1
|
Dou T, Chen Y, Liu L, Zhang Y, Pei W, Li J, Lei Y, Wang Y, Jia H. Radiogenomic analysis of clinical and ultrasonic characteristics in correlation to immune-related genes in breast cancer. Sci Rep 2025; 15:15918. [PMID: 40335526 PMCID: PMC12058982 DOI: 10.1038/s41598-025-00891-w] [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: 10/23/2024] [Accepted: 05/02/2025] [Indexed: 05/09/2025] Open
Abstract
Breast ultrasound plays a significant role in the non-invasive screening and diagnosis of breast cancer. The application of immunotherapy for breast cancer can significantly prolong the overall survival of advanced patients, which is an important research area of breast cancer treatment. The combination of ultrasound and immunotherapy helps patients diagnose and predict survival and develop a personalized treatment plan. This study analyzed the correlation between the clinical and ultrasonic characteristics of breast cancer and immune-related genes. First, the differential expression of immune-related genes was obtained using the GEO and IMMPORT database. Then, differentially expressed immune-related genes related to the overall survival of breast cancer were obtained using the GEPIA and Kaplan-Meier plotter platforms. Additionally, clinical, ultrasonic characteristics and pathological specimens of breast cancer patients' tumors were collected. Whole transcriptome sequencing and immunohistochemical staining were performed on the tumor specimens to obtain gene expression. CXCL2, MIA, NR3C2, PTX3, S100B, SAA1, SAA2, and CXCL9 genes were correlated with each other and with clinical and ultrasonic characteristics. The high expression of MIA was related to the positive expression of PR in breast cancer. The low expression of NR3C2 was correlated with the clinical characteristics of tumor size ≥ 20 mm, later stage, Her-2 positive, Ki-67 ≥ 20%. NR3C2 was negatively correlated with the value of PKI and AUC in contrast-enhanced ultrasound parameters, and positively correlated with the value of AT and TTP. The expression of the PTX3 gene was also negatively correlated with the value of PKI and Emax of shear wave elastography. SAA2 was related to the presence or absence of edge burrs characterized by ultrasound. The expression of the CXCL9 gene was associated with the age of onset and tumor stage. In this study, 8 differentially expressed immune-related genes related to the overall survival of breast cancer were screened, which had been proved to be associated with some characteristics of cancer in previous studies, and could be further studied in the subsequent immunotherapy of breast cancer. Some clinical and ultrasonic characteristics of breast cancer were significantly correlated with immune-related genes, such as NR3C2, SAA2, and CXCL9. Further analysis of these genes provides new ideas for the diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Tingyao Dou
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaodong Chen
- Department of Ultrasonic Imaging, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Lunhang Liu
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaochen Zhang
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wanru Pei
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Li
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yan Lei
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, ShanxiMedical University, Taiyuan, Shanxi, China.
- key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China.
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Hongyan Jia
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China.
| |
Collapse
|
2
|
Dou T, Li J, Zhang Y, Pei W, Zhang B, Wang B, Wang Y, Jia H. The cellular composition of the tumor microenvironment is an important marker for predicting therapeutic efficacy in breast cancer. Front Immunol 2024; 15:1368687. [PMID: 38487526 PMCID: PMC10937353 DOI: 10.3389/fimmu.2024.1368687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
At present, the incidence rate of breast cancer ranks first among new-onset malignant tumors in women. The tumor microenvironment is a hot topic in tumor research. There are abundant cells in the tumor microenvironment that play a protumor or antitumor role in breast cancer. During the treatment of breast cancer, different cells have different influences on the therapeutic response. And after treatment, the cellular composition in the tumor microenvironment will change too. In this review, we summarize the interactions between different cell compositions (such as immune cells, fibroblasts, endothelial cells, and adipocytes) in the tumor microenvironment and the treatment mechanism of breast cancer. We believe that detecting the cellular composition of the tumor microenvironment is able to predict the therapeutic efficacy of treatments for breast cancer and benefit to combination administration of breast cancer.
Collapse
Affiliation(s)
- Tingyao Dou
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaochen Zhang
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Wanru Pei
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Binyue Zhang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Bin Wang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi, China
| | - Hongyan Jia
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi, China
| |
Collapse
|
3
|
Yihunie W, Nibret G, Aschale Y. Recent Advances in Messenger Ribonucleic Acid (mRNA) Vaccines and Their Delivery Systems: A Review. Clin Pharmacol 2023; 15:77-98. [PMID: 37554660 PMCID: PMC10405914 DOI: 10.2147/cpaa.s418314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/28/2023] [Indexed: 08/10/2023] Open
Abstract
Messenger ribonucleic acid (mRNA) was found as the intermediary that transfers genetic information from DNA to ribosomes for protein synthesis in 1961. The emergency use authorization of the two covid-19 mRNA vaccines, BNT162b2 and mRNA-1273, is a significant achievement in the history of vaccine development. Because they are generated in a cell-free environment using the in vitro transcription (IVT) process, mRNA vaccines are risk-free. Moreover, chemical modifications to the mRNA molecule, such as cap structures and changed nucleosides, have proved critical in overcoming immunogenicity concerns, achieving sustained stability, and achieving effective, accurate protein production in vivo. Several vaccine delivery strategies (including protamine, lipid nanoparticles (LNPs), polymers, nanoemulsions, and cell-based administration) were also optimized to load and transport RNA into the cytosol. LNPs, which are composed of a cationic or a pH-dependent ionizable lipid layer, a polyethylene glycol (PEG) component, phospholipids, and cholesterol, are the most advanced systems for delivering mRNA vaccines. Moreover, modifications of the four components that make up the LNPs showed to increase vaccine effectiveness and reduce side effects. Furthermore, the introduction of biodegradable lipids improved LNP biocompatibility. Furthermore, mRNA-based therapies are expected to be effective treatments for a variety of refractory conditions, including infectious diseases, metabolic genetic diseases, cancer, cardiovascular and cerebrovascular diseases. Therefore, the present review aims to provide the scientific community with up-to-date information on mRNA vaccines and their delivery systems.
Collapse
Affiliation(s)
- Wubetu Yihunie
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Getinet Nibret
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Aschale
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
4
|
Abbaspour M, Akbari V. Cancer vaccines as a targeted immunotherapy approach for breast cancer: an update of clinical evidence. Expert Rev Vaccines 2021; 21:337-353. [PMID: 34932427 DOI: 10.1080/14760584.2022.2021884] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Breast cancer (BC) is the first common neoplastic malignancy and the second leading cause of death in women worldwide. Conventional treatments for BC are often associated with severe side effects and may even lead to late recurrence. For this reason, in recent years, cancer immunotherapy (e.g., cancer vaccines), a novel approach based on the specificity and amplification of acquired immune responses, has been considered as a potential candidate in particular to treat metastatic BC. AREAS COVERED In this review, we summarize and discuss the recent development of therapeutic vaccines for BC, use of specific BC cellular antigens, antigen selection, and probable causes for their insufficient effectiveness. EXPERT OPINION Despite development of several different BC vaccines strategies including protein/peptide, dendritic cell, and genetic vaccines, until now, no BC vaccine has been approved for clinical use. Most of the current BC vaccines themselves fail to bring clinical benefit to BC patients and are applied in combination with radiotherapy, chemotherapy, or targeted therapy. It is hoped that with advances in our knowledge about tumor microenvironment and the development of novel combination strategies, the tumor immunosuppressive mechanisms can be overcome and prolonged immunologic and effective anti-tumor response can be developed in patients.
Collapse
Affiliation(s)
- Maryam Abbaspour
- Department of pharmaceutical biotechnology, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vajihe Akbari
- Department of pharmaceutical biotechnology, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.,Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
5
|
The Efficacy of Dendritic Cell Vaccine for Newly Diagnosed Glioblastoma: A Meta-analysis of Randomized Controlled Studies. Clin Neuropharmacol 2021; 44:216-221. [PMID: 34767325 DOI: 10.1097/wnf.0000000000000452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
ABSTRACT The efficacy of dendritic cell vaccine for newly diagnosed glioblastoma remains controversial. We conduct a systematic review and meta-analysis to explore the influence of dendritic cell vaccine on treatment efficacy for newly diagnosed glioblastoma. We search PubMed, EMBASE, Web of science, EBSCO, and Cochrane library databases through December 2019 for randomized controlled trials assessing the efficacy and safety of dendritic cell vaccine for newly diagnosed glioblastoma. This meta-analysis is performed using the random effect model. Three randomized controlled trials are included in the meta-analysis. Overall, compared with control group for newly diagnosed glioblastoma, dendritic cell vaccine shows no substantial effect on median overall survival [standard mean difference, 0.11; 95% confidence interval (CI), -0.18 to 0.41; P = 0.45], median progression-free survival (standard mean difference, 0.12; 95% CI, -0.24 to 0.48; P = 0.50), progression-free survival rate [risk ratio (RR), 1.29; 95% CI, 0.82-2.04; P = 0.27], overall survival rate (RR, 1.29; 95% CI, 0.61-2.72; P = 0.50), or nervous system disorders (RR, 0.80; 95% CI, 0.59-1.08; P = 0.14). Dendritic cell vaccine may provide no obvious benefits for the newly diagnosed glioblastoma.
Collapse
|
6
|
Ferdowsi S, Abbasi-Malati Z, Pourfathollah AA. Leukocyte reduction filters as an alternative source of peripheral blood leukocytes for research. Hematol Transfus Cell Ther 2021; 43:494-498. [PMID: 33422490 PMCID: PMC8573042 DOI: 10.1016/j.htct.2020.10.963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/11/2020] [Accepted: 10/01/2020] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION Peripheral blood leukocytes are a suitable cell model for science research. However, blood samples from healthy volunteers are limited in volume and difficult to obtain due to the complexity of volunteer recruitment. OBJECTIVE Therefore, it is urgent to find an alternative source of peripheral blood leukocytes. METHOD One of the possibilities is the use of leukocyte reduction filters (LRFs) in blood banks that is used for preparation of leukoreduced blood products. More than 90% of the leukocytes are trapped in the leukofilters allowing the desired blood product to pass through. RESULTS It has been reported that the biological function of leukocytes collected from the filters are no different from those isolated from buffy coats, leukapheresis products and whole blood (WB) cells. Moreover, LRFs are waste products that are discarded after leukoreduction. CONCLUSION Thus, leukofilters represent an economic source of human cell populations that can be used for a variety of investigative purposes, with no cost. In the present study, we reviewed the different usage of LRFs in the research, clinical and commercial applications.
Collapse
Affiliation(s)
- Shirin Ferdowsi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Zahra Abbasi-Malati
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Ali Akbar Pourfathollah
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran; Tarbiat Modares University, Faculty of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Qian B, Termer A, Sommer CM, Mehrabi A, Ryschich E. Low efficiency of leucocyte plugging-based drug delivery to cancer in mice. Drug Deliv Transl Res 2021; 12:1475-1487. [PMID: 34319578 PMCID: PMC9061658 DOI: 10.1007/s13346-021-01028-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 12/02/2022]
Abstract
Cells of the immune system were proposed for use as Trojan horse for tumour-specific drug delivery. The efficacy of such cell-based drug delivery depends on the site-specific cell homing. This present study was aimed to investigate the potential of leucocytes for intratumoural site-specific enrichment using a locoregional application route in experimental liver tumours. Human neutrophils were isolated from peripheral blood and directly labelled with calcein AM or loaded with doxorubicin. The neutrophil loading and release of doxorubicin and the migration and adhesion to ICAM-1 were analysed in vitro. Macrophages were isolated and activated in vitro. Leucocyte plugging and the distribution pattern in the liver microvasculature were studied ex vivo, and the efficacy of leucocyte plugging in tumour blood vessels was analysed in vivo after superselective intra-arterial injection in mouse liver tumour models. Neutrophils were characterised by the high dose-dependent uptake and rapid release of doxorubicin. Doxorubicin loading did not affect neutrophil migration function. Neutrophil plugging in liver microvasculature was very high (> 90%), both after ex vivo perfusion and after injection in vivo. However, neutrophils as well as activated macrophages plugged insufficiently in tumour blood vessels and passed through the tumour microvasculture with a very low sequestration rate in vivo. Neutrophils possess several properties to function as potentially effective drug carriers; however, the tumour site-specific drug delivery after selective locoregional injection was observed to be insufficient owing to low intratumoural microvascular plugging.
