in adult life. We summarize here the current knowledge about expression and function of Wt1 in heart development and disease and point out controversies to further stimulate additional research in the areas of cardiac development and pathophysiology. As re-activation of developmental programs is considered as paradigm for regeneration in response to injury, understanding of these processes and the molecules involved therein is essential for the development of therapeutic strategies, which we discuss on the example of WT1.
Collapse
|
27
|
Implications of the Wilms' Tumor Suppressor Wt1 in Cardiomyocyte Differentiation. Int J Mol Sci 2021; 22:ijms22094346. [PMID: 33919406 PMCID: PMC8122684 DOI: 10.3390/ijms22094346] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
The Wilms’ tumor suppressor Wt1 is involved in multiple developmental processes and adult tissue homeostasis. The first phenotypes recognized in Wt1 knockout mice were developmental cardiac and kidney defects. Wt1 expression in the heart has been described in epicardial, endothelial, smooth muscle cells, and fibroblasts. Expression of Wt1 in cardiomyocytes has been suggested but remained a controversial issue, as well as the role of Wt1 in cardiomyocyte development and regeneration after injury. We determined cardiac Wt1 expression during embryonic development, in the adult, and after cardiac injury by quantitative RT-PCR and immunohistochemistry. As in vitro model, phenotypic cardiomyocyte differentiation, i.e., the appearance of rhythmically beating clones from mouse embryonic stem cells (mESCs) and associated changes in gene expression were analyzed. We detected Wt1 in cardiomyocytes from embryonic day (E10.5), the first time point investigated, until adult age. Cardiac Wt1 mRNA levels decreased during embryonic development. In the adult, Wt1 was reactivated in cardiomyocytes 48 h and 3 weeks following myocardial infarction. Wt1 mRNA levels were increased in differentiating mESCs. Overexpression of Wt1(-KTS) and Wt1(+KTS) isoforms in ES cells reduced the fraction of phenotypically cardiomyocyte differentiated clones, which was preceded by a temporary increase in c-kit expression in Wt1(-KTS) transfected ES cell clones and induction of some cardiomyocyte markers. Taken together, Wt1 shows a dynamic expression pattern during cardiomyocyte differentiation and overexpression in ES cells reduces their phenotypical cardiomyocyte differentiation.
Collapse
|
28
|
WT1 activates transcription of the splice factor kinase SRPK1 gene in PC3 and K562 cancer cells in the absence of corepressor BASP1. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2020; 1863:194642. [PMID: 33017668 DOI: 10.1016/j.bbagrm.2020.194642] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/14/2020] [Accepted: 09/24/2020] [Indexed: 12/18/2022]
Abstract
Dysregulated alternative splicing plays a prominent role in all hallmarks of cancer. The splice factor kinase SRPK1 drives the activity of oncogenic splice factors such as SRSF1. SRSF1 in turn promotes the expression of splice isoforms that favour tumour growth, including proangiogenic VEGF. Knockdown (with siRNA) or chemical inhibition (using SPHINX) of SRPK1 in K562 leukemia and PC3 prostate cancer cell lines reduced cell proliferation, invasion and migration. In glomerular podocytes, the Wilms tumour suppressor zinc-finger transcription factor WT1 represses SRPK1 transcription. Here we show that in cancer cells WT1 activates SRPK1 transcription, unless a canonical WT1 binding site adjacent to the transcription start site is mutated. The ability of WT1 to activate SRPK1 transcription was reversed by the transcriptional corepressor BASP1, and both WT1 and BASP1 co-precipitated with the SRPK1 promoter. BASP1 significantly increased the expression of the antiangiogenic VEGF165b splice isoform. We propose that by upregulating SRPK1 transcription WT1 can direct an alternative splicing landscape that facilitates tumour growth.
Collapse
|
29
|
The Emerging Role of PPAR Beta/Delta in Tumor Angiogenesis. PPAR Res 2020; 2020:3608315. [PMID: 32855630 PMCID: PMC7443046 DOI: 10.1155/2020/3608315] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/24/2020] [Indexed: 12/31/2022] Open
Abstract
PPARs are ligand-activated transcriptional factors that belong to the nuclear receptor superfamily. Among them, PPAR alpha and PPAR gamma are prone to exert an antiangiogenic effect, whereas PPAR beta/delta has an opposite effect in physiological and pathological conditions. Angiogenesis has been known as a hallmark of cancer, and our recent works also demonstrate that vascular-specific PPAR beta/delta overexpression promotes tumor angiogenesis and progression in vivo. In this review, we will mainly focus on the role of PPAR beta/delta in tumor angiogenesis linked to the tumor microenvironment to further facilitate tumor progression and metastasis. Moreover, the crosstalk between PPAR beta/delta and its downstream key signal molecules involved in tumor angiogenesis will also be discussed, and the network of interplay between them will further be established in the review.
Collapse
|
30
|
Vanni I, Tanda ET, Dalmasso B, Pastorino L, Andreotti V, Bruno W, Boutros A, Spagnolo F, Ghiorzo P. Non-BRAF Mutant Melanoma: Molecular Features and Therapeutical Implications. Front Mol Biosci 2020; 7:172. [PMID: 32850962 PMCID: PMC7396525 DOI: 10.3389/fmolb.2020.00172] [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] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
Melanoma is one of the most aggressive tumors of the skin, and its incidence is growing worldwide. Historically considered a drug resistant disease, since 2011 the therapeutic landscape of melanoma has radically changed. Indeed, the improved knowledge of the immune system and its interactions with the tumor, and the ever more thorough molecular characterization of the disease, has allowed the development of immunotherapy on the one hand, and molecular target therapies on the other. The increased availability of more performing technologies like Next-Generation Sequencing (NGS), and the availability of increasingly large genetic panels, allows the identification of several potential therapeutic targets. In light of this, numerous clinical and preclinical trials are ongoing, to identify new molecular targets. Here, we review the landscape of mutated non-BRAF skin melanoma, in light of recent data deriving from Whole-Exome Sequencing (WES) or Whole-Genome Sequencing (WGS) studies on melanoma cohorts for which information on the mutation rate of each gene was available, for a total of 10 NGS studies and 992 samples, focusing on available, or in experimentation, targeted therapies beyond those targeting mutated BRAF. Namely, we describe 33 established and candidate driver genes altered with frequency greater than 1.5%, and the current status of targeted therapy for each gene. Only 1.1% of the samples showed no coding mutations, whereas 30% showed at least one mutation in the RAS genes (mostly NRAS) and 70% showed mutations outside of the RAS genes, suggesting potential new roads for targeted therapy. Ongoing clinical trials are available for 33.3% of the most frequently altered genes.
