1
|
Fu R, Zhou S, Liu C, Zhou J, Li Q. Administration of a combination of COX-2/TGF-β1 siRNAs induces hypertrophic scar fibroblast apoptosis through a TP53 mediated caspase pathway. Sci Rep 2024; 14:26427. [PMID: 39488600 PMCID: PMC11531465 DOI: 10.1038/s41598-024-77756-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 10/24/2024] [Indexed: 11/04/2024] Open
Abstract
Hypertrophic scar (HTS) formation is a pathological fibrotic skin disease, with no satisfactory treatments available currently. Inducing apoptosis of HTS-derived fibroblasts (HSFs) are becoming promising approaches. In this research, we aim to improve the technology with co-delivery COX-2 and TGF-β1 siRNAs and further investigate the underlying mechanism. Firstly, the HSFs were transfected with 1 µg/ml COX-2 and/or TGF-β1 siRNAs, and proved that the apoptosis of HSFs was greater induced by COX-2/TGF-β1 siRNAs than either COX-2 or TGF-β1 siRNA alone by flow cytometry. To investigate the impact of co-silencing TGF-β1 and COX-2 mRNA expression in vivo, we established HTSs model in rat tails. Our results confirmed that co-silencing of TGF-β1 and COX-2 mRNA expression could significantly alleviate the HTS formation in vivo. Furthermore, we explored the potential molecular mechanism and revealed that the protein levels of TP53, Bcl-2 and Caspase-3 were downregulated while Bax and Cleaved Caspase-3 were upregulated in the COX-2/TGF-β1 siRNA groups compared with HKP group. Taken together, our results demonstrated that simultaneous silencing of COX-2 and TGF-β1 expression by siRNAs induced HSF apoptosis through a TP53 mediated caspase pathway. Therefore, COX-2/TGF-β1 siRNAs might serve as a novel and effective therapeutic alternative for HTSs treatments.
Collapse
Affiliation(s)
- Rao Fu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Sizheng Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Chuanqi Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Jia Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
| |
Collapse
|
2
|
Li Y, Zhang C, Feng L, Shen Q, Liu F, Jiang X, Pang B. Application of natural polysaccharides and their novel dosage forms in gynecological cancers: therapeutic implications from the diversity potential of natural compounds. Front Pharmacol 2023; 14:1195104. [PMID: 37383719 PMCID: PMC10293794 DOI: 10.3389/fphar.2023.1195104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/02/2023] [Indexed: 06/30/2023] Open
Abstract
Cancer is one of the most lethal diseases. Globally, the number of cancers is nearly 10 million per year. Gynecological cancers (for instance, ovarian, cervical, and endometrial), relying on hidden diseases, misdiagnoses, and high recurrence rates, have seriously affected women's health. Traditional chemotherapy, hormone therapy, targeted therapy, and immunotherapy effectively improve the prognosis of gynecological cancer patients. However, with the emergence of adverse reactions and drug resistance, leading to the occurrence of complications and poor compliance of patients, we have to focus on the new treatment direction of gynecological cancers. Because of the potential effects of natural drugs in regulating immune function, protecting against oxidative damage, and improving the energy metabolism of the body, natural compounds represented by polysaccharides have also attracted extensive attention in recent years. More and more studies have shown that polysaccharides are effective in the treatment of various tumors and in reducing the burden of metastasis. In this review, we focus on the positive role of natural polysaccharides in the treatment of gynecologic cancer, the molecular mechanisms, and the available evidence, and discuss the potential use of new dosage forms derived from polysaccharides in gynecologic cancer. This study covers the most comprehensive discussion on applying natural polysaccharides and their novel preparations in gynecological cancers. By providing complete and valuable sources of information, we hope to promote more effective treatment solutions for clinical diagnosis and treatment of gynecological cancers.