Collapse
Affiliation(s)
- Baifeng Qian
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 365, 69120, Heidelberg, Germany
| | - Andreas Termer
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 365, 69120, Heidelberg, Germany
| | - Christof M Sommer
- Clinic for Diagnostic and Interventional Radiology and Department of Nuclear Medicine; Clinic of Radiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Arianeb Mehrabi
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 365, 69120, Heidelberg, Germany
| | - Eduard Ryschich
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 365, 69120, Heidelberg, Germany.
| |
Collapse
|
8
|
Liu Z, Gao C, Tian J, Ma T, Cao X, Li A. The efficacy of dendritic cell vaccine for newly diagnosed glioblastoma: A meta-analysis of randomized controlled studies. Neurochirurgie 2021; 67:433-438. [PMID: 33915151 DOI: 10.1016/j.neuchi.2021.04.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 03/21/2021] [Accepted: 04/11/2021] [Indexed: 11/25/2022]
Abstract
INTRODUCTION The efficacy of dendritic cell vaccine to treat glioblastoma remained elusive and therefore we conducted a meta-analysis to explore the influence of dendritic cell vaccine on treatment efficacy of glioblastoma. METHODS PubMed, EMbase, Web of science, EBSCO and Cochrane library databases have been searched through October 2020, and we included randomized controlled trials (RCTs) assessing the efficacy of dendritic cell vaccine for glioblastoma. RESULTS Four RCTs and 267 patients were included in the meta-analysis. Compared to control group for glioblastoma, dendritic cell vaccine demonstrated no obvious impact on overall survival (HR=0.59; 95% CI=0.34 to 1.04; P=0.07), progression-free survival (PFS, HR=0.72; 95% CI=0.52 to 1.00; P=0.05), nervous system disorders (OR=0.61; 95% CI=0.29 to 1.29; P=0.20), or adverse events (OR=1.44; 95% CI=0.82 to 2.50; P=0.20). CONCLUSIONS Dendritic cell vaccine may be not effective to treat glioblastoma.
Collapse
Affiliation(s)
- Z Liu
- Department of neurosurgery, the general hospital of Ningxia Medical University, Ningxia, China.
| | - C Gao
- Ningxia Medical University, Ningxia, China.
| | - J Tian
- Department of neurosurgery, the general hospital of Ningxia Medical University, Ningxia, China.
| | - T Ma
- Department of neurosurgery, the People's Hospital of Tongxin County, Niangxia,China.
| | - X Cao
- Department of pathology, Ningxia Medical University, Niangxia, China.
| | - A Li
- Department of Pharmaceutical science, North Carolina Central University, Carolina, USA.
| |
Collapse
|
9
|
Rahman MM, Zhou N, Huang J. An Overview on the Development of mRNA-Based Vaccines and Their Formulation Strategies for Improved Antigen Expression In Vivo. Vaccines (Basel) 2021; 9:244. [PMID: 33799516 PMCID: PMC8001631 DOI: 10.3390/vaccines9030244] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 12/25/2022] Open
Abstract
The mRNA-based vaccine approach is a promising alternative to traditional vaccines due to its ability for prompt development, high potency, and potential for secure administration and low-cost production. Nonetheless, the application has still been limited by the instability as well as the ineffective delivery of mRNA in vivo. Current technological improvements have now mostly overcome these concerns, and manifold mRNA vaccine plans against various forms of malignancies and infectious ailments have reported inspiring outcomes in both humans and animal models. This article summarizes recent mRNA-based vaccine developments, advances of in vivo mRNA deliveries, reflects challenges and safety concerns, and future perspectives, in developing the mRNA vaccine platform for extensive therapeutic use.
Collapse
Affiliation(s)
- Md. Motiar Rahman
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China; (N.Z.); (J.H.)
| | - Nan Zhou
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China; (N.Z.); (J.H.)
| | - Jiandong Huang
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China; (N.Z.); (J.H.)
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
10
|
Zhou H, He H, Liang R, Pan H, Chen Z, Deng G, Zhang S, Ma Y, Liu L, Cai L. In situ poly I:C released from living cell drug nanocarriers for macrophage-mediated antitumor immunotherapy. Biomaterials 2021; 269:120670. [PMID: 33485214 DOI: 10.1016/j.biomaterials.2021.120670] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/17/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022]
Abstract
Immunotherapy is one of the most promising approaches to inhibit tumor growth and metastasis by activating host immune functions. However, the arising problems such as low immune response caused by complex tumor microenvironment and extremely systemic immune storm still limit the clinical applications of immunotherapy. Here, we construct Poly I: C-encapsulated poly (lactic-co-glycolic acid) nanoparticles (PLP NPs) with a slow release profile. A biomimetic system (MPLP), which loads PLP NPs on the surface of bone marrow-derived macrophage (BMDM) via the maleimide-thiol conjugation, is synthesized to effectively deliver PLP, control drug release and activate the tumor-specific immune response in situ. The results show that PLP NPs loading does not affect the activity and function of BMDM. Then, BMDM acts as a living cell drug vehicle and promotes the accumulation of PLP NPs in tumors, where Poly I: C is released from PLP NPs and reprograms BMDM into tumoricidal M1 macrophage. Furthermore, MPLP triggers potent antitumor immune responses in vivo and effectively inhibits local and metastatic tumors without causing adverse pathological immune reactions. This study offers an inspiration to facilitate clinical translation through the delivery of drugs by living immune cells for future anticancer therapy.
Collapse
Affiliation(s)
- Haimei Zhou
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China; Nano Science and Technology Institute, University of Science &Technology of China, Suzhou, 215123, PR China
| | - Huamei He
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Ruijing Liang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Ze Chen
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Guanjun Deng
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Shengping Zhang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China; Nano Science and Technology Institute, University of Science &Technology of China, Suzhou, 215123, PR China
| | - Yifan Ma
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China; HRYZ Biotech Co., Shenzhen, 518057, PR China.
| | - Lanlan Liu
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; Zhuhai Institute of Advanced Technology Chinese Academy of Sciences, Zhuhai, 519000, PR China.
| |
Collapse
|
11
|
Nasirmoghadas P, Mousakhani A, Behzad F, Beheshtkhoo N, Hassanzadeh A, Nikoo M, Mehrabi M, Kouhbanani MAJ. Nanoparticles in cancer immunotherapies: An innovative strategy. Biotechnol Prog 2020; 37:e3070. [PMID: 32829506 DOI: 10.1002/btpr.3070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/14/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022]
Abstract
Cancer has been one of the most significant causes of mortality, worldwide. Cancer immunotherapy has recently emerged as a competent, cancer-fighting clinical strategy. Nevertheless, due to the difficulty of such treatments, costs, and off-target adverse effects, the implementation of cancer immunotherapy described by the antigen-presenting cell (APC) vaccine and chimeric antigen receptor T cell therapy ex vivo in large clinical trials have been limited. Nowadays, the nanoparticles theranostic system as a promising target-based modality provides new opportunities to improve cancer immunotherapy difficulties and reduce their adverse effects. Meanwhile, the appropriate engineering of nanoparticles taking into consideration nanoparticle characteristics, such as, size, shape, and surface features, as well as the use of these physicochemical properties for suitable biological interactions, provides new possibilities for the application of nanoparticles in cancer immunotherapy. In this review article, we focus on the latest state-of-the-art nanoparticle-based antigen/adjuvant delivery vehicle strategies to professional APCs and engineering specific T lymphocyte required for improving the efficiency of tumor-specific immunotherapy.
Collapse
Affiliation(s)
- Pourya Nasirmoghadas
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Akbar Mousakhani
- Department of Plant Sciences, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Farahnaz Behzad
- Research Institute for Fundamental Sciences (RIFS), University of Tabriz, Tabriz, Iran
| | - Nasrin Beheshtkhoo
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Hassanzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Nikoo
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Helal Iran Pharmaceutical and Clinical Complex, Tehran, Iran
| | - Mohsen Mehrabi
- Department of Medical Nanotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Amin Jadidi Kouhbanani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Testori AAE, Ribero S, Indini A, Mandalà M. Adjuvant Treatment of Melanoma: Recent Developments and Future Perspectives. Am J Clin Dermatol 2019; 20:817-827. [PMID: 31177507 DOI: 10.1007/s40257-019-00456-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Surgical excision is the treatment of choice for early stage melanoma, and this strategy is initially curative for the vast majority of patients. However, only approximately 40-60% of high-risk patients who undergo surgery alone will be disease-free at 5 years. These patients will ultimately experience loco-regional relapse or relapse at distant sites. The main aim of adjuvant therapies is to reduce the recurrence rate of radically operated patients at high risk and to potentially improve survival. Recent practice changing results with immune checkpoint inhibitors and targeted therapies have been published in stage III/IV melanoma patients, after surgical complete resection, and have dramatically improved the landscape of adjuvant therapy. Interferon-α, ipilimumab, and more recently anti-programmed cell death protein-1 antibodies and BRAF inhibitors plus MEK inhibitors have been approved in the adjuvant setting by the US Food and Drug Administration; similarly, the same drugs are approved by the European Medicines Agency with the exception of ipilimumab. A completely new scenario is emerging in the neoadjuvant setting as well: in locally advanced or metastatic disease, patients may partially respond to neoadjuvant therapy and become virtually resectable with systemic control of disease. This review summarizes the current state of the field and describes new strategies tracing the history of adjuvant therapy in melanoma, with a view on future projects.
Collapse
Affiliation(s)
| | - Simone Ribero
- Medical Sciences Department, Dermatologic Clinic, University of Turin, Turin, Italy
| | - Alice Indini
- Melanoma Unit, Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Mario Mandalà
- Melanoma Unit, Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| |
Collapse
|
13
|
Wang L, Pegram MD, Wu JC. Induced pluripotent stem cells as a novel cancer vaccine. Expert Opin Biol Ther 2019; 19:1191-1197. [PMID: 31364894 PMCID: PMC12006513 DOI: 10.1080/14712598.2019.1650909] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/29/2019] [Indexed: 12/30/2022]
Abstract
Introduction: Although many current cancer therapies are effective, the mortality rate globally is unacceptably high. Cancer remains the second leading cause of death worldwide after heart disease and has caused nearly 10 million deaths in 2018. Additionally, current preventive therapies for cancer are underdeveloped, undermining the quality of life of high-risk individuals. Therefore, new treatment options for targeting cancer are urgently needed. In a recent study, researchers adopted an autologous iPSC-based vaccine to present a broad spectrum of tumor antigens to the immune system and succeeded in orchestrating a strong prophylactic immunity towards multiple types of cancer in mice. Areas covered: In this review, we provide an overview of how cancer develops, the role of immune surveillance in cancer progression, the current status and challenges of cancer immunotherapy as well as the genetic overlap between pluripotent stem cells and cancer cells. Finally, we discuss the rationale for an autologous iPSC-based vaccine and its applications in murine cancer models. Expert opinion: The autologous iPSC-based vaccine is a promising preventive and therapeutic strategy for fighting various types of cancers. Continuing efforts and clinical/translational follow-up studies may bring an autologous iPSC-based cancer vaccination approach from bench to bedside.