Collapse
Affiliation(s)
- Irene Vanni
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
| | | | - Bruna Dalmasso
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
| | - Lorenza Pastorino
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
| | - Virginia Andreotti
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
| | - William Bruno
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
| | - Andrea Boutros
- Medical Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Paola Ghiorzo
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
| |
Collapse
|
31
|
Wagner N, Wagner KD. PPAR Beta/Delta and the Hallmarks of Cancer. Cells 2020; 9:cells9051133. [PMID: 32375405 PMCID: PMC7291220 DOI: 10.3390/cells9051133] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear hormone receptor family. Three different isoforms, PPAR alpha, PPAR beta/delta and PPAR gamma have been identified. They all form heterodimers with retinoic X receptors to activate or repress downstream target genes dependent on the presence/absence of ligands and coactivators or corepressors. PPARs differ in their tissue expression profile, ligands and specific agonists and antagonists. PPARs attract attention as potential therapeutic targets for a variety of diseases. PPAR alpha and gamma agonists are in clinical use for the treatment of dyslipidemias and diabetes. For both receptors, several clinical trials as potential therapeutic targets for cancer are ongoing. In contrast, PPAR beta/delta has been suggested as a therapeutic target for metabolic syndrome. However, potential risks in the settings of cancer are less clear. A variety of studies have investigated PPAR beta/delta expression or activation/inhibition in different cancer cell models in vitro, but the relevance for cancer growth in vivo is less well documented and controversial. In this review, we summarize critically the knowledge of PPAR beta/delta functions for the different hallmarks of cancer biological capabilities, which interplay to determine cancer growth.
Collapse
|
32
|
Nikas IP, Themistocleous SC, Paschou SA, Tsamis KI, Ryu HS. Serine-Arginine Protein Kinase 1 (SRPK1) as a Prognostic Factor and Potential Therapeutic Target in Cancer: Current Evidence and Future Perspectives. Cells 2019; 9:cells9010019. [PMID: 31861708 PMCID: PMC7017105 DOI: 10.3390/cells9010019] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023] Open
Abstract
Cancer, a heterogeneous disease composed of tumor cells and microenvironment, is driven by deregulated processes such as increased proliferation, invasion, metastasis, angiogenesis, and evasion of apoptosis. Alternative splicing, a mechanism led by splicing factors, is implicated in carcinogenesis by affecting any of the processes above. Accumulating evidence suggests that serine-arginine protein kinase 1 (SRPK1), an enzyme that phosphorylates splicing factors rich in serine/arginine domains, has a prognostic and potential predictive role in various cancers. Its upregulation is correlated with higher tumor staging, grading, and shorter survival. SRPK1 is also highly expressed in the premalignant changes of some cancers, showing a potential role in the early steps of carcinogenesis. Of interest, its downregulation in preclinical models has mostly been tumor-suppressive and affected diverse processes heterogeneously, depending on the oncogenic context. In addition, targeting SRPK1 has enhanced sensitivity to platinum-based chemotherapy in some cancers. Lastly, its aberrant function has been noted not only in cancer cells but also in the endothelial cells of the microenvironment. Although the aforementioned evidence seems promising, more studies are needed to reinforce the use of SRPK1 inhibitors in clinical trials.
Collapse
Affiliation(s)
- Ilias P. Nikas
- School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus; (S.C.T.); (S.A.P.); (K.I.T.)
- Correspondence: ; Tel.: +357-22559633
| | - Sophie C. Themistocleous
- School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus; (S.C.T.); (S.A.P.); (K.I.T.)
| | - Stavroula A. Paschou
- School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus; (S.C.T.); (S.A.P.); (K.I.T.)
- Division of Endocrinology and Diabetes, “Aghia Sophia” Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Konstantinos I. Tsamis
- School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus; (S.C.T.); (S.A.P.); (K.I.T.)
- Neurosurgical Institute, Medical School, University of Ioannina, 45500 Ioannina, Greece
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, 03080 Seoul, Korea;
| |
Collapse
|
33
|
Immunohistochemical Expression of Wilms’ Tumor 1 Protein in Human Tissues: From Ontogenesis to Neoplastic Tissues. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app10010040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The human Wilms’ tumor gene (WT1) was originally isolated in a Wilms’ tumor of the kidney as a tumor suppressor gene. Numerous isoforms of WT1, by combination of alternative translational start sites, alternative RNA splicing and RNA editing, have been well documented. During human ontogenesis, according to the antibodies used, anti-C or N-terminus WT1 protein, nuclear expression can be frequently obtained in numerous tissues, including metanephric and mesonephric glomeruli, and mesothelial and sub-mesothelial cells, while cytoplasmic staining is usually found in developing smooth and skeletal cells, myocardium, glial cells, neuroblasts, adrenal cortical cells and the endothelial cells of blood vessels. WT1 has been originally described as a tumor suppressor gene in renal Wilms’ tumor, but more recent studies emphasized its potential oncogenic role in several neoplasia with a variable immunostaining pattern that can be exclusively nuclear, cytoplasmic or both, according to the antibodies used (anti-C or N-terminus WT1 protein). With the present review we focus on the immunohistochemical expression of WT1 in some tumors, emphasizing its potential diagnostic role and usefulness in differential diagnosis. In addition, we analyze the WT1 protein expression profile in human embryonal/fetal tissues in order to suggest a possible role in the development of organs and tissues and to establish whether expression in some tumors replicates that observed during the development of tissues from which these tumors arise.
Collapse
|
34
|
Wagner KD, Du S, Martin L, Leccia N, Michiels JF, Wagner N. Vascular PPARβ/δ Promotes Tumor Angiogenesis and Progression. Cells 2019; 8:cells8121623. [PMID: 31842402 PMCID: PMC6952835 DOI: 10.3390/cells8121623] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/01/2019] [Accepted: 12/11/2019] [Indexed: 01/20/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors, which function as transcription factors. Among them, PPARβ/δ is highly expressed in endothelial cells. Pharmacological activation with PPARβ/δ agonists had been shown to increase their angiogenic properties. PPARβ/δ has been suggested to be involved in the regulation of the angiogenic switch in tumor progression. However, until now, it is not clear to what extent the expression of PPARβ/δ in tumor endothelium influences tumor progression and metastasis formation. We addressed this question using transgenic mice with an inducible conditional vascular-specific overexpression of PPARβ/δ. Following specific over-expression of PPARβ/δ in endothelial cells, we induced syngenic tumors. We observed an enhanced tumor growth, a higher vessel density, and enhanced metastasis formation in the tumors of animals with vessel-specific overexpression of PPARβ/δ. In order to identify molecular downstream targets of PPARβ/δ in the tumor endothelium, we sorted endothelial cells from the tumors and performed RNA sequencing. We identified platelet-derived growth factor receptor beta (Pdgfrb), platelet-derived growth factor subunit B (Pdgfb), and the tyrosinkinase KIT (c-Kit) as new PPARβ/δ -dependent molecules. We show here that PPARβ/δ activation, regardless of its action on different cancer cell types, leads to a higher tumor vascularization which favors tumor growth and metastasis formation.