Collapse
Affiliation(s)
- Yi Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chuanlong Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lu Feng
- College of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qian Shen
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fudong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochen Jiang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bo Pang
- International Medical Department of Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Szczygieł A, Węgierek-Ciura K, Wróblewska A, Mierzejewska J, Rossowska J, Szermer-Olearnik B, Świtalska M, Anger-Góra N, Goszczyński TM, Pajtasz-Piasecka E. Combined therapy with methotrexate nanoconjugate and dendritic cells with downregulated IL-10R expression modulates the tumor microenvironment and enhances the systemic anti-tumor immune response in MC38 murine colon carcinoma. Front Immunol 2023; 14:1155377. [PMID: 37033926 PMCID: PMC10078943 DOI: 10.3389/fimmu.2023.1155377] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Background Understanding the negative impact of the tumor microenvironment on the creation of an effective immune response has contributed to the development of new therapeutic anti-cancer strategies. One such solution is combined therapy consisting of chemotherapeutic administration followed by dendritic cell (DC)-based vaccines. The use of cytostatic leads to the elimination of cancer cells, but can also modulate the tumor milieu. Moreover, great efforts are being made to increase the therapeutic outcome of immunotherapy, e.g. by enhancing the ability of DCs to generate an efficient immune response, even in the presence of immunosuppressive cytokines such as IL-10. The study aimed to determine the effectiveness of combined therapy with chemotherapeutic with immunomodulatory potential - HES-MTX nanoconjugate (composed of methotrexate (MTX) and hydroxyethyl starch (HES)) and DCs with downregulated expression of IL-10 receptor stimulated with tumor antigens (DC/shIL-10R/TAg) applied in MC38 murine colon carcinoma model. Methods With the use of lentiviral vectors the DCs with decreased expression of IL-10R were obtained and characterized. During in vivo studies MC38-tumor bearing mice received MTX or HES-MTX nanoconjugate as a sole treatment or combined with DC-based immunotherapy containing unmodified DCs or DCs transduced with shRNA against IL-10R (or control shRNA sequence). Tumor volume was monitored during the experiment. One week after the last injection of DC-based vaccines, tumor nodules and spleens were dissected for ex vivo analysis. The changes in the local and systemic anti-tumor immune response were estimated with the use of flow cytometry and ELISA methods. Results and conclusions In vitro studies showed that the downregulation of IL-10R expression in DCs enhances their ability to activate the specific anti-tumor immune response. The use of HES-MTX nanoconjugate and DC/shIL-10R/TAg in the therapy of MC38-tumor bearing mice resulted in the greatest tumor growth inhibition. At the local anti-tumor immune response level a decrease in the infiltration of cells with suppressor activity and an increase in the influx of effector cells into MC38 tumor tissue was observed. These changes were crucial to enhance the effective specific immune response at the systemic level, which was revealed in the greatest cytotoxic activity of spleen cells against MC38 cells.
Collapse
|
4
|
Wu J, Peng J, Zhou Y, Zhang R, Wang Z, Hu N, Zhang D, Quan G, Wu Y, Feng J, Shen B, Zhao J, Zhang Y, Yang K, Luo L. Screening and Identification of a Novel Anti-Siglec-15 Human Antibody 3F1 and Relevant Antitumor Activity. Mol Pharmacol 2022; 102:161-171. [PMID: 35764384 DOI: 10.1124/molpharm.121.000470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/19/2022] [Indexed: 11/22/2022] Open
Abstract
Sialic acid-binding Ig-like lectin-15 is an important immunosuppressive molecule considered to be a key target in next-generation tumor immunotherapy. In this study, we screened 22 high-affinity antibodies that specifically recognize human Siglec-15 by using a large human phage antibody library, and five representative sequences were selected for further study. The results showed the binding activity of five antibodies to Siglec-15 (EC50 ranged from 0.02368 μg/mL to 0.07949 μg/mL), and in two Siglec-15-overexpressed cell lines, three antibodies had the strongest binding activity, so the two clones were discarded for further study. Subsequently, the affinity of three antibodies were measured by bio-layer interferometry technology (5-9 × 10E-09M). As the reported ligands of Siglec-15, the binding activity of Siglec-15 and sialyl-Tn, cluster of differentiation 44, myelin-associated glycoprotein, and leucine-rich repeat-containing protein 4C can be blocked by three of the antibodies. Among these, 3F1 had a competitive advantage. Then, the antibody 3F1 showed an obvious antibody-dependent cell-mediated cytotoxicity effect (EC50 was 0.85 μg/mL). Further, antibody 3F1 can reverse the inhibitory effect of Siglec-15 on lymphocyte proliferation (especially CD4+T and CD8+T) and cytokine release Interferon-γ. Given the above results, 3F1 was selected as a candidate for the in vivo pharmacodynamics study. In the tumor model of Balb/c Nude mice, 3F1 (10 mg/kg) showed certain antitumor effects [tumor growth inhibition (TGI) was 31.5%], while the combination of 3F1 (5 mg/kg) and Erbitux (5 mg/kg) showed significant antitumor effects (TGI was 48.7%) compared with the PBS group. In conclusion, novel human antibody 3F1 has antitumor activity and is expected to be an innovative candidate drug targeting Siglec-15 for tumor immunotherapy. SIGNIFICANCE STATEMENT: Siglec-15 is considered as an important target in the next generation of tumor immunotherapy. 3F1 is expected to be the most promising potential candidate for targeting Siglec-15 for cancer treatment and could provide a reference for the development of antitumor drugs.