Collapse
Affiliation(s)
- Lin Wang
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark D. Pegram
- Stanford Women’s Cancer Center, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Joseph C. Wu
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University, Stanford, CA, USA
| |
Collapse
|
14
|
Nigro A, Montico B, Casolaro V, Dal Col J. A Novel Dendritic Cell-Based Vaccination Protocol to Stimulate Immunosurveillance of Aggressive Cancers. Methods Mol Biol 2019; 1884:317-333. [PMID: 30465213 DOI: 10.1007/978-1-4939-8885-3_22] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A major challenge in the development of a successful tumor vaccination is to break immune tolerance and to sensitize efficiently the immune system toward relevant tumor antigens, thus enabling T-cell-mediated antitumor responses in vivo. Dendritic cell (DC)-based immunotherapy shows the advantage to induce an adaptive immune response against the tumor, with the potential to generate a long-lasting immunological memory able to prevent further relapses and hopefully metastasis. Recently different preclinical studies highlighted the golden opportunity to exploit the features of immunogenic cell death (ICD) to generate ex vivo a highly immunogenic tumor cell lysate as potent antigen formulation for improved DC-based vaccine against aggressive cancers. This chapter focuses on the methods to obtain tumor lysates from cells undergoing ICD to be used for DC pulsing and to test the functionality of the generated DCs for antitumor vaccine development.
Collapse
Affiliation(s)
- Annunziata Nigro
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Baronissi, Salerno, Italy
| | - Barbara Montico
- Immunopathology and Cancer Biomarkers Unit, Department of Translational Research, CRO National Cancer Institute - IRCCS, Aviano, Pordenone, Italy
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Baronissi, Salerno, Italy
| | - Jessica Dal Col
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Baronissi, Salerno, Italy.
| |
Collapse
|
15
|
Zhen S, Lu J, Chen W, Zhao L, Li X. Synergistic Antitumor Effect on Bladder Cancer by Rational Combination of Programmed Cell Death 1 Blockade and CRISPR-Cas9-Mediated Long Non-Coding RNA Urothelial Carcinoma Associated 1 Knockout. Hum Gene Ther 2018; 29:1352-1363. [PMID: 30457360 DOI: 10.1089/hum.2018.048] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Targeted therapy produces objective responses in bladder cancer patients, although the responses can be short. Meanwhile, response rates to immune therapy are lower, but the effects are more durable. Based on these findings, it was hypothesized that urothelial carcinoma associated 1 (UCA1)-targeted therapy could synergize with programmed cell death 1 (PD-1) blockade to enhance antitumor activity. To test this hypothesis, the effects of CRISPR-Cas9 targeting of UCA1 and PD-1 were assessed in vitro and in vivo. It was found that gRNA/cas9-targeted UCA1 induced apoptosis of 5637 bladder cancer cells, whereas PD-1 gene knockout could be achieved by electroporation of gRNA/cas9 targeting PD-1, as detected by polymerase chain reaction. In 5637 cell-xenografted humanized SCID mice, stimulation with CRISPR-Cas9 systems, immune phenotypes, and cytokine expression of human dendritic cells (DCs) was detected by flow cytometry, and polymerase chain reaction, respectively. The results of these assays suggested that the gRNA/cas9 treatment upregulated expression of CD80, CD83, and CD86 and significantly increased interleukin (IL)-6, IL-12, and IL-23 and tumor necrosis factor alpha mRNA levels. Co-administration of anti-PD-1 and anti-UCA1 treatment suppressed tumor growth and markedly improved survival of 5637 xenografted mice. Additionally, the combination treatment increased interferon gamma production by T cells that subsequently enhanced the expression of Th1-associated immune-stimulating genes to reduce transcription of regulatory/suppressive immune genes and reshape the tumor microenvironment from an immunosuppressive to a stimulatory state. Finally, anti-UCA1 treatment was shown to induce interferon gamma-dependent programmed cell death ligand 1 expression within 5637 xenograft tumors in vivo. Together, these results demonstrate potent synergistic effects of a combination therapy using LncRNA UCA1-targeted therapy and immune checkpoint blockade of PD-1, thus supporting the translational potential of this combination strategy for clinical treatment of bladder cancer.
Collapse
Affiliation(s)
- Shuai Zhen
- 1 Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, P.R. China
- 2 Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, P.R. China
| | - Jiaojiao Lu
- 1 Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, P.R. China
- 2 Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, P.R. China
| | - Wei Chen
- 3 Center of Laboratory Medicine, the First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, P.R. China
| | - Le Zhao
- 1 Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, P.R. China
- 2 Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, P.R. China
| | - Xu Li
- 1 Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, P.R. China
- 2 Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, P.R. China
| |
Collapse
|
16
|
Ren S, Wang Q, Zhang Y, Song Y, Dong X, Zhang W, Qin X, Liu M, Yu T. Imiquimod enhances the potency of an exogenous BM-DC based vaccine against mouse melanoma. Int Immunopharmacol 2018; 64:69-77. [PMID: 30149266 DOI: 10.1016/j.intimp.2018.08.026] [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: 06/07/2018] [Revised: 08/08/2018] [Accepted: 08/20/2018] [Indexed: 12/19/2022]
Abstract
Dendritic cell (DC) vaccine is a potent immunotherapeutic approach for cancer treatment, but the clinical efficacy needs to be improved. In this study, we evaluated the combinational effect of Toll-like receptor 7 (TLR7) agonist Imiquimod and BM-DC vaccine against mouse melanoma and explored the potential mechanisms. We found that topical application of Imiquimod cream caused skin inflammation and enhanced exogenous BM-DC homing to draining lymph nodes. Imiquimod treatment enhanced DC vaccine efficacy against B16-OVA melanoma. The combinational modality enhanced cytotoxicity of splenic lymphocyte to tumor cells and inhibited CD4+FOXP3+Treg cell production. TLR7 mRNA expression was confirmed in both MC/9 mast cells and DCs. MC/9 cells treated by R837 (soluble form of Imiquimod) enhanced CD80, CD86, MHC-II and CCR7 expression on DCs. R837 inhibited B16-OVA cell growth in vitro. Our findings suggest that Imiquimod can be used as a potent adjuvant in the formulation of a DC-based tumor fighting vaccine. The mechanisms underlying these effects of Imiquimod are related with enhanced DC homing to DLNs, inhibition of Treg's production, direct tumor cell toxicity and synergistic function with mast cell in enhancing DC activation.
Collapse
Affiliation(s)
- Shurong Ren
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China.
| | - Qiubo Wang
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Yanli Zhang
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Yancheng Song
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Xue Dong
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Wendi Zhang
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Xianfei Qin
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Mingyue Liu
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| | - Ting Yu
- Department of Immunology, Basic Medical College of Qingdao University, Qingdao 266071, China
| |
Collapse
|
17
|
Han HD, Byeon Y, Jang JH, Jeon HN, Kim GH, Kim MG, Pack CG, Kang TH, Jung ID, Lim YT, Lee YJ, Lee JW, Shin BC, Ahn HJ, Sood AK, Park YM. In vivo stepwise immunomodulation using chitosan nanoparticles as a platform nanotechnology for cancer immunotherapy. Sci Rep 2016; 6:38348. [PMID: 27910914 PMCID: PMC5133713 DOI: 10.1038/srep38348] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 11/09/2016] [Indexed: 12/22/2022] Open
Abstract
Dentritic cell (DC)-based cancer immunotherapy faces challenges in both efficacy and practicality. However, DC-based vaccination requires multiple injections and elaborates ex vivo manipulation, which substantially limits their use. Therefore, we sought to develop a chitosan nanoparticle (CH-NP)-based platform for the next generation of vaccines to bypass the ex vivo manipulation and induce immune responses via active delivery of polyinosinic-polycytidylic acid sodium salt (poly I:C) to target Toll-like receptor 3 (TLR3) in endosomes. We developed CH-NPs encapsulating ovalbumin (OVA) as a model antigen and poly I:C as the adjuvant in an ionic complex. These CH-NPs showed increased in vivo intracellular delivery to the DCs in comparison with controls after injection into tumor-bearing mice, and promoted DC maturation, leading to emergence of antigen-specific cytotoxic CD8+ T cells. Finally, the CH-NPs showed significantly greater antitumor efficacy in EG.7 and TC-1 tumor-bearing mice compared to the control (p < 0.01). Taken together, these data show that the CH-NP platform can be used as an immune response modulatory vaccine for active cancer immunotherapy without ex vivo manipulation, thus resulting in increased anticancer efficacy.
Collapse
Affiliation(s)
- Hee Dong Han
- Department of Immunology, School of Medicine, Konkuk University, Chungju 380-701, South Korea
| | - Yeongseon Byeon
- Department of Immunology, School of Medicine, Konkuk University, Chungju 380-701, South Korea
| | - Jong-Hwa Jang
- Department of Dental Hygiene, Hanseo University, Seosan 31962, South Korea
| | - Hat Nim Jeon
- Department of Immunology, School of Medicine, Konkuk University, Chungju 380-701, South Korea
| | - Ga Hee Kim
- Department of Immunology, School of Medicine, Konkuk University, Chungju 380-701, South Korea
| | - Min Gi Kim
- Department of Immunology, School of Medicine, Konkuk University, Chungju 380-701, South Korea
| | - Chan-Gi Pack
- Department of Convergence Medicine, University of Ulsan College of Medicine &Asan Institute for Life Sciences, Asan Medical Center, Seoul 055-05, South Korea
| | - Tae Heung Kang
- Department of Immunology, School of Medicine, Konkuk University, Chungju 380-701, South Korea
| | - In Duk Jung
- Department of Immunology, School of Medicine, Konkuk University, Chungju 380-701, South Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), School of Chemical Engineering, Sungkyunkwan University, Suwon 25-2, South Korea
| | - Young Joo Lee
- Department of Bioscience and Biotechnology, Sejong University, Kwang-Jin-Gu, Seoul 143-747, South Korea
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sunkyunkwan University School of Medicine, Seoul 06531, South Korea
| | - Byung Cheol Shin
- Bio/Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 305-600, South Korea
| | - Hyung Jun Ahn
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 136-791, South Korea
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, the University of Texas M.D. Anderson Cancer Center, Texas, USA.,Department of Cancer Biology, the University of Texas M.D. Anderson Cancer Center, Texas, USA.,Center for RNA Interference and Non-coding RNA, The University of Texas M.D. Anderson Cancer Center, Texas, USA
| | - Yeong-Min Park
- Department of Immunology, School of Medicine, Konkuk University, Chungju 380-701, South Korea
| |
Collapse
|
18
|
Yan LH, Chen ZN, Li L, Chen J, Mo XW, Qin YZ, Wei WE, Qin HQ, Lin Y, Chen JS. E2F-1 promotes DAPK2-induced anti-tumor immunity of gastric cancer cells by targeting miR-34a. Tumour Biol 2016; 37:15925–15936. [PMID: 27704360 DOI: 10.1007/s13277-016-5446-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 09/23/2016] [Indexed: 12/20/2022] Open
Abstract
Activation of the transcription factor E2F-1 gene is a negative event in dendritic cell (DC) maturation process. Down-regulation of E2F1 causes immaturity of DC thereby stopping antigen production which in turn leads to inhibition of immune responses. E2F-1-free stimulates the NF-kB signaling pathway, leading to activation of monocytes and several other transcription factor genes. In the study, we report that down-regulation of E2F-1 in DCs promote anti-tumor immune response in gastric cancer (GC) cells through a novel mechanism. DCs were isolated from peripheral blood mononuclear cells. E2F-1 small interfering RNA (E2F-1-shRNA) induced down-regulation of E2F-1 mRNA and protein expression in DCs. Furthermore, we identified the E2F-1-shRNA targeted the CD80, CD83, CD86, and MHC II molecules, promoted their expression, and induced T lymphocytes proliferation activity and up-regulation of IFN-γ production and GC cell killing effect, which significantly correlated with the cytotoxic T lymphocytes activated by E2F-1-shRNA DCs. The higher expression of miR-34a was found which was significantly correlated with the DC enhancing anti-tumor immunity against gastric cancer cell, and miR-34a potently targeted DAPK2 and Sp1, both of which were involved in the deactivation of E2F-1. Moreover, in E2F-1-DC-down-regulation in mice, GC transplantation tumors displayed down-regulation of Sp1, DAPK2, Caspase3, and Caspase7 and progressed to anti-tumor immunity. Collectively, our data uncover an E2F-1-mediated mechanism for the control of DC anti-tumor immunity via miR-34a-dependent down-regulation of E2F-1 expression and suggest its contribution to GC immunotherapy.