Collapse
Affiliation(s)
- Kay-Dietrich Wagner
- Université Côte d’Azur, CNRS, INSERM, iBV, 06107 Nice, France; (K.-D.W.); (S.D.); (L.M.)
| | - Siyue Du
- Université Côte d’Azur, CNRS, INSERM, iBV, 06107 Nice, France; (K.-D.W.); (S.D.); (L.M.)
| | - Luc Martin
- Université Côte d’Azur, CNRS, INSERM, iBV, 06107 Nice, France; (K.-D.W.); (S.D.); (L.M.)
| | - Nathalie Leccia
- Department of Pathology, CHU Nice, 06107 Nice, France; (N.L.); (J.-F.M.)
| | | | - Nicole Wagner
- Université Côte d’Azur, CNRS, INSERM, iBV, 06107 Nice, France; (K.-D.W.); (S.D.); (L.M.)
- Correspondence: ; Tel.: +33-493-377665
| |
Collapse
|
35
|
Mazzei L, Sanz R, Manucha W. Alterations on a key nephrogenic/cardiogenic gene expression linked to hypertension development. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2019; 32:70-78. [PMID: 31472952 DOI: 10.1016/j.arteri.2019.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/11/2019] [Indexed: 10/26/2022]
Abstract
The elevation of blood pressure produces specific organic lesions, including kidney and cardiac damage. On the other hand, cardiovascular disease usually leads to the development of hypertension. Thus, hypertension could be both a cause and a consequence of cardiovascular disease. Previous studies linked the lack of nitric oxide to cardiovascular abnormalities, including hypertension, arteriosclerosis, myocardial infarction, cardiac hypertrophy, diastolic heart failure, and reduced endothelium-derived hyperpolarizing factor responses, with shorter survival. The lack of this gas also leads to renal/cardiac abnormalities. It is widely known that nephrogenic deficiency is a risk factor for kidney disease. Besides, recent evidence suggests that alterations in WT-1, a key nephrogenic factor, could contribute to the development of hypertension. Moreover, some genes involved in the development of hypertension depend on WT-1. This knowledge makes it essential to investigate and understand the mechanisms regulating the expression of these genes during renal/cardiac development, and hypertension. As a consequence, the most in-depth knowledge of the complex aetiopathogenic mechanism responsible for the hypertensive disease will allow us to propose novel therapeutic tools.
Collapse
Affiliation(s)
- Luciana Mazzei
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica (IMBECU-CONICET), Mendoza, Argentina; Laboratorio de Farmacología Experimental Básica y Traslacional, Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Universitario, CP 5500 Mendoza, Argentina
| | - Raúl Sanz
- Laboratorio de Farmacología Experimental Básica y Traslacional, Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Universitario, CP 5500 Mendoza, Argentina
| | - Walter Manucha
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica (IMBECU-CONICET), Mendoza, Argentina; Laboratorio de Farmacología Experimental Básica y Traslacional, Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Universitario, CP 5500 Mendoza, Argentina.
| |
Collapse
|
36
|
Nishida S, Tsuboi A, Tanemura A, Ito T, Nakajima H, Shirakata T, Morimoto S, Fujiki F, Hosen N, Oji Y, Kumanogoh A, Kawase I, Oka Y, Azuma I, Morita S, Sugiyama H. Immune adjuvant therapy using Bacillus Calmette-Guérin cell wall skeleton (BCG-CWS) in advanced malignancies: A phase 1 study of safety and immunogenicity assessments. Medicine (Baltimore) 2019; 98:e16771. [PMID: 31415377 PMCID: PMC6831317 DOI: 10.1097/md.0000000000016771] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The cell wall skeleton of Bacillus Calmette-Guérin (BCG-CWS) is a bioactive component that is a strong immune adjuvant for cancer immunotherapy. BCG-CWS activates the innate immune system through various pattern recognition receptors and is expected to elicit antigen-specific cellular immune responses when co-administered with tumor antigens. To determine the recommended dose (RD) of BCG-CWS based on its safety profile, we conducted a phase I dose-escalation study of BCG-CWS in combination with WT1 peptide for patients with advanced cancer.The primary endpoint was the proportion of treatment-related adverse events (AEs) at each BCG-CWS dose. The secondary endpoints were immune responses and clinical effects. A BCG-CWS dose of 50, 100, or 200 μg/body was administered intradermally on days 0, 7, 21, and 42, followed by 2 mg of WT1 peptide on the next day. For the escalation of a dose level, 3 + 3 design was used.Study subjects were 18 patients with advanced WT1-expressing cancers refractory to standard anti-cancer therapies (7 melanoma, 5 colorectal, 4 hepatobiliary, 1 ovarian, and 1 lung). Dose-limiting toxicity occurred in the form of local skin reactions in 2 patients at a dose of 200 μg although no serious treatment-related systemic AEs were observed. Neutrophils and monocytes transiently increased in response to BCG-CWS. Some patients demonstrated the induction of the CD4 T cell subset and its differentiation from the naïve to memory phenotype, resulting in a tumor response.The RD of BCG-CWS was determined to be 100 μg/body. This dose was well tolerated and showed promising clinical effects with the induction of an appropriate immune response.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yusuke Oji
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita, Osaka
| | - Ichiro Kawase
- Department of Respiratory Medicine and Clinical Immunology
| | - Yoshihiro Oka
- Department of Respiratory Medicine and Clinical Immunology
- Department of Cancer Stem Cell Biology
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita, Osaka
| | | | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | |
Collapse
|
37
|
Cherfils-Vicini J, Iltis C, Cervera L, Pisano S, Croce O, Sadouni N, Győrffy B, Collet R, Renault VM, Rey-Millet M, Leonetti C, Zizza P, Allain F, Ghiringhelli F, Soubeiran N, Shkreli M, Vivier E, Biroccio A, Gilson E. Cancer cells induce immune escape via glycocalyx changes controlled by the telomeric protein TRF2. EMBO J 2019; 38:embj.2018100012. [PMID: 31000523 DOI: 10.15252/embj.2018100012] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 03/10/2019] [Accepted: 03/15/2019] [Indexed: 12/15/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells with strong immunosuppressive activity that promote tumor growth. In this study, we describe a mechanism by which cancer cells control MDSCs in human cancers by upregulating TRF2, a protein required for telomere stability. Specifically, we showed that the TRF2 upregulation in cancer cells has extratelomeric roles in activating the expression of a network of genes involved in the biosynthesis of heparan sulfate proteoglycan, leading to profound changes in glycocalyx length and stiffness, as revealed by atomic force microscopy. This TRF2-dependent regulation facilitated the recruitment of MDSCs, their activation via the TLR2/MyD88/IL-6/STAT3 pathway leading to the inhibition of natural killer recruitment and cytotoxicity, and ultimately tumor progression and metastasis. The clinical relevance of these findings is supported by our analysis of cancer cohorts, which showed a correlation between high TRF2 expression and MDSC infiltration, which was inversely correlated with overall patient survival.