Collapse
Affiliation(s)
- Jiaguo Wu
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Jingyi Peng
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Yangyihua Zhou
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Ran Zhang
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Zhihong Wang
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Naijing Hu
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Dingmu Zhang
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Guiqi Quan
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Yuanyu Wu
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Jiannan Feng
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Beifen Shen
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Jian Zhao
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Yan Zhang
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Kaiming Yang
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| | - Longlong Luo
- Department of Anatomy, School of Basic Medical Sciences of Dali University, Dali, China (J.W., K.Y.); State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China (J.W., J.P., Y.Zho., Z.W., N.H., D.Z., G.Q., Y.W., J.F., B.S., L.L.); Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China (J.P.); Hunan Normal University School of Medicine, Changsha, China (Y.Zho., R.Z., G.Q.); JOINN Biologics, Co., Ltd, Beijing, China (J.Z.); and Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China (Y.Zha.)
| |
Collapse
|
5
|
Zhang Z, Yao S, Hu Y, Zhao X, Lee RJ. Application of lipid-based nanoparticles in cancer immunotherapy. Front Immunol 2022; 13:967505. [PMID: 36003395 PMCID: PMC9393708 DOI: 10.3389/fimmu.2022.967505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy is revolutionizing the clinical management of patients with different cancer types by sensitizing autologous or allogenic immune cells to the tumor microenvironment which eventually leads to tumor cell lysis without rapidly killing normal cells. Although immunotherapy has been widely demonstrated to be superior to chemotherapies, only a few populations of patients with specific cancer types respond to such treatment due to the failure of systemic immune activation. In addition, severe immune-related adverse events are rapidly observed when patients with very few responses are given higher doses of such therapies. Recent advances of lipid-based nanoparticles (NPs) development have made it possible to deliver not only small molecules but also mRNAs to achieve systemic anticancer immunity through cytotoxic immune cell activation, checkpoint blockade, and chimeric antigen receptor cell therapies, etc. This review summarized recent development and applications of LNPs in anticancer immunotherapy. The diversity of lipid-based NPs would encapsulate payloads with different structures and molecular weights to achieve optimal antitumor immunity through multiple mechanisms of action. The discussion about the components of lipid-based NPs and their immunologic payloads in this review hopefully shed more light on the future direction of anticancer immunotherapy.
Collapse
Affiliation(s)
- Zhongkun Zhang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Siyu Yao
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
| | - Yingwen Hu
- The Whiteoak Group, Inc., Rockville, MD, United States
| | - Xiaobin Zhao
- The Whiteoak Group, Inc., Rockville, MD, United States
| | - Robert J. Lee
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
6
|
Zhong W, Fang C, Liu H, Zhang L, Zhang X, Zhong J, He X, Zhang L. LAP+CD4+T cells regulate the anti-tumor role of CIK cells in colorectal cancer through IL-10 and TGF-β. Am J Transl Res 2022; 14:3716-3728. [PMID: 35836905 PMCID: PMC9274552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 12/29/2021] [Indexed: 06/15/2023]
Abstract
The rate of colorectal cancer (CRC) is increasing. Adoptive immune cell therapy (ACT) is a research hotspot in CRC treatment, and the common adoptive cells are cytokine-induced killer cells (CIK). The problem of ACT is that some regulatory T cells (Treg) will affect the efficacy. Latent associated polypeptide (LAP)+CD4+T is a new Treg, and its immunosuppressive effect is much higher than that of traditional Tregs. This research mainly explored the influence of LAP+CD4+T cells on anti-tumor lethality of CIK cells, so as to fill this gap. The LAP+CD4+T CIK cells and LAP-CD4+T CIK cells were sorted by immunomagnetic beads. LAP+CD4+T cells were expanded in vitro, and high expression cytokine genes were screened by RT-qPCR. LAP+CD4+T and LAP-CD4+T CIK cells were co-cultured to test cyto-activity. Transplanted tumor models of CRC were established in nude mice, which were randomized into a control group (CG), CIK group, LAP (-) group, LAP (+) group, IL-10 siRNA group, and TGF-siRNA group, and the tumor growth in each group was observed. The research results revealed that interleukin-10 (IL-10) and transforming growth factor-β (TGF-β) were highly expressed in LAP+CD4+T cells. LAP+CD4+T could effectively suppress CIK cell proliferation and activity. LAP-CD4+T could suppress IL-10 and TGF-β, and inhibit CIK cell apoptosis, proliferation, and tumor growth, thus improving their anti-tumor lethality. LAP+CD4+T cells regulate the anti-tumor role of CIK cells in CRC through IL-10 and TGF-β.