Collapse
Affiliation(s)
- Lin-Hai Yan
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
| | - Zhi-Ning Chen
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Li Li
- Department of Pharmacy, The People Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Jia Chen
- Department of Medical Image Center, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xian-Wei Mo
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yu-Zhou Qin
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Wen-E Wei
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Hai-Quan Qin
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yuan Lin
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Jian-Si Chen
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| |
Collapse
|
19
|
Fliervoet LAL, Mastrobattista E. Drug delivery with living cells. Adv Drug Deliv Rev 2016; 106:63-72. [PMID: 27129442 DOI: 10.1016/j.addr.2016.04.021] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 12/25/2022]
Abstract
The field of drug delivery has grown tremendously in the past few decades by developing a wide range of advanced drug delivery systems. An interesting category is cell-based drug delivery, which includes encapsulation of drugs inside cells or attached to the surface and subsequent transportation through the body. Another approach involves genetic engineering of cells to secrete therapeutic molecules in a controlled way. The next-generation systems integrate expertise from synthetic biology to generate therapeutic gene networks for highly advanced sensory and output devices. These developments are very exciting for the drug delivery field and could radically change the way we administer biological medicines to chronically ill patients. This review is covering the use of living cells, either as transport system or production-unit, to deliver therapeutic molecules and bioactive proteins inside the body. It describes a wide range of approaches in cell-based drug delivery and highlights exceptional examples.
Collapse
Affiliation(s)
- Lies A L Fliervoet
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands.
| |
Collapse
|
20
|
Pham PV, Le HT, Vu BT, Pham VQ, Le PM, Phan NLC, Trinh NV, Nguyen HTL, Nguyen ST, Nguyen TL, Phan NK. Targeting breast cancer stem cells by dendritic cell vaccination in humanized mice with breast tumor: preliminary results. Onco Targets Ther 2016; 9:4441-51. [PMID: 27499638 PMCID: PMC4959598 DOI: 10.2147/ott.s105239] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background Breast cancer (BC) is one of the leading cancers in women. Recent progress has enabled BC to be cured with high efficiency. However, late detection or metastatic disease often renders the disease untreatable. Additionally, relapse is the main cause of death in BC patients. Breast cancer stem cells (BCSCs) are considered to cause the development of BC and are thought to be responsible for metastasis and relapse. This study aimed to target BCSCs using dendritic cells (DCs) to treat tumor-bearing humanized mice models. Materials and methods NOD/SCID mice were used to produce the humanized mice by transplantation of human hematopoietic stem cells. Human BCSCs were injected into the mammary fat pad to produce BC humanized mice. Both hematopoietic stem cells and DCs were isolated from the human umbilical cord blood, and immature DCs were produced from cultured mononuclear cells. DCs were matured by BCSC-derived antigen incubation for 48 hours. Mature DCs were vaccinated to BC humanized mice with a dose of 106 cells/mice, and the survival percentage was monitored in both treated and untreated groups. Results The results showed that DC vaccination could target BCSCs and reduce the tumor size and prolong survival. Conclusion These results suggested that targeting BCSCs with DCs is a promising therapy for BC.
Collapse
Affiliation(s)
- Phuc Van Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City
| | - Hanh Thi Le
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City
| | - Binh Thanh Vu
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City
| | - Viet Quoc Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City
| | - Phong Minh Le
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City
| | - Nhan Lu-Chinh Phan
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City
| | - Ngu Van Trinh
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City
| | - Huyen Thi-Lam Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City
| | - Sinh Truong Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City
| | | | - Ngoc Kim Phan
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City
| |
Collapse
|
21
|
Improvement of DC vaccine with ALA-PDT induced immunogenic apoptotic cells for skin squamous cell carcinoma. Oncotarget 2016; 6:17135-46. [PMID: 25915530 PMCID: PMC4627297 DOI: 10.18632/oncotarget.3529] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/08/2015] [Indexed: 02/07/2023] Open
Abstract
Dendritic cell (DC) based vaccines have emerged as a promising immunotherapy for cancers. However, most DC vaccines so far have achieved only limited success in cancer treatment. Photodynamic therapy (PDT), an established cancer treatment strategy, can cause immunogenic apoptosis to induce an effective antitumor immune response. In this study, we developed a DC-based cancer vaccine using immunogenic apoptotic tumor cells induced by 5-aminolevulinic acid (ALA) mediated PDT. The maturation of DCs induced by PDT-treated apoptotic cells was evaluated using electron microscopy, FACS, and ELISA. The anti-tumor immunity of ALA-PDT-DC vaccine was tested with a mouse model. We observed the maturations of DCs potentiated by ALA-PDT treated tumor cells, including morphology maturation (enlargement of dendrites and increase of lysosomes), phenotypic maturation (upregulation of surface expression of MHC-II, DC80, and CD86), and functional maturation (enhanced capability to secrete IFN-γ and IL-12, and to induce T cell proliferation). Most interestingly, PDT-induced apoptotic tumor cells are more capable of potentiating maturation of DCs than PDT-treated or freeze/thaw treated necrotic tumor cells. ALA-PDT-DC vaccine mediated by apoptotic cells provided protection against tumors in mice, far stronger than that of DC vaccine obtained from freeze/thaw treated tumor cells. Our results indicate that immunogenic apoptotic tumor cells can be more effective in enhancing a DC-based cancer vaccine, which could improve the clinical application of PDT-DC vaccines.
Collapse
|
22
|
Wojas-Turek J, Szczygieł A, Kicielińska J, Rossowska J, Piasecki E, Pajtasz-Piasecka E. Treatment with cyclophosphamide supported by various dendritic cell-based vaccines induces diversification in CD4⁺ T cell response against MC38 colon carcinoma. Int J Oncol 2015; 48:493-505. [PMID: 26648160 PMCID: PMC4725454 DOI: 10.3892/ijo.2015.3278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/26/2015] [Indexed: 12/18/2022] Open
Abstract
The present study shows that an application of cyclophosphamide (CY) supported by dendritic cell (DC)-based vaccines affected differentiation of the activity of CD4+ T cell subpopulations accompanied by an alteration in CD8+ cell number. Vaccines were composed of bone marrow-derived DCs activated with tumor cell lysate (BM-DC/TAgTNF-α) and/or genetically modified DCs of JAWS II line (JAWS II/ Neo or JAWS II/IL-2 cells). Compared to untreated or CY-treated mice, the combined treatment of MC38 colon carcinoma-bearing mice resulted in significant tumor growth inhibition associated with an increase in influx of CD4+ and CD8+ T cells into tumor tissue. Whereas, the division of these cell population in spleen was not observed. Depending on the nature of DC-based vaccines and number of their applications, both tumor infiltrating cells and spleen cells were able to produce various amount of IFN-γ, IL-4 and IL-10 after mitogenic ex vivo stimulation. The administration of CY followed by BM-DC/TAgTNF-α and genetically modified JAWS II cells, increased the percentage of CD4+T-bet+ and CD4+GATA3+ cells and decreased the percentage of CD4+RORγt+ and CD4+FoxP3+ lymphocytes. However, the most intensive response against tumor was noted after the ternary treatment with CY + BM-DC/TAgTNF-α + JAWS II/IL-2 cells. Thus, the administration of various DC-based vaccines was responsible for generation of the diversified antitumor response. These findings demonstrate that the determination of the size of particular CD4+ T cell subpopulations may become a prognostic factor and be the basis for future development of anticancer therapy.
Collapse
Affiliation(s)
- Justyna Wojas-Turek
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Agnieszka Szczygieł
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Jagoda Kicielińska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Joanna Rossowska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Egbert Piasecki
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Elżbieta Pajtasz-Piasecka
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| |
Collapse
|
23
|
Gutknecht M, Geiger J, Joas S, Dörfel D, Salih HR, Müller MR, Grünebach F, Rittig SM. The transcription factor MITF is a critical regulator of GPNMB expression in dendritic cells. Cell Commun Signal 2015; 13:19. [PMID: 25889792 PMCID: PMC4422548 DOI: 10.1186/s12964-015-0099-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 03/10/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Dendritic cells (DC) are the most potent antigen-presenting cells (APC) with the unique ability to activate naïve T cells and to initiate and maintain primary immune responses. Immunosuppressive and anti-inflammatory stimuli on DC such as the cytokine IL-10 suppress the activity of the transcription factor NF-κB what results in downregulation of costimulatory molecules, MHC and cytokine production. Glycoprotein NMB (GPNMB) is a transmembrane protein, which acts as a coinhibitory molecule strongly inhibiting T cell responses if present on APC. Interestingly, its expression on human monocyte-derived dendritic cells (moDC) is dramatically upregulated upon treatment with IL-10 but also by the BCR-ABL tyrosine kinase inhibitors (TKI) imatinib, nilotinib or dasatinib used for the treatment of chronic myeloid leukemia (CML). However, the molecular mechanisms responsible for GPNMB overexpression are yet unknown. RESULTS The immunosuppressive cytokine IL-10 and the BCR-ABL TKI imatinib or nilotinib, that were examined here, concordantly inhibit the PI3K/Akt signaling pathway, thereby activating the downstream serine/threonine protein kinase GSK3ß, and subsequently the microphthalmia-associated transcription factor (MITF) that is phosphorylated and translocated into the nucleus. Treatment of moDC with a small molecule inhibitor of MITF activity reduced the expression of GPNMB at the level of mRNA and protein, indicating that GPNMB expression is in fact facilitated by MITF activation. In line with these findings, PI3K/Akt inhibition was found to result in GPNMB overexpression accompanied by reduced stimulatory capacity of moDC in mixed lymphocyte reactions (MLR) with allogeneic T cells that could be restored by addition of the GPNMB T cell ligand syndecan-4 (SD-4). CONCLUSIONS In summary, imatinib, nilotinib or IL-10 congruently inhibit the PI3K/Akt signaling pathway thereby activating MITF in moDC, resulting in a tolerogenic phenotype. These findings extend current knowledge on the molecular mechanisms balancing activating and inhibitory signals in human DC and may facilitate the targeted manipulation of T cell responses in the context of DC-based immunotherapeutic interventions.