Collapse
Affiliation(s)
- Julien Cherfils-Vicini
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Charlene Iltis
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Ludovic Cervera
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Sabrina Pisano
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Olivier Croce
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Nori Sadouni
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary.,2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Romy Collet
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Valérie M Renault
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Martin Rey-Millet
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Carlo Leonetti
- IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Pasquale Zizza
- IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Fabrice Allain
- CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, University of Lille, Villeneuve d'Ascq, Lille, France
| | - Francois Ghiringhelli
- INSERM, U866, UFR des Sciences de Sante, Universite de Bourgogne-Franche Comte, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| | - Nicolas Soubeiran
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Marina Shkreli
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Eric Vivier
- Aix Marseille Univ, APHM, CNRS, INSERM, CIML, Hôpital de la Timone, Marseille-Immunopole, Marseille, France.,Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | - Eric Gilson
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR7284, Institut National de la Santé et de la Recherche Médicale (INSERM) U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France .,Department of Medical Genetics, Archet 2 Hospital, CHU of Nice, FHU Oncoage, Nice, France
| |
Collapse
|
38
|
G-quadruplex forming region within WT1 promoter is selectively targeted by daunorubicin and mitoxantrone: A possible mechanism for anti-leukemic effect of drugs. J Biosci 2019. [DOI: 10.1007/s12038-018-9837-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Altered VEGF Splicing Isoform Balance in Tumor Endothelium Involves Activation of Splicing Factors Srpk1 and Srsf1 by the Wilms' Tumor Suppressor Wt1. Cells 2019; 8:cells8010041. [PMID: 30641926 PMCID: PMC6356959 DOI: 10.3390/cells8010041] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 12/27/2018] [Accepted: 01/08/2019] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is one hallmark of cancer. Vascular endothelial growth factor (VEGF) is a known inducer of angiogenesis. Many patients benefit from antiangiogenic therapies, which however have limitations. Although VEGF is overexpressed in most tumors, different VEGF isoforms with distinct angiogenic properties are produced through alternative splicing. In podocytes, the Wilms' tumor suppressor 1 (WT1) suppresses the Serine/arginine-rich protein-specific splicing factor kinase (SRPK1), and indirectly Serine/arginine-rich splicing factor 1 (Srsf1) activity, and alters VEGF splicing. We analyzed VEGF isoforms, Wt1, Srpk1, and Srsf1 in normal and tumor endothelium. Wt1, Srpk1, Srsf1, and the angiogenic VEGF164a isoform were highly expressed in tumor endothelium compared to normal lung endothelium. Nuclear expression of Srsf1 was detectable in the endothelium of various tumor types, but not in healthy tissues. Inducible conditional vessel-specific knockout of Wt1 reduced Wt1, Srpk1, and Srsf1 expression in endothelial cells and induced a shift towards the antiangiogenic VEGF120 isoform. Wt1(-KTS) directly binds and activates both the promoters of Srpk1 and Srsf1 in endothelial cells. In conclusion, Wt1 activates Srpk1 and Srsf1 and induces expression of angiogenic VEGF isoforms in tumor endothelium.
Collapse
|
40
|
McGregor RJ, Chau YY, Kendall TJ, Artibani M, Hastie N, Hadoke PWF. WT1 expression in vessels varies with histopathological grade in tumour-bearing and control tissue from patients with breast cancer. Br J Cancer 2018; 119:1508-1517. [PMID: 30374123 PMCID: PMC6288121 DOI: 10.1038/s41416-018-0317-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 09/28/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The Wilms' tumour protein (WT1), which influences tumour development and angiogenesis, is a promising therapeutic target in breast cancer. We hypothesised that WT1 expression would vary in endothelial cells in distinct sub-classifications of breast cancer. METHODS WT1 expression and vascular density were quantified by immunohistochemical analysis of human (n = 57) and murine breast cancers. Human tumours were sub-classified by histopathological grade, ER status and HER2 enrichment. RESULTS WT1 was identified in endothelial (and epithelial and smooth muscle) cells in tumours and tumour-free tissues (controls) from patients and mice with breast cancer. WT1 expression was higher in tumours than in controls, but this was not due to increased endothelial WT1. Vascular WT1 in cancers decreased as histopathological grade increased. WT1 was higher in ER-positive versus ER-negative cancers. Strikingly, reduced WT1 expression in controls correlated with an increased Nottingham Prognostic Index score. CONCLUSIONS Expression of WT1 is increased in breast cancers but this is not limited to the vascular compartment. The association between reduced WT1 in tumour-free tissue and poor prognosis suggests a protective role for WT1 in the healthy breast.