Collapse
Affiliation(s)
- Michael Gutknecht
- Department of Internal Medicine II, Oncology, Hematology, Immunology, Rheumatology and Pulmology, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
| | - Julian Geiger
- Department of Internal Medicine II, Oncology, Hematology, Immunology, Rheumatology and Pulmology, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
| | - Simone Joas
- Department of Internal Medicine II, Oncology, Hematology, Immunology, Rheumatology and Pulmology, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
| | - Daniela Dörfel
- Department of Internal Medicine II, Oncology, Hematology, Immunology, Rheumatology and Pulmology, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
| | - Helmut R Salih
- Department of Internal Medicine II, Oncology, Hematology, Immunology, Rheumatology and Pulmology, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
| | - Martin R Müller
- Department of Internal Medicine II, Oncology, Hematology, Immunology, Rheumatology and Pulmology, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
| | - Frank Grünebach
- Department of Internal Medicine II, Oncology, Hematology, Immunology, Rheumatology and Pulmology, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
| | - Susanne M Rittig
- Department of Internal Medicine II, Oncology, Hematology, Immunology, Rheumatology and Pulmology, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
| |
Collapse
|
24
|
Gelao L, Criscitiello C, Esposito A, De Laurentiis M, Fumagalli L, Locatelli MA, Minchella I, Santangelo M, De Placido S, Goldhirsch A, Curigliano G. Dendritic cell-based vaccines: clinical applications in breast cancer. Immunotherapy 2015; 6:349-60. [PMID: 24762078 DOI: 10.2217/imt.13.169] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Recent evidence suggests that the immune system is involved in the carcinogenesis process and the antitumor immune responses impact the clinical outcome, thus emphasizing the concept of cancer immune surveillance. In this context, dendritic cells (DCs) seem to play a crucial role, as they are the most potent antigen-presenting cells (APCs) and are able to stimulate naive T lymphocytes and to generate memory T lymphocytes. Immunotherapy with DC-based vaccines is a very attractive approach to treat cancer, offering the potential for high tumor-specific cytotoxicity. Although breast cancer (BC) is traditionally considered a poorly immunogenic tumor, increasing numbers of both preclinical and clinical studies demonstrate that vaccination with DCs is capable of inducing an antitumor-specific response, while being well tolerated and safe. However, clinical objective responses are still disappointing and many reasons may explain the difficulty of developing effective DC-based therapies for BC. In this review, we discuss the characteristics of DCs, and the major clinical indications for DC-based immunotherapy in BC with related drawbacks.
Collapse
Affiliation(s)
- Lucia Gelao
- Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Yang YF, Xue SY, Lu ZZ, Xiao FJ, Yin Y, Zhang QW, Wu CT, Wang H, Wang LS. Antitumor effects of oncolytic adenovirus armed with PSA-IZ-CD40L fusion gene against prostate cancer. Gene Ther 2014; 21:723-31. [DOI: 10.1038/gt.2014.46] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 03/25/2014] [Accepted: 04/09/2014] [Indexed: 11/10/2022]
|
26
|
Pyzer AR, Avigan DE, Rosenblatt J. Clinical trials of dendritic cell-based cancer vaccines in hematologic malignancies. Hum Vaccin Immunother 2014; 10:3125-31. [PMID: 25625926 PMCID: PMC4514037 DOI: 10.4161/21645515.2014.982993] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/17/2014] [Accepted: 10/05/2014] [Indexed: 11/19/2022] Open
Abstract
The potential for the immune system to target hematological malignancies is demonstrated in the allogeneic transplant setting, where durable responses can be achieved. However, allogeneic transplantation is associated with significant morbidity and mortality related to graft versus host disease. Cancer immunotherapy has the capacity to direct a specific cytotoxic immune response against cancer cells, particularly residual cancer cells, in order to reduce the likelihood of disease relapse in a more targeted and tolerated manner. Ex vivo dendritic cells can be primed in various ways to present tumor associated antigen to the immune system, in the context of co-stimulatory molecules, eliciting a tumor specific cytotoxic response in patients. Several approaches to prime dendritic cells and overcome the immunosuppressive microenvironment have been evaluated in pre-clinical and early clinical trials with promising results. In this review, we summarize the clinical data evaluating dendritic cell based vaccines for the treatment of hematological malignancies.
Collapse
Key Words
- AML, Acute Myeloid Leukemia
- ASCT, Autologous Stem Cell Transplant
- Apo-DC, Apoptotic body loaded- dendritic cells
- CML, Chronic Myeloid Leukemia
- CR, Complete response
- CTLA-4, Cytotoxic T-Lymphocyte Antigen 4
- DC/AML, Dendritic cell Acute Myeloid Leukemia fusion vaccine
- DC/MM, Dendritic cell Multiple Myeloma fusion vaccine
- DNA Deoxyribonucleic acid
- FLT-ITD, Fms-like Tyrosine Kinase with Internal Tandem Duplication
- GMCSF, Granulocyte macrophage colony-stimulating factor
- GVHD, Graft vs Host Disease
- HLA-A*2402, Human Leukocyte antigen A*2402
- IFN, Interferon
- IFNg, Interferon gamma
- IL, Interleukin
- Id, Idiotype
- KLH, Keyhole limpet hemocyanin
- MDS, Myelodysplastic syndrome
- MHC, Major histocompatibility complex
- OS, Overall Survival
- PD-1, Programmed death 1
- PD-L1, Programmed death-ligand 1
- PR, Partial response
- PRR, Pathogen recognition receptor
- RNA, Ribonucleic acid
- SCT, Stem cell transplant
- TGFB, Transforming growth factor β
- TNFα, Tumor necrosis factor α
- VEGF, Vascular endothelial growth factor
- VGPR, Very good partial response
- WT-1, Wilm's tumor suppressor gene 1
- cancer
- dendritic cell
- immunotherapy
- leukemia
- mRNA, mRNA
- myeloma
- pDCs, Plasmacytoid Dendritic cell
- trial
- vaccine
Collapse
Affiliation(s)
- Athalia R Pyzer
- Beth Israel Deaconess Medical Center; Harvard Medical School; Boston, MA USA
| | - David E Avigan
- Beth Israel Deaconess Medical Center; Harvard Medical School; Boston, MA USA
| | - Jacalyn Rosenblatt
- Beth Israel Deaconess Medical Center; Harvard Medical School; Boston, MA USA
| |
Collapse
|
27
|
Gottschalk S, Yu F, Ji M, Kakarla S, Song XT. A vaccine that co-targets tumor cells and cancer associated fibroblasts results in enhanced antitumor activity by inducing antigen spreading. PLoS One 2013; 8:e82658. [PMID: 24349329 PMCID: PMC3861387 DOI: 10.1371/journal.pone.0082658] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/25/2013] [Indexed: 11/18/2022] Open
Abstract
Dendritic cell (DC) vaccines targeting only cancer cells have produced limited antitumor activity in most clinical studies. Targeting cancer-associated fibroblasts (CAFs) in addition to cancer cells may enhance antitumor effects, since CAFs, the central component of the tumor stroma, directly support tumor growth and contribute to the immunosuppressive tumor microenvironment. To co-target CAFs and tumor cells we developed a new compound DC vaccine that encodes an A20-specific shRNA to enhance DC function, and targets fibroblast activation protein (FAP) expressed in CAFs and the tumor antigen tyrosine-related protein (TRP)2 (DC-shA20-FAP-TRP2). DC-shA20-FAP-TRP2 vaccination induced robust FAP- and TRP2-specific T-cell responses, resulting in greater antitumor activity in the B16 melanoma model in comparison to monovalent vaccines or a vaccine encoding antigens and a control shRNA. DC-shA20-FAP-TRP2 vaccination enhanced tumor infiltration of CD8-positive T cells, and induced antigen-spreading resulting in potent antitumor activity. Thus, co-targeting of tumor cells and CAFs results in the induction of broad-based tumor-specific T-cell responses and has the potential to improve current vaccine approaches for cancer.
Collapse
Affiliation(s)
- Stephen Gottschalk
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (XTS); (SG)
| | - Feng Yu
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Minjun Ji
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sunitha Kakarla
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, United States of America
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Xiao-Tong Song
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (XTS); (SG)
| |
Collapse
|
28
|
Orsini G, Legitimo A, Failli A, Ferrari P, Nicolini A, Spisni R, Miccoli P, Consolini R. Defective generation and maturation of dendritic cells from monocytes in colorectal cancer patients during the course of disease. Int J Mol Sci 2013; 14:22022-41. [PMID: 24213603 PMCID: PMC3856049 DOI: 10.3390/ijms141122022] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/09/2013] [Accepted: 10/17/2013] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the second-leading cause of cancer-related deaths in Western countries. Today, the role of the host’s immune system in controlling the progression and spread of solid tumors is broadly established. Tumor immunosurveillance escape mechanisms, such as those involving dendritic cells (DCs), the most important antigen-presenting cells, are likewise recognized processes involved in cancer. The present study evaluates the ability of CRC patients to generate DCs in vitro from circulating monocytes at both pre- and post-operative timepoints; the results are correlated with the stage of disease to shed light on the systemic immune statuses of CRC patients. Our data showed that patients’ DCs had lower co-stimulatory molecule expression and were less able to present antigens to allogeneic T cells compared to healthy controls’ (HC) DCs. Furthermore altered cytokine secretion, such as increased IL-10 and reduced IL-12 and TNF-α, was observed. At the post-operative timepoints we observed a recovery of the patients’ ability to generate immature DCs, compared to HCs, but the maturational capacity remained affected. Our study conclusively highlights the persistently impaired in vitro generation of fully mature and functional DCs, which appears to be more altered during advanced stages. This work sheds light on a dendritic cell-based tumor immune escape mechanism that could be useful for the development of more effective immunotherapeutic strategies.
Collapse
Affiliation(s)
- Giulia Orsini
- Laboratory of Immunology, Department of Clinical and Experimental Medicine, University of Pisa, via Roma, 67, Pisa 56126, Italy; E-Mails: (A.L.); (A.F.); (R.C.)
- Author to whom correspondence should be addressed; E-Mail: or ; Tel.: +39-050-992-222
| | - Annalisa Legitimo
- Laboratory of Immunology, Department of Clinical and Experimental Medicine, University of Pisa, via Roma, 67, Pisa 56126, Italy; E-Mails: (A.L.); (A.F.); (R.C.)
| | - Alessandra Failli
- Laboratory of Immunology, Department of Clinical and Experimental Medicine, University of Pisa, via Roma, 67, Pisa 56126, Italy; E-Mails: (A.L.); (A.F.); (R.C.)
| | - Paola Ferrari
- Department of Clinical and Experimental Medicine, Azienda Ospedaliero-Universitaria Pisana (AOUP), via Roma, 67, Pisa 56126, Italy; E-Mail:
| | - Andrea Nicolini
- Department of Clinical and Experimental Medicine, Section Medical Oncology, University of Pisa, via Roma, 67, Pisa 56126, Italy; E-Mail:
| | - Roberto Spisni
- Department of Surgery, Molecular, Medical and Critical Area Pathology, University of Pisa, via Paradisa, 2, Pisa 56126, Italy; E-Mails: (R.S.) (P.M.)
| | - Paolo Miccoli
- Department of Surgery, Molecular, Medical and Critical Area Pathology, University of Pisa, via Paradisa, 2, Pisa 56126, Italy; E-Mails: (R.S.) (P.M.)
| | - Rita Consolini
- Laboratory of Immunology, Department of Clinical and Experimental Medicine, University of Pisa, via Roma, 67, Pisa 56126, Italy; E-Mails: (A.L.); (A.F.); (R.C.)
| |
Collapse
|
29
|
Hunyadi J, András C, Szabó I, Szántó J, Szluha K, Sipka S, Kovács P, Kiss A, Szegedi G, Altorjay I, Sápy P, Antal-Szalmás P, Tóth L, Fazekas G, Rajnavölgyi É. Autologous dendritic cell based adoptive immunotherapy of patients with colorectal cancer-A phase I-II study. Pathol Oncol Res 2013; 20:357-65. [PMID: 24163303 DOI: 10.1007/s12253-013-9704-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 10/02/2013] [Indexed: 12/17/2022]
Abstract
Dendritic cell-based active immunotherapies of cancer patients are aimed to provoke the proliferation and differentiation of tumor-specific CD4(+) and CD8(+) T-lymphocytes towards protective effector cells. Isolation and in vitro differentiation of circulating blood monocytes has been established a reasonable platform for adoptively transferred DC-based immunotherapies. In the present study the safety and tolerability of vaccination by autologous tumor cell lysates (oncolysate)- or carcinoembriogenic antigen (CEA)-loaded DCs in patients with colorectal cancer was investigated in a phase I-II trial. The study included 12 patients with histologically confirmed colorectal cancer (Dukes B2-C stages). Six of the patients received oncolysate-pulsed, whereas the other six received recombinant CEA-loaded autologous DCs. The potential of the tumor antigen-loaded DCs to provoke the patient's immune system was studied both in vivo and in vitro. The clinical outcome of the therapy evaluated after 7 years revealed that none of the six patients treated with oncolysate-loaded DCs showed relapse of colorectal cancer, whereas three out of the six patients treated with CEA-loaded DCs died because of tumor relapse. Immunization with both the oncolysate- and the CEA-loaded autologous DCs induced measurable immune responses, which could be detected in vivo by cutaneous reactions and in vitro by lymphocyte proliferation assay. Our results show that vaccination by autologous DCs loaded with autologous oncolysates containing various tumor antigens represents a well tolerated therapeutic modality in patients with colorectal cancer without any detectable adverse effects. Demonstration of the efficacy of such therapy needs further studies with increased number of patients.
Collapse
Affiliation(s)
- János Hunyadi
- Clinical Centre for Cell Therapy, Medical and Health Science Center, University of Debrecen, Nagyerdei krt. 98, 4032, Debrecen, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Gene therapy as a treatment for cancer is regarded as high in promise, but low in delivery, a deficiency that has become more obvious with ever-increasing reports of the successful correction of monogenic disorders by this approach. We review the commercial and scientific obstacles that have led to these delays and describe how they are progressively being overcome. Recent and striking successes and correspondingly increased commercial involvement suggest that gene transfer could finally become a powerful method for development of safe and effective cancer therapeutic drugs.
Collapse
Affiliation(s)
- Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, TX, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Stephen Gottschalk
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, TX, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Ann M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, TX, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Juan F Vera
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
31
|
Quantification of blood dendritic cells in colorectal cancer patients during the course of disease. Pathol Oncol Res 2013; 20:267-76. [PMID: 24022399 DOI: 10.1007/s12253-013-9691-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 08/30/2013] [Indexed: 01/07/2023]
Abstract
Colorectal cancer is a malignancy with poor prognosis that might be associated with defective immune function. The aim of the present study was to investigate circulating dendritic cells in colorectal cancer patients, in order to contribute to elucidate tumor-escape mechanisms and to point out a possible correlation with the clinical condition of the disease. Therefore, we enumerated ex vivo myeloid and plasmacytoid dendritic cells, through multicolor flow cytometry, in 26 colorectal patients and 33 healthy controls. Furthermore we performed several analyses at determined time points in order to define the immunological trend of cancer patients after surgery and other conventional treatments. At the pre-operative time point the absolute number of plasmacytoid dendritic cells in cancer patients was significantly reduced in comparison to controls, this result being mainly referred to stage III-IV patients. The number of myeloid dendritic cells did not show any significant difference compared to healthy controls; interestingly the expression of the tolerogenic antigen CD85k was significantly higher on cancer patients' myeloid dendritic cells than controls'. At the following samplings, circulating dendritic cell absolute number did not show any difference compared to controls. Conclusively the impairment of the number of circulating dendritic cells may represent one of the tumor escape mechanisms occurring in colorectal cancer. These alterations seem to be correlated to cancer progression. Our work sheds light on one of dendritic cell-based tumor immune escape mechanisms. This knowledge may be useful to the development of more effective immunotherapeutic strategies.
Collapse
|
32
|
Davison GM, Novitzky N, Abdulla R. Monocyte derived dendritic cells have reduced expression of co-stimulatory molecules but are able to stimulate autologous T-cells in patients with MDS. Hematol Oncol Stem Cell Ther 2013; 6:49-57. [DOI: 10.1016/j.hemonc.2013.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2013] [Indexed: 01/21/2023] Open
|
33
|
Flörcken A, Kopp J, van Lessen A, Movassaghi K, Takvorian A, Jöhrens K, Möbs M, Schönemann C, Sawitzki B, Egerer K, Dörken B, Pezzutto A, Westermann J. Allogeneic partially HLA-matched dendritic cells pulsed with autologous tumor cell lysate as a vaccine in metastatic renal cell cancer: a clinical phase I/II study. Hum Vaccin Immunother 2013; 9:1217-27. [PMID: 23458999 DOI: 10.4161/hv.24149] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Multi-kinase inhibitors have been established for the treatment of advanced renal cell cancer, but long-term results are still disappointing and immunotherapeutic approaches remain an interesting experimental option particularly in patients with a low tumor burden. DC are crucial for antigen-specific MHC-restricted T cell immunity. Furthermore, allogeneic HLA-molecules pose a strong immunogenic signal and may help to induce tumor-specific T cell responses. In this phase I/II trial, 7 patients with histologically confirmed progressive metastatic RCC were immunized repetitively with 1 × 10 (7) allogeneic partially HLA-matched DC pulsed with autologous tumor lysate following a schedule of 8 vaccinations over 20 weeks. Patients also received 3 Mio IE IL-2 s.c. once daily starting in week 4. Primary endpoints of the study were feasibility and safety. Secondary endpoints were immunological and clinical responses. Vaccination was feasible and safe with no severe toxicity being observed. No objective response could be documented. However, while all patients had documented progress at study entry, 29% of the patients showed SD throughout the study with a mean TTP of 24.6 weeks (range 5 to 96 weeks). In 3/7 patients, TH1-polarized immune responses against RCC-associated antigens were observed. In one patient showing a minimal clinical response and a TTP of 96 weeks, clonally proliferated T cells against yet undefined antigens were induced by the vaccine. Vaccination with tumor antigen loaded DC remains an interesting experimental approach, but should rather be applied in the situation of minimal residual disease after systemic therapy. Additional depletion of regulatory cells might be a promising strategy.
Collapse
Affiliation(s)
- Anne Flörcken
- Department of Hematology; Oncology, and Tumor Immunology; Charité University Medicine Berlin; Campus Virchow- Klinikum; Berlin, Germany; Department of Hematology; Oncology, and Tumor Immunology; Charité University Medicine Berlin; Campus Benjamin Franklin; Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Dillman RO, Cornforth AN, Nistor G. Cancer stem cell antigen-based vaccines: the preferred strategy for active specific immunotherapy of metastatic melanoma? Expert Opin Biol Ther 2013; 13:643-56. [PMID: 23451922 DOI: 10.1517/14712598.2013.759556] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION There are now two chemotherapy agents, one tyrosine kinase inhibitor and three immunotherapy products approved for the treatment of metastatic melanoma, but an unmet need persists because these options are toxic and of limited therapeutic benefit. Active specific immunotherapy with therapeutic vaccines could be a useful addition to the therapeutic armamentarium, especially in patients whose tumor burden has been reduced by other treatment modalities. AREAS COVERED This article reviews various sources of melanoma antigens, such as peptides, gangliosides, autologous tumor and cancer stem cells including allogeneic and autologous cell lines. The advantages and disadvantages of various antigen sources and allogeneic and autologous approaches are discussed with an emphasis on the theoretical benefits of immunizing against cancer stem cells. The results from published randomized trials testing the benefit of various vaccine approaches are summarized, as well as promising results from three Phase II trials (one randomized) of patient-specific stem cell antigen-based products. EXPERT OPINION Immune responses directed toward the unique neoantigens and stem cell antigens expressed on continuously proliferating, self-renewing, autologous tumor cells could potentially overcome the limitations inherent in these other antigen-based approaches, that to date, have yielded disappointing results in randomized trials.
Collapse
Affiliation(s)
- Robert O Dillman
- Hoag Institute for Research and Education, Hoag Hospital, One Hoag Dr, Bldg 44 Suite 210, Newport Beach, California 92663, USA.
| | | | | |
Collapse
|
35
|
Lundberg K, Albrekt AS, Nelissen I, Santegoets S, de Gruijl TD, Gibbs S, Lindstedt M. Transcriptional profiling of human dendritic cell populations and models--unique profiles of in vitro dendritic cells and implications on functionality and applicability. PLoS One 2013; 8:e52875. [PMID: 23341914 PMCID: PMC3544800 DOI: 10.1371/journal.pone.0052875] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 11/22/2012] [Indexed: 01/12/2023] Open
Abstract
Background Dendritic cells (DCs) comprise heterogeneous populations of cells, which act as central orchestrators of the immune response. Applicability of primary DCs is restricted due to their scarcity and therefore DC models are commonly employed in DC-based immunotherapy strategies and in vitro tests assessing DC function. However, the interrelationship between the individual in vitro DC models and their relative resemblance to specific primary DC populations remain elusive. Objective To describe and assess functionality and applicability of the available in vitro DC models by using a genome-wide transcriptional approach. Methods Transcriptional profiling was performed with four commonly used in vitro DC models (MUTZ-3-DCs, monocyte-derived DCs, CD34-derived DCs and Langerhans cells (LCs)) and nine primary DC populations (dermal DCs, LCs, blood and tonsillar CD123+, CD1c+ and CD141+ DCs, and blood CD16+ DCs). Results Principal Component Analysis showed that transcriptional profiles of each in vitro DC model most closely resembled CD1c+ and CD141+ tonsillar myeloid DCs (mDCs) among primary DC populations. Thus, additional differentiation factors may be required to generate model DCs that more closely resemble other primary DC populations. Also, no model DC stood out in terms of primary DC resemblance. Nevertheless, hierarchical clustering showed clusters of differentially expressed genes among individual DC models as well as primary DC populations. Furthermore, model DCs were shown to differentially express immunologically relevant transcripts and transcriptional signatures identified for each model DC included several immune-associated transcripts. Conclusion The unique transcriptional profiles of in vitro DC models suggest distinct functionality in immune applications. The presented results will aid in the selection of an appropriate DC model for in vitro assays and assist development of DC-based immunotherapy.