Collapse
Affiliation(s)
| | - You-Ying Chau
- University/BHF Centre for Cardiovascular Science, Edinburgh, UK.,MRC HGU at the MRC Institute of Genetics and Molecular Medicine (IGMM), Edinburgh, UK
| | - Timothy J Kendall
- MRC HGU at the MRC Institute of Genetics and Molecular Medicine (IGMM), Edinburgh, UK.,Division of Pathology, University of Edinburgh, Edinburgh, UK
| | - Mara Artibani
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nicholas Hastie
- MRC HGU at the MRC Institute of Genetics and Molecular Medicine (IGMM), Edinburgh, UK
| | | |
Collapse
|
41
|
Krivtsova O, Makarova A, Lazarevich N. Aberrant expression of alternative isoforms of transcription factors in hepatocellular carcinoma. World J Hepatol 2018; 10:645-661. [PMID: 30386458 PMCID: PMC6206146 DOI: 10.4254/wjh.v10.i10.645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/08/2018] [Accepted: 06/28/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies worldwide and the second leading cause of death among all cancer types. Deregulation of the networks of tissue-specific transcription factors (TFs) observed in HCC leads to profound changes in the hepatic transcriptional program that facilitates tumor progression. In addition, recent reports suggest that substantial aberrations in the production of TF isoforms occur in HCC. In vitro experiments have identified distinct isoform-specific regulatory functions and related biological effects of liver-specific TFs that are implicated in carcinogenesis, which may be relevant for tumor progression and clinical outcome. This study reviews available data on the expression of isoforms of liver-specific and ubiquitous TFs in the liver and HCC and their effects, including HNF4α, C/EBPs, p73 and TCF7L2, and indicates that assessment of the ratio of isoforms and targeting specific TF variants may be beneficial for the prognosis and treatment of HCC.
Collapse
Affiliation(s)
- Olga Krivtsova
- Federal State Budgetary Institution, “N. N. Blokhin Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Russian
- M. V. Lomonosov Moscow State University, Moscow 119991, Russian
| | - Anna Makarova
- Federal State Budgetary Institution, “N. N. Blokhin Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Russian
| | - Natalia Lazarevich
- Federal State Budgetary Institution, “N. N. Blokhin Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Russian
- M. V. Lomonosov Moscow State University, Moscow 119991, Russian
| |
Collapse
|
42
|
Hanada S, Tsuruta T, Haraguchi K, Okamoto M, Sugiyama H, Koido S. Long-term survival of pancreatic cancer patients treated with multimodal therapy combined with WT1-targeted dendritic cell vaccines. Hum Vaccin Immunother 2018; 15:397-406. [PMID: 30230959 DOI: 10.1080/21645515.2018.1524238] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND/AIM Pancreatic ductal adenocarcinoma (PDA) remains one of the most aggressive tumors with a dismally poor prognosis. Although surgical resection remains the only potentially curative treatment, most PDAs are not surgically resectable at diagnosis. Therefore, multimodal therapy is urgently needed to improve the long-term survival of PDA patients. METHODS Six eligible PDA patients underwent multimodal therapy comprising dendritic cells (DCs) pulsed with Wilms' tumor 1 (WT1) peptide (DC/WT1-I) restricted by the human leukocyte antigen (HLA) class I (A*24:02 or A*02:06) allele, chemotherapy, radiation, and/or surgery. Patient laboratory data, DC/WT1-I-specific delayed-type hypersensitivity (DTH) reactions, and WT1-specific immune responses were analyzed to assess the prognostic markers of multimodal therapy. RESULTS Compared to 2-treatment type combinations, multimodal therapy involving 3 to 4 treatment types was significantly associated with longer overall survival (p = 0.0177). Moreover, after 7 DC/WT1-I vaccinations, the progression-free survival (PFS) of PDA patients with a neutrophil to lymphocyte ratio (NLR) or C-reactive protein (CRP) level less than the median was superior to that of PDA patients with values above the median (p = 0.0246). PDA patients with an overall survival (OS)>1000 days had significantly more lymphocytes after one DC/WT1-I vaccination course than did those with an OS<1000 days. CONCLUSION Multimodal therapy involving the DC/WT1-I vaccination may benefit patients with advanced PDA. However, comparing the limited number of PDA patients in terms of survival is difficult because the patients were at different disease stages and received different treatments. Further studies are needed to evaluate the clinical benefits of this multimodal therapy.
Collapse
Affiliation(s)
- Shuichi Hanada
- a Department of Hematology , National Hospital Organization Kagoshima Medical Center , Kagoshima , Japan
| | - Tomoko Tsuruta
- a Department of Hematology , National Hospital Organization Kagoshima Medical Center , Kagoshima , Japan
| | - Kouichi Haraguchi
- a Department of Hematology , National Hospital Organization Kagoshima Medical Center , Kagoshima , Japan
| | - Masato Okamoto
- b Department of Advanced Immunotherapeutics, Graduate School of Pharmaceutical Sciences , Osaka University , Suita , Osaka , Japan
| | - Haruo Sugiyama
- c Department of Functional Diagnostic Science , Osaka University Graduate School of Medicine , Suita , Osaka , Japan
| | - Shigeo Koido
- d Division of Gastroenterology and Hepatology, Department of Hematology , The Jikei University School of Medicine , Kashiwa City , Chiba , Japan.,e Institute of Clinical Medicine and Research , The Jikei University School of Medicine , Kashiwa City , Chiba , Japan
| |
Collapse
|
43
|
Takizawa N, Tanaka S, Oe S, Koike T, Yoshida T, Hirahara Y, Matsuda T, Yamada H. Involvement of DHH and GLI1 in adrenocortical autograft regeneration in rats. Sci Rep 2018; 8:14542. [PMID: 30266964 PMCID: PMC6162278 DOI: 10.1038/s41598-018-32870-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 09/14/2018] [Indexed: 01/20/2023] Open
Abstract
Bilateral adrenalectomy forces the patient to undergo glucocorticoid replacement therapy and bear a lifetime risk of adrenal crisis. Adrenal autotransplantation is considered useful to avoid adrenal crisis and glucocorticoid replacement therapy. However, the basic process of regeneration in adrenal autografts is poorly understood. Here, we investigated the essential regeneration factors in rat adrenocortical autografts, with a focus on the factors involved in adrenal development and steroidogenesis, such as Hh signalling. A remarkable renewal in cell proliferation and increase in Cyp11b1, which encodes 11-beta-hydroxylase, occurred in adrenocortical autografts from 2-3 weeks after autotransplantation. Serum corticosterone and adrenocorticotropic hormone levels were almost recovered to sham level at 4 weeks after autotransplantation. The adrenocortical autografts showed increased Dhh expression at 3 weeks after autotransplantation, but not Shh, which is the only Hh family member to have been reported to be expressed in the adrenal gland. Increased Gli1 expression was also found in the regenerated capsule at 3 weeks after autotransplantation. Dhh and Gli1 might function in concert to regenerate adrenocortical autografts. This is the first report to clearly show Dhh expression and its elevation in the adrenal gland.