Collapse
|
36
|
Youlin K, Li Z, Xin G, Mingchao X, Xiuheng L, Xiaodong W. Enhanced function of cytotoxic T lymphocytes induced by dendritic cells modified with truncated PSMA and 4-1BBL. Hum Vaccin Immunother 2013; 9:766-72. [PMID: 23295983 DOI: 10.4161/hv.23116] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Interactions between costimulatory molecules and their receptors are vital for Ag-presenting dendritic cells (DCs) to initiate T cells activation, expansion and their antitumor immune responses. Augmentation of costimulatory signal due to the interaction of DCs and T cells may amplify, sustain and drive diversity of cytotoxic T lymphocytes (CTLs) and consequently enhance the antitumor response. 4-1BBL/4-1BB is such a pair of costimulatory ligand and receptor, playing an important role in the co-stimulation of CTLs. Previously, we demonstrated that DCs transduced with recombinant adenovirus encoding truncated PSMA (tPSMA) and m4-1BBL could induce prostate cancer regression in mouse models. In the present study, we further explored the adjuvant role of 4-1BBL in modulating CTLs activation induced by tPSMA gene-pulsed DCs. The apoptosis and cytotoxicity against tPSMA expressing RM-1 cells of CTLs were determined. Results showed that tPSMA gene-pulsed DCs effectively induced T lymphocyte activation and cytotoxicity, which was enhanced by upregulated expression of 4-1BBL, displaying better cell viability, lower CTLs apoptosis, higher expression anti-apoptotic protein of Bcl-xL and phosphorylation of P38, enhanced NF-κB activation, as well as more IFN-γ production. These results demonstrated that 4-1BBL may play a significant role in the co-stimulation pathway for Ag-presenting DCs-mediated CTLs activity, which might be a beneficial adjuvant factor for DCs-based cancer immunotherapy.
Collapse
Affiliation(s)
- Kuang Youlin
- Department of Urology; The First Affiliated Hospital; Chongqing Medical University; Chongqing, P.R. China
| | | | | | | | | | | |
Collapse
|
37
|
Youlin K, Jianwei Z, Xin G, Li Z, Xiaodong W, Xiuheng L, Hengchen Z, Zhiyuan C. 4-1BB protects dendritic cells from prostate cancer-induced apoptosis. Pathol Oncol Res 2012; 19:177-81. [PMID: 23070584 DOI: 10.1007/s12253-012-9566-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 08/15/2012] [Indexed: 10/27/2022]
Abstract
It has been shown that human prostate cancer (PCa) cells induced apoptotic death of the most potent antigen-presenting cells, dendritic cells (DCs), which are responsible for the induction of specific antitumor immune responses. Here, we investigated the function of 4-1BB on protecting DCs from prostate cancer-induced apoptosis with an agonistic mAb to 4-1BB. RM-1 cells and DCs were co-incubated for 48 h and DC apoptosis was assessed by Annexin Vassay. TNF-α and IL-12 production were assessed by enzyme-linked immunosorbent assay (ELISA) and Bcl-2 and Bcl-xL on DCs were analyzed by Western blot. We have shown that co-incubation of RM-1 cells with DCs is accompanied by an increased level of DCs apoptosis. Triggering 4-1BB on DCs resulted in increased resistance of DCs to RM-1 cells-induced apoptosis, which was owing to the up-regulated expression of Bcl-2 and Bcl-xL, and increased secretion of TNF-αand IL-12. These results demonstrate that triggering 4-1BB on DCs could increased resistance of DCs to PCa-induced apoptosis.
Collapse
Affiliation(s)
- Kuang Youlin
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Martorelli D, Coppotelli G, Muraro E, Dolcetti R, Masucci MG. Remodeling of the epitope repertoire of a candidate idiotype vaccine by targeting to lysosomal degradation in dendritic cells. Cancer Immunol Immunother 2012; 61:881-92. [PMID: 22089857 PMCID: PMC11028998 DOI: 10.1007/s00262-011-1157-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Accepted: 11/03/2011] [Indexed: 02/05/2023]
Abstract
The generation of efficacious vaccines against self-antigens expressed in tumor cells requires breakage of tolerance, and the refocusing of immune responses toward epitopes for which tolerance may not be established. While the presentation of tumor antigens by mature dendritic cells (mDC) may surpass tolerance, broadening of the antigenic repertoire remains an issue. We report that fusion of the candidate idiotype vaccine IGKV3-20 to the Gly-Ala repeat (GAr) of the Epstein-Barr virus nuclear antigen (EBNA)-1 inhibits degradation by the proteasome and redirects processing to the lysosome. mDCs transduced with a recombinant lentivirus encoding the chimeric idiotype efficiently primed CD4+ and CD8+ cytotoxic T-cell (CTL) responses that lysed autologous blasts expressing IGKV3-20 or pulsed with IGKV3-20 synthetic peptides, and HLA-matched IGKV3-20-positive tumor cell lines. Comparison of the cytotoxic response of CD4+ and CD8+ T lymphocytes activated by mDCs expressing the wild-type or chimeric IGKV3-20 reveled largely non-overlapping epitope repertoires in both CD4+ and CD8+ effectors. Thus, fusion to the GAr may provide an effective means to broaden the immune response to an endogenous protein by promoting the presentation of antigenic epitopes that require a lysosome-dependent processing step.
Collapse
Affiliation(s)
- Debora Martorelli
- Cancer Bio-immunotherapy Unit, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Giuseppe Coppotelli
- Department of Cell and Molecular Biology, Karolinska Institute, Box 285, SE-171 77 Stockholm, Sweden
| | - Elena Muraro
- Cancer Bio-immunotherapy Unit, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Riccardo Dolcetti
- Cancer Bio-immunotherapy Unit, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Maria G. Masucci
- Department of Cell and Molecular Biology, Karolinska Institute, Box 285, SE-171 77 Stockholm, Sweden
| |
Collapse
|
39
|
Xu C, Mu L, Roes I, Miranda-Nieves D, Nahrendorf M, Ankrum JA, Zhao W, Karp JM. Nanoparticle-based monitoring of cell therapy. NANOTECHNOLOGY 2011; 22:494001. [PMID: 22101191 PMCID: PMC3334527 DOI: 10.1088/0957-4484/22/49/494001] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Exogenous cell therapy aims to replace/repair diseased or dysfunctional cells and promises to revolutionize medicine by restoring tissue and organ function. To develop effective cell therapy, the location, distribution and long-term persistence of transplanted cells must be evaluated. Nanoparticle (NP) based imaging technologies have the potential to track transplanted cells non-invasively. Here we summarize the most recent advances in NP-based cell tracking with emphasis on (1) the design criteria for cell tracking NPs, (2) protocols for cell labeling, (3) a comparison of available imaging modalities and their corresponding contrast agents, (4) a summary of preclinical studies on NP-based cell tracking and finally (5) perspectives and future directions.
Collapse
Affiliation(s)
- Chenjie Xu
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Luye Mu
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Isaac Roes
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - David Miranda-Nieves
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| | - James A Ankrum
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Weian Zhao
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Jeffrey M Karp
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA
| |
Collapse
|
40
|
Immunotherapy of lung adenocarcinoma patient with Peptide-pulsed dendritic cells: a case report. Arch Immunol Ther Exp (Warsz) 2011; 60:69-77. [PMID: 22143160 DOI: 10.1007/s00005-011-0157-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 07/14/2011] [Indexed: 10/15/2022]
Abstract
Immunotherapy with ex vivo generated dendritic cells (DCs) is reported to be of low toxicity and of diverse effectiveness in cancer treatment. The synthetic antigens are frequently used for immunotherapy especially for patients with stable disease after prior treatment. We described the effect of peptide-loaded DCs-based immunotherapy on patient with recurrent surgically resected adenocarcinoma with bronchoalveolar feature with co-existing of Takayasu arteritis and chronic hepatitis B. In January 2010, 61-year-old patient received subcutaneously four bi-weekly vaccinations of DCs loaded with MUC1 and MAGE-3 epitopes. Additionally, he received three bi-weekly booster vaccinations after 7 months from the first course of immunotherapy. Delayed-type hypersensitivity test was positive only for MAGE-3 antigen. The evidence expansion of MAGE-3-specific CD8(+) cells after first vaccination and after third vaccination during boosters injections was observed (from 0.08% before vaccination to 0.5% after first vaccination; from 0.05% before booster vaccination to 0.24% after third injection). Computed tomography scans performed after first course and after booster course of vaccination until April 2011 did not shown any presence of lung tumour or metastases. Based on clinical factors (no completed wedge-resection and recurrent character of cancer) as well as on the presence of the tumour-antigen-specific immunological response, we could speculated that immunotherapy prolonged disease free-survival in our patient. Over 16 months from first vaccination, the patient remains without symptoms of cancer.
Collapse
|
41
|
Kuang Y, Weng X, Liu X, Zhu H, Chen Z, Chen H. Effects of 4-1BB signaling on the biological function of murine dendritic cells. Oncol Lett 2011; 3:477-481. [PMID: 22740935 DOI: 10.3892/ol.2011.506] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 11/14/2011] [Indexed: 11/06/2022] Open
Abstract
4-1BB signaling has profound effects on the T cell-induced cell immune response, but its biological function in dendritic cells (DCs) has remained largely uncharacterized. In this study, we investigated the function of 4-1BB in murine DCs with an agonistic mAb to 4-1BB. Interleukin (IL)-6 and IL-12 production was assessed by an enzyme-linked immunosorbent assay (ELISA). Co-stimulatory molecules (CD80 and CD86) in DCs were analyzed by flow cytometry. The results showed that 4-1BB was strongly expressed in DCs during the maturation process. Triggering 4-1BB increased the secretion of IL-6 and IL-12 and the upregulation of co-stimulatory molecules (CD80 and CD86) from DCs, indicating that agonistic mAb to 4-1BB directly improves the activation of DCs. Moreover, triggering 4-1BB induced a higher survival rate of DCs compared to that of hamster IgG isotype control, due to the upregulated expression of Bcl-2 and Bcl-xL. To further assess the role of 4-1BB on DCs stimulating T-cell proliferation, allogeneic mixed lymphocyte reactions were analyzed. The agonistic anti-4-1BB mAb induced a higher T-cell proliferation. These results suggest that 4-1BB affects the duration, DC-T interaction and immunogenicity of DCs.
Collapse
Affiliation(s)
- Youlin Kuang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China
| | | | | | | | | | | |
Collapse
|
42
|
Di Caro V, D'Anneo A, Phillips B, Engman C, Harnaha J, Trucco M, Giannoukakis N. Phosphatidylinositol-3-kinase activity during in vitro dendritic cell generation determines suppressive or stimulatory capacity. Immunol Res 2011; 50:130-52. [PMID: 21476100 DOI: 10.1007/s12026-011-8206-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Modulating PI3K at different stages of dendritic cells (DC) generation could be a novel means to balance the generation of immunosuppressive versus immunostimulatory DC. We show that PI3K inhibition during mouse DC generation in vitro results in cells that are potently immunosuppressive and characteristic of CD8alpha- CD11c+ CD11b+ DC. These DC exhibited low surface class I and class II MHC, CD40, and CD86 and did not produce TNF-alpha. In allogeneic MLR, these DC were suppressive. Although in these mixed cultures, there was no increase in the frequency of CD4+ CD25+ Foxp3+ cells, the Foxp3 content on a per cell basis was significantly increased. Sustained TLR9 signaling in the presence of PI3K inhibition during DC generation overrode the cells' suppressive phenotype.