Collapse
Affiliation(s)
- Nae Takizawa
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
- Department of Urology and Andrology, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Susumu Tanaka
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan.
| | - Souichi Oe
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Taro Koike
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Takashi Yoshida
- Department of Urology and Andrology, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Yukie Hirahara
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Tadashi Matsuda
- Department of Urology and Andrology, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Hisao Yamada
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| |
Collapse
|
44
|
Niderla-Bielińska J, Bartkowiak K, Ciszek B, Jankowska-Steifer E, Krejner A, Ratajska A. Sulodexide inhibits angiogenesis via decreasing Dll4 and Notch1 expression in mouse proepicardial explant cultures. Fundam Clin Pharmacol 2018; 33:159-169. [PMID: 30246884 DOI: 10.1111/fcp.12418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/17/2018] [Accepted: 09/19/2018] [Indexed: 12/28/2022]
Abstract
Sulodexide (SDX) is a mixed drug containing low-molecular-weight heparin sulfate and dermatan sulfate. It exerts mild anticoagulant action but can also affect leukocytes, macrophages, and cell-cell adhesion and may interact with growth factors although its direct influence on endothelial cells is not well described. Clinically, SDX is used for the treatment of cardiovascular diseases, where it exerts anti-inflammatory and endothelial protective effects. The aim of this study was to determine the influence of SDX on tubule formation and angiogenesis-related proteins' mRNA expression in endothelial cell line C166 and mouse proepicardial explants. C166 cells and explants were stimulated with a proangiogenic cocktail containing bFGF/VEGF-A120 /VEGF-A164 enriched with SDX. After stimulation, the number and morphology of tubules stained with anti-CD31 antibody were examined under confocal microscope and expression of mRNA for VEGF-A, VEGF-B, VEGF-C, bFGF, IGF-1, Dll4, and Notch1 was measured with real-time PCR. In C166 cell line, there was no difference in tubule formation and mRNA expression, but in proepicardial explants, we observed reduction in tubule number and in mRNA level for DLL4 and Notch1 after SDX administration. In conclusion, SDX indirectly inhibits angiogenesis in mouse proepicardial explant cultures but has no direct effect on the C166 endothelial cell line.
Collapse
Affiliation(s)
- Justyna Niderla-Bielińska
- Department of Histology and Embryology, Medical Univertiry of Warsaw, Chałubinskiego 5 St, Warsaw, 02-004, Poland
| | - Krzysztof Bartkowiak
- Department of Histology and Embryology, Medical Univertiry of Warsaw, Chałubinskiego 5 St, Warsaw, 02-004, Poland
| | - Bogdan Ciszek
- Department of Clinical Anatomy, Medical Univertiry of Warsaw, Chałubinskiego 5 St, Warsaw, 02-004, Poland
| | - Ewa Jankowska-Steifer
- Department of Histology and Embryology, Medical Univertiry of Warsaw, Chałubinskiego 5 St, Warsaw, 02-004, Poland
| | - Alicja Krejner
- Department of Histology and Embryology, Medical Univertiry of Warsaw, Chałubinskiego 5 St, Warsaw, 02-004, Poland
| | - Anna Ratajska
- Department of Pathology, Medical University of Warsaw, Chałubinskiego 5 St, Warsaw, 02-004, Poland
| |
Collapse
|
45
|
Wu R, Liao Y, Shen W, Liu Y, Zhang J, Zheng M, Chen G, Su Y, Zhao M, Lu Q. Overexpression of Wilms' tumor 1 in skin lesions of psoriasis is associated with abnormal proliferation and apoptosis of keratinocytes. Mol Med Rep 2018; 18:3973-3982. [PMID: 30132523 DOI: 10.3892/mmr.2018.9391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/12/2018] [Indexed: 11/05/2022] Open
Abstract
Psoriasis vulgaris (PV) is a chronic inflammatory skin disease, which is characterized by the abnormal proliferation and apoptosis of keratinocytes. Previous studies have demonstrated that transcription factor Wilms' tumor 1 (WT1) is involved in a number of pathophysiological processes, including organ development, tumorigenesis and cell proliferation. However, the role of WT1 in PV remains unclear. In the present study, WT1 expression was analyzed by reverse transcription‑quantitative polymerase chain reaction and western blot analyses. WT1 was stably overexpressed or inhibited in HaCaT cells using Lipofectamine® 2000, and cell proliferation and apoptosis were determined using a Cell Counting Kit‑8 or Fluorescein Isothiocyanate Annexin V Apoptosis Detection kit II, respectively. We demonstrated that compared with normal controls, the mRNA and protein expression levels of WT1 were significantly increased in non‑lesional skins (human, P<0.0001 and P=0.0291, respectively; mouse, P=0.0020 and P=0.0150, respectively) and lesional skins (human, P<0.0001 and P=0.0060, respectively; mouse, P=0.0010 and P=0.0172, respectively) of patients with PV, in addition to the imiquimod (IMQ)‑induced psoriasis‑like mouse model. WT1 mRNA and protein expression levels in lesional skins were slightly increased compared with those in non‑lesional skins from patients with psoriasis (P=0.2510 and P=0.1690, respectively) and IMQ‑treated mice (P=0.9590 and P=0.2552, respectively), although there were no statistical differences. Knockdown of WT1 inhibited the proliferation of HaCaT cells [day (D)4, P=0.0454; D5, P=0.0021] and promoted their apoptosis (P=0.0007), while overexpressing WT1 exhibited the opposite effects (proliferation D3, P=0.0216; D4, P=0.0356; D5, P=0.0188; apoptosis, P=0.0003). Furthermore, it was identified that the inflammatory cytokines interleukin‑17A (IL‑17A), interferon‑γ and IL‑22 induced the overexpression of WT1 in HaCaT cells. The results of the present study suggested that inflammatory cytokine‑induced WT1 overexpression may promote the formation of psoriatic skin lesions via regulation of the proliferation and apoptosis of keratinocytes.