Collapse
Affiliation(s)
- Valentina Di Caro
- Department of Pediatrics, Division of Immunogenetics, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Chia WK, Wang WW, Teo M, Tai WM, Lim WT, Tan EH, Leong SS, Sun L, Chen JJ, Gottschalk S, Toh HC. A phase II study evaluating the safety and efficacy of an adenovirus-ΔLMP1-LMP2 transduced dendritic cell vaccine in patients with advanced metastatic nasopharyngeal carcinoma. Ann Oncol 2011; 23:997-1005. [PMID: 21821548 DOI: 10.1093/annonc/mdr341] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Individuals with metastatic Epstein-Barr virus (EBV)-positive nasopharyngeal carcinoma (NPC) continue to have poor outcomes. To evaluate the ability of a dendritic cell (DC) vaccine to target subdominant EBV antigens LMP1 and LMP2 expressed by NPC cells, we vaccinated patients using autologous DCs transduced with an adenovirus encoding a truncated LMP1 (ΔLMP1) and full-length LMP2 (Ad-ΔLMP1-LMP2). MATERIALS AND METHODS Sixteen subjects with metastatic NPC received Ad-ΔLMP1-LMP2 DC vaccines i.d. biweekly for up to five doses. Toxicity, immune responses and clinical responses were determined. RESULTS Most patients had extensive disease, with a median of three visceral sites of involvement (range 1-7). No significant toxicity was observed. Ad-ΔLMP1-LMP2 DCs induced delayed type hypersensitivity responses in 9 out of 12 patients, but although these DCs activated LMP1/2-specific T cells in vitro, no such increase in the frequency of peripheral LMP1/2-specific T cells was detected. Three patients had clinical responses including one with partial response (for 7½ months) and two with stable disease (for 6½ and 7½ months). CONCLUSIONS Ad-ΔLMP1-LMP2 transduced DCs can be successfully generated and safely administered to patients with advanced NPC. Since efficacy was limited, future studies should focus on DC vaccines with greater potency administered to subjects with less tumor burden.
Collapse
Affiliation(s)
- W K Chia
- Department of Medical Oncology, National Cancer Centre, Singapore
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Shurin MR, Gregory M, Morris JC, Malyguine AM. Genetically modified dendritic cells in cancer immunotherapy: a better tomorrow? Expert Opin Biol Ther 2011; 10:1539-53. [PMID: 20955111 DOI: 10.1517/14712598.2010.526105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
IMPORTANCE OF THE FIELD Dendritic cells (DC) are powerful antigen-presenting cells that induce and maintain primary cytotoxic T lymphocyte (CTL) responses directed against tumor antigens. Consequently, there has been much interest in their application as antitumor vaccines. AREAS COVERED IN THIS REVIEW A large number of DC-based vaccine trials targeting a variety of cancers have been conducted; however, the rate of reported clinically significant responses remains low. Modification of DC to express tumor antigens or immunostimulatory molecules through the transfer of genes or mRNA transfection offers a logical alternative with potential advantages over peptide- or protein antigen-loaded DC. In this article, we review the current results and future prospects for genetically modified DC vaccines for the treatment of cancer. WHAT THE READER WILL GAIN Genetically-modified dendritic cell-based vaccines represent a powerful tool for cancer therapy. Numerous preclinical and clinical studies have demonstrated the potential of dendritic cell vaccines alone or in combination with other therapeutic modalities. TAKE HOME MESSAGE Genetically modified DC-based anti-cancer vaccination holds promise, perhaps being best employed in the adjuvant setting with minimal residual disease after primary therapy, or in combination with other antitumor or immune-enhancing therapies.
Collapse
Affiliation(s)
- Michael R Shurin
- Department of Pathology and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
45
|
Abstract
Explorative knowledge of cellular and molecular mechanisms of immune function and regulation has provided optimism in developing cancer immunotherapy. However, three decades of experimental and clinical investigations to offer powerful immunotherapeutic strategies against solid tumors, with the possible exception of monoclonal antibody-targeted therapies, have not succeeded in significantly prolonging patient survival. Nonspecific immune approaches, including cytokine-based therapies and allogeneic hematopoietic stem cell transplantation, have so far produced consistent, although limited, results. In this review, we present the developments of cell transfer-based strategies that, in preclinical studies, have demonstrated potential efficacy, but have only established tumor regression in limited numbers of patients. The key to success demands creative combinations of tumor antigens, adjuvance, gene modification and various administration strategies in the development of cell-based therapies together with other cancer-treatment principles, often in a stepwise 'space-rocket-type' approach. Combined efforts of several scientific disciplines, such as tumor biology and immunology, as well as cell and gene research in transplantation, will open new venues. New regulation for clinical trials with advanced therapy medicine products to ensure patient safety will be highlighted.
Collapse
Affiliation(s)
- Lisbeth Barkholt
- Division of Clinical Immunology & Transfusion Medicine, Department of Laboratory Medicine, Karolinska University Hospital Huddinge F79, Stockholm, Sweden.
| | | |
Collapse
|
46
|
Weng X, Kuang Y, Liu X, Chen Z, Zhu H, Chen H, Jiang B, Shen H. Construction of a recombinant adenovirus co-expressing truncated human prostate-specific membrane antigen and mouse 4-1BBL genes and its effect on dendritic cells. Braz J Med Biol Res 2011; 44:186-92. [PMID: 21243315 DOI: 10.1590/s0100-879x2011007500002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 12/17/2010] [Indexed: 11/21/2022] Open
Abstract
Our aim was to construct a recombinant adenovirus co-expressing truncated human prostate-specific membrane antigen (tPSMA) and mouse 4-1BBL genes and to determine its effect on dendritic cells (DCs) generated from bone marrow suspensions harvested from C57BL/6 mice for which the effect of 4-1BBL on DCs is not clear, especially during DCs processing tumor-associated antigen. Replication deficient adenovirus AdMax™ Expression System was used to construct recombinant adenovirus Ad-tPSMA-internal ribosome entry site-mouse 4-1BBL (Ad-tPSMA-IRES-m4-1BBL) and Ad-enhanced green fluorescent protein. Day 7 proliferating DC aggregates generated from C57BL/6 mice were collected as immature DCs and further mature DCs were obtained by lipopolysaccharide activated immature DCs. After DCs were exposed to the recombinant adenovirus with 250 multiplicity of infection, the expression of tPSMA and m4-1BBL proteins were detected by Western blot, and the apoptosis and phenotype of DCs were analyzed by flow cytometry. Cytokines (IL-6 and IL-12) in the supernatant were detected by enzyme-linked immunosorbent assay (ELISA). Proliferation of T cells was detected by allogeneic mixed lymphocyte reactions. The tPSMA and m4-1BBL proteins were expressed correctly. The apoptosis rate of DCs transfected with Ad-tPSMA-IRES-m4-1BBL was 14.6%, lower than that of control DCs. The expression of co-stimulatory molecules [CD80 (81.6 ± 5.4%) and CD86 (80.13 ± 2.81%)] up-regulated in Ad-tPSMA-IRES-m4-1BBL-pulsed DCs, and the level of IL-6 (3960.2 ± 50.54 pg/mL) and IL-12 (249.57 ± 12.51 pg/mL) production in Ad-tPSMA-IRES-m4-1BBL-transduced DCs were significantly higher (P < 0.05) than those in control DCs. Ad-tPSMA-IRES-m4-1BBL induced higher T-cell proliferation (OD(450) = 0.614 ± 0.018), indicating that this recombinant adenovirus can effectively enhance the activity of DCs.
Collapse
Affiliation(s)
- Xiaodong Weng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Song XT, Turnis ME, Zhou X, Zhu W, Hong BX, Rollins L, Rabinovich B, Chen SY, Rooney CM, Gottschalk S. A Th1-inducing adenoviral vaccine for boosting adoptively transferred T cells. Mol Ther 2011; 19:211-7. [PMID: 20959814 PMCID: PMC3017450 DOI: 10.1038/mt.2010.223] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 09/20/2010] [Indexed: 01/08/2023] Open
Abstract
Although the benefits of adoptive T-cell therapy can be increased by prior lymphodepletion of the recipient, this process usually requires chemotherapy or radiation. Vaccination with antigens to which the transferred T cells respond should be a less toxic means of enhancing their activity, but to date such vaccines have not been effective. We, therefore, determined which characteristics an adenoviral vaccine has to fulfill to optimally activate and expand adoptively transferred antigen-specific T cells in vivo. We evaluated (i) antigen, (ii) flagellin, a Toll-like receptor (TLR) 5 ligand, and (iii) an inhibitor of the antigen-presenting attenuator A20. Vaccination of mice before T-cell transfer with a vaccine that contained all three components dramatically enhanced the effector function of ovalbumin (OVA)-specific T cells as judged by the regression of established B16-OVA tumors compared to one- and two-component vaccines. Immunization with the three-component vaccine induced a strong Th1 environment, which was critical for the observed synergy and proved as effective as cytoxan-induced lymphodepletion in enhancing in vivo T-cell expansion. Thus, the combination of our vaccine with T-cell therapy has the potential to enhance and broaden adoptive cellular immunotherapy.
Collapse
Affiliation(s)
- Xiao-Tong Song
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, The Methodist Hospital, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Pajtasz-Piasecka E, Indrová M. Dendritic cell-based vaccines for the therapy of experimental tumors. Immunotherapy 2010; 2:257-68. [PMID: 20635932 DOI: 10.2217/imt.10.7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Dendritic cells (DCs) are believed to be the most potent antigen-presenting cells able to link the innate and adaptive immune systems. Many studies have focused on different immunotherapeutic approaches to applying DCs as tools to improve anticancer therapy. Although a number of investigations suggesting the benefit of DC-based vaccination during anticancer therapy have been reported, the general knowledge regarding the ultimate methods of DC-vaccine preparation is still unsatisfactory. In this article, the perspectives of DC-based anti-tumor immunotherapy and optimizing strategies of DC vaccination in humans in light of results obtained in mouse models are discussed.
Collapse
Affiliation(s)
- Elzbieta Pajtasz-Piasecka
- Ludwik Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland.
| | | |
Collapse
|
49
|
Abstract
The overexpression and aberrant glycosylation of MUC1 is associated with a wide variety of cancers, making it an ideal target for immunotherapeutic strategies. This review highlights the main avenues of research in this field, focusing on adenocarcinomas, from the preclinical to clinical; the problems and possible solutions associated with each approach; and speculates on the direction of MUC1 immunotherapeutic research over the next 5-10 years.
Collapse
Affiliation(s)
- Richard E Beatson
- Breast Cancer Biology Group, King's College London, Guy's Hospital, London SE1 9RT, UK
| | | | | |
Collapse
|
50
|
Saka M, Amano T, Kajiwara K, Yoshikawa K, Ideguchi M, Nomura S, Fujisawa H, Kato S, Fujii M, Ueno K, Hinoda Y, Suzuki M. Vaccine therapy with dendritic cells transfected with Il13ra2 mRNA for glioma in mice. J Neurosurg 2010; 113:270-9. [PMID: 19895199 DOI: 10.3171/2009.9.jns09708] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The Il13ra2 gene is often overexpressed in brain tumors, making Il13ra2 one of the vaccine targets for immunotherapy of glioma. In this study, using a mouse glioma model, the authors tested the hypothesis that vaccination using dendritic cells transfected with Il13ra2 mRNA induces strong immunological antitumor effects. METHODS A plasmid was constructed for transduction of the mRNAs transcribed in vitro into dendritic cells. This was done to transport the intracellular protein efficiently into major histocompatibility complex class II compartments by adding a late endosomal/lysosomal sorting signal to the Il13ra2 gene. The dendritic cells transfected with this Il13ra2 mRNA were injected intraperitoneally into the mouse glioma model at 3 and 10 days after tumor cell implantation. The antitumor effects were estimated based on the survival rate, results of histological analysis, and immunohistochemical findings for immune cells. RESULTS The group treated by vaccination therapy with dendritic cells transfected with Il13ra2 mRNA survived significantly longer than did the control groups. Immunohistochemical analysis revealed that greater numbers of T lymphocytes containing CD4+ and CD8+ T cells were found in the group vaccinated with dendritic cells transfected with Il13ra2 mRNA. CONCLUSIONS These results demonstrate the therapeutic potential of vaccination with dendritic cells transfected with Il13ra2 mRNA for the treatment of malignant glioma.
Collapse
Affiliation(s)
- Makoto Saka
- Department of Neurosurgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|