Collapse
Affiliation(s)
- Ruifang Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yuan Liao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Weiyun Shen
- Department of Anesthesiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yu Liu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Jianzhong Zhang
- Department of Dermatology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Min Zheng
- Department of Dermatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Genghui Chen
- Beijing Wenfeng Tianji Pharmaceuticals Ltd., Beijing 100027, P.R. China
| | - Yuwen Su
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
46
|
Kanai T, Ito Z, Oji Y, Suka M, Nishida S, Takakura K, Kajihara M, Saruta M, Fujioka S, Misawa T, Akiba T, Yanagisawa H, Shimodaira S, Okamoto M, Sugiyama H, Koido S. Prognostic significance of Wilms' tumor 1 expression in patients with pancreatic ductal adenocarcinoma. Oncol Lett 2018; 16:2682-2692. [PMID: 30008944 DOI: 10.3892/ol.2018.8961] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 05/16/2018] [Indexed: 02/07/2023] Open
Abstract
The only current curative treatment for patients with pancreatic ductal adenocarcinoma (PDA) is surgical resection, and certain patients still succumb to disease shortly after complete surgical resection. Wilms' tumor 1 (WT1) serves an oncogenic role in various types of tumors; therefore, in the present study, WT1 protein expression in patients with PDA was analyzed and the association with overall survival (OS) and disease-free survival (DFS) time in patients with PDA was assessed following surgical resection. A total of 50 consecutive patients with PDA who received surgical resection between January 2005 and December 2015 at the Jikei University Kashiwa Hospital (Kashiwa, Chiba, Japan) were enrolled. WT1 protein expression in PDA tissue was measured using immunohistochemical staining. Furthermore, laboratory parameters were measured within 2 weeks of surgery, and systemic inflammatory response markers were evaluated. WT1 protein expression was detected in the nucleus and cytoplasm of all PDA cells and in tumor vessels. WT1 exhibited weak staining in the nuclei of all PDA cells; however, the cytoplasmic expression of WT1 levels was classified into four groups: Negative (n=0), weak (n=19), moderate (n=23) and strong (n=8). In patients with PDA, it was demonstrated that the OS and DFS times of patients with weak cytoplasmic WT1 expression were significantly prolonged compared with those of patients with moderate-to-strong cytoplasmic WT1 expression, as determined by log-rank test (P=0.0005 and P=0.0001, respectively). Furthermore, an association between the density of WT1-expressing tumor vessels and worse OS/DFS times was detected. Multivariate analysis also indicated a significant association between the overexpression of WT1 in PDA tissue and worse OS/DFS times. To the best of our knowledge, the present study is the first to demonstrate that moderate-to-strong overexpression of WT1 in the cytoplasm of PDA cells is significantly associated with worse OS/DFS times. Therefore, overexpression of WT1 in the cytoplasm of PDA cells may impact the recurrence and prognosis of patients with PDA following surgical resection. The results further support the development of WT1-targeted therapies to prolong survival in all patients with PDA.
Collapse
Affiliation(s)
- Tomoya Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital, Kashiwa, Chiba 277-8567, Japan
| | - Zensho Ito
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital, Kashiwa, Chiba 277-8567, Japan
| | - Yusuke Oji
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Machi Suka
- Department of Public Health and Environmental Medicine, The Jikei University School of Medicine, Tokyo 105-8571, Japan
| | - Sumiyuki Nishida
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kazuki Takakura
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital, Kashiwa, Chiba 277-8567, Japan
| | - Mikio Kajihara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital, Kashiwa, Chiba 277-8567, Japan
| | - Masayuki Saruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8571, Japan
| | - Shuichi Fujioka
- Department of Surgery, The Jikei University School of Medicine, Kashiwa Hospital, Kashiwa, Chiba 277-8567, Japan
| | - Takeyuki Misawa
- Department of Surgery, The Jikei University School of Medicine, Kashiwa Hospital, Kashiwa, Chiba 277-8567, Japan
| | - Tadashi Akiba
- Department of Surgery, The Jikei University School of Medicine, Kashiwa Hospital, Kashiwa, Chiba 277-8567, Japan
| | - Hiroyuki Yanagisawa
- Department of Public Health and Environmental Medicine, The Jikei University School of Medicine, Tokyo 105-8571, Japan
| | - Shigetaka Shimodaira
- Department of Regenerative Medicine, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Masato Okamoto
- Department of Advanced Immunotherapeutics, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Shigeo Koido
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital, Kashiwa, Chiba 277-8567, Japan.,Institute of Clinical Medicine and Research, The Jikei University School of Medicine, Kashiwa, Chiba 277-8567, Japan
| |
Collapse
|
47
|
Peptide-pulsed dendritic cell vaccine in combination with carboplatin and paclitaxel chemotherapy for stage IV melanoma. Melanoma Res 2018; 27:326-334. [PMID: 28263240 DOI: 10.1097/cmr.0000000000000342] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this study, we aimed to evaluate the feasibility and efficacy of peptide-pulsed dendritic cell (DC) vaccine in combination with carboplatin and paclitaxel chemotherapy (DCCP) for patients with stage IV melanoma previously treated with dacarbazine-containing regimen. Six HLA-A24 and 3 HLA-A02 patients were treated with carboplatin (area under the curve 5) and paclitaxel (175 mg/m) on day 1 and DCs (2×10 cells) pulsed with Wilms tumor gene 1 (WT1), gp100, tyrosinase, and either MAGE-A3 (for HLA-A24) or MAGE-A2 (for HLA-A02) peptides on days 8 and 22 in 28-day cycle for up to three cycles. DCCP was well tolerated, and median progression-free survival and median overall survival were 2.3 and 12.0 months, respectively. In four of nine patients, a WT1-specific immune response (WT1-IR) was detected using the interferon-γ enzyme-linked ImmunoSpot assay and WT1/HLA tetramer assay. DCCP was more likely to elicit a WT1-IR in patients who received DCs pulsed with the HLA-A24-restricted peptide (75%) compared with patients who received DCs pulsed with the HLA-A02-restricted peptide (0%, P=0.058). Furthermore, three (75%) of four patients with a WT1-IR survived longer than 12 months, whereas only one (20%) of five patients without a WT1-IR who received the BRAF inhibitor after DCCP survived longer than 12 months. These results suggest that DCCP may be beneficial for HLA-A24 melanoma patients with a WT1-IR.
Collapse
|
48
|
Peng F, Li Q, Sun JY, Luo Y, Chen M, Bao Y. PFKFB3 is involved in breast cancer proliferation, migration, invasion and angiogenesis. Int J Oncol 2018; 52:945-954. [PMID: 29393396 DOI: 10.3892/ijo.2018.4257] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 12/29/2017] [Indexed: 11/05/2022] Open
Abstract
6-Phosphofructo 2-kinase/fructose 2, 6-bisphosphatase 3 (PFKFB3) has been reported to be overexpressed in human cancer tissues and to promote the proliferation and migration of cancer cells. However, the role of PFKFB3 in the progression and prognosis of breast cancer is not yet fully understood. In the present study, we investigated the specific role of PFKFB3 in breast cancer progression and its preliminary mechanisms of action. We first used an immunohistochemistry assay to determine that PFKFB3 was highly expressed in breast cancer tissues and that this high level of expression was involved in the poor overall survival of patients with breast cancer. In addition, the suppression of PFKFB3 by lentiviruses carrying shRNA against PFKFB3 (shPFKFB3) subsequently inhibited breast cancer cell (MDA-MB-231 and MDA-MB-468) proliferation, migration and invasion, and induced cell cycle G1 and S phase arrest in vitro. Moreover, PFKFB3 inhibition decreased p-AKT and increased p27 expression levels in breast cancer cells. Furthermore, PFKFB3 suppression inhibited breast cancer cell tumor xenograft growth in nude mice. We also found that PFKFB3 inhibition suppressed vascular endothelial growth factor α (VEGFα) protein expression and inhibited the angiogenic activity of human umbilical vein endothelial cells (HUVECs). On the whole, our results indicate that PFKFB3 is involved in the proliferation, migration, invasion and angiogenesis of breast cancer.
Collapse
Affiliation(s)
- Fang Peng
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Qiang Li
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jia-Yuan Sun
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of Radiation Oncology, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Ying Luo
- Department of Clinical Laboratory, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Ming Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022, P.R. China
| | - Yong Bao
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
49
|
Nishida S, Ishikawa T, Egawa S, Koido S, Yanagimoto H, Ishii J, Kanno Y, Kokura S, Yasuda H, Oba MS, Sato M, Morimoto S, Fujiki F, Eguchi H, Nagano H, Kumanogoh A, Unno M, Kon M, Shimada H, Ito K, Homma S, Oka Y, Morita S, Sugiyama H. Combination Gemcitabine and WT1 Peptide Vaccination Improves Progression-Free Survival in Advanced Pancreatic Ductal Adenocarcinoma: A Phase II Randomized Study. Cancer Immunol Res 2018; 6:320-331. [PMID: 29358173 DOI: 10.1158/2326-6066.cir-17-0386] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/17/2017] [Accepted: 01/09/2018] [Indexed: 11/16/2022]
Abstract
We investigated the efficacy of a Wilms' tumor gene 1 (WT1) vaccine combined with gemcitabine (GEMWT1) and compared it with gemcitabine (GEM) monotherapy for advanced pancreatic ductal adenocarcinoma (PDAC) in a randomized phase II study. We randomly assigned HLA-A*02:01- or HLA-A*24:02-positive patients with advanced PDAC to receive GEMWT1 or GEM. We assessed WT1-specific immune responses via delayed-type hypersensitivity (DTH) to the WT1 peptide and a tetramer assay to detect WT1-specific cytotoxic T lymphocytes (WT1-CTL). Of 91 patients enrolled, 85 were evaluable (GEMWT1: n = 42; GEM: n = 43). GEMWT1 prolonged progression-free survival [PFS; hazard ratio (HR), 0.66; P = 0.084] and improved overall survival rate at 1 year (1-year OS%; GEMWT1: 35.7%; GEM: 20.9%). However, the difference in OS was not significant (HR: 0.82; P = 0.363). These effects were particularly evident in metastatic PDAC (PFS: HR 0.51, P = 0.0017; 1-year OS%: GEMWT1 27.3%; GEM 11.8%). The combination was well tolerated, with no unexpected serious adverse events. In patients with metastatic PDAC, PFS in the DTH-positive GEMWT1 group was significantly prolonged, with a better HR of 0.27 compared with the GEM group, whereas PFS in the DTH-negative GEMWT1 group was similar to that in the GEM group (HR 0.86; P = 0.001). DTH positivity was associated with an increase in WT1-CTLs induced by the WT1 vaccine. GEM plus the WT1 vaccine prolonged PFS and may improve 1-year OS% in advanced PDAC. These clinical effects were associated with the induction of WT1-specific immune responses. Cancer Immunol Res; 6(3); 320-31. ©2018 AACR.
Collapse
Affiliation(s)
- Sumiyuki Nishida
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Takeshi Ishikawa
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shinichi Egawa
- Division of International Cooperation for Disaster Medicine, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Shigeo Koido
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Jun Ishii
- Division of General and Gastroenterological Surgery, Department of Surgery, Toho University Faculty of Medicine, Tokyo, Japan
| | - Yoshihide Kanno
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Satoshi Kokura
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroaki Yasuda
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mari Saito Oba
- Department of Biostatistics, Yokohama City University, Yokohama, Japan
| | - Maho Sato
- Department of Biostatistics, Yokohama City University, Yokohama, Japan
| | - Soyoko Morimoto
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Fumihiro Fujiki
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masanori Kon
- Department of Surgery, Kansai Medical University, Hirakata, Japan
| | - Hideaki Shimada
- Division of General and Gastroenterological Surgery, Department of Surgery, Toho University Faculty of Medicine, Tokyo, Japan
| | - Kei Ito
- Department of Gastroenterology, Sendai City Medical Center, Sendai, Japan
| | - Sadamu Homma
- Division of Oncology, Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshihiro Oka
- Department of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
50
|
STAT3 Gene Silencing by Aptamer-siRNA Chimera as Selective Therapeutic for Glioblastoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 10:398-411. [PMID: 29499951 PMCID: PMC5862137 DOI: 10.1016/j.omtn.2017.12.021] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 12/28/2017] [Accepted: 12/28/2017] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is the most frequent and aggressive primary brain tumor in adults, and despite advances in neuro-oncology, the prognosis for patients remains dismal. The signal transducer and activator of transcription-3 (STAT3) has been reported as a key regulator of the highly aggressive mesenchymal GBM subtype, and its direct silencing (by RNAi oligonucleotides) has revealed a great potential as an anti-cancer therapy. However, clinical use of oligonucleotide-based therapies is dependent on safer ways for tissue-specific targeting and increased membrane penetration. The objective of this study is to explore the use of nucleic acid aptamers as carriers to specifically drive a STAT3 siRNA to GBM cells in a receptor-dependent manner. Using an aptamer that binds to and antagonizes the oncogenic receptor tyrosine kinase PDGFRβ (Gint4.T), here we describe the design of a novel aptamer-siRNA chimera (Gint4.T-STAT3) to target STAT3. We demonstrate the efficient delivery and silencing of STAT3 in PDGFRβ+ GBM cells. Importantly, the conjugate reduces cell viability and migration in vitro and inhibits tumor growth and angiogenesis in vivo in a subcutaneous xenograft mouse model. Our data reveals Gint4.T-STAT3 conjugate as a novel molecule with great translational potential for GBM therapy.
Collapse
|