Collapse
Affiliation(s)
- Wu Zhong
- Department of Gastrointestinal Surgery, Ganzhou Hospital Affiliated to Nanchang UniversityGanzhou 341000, Jiangxi, China
| | - Chuanfa Fang
- Department of Gastrointestinal Surgery, Ganzhou Hospital Affiliated to Nanchang UniversityGanzhou 341000, Jiangxi, China
| | - Hongquan Liu
- Department of Gastrointestinal Surgery, Ganzhou Hospital Affiliated to Nanchang UniversityGanzhou 341000, Jiangxi, China
| | - Lei Zhang
- Department of Gastrointestinal Surgery, Ganzhou Hospital Affiliated to Nanchang UniversityGanzhou 341000, Jiangxi, China
| | - Xiaofei Zhang
- Department of Anorectal, Ganzhou Traditional Chinese Medicine HospitalGanzhou 341000, Jiangxi, China
| | - Junqiao Zhong
- Department of Gastrointestinal Surgery, Ganzhou Hospital Affiliated to Nanchang UniversityGanzhou 341000, Jiangxi, China
| | - Xianping He
- Department of Gastrointestinal Surgery, Ganzhou Hospital Affiliated to Nanchang UniversityGanzhou 341000, Jiangxi, China
| | - Leichang Zhang
- Department of Anorectal, Affiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchang 330006, Jiangxi, China
| |
Collapse
|
7
|
Jasrotia R, Dhanjal DS, Bhardwaj S, Sharma P, Chopra C, Singh R, Kumar A, Mubayi A, Kumar D, Kumar R, Goyal A. Nanotechnology based vaccines: Cervical cancer management and perspectives. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
8
|
Phung CD, Tran TH, Nguyen HT, Nguyen TT, Jeong JH, Ku SK, Yong CS, Choi HG, Kim JO. Nanovaccines silencing IL-10 production at priming phase for boosting immune responses to melanoma. J Control Release 2021; 338:211-223. [PMID: 34419495 DOI: 10.1016/j.jconrel.2021.08.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 08/14/2021] [Accepted: 08/18/2021] [Indexed: 01/15/2023]
Abstract
Despite the significant efforts in developing cancer vaccines, there are still numerous challenges that need to be addressed to ensure their clinical efficacy. Herein, a lymphatic dendritic cell (DC)-targeted artificial nanovaccine mimicking tumor cell membrane (ATM-NV) is developed to boost effector immune response and control immunosuppression simultaneously. The NVs are formulated with lipids, tumor cell membrane proteins, imiquimod (IMQ), and IL-10 siRNA. IL-10 siRNA is incorporated to inhibit the secretion of IL-10, an immunosuppressive cytokine, of maturated DCs upon IMQ. To enhance the DC targeting ability, the nanovaccine surface was non-covalently conjugated with the anti-CD205 antibody. The IMQ and IL-10 siRNA co-loaded, CD205 receptor-targeted artificial tumor membrane NVs (IMQ/siR@ATM-NVs) efficiently migrate to the tumor-draining lymph node and target DCs. Furthermore, immunization with IMQ/siR@ATM-NVs reduces the production of IL-10 and increases Th1-driven antitumor immunity resulted in a great tumor inhibition efficacy. Our results suggest a potential strategy to promote the vaccination's antitumor efficacy by blocking the intrinsic negative regulators in DCs.
Collapse
Affiliation(s)
- Cao Dai Phung
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi 12116, Viet Nam; PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No.167 Hoang Ngan, Trung Hoa, Cau Giay, Hanoi 11313, Viet Nam
| | - Hanh Thuy Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Tien Tiep Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 15588, Gyeonggi-do, Republic of Korea.
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
9
|
Pellom ST, Smalley Rumfield C, Morillon YM, Roller N, Poppe LK, Brough DE, Sabzevari H, Schlom J, Jochems C. Characterization of recombinant gorilla adenovirus HPV therapeutic vaccine PRGN-2009. JCI Insight 2021; 6:141912. [PMID: 33651712 PMCID: PMC8119209 DOI: 10.1172/jci.insight.141912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 02/25/2021] [Indexed: 12/31/2022] Open
Abstract
There are approximately 44,000 cases of human papillomavirus-associated (HPV-associated) cancer each year in the United States, most commonly caused by HPV types 16 and 18. Prophylactic vaccines successfully prevent healthy people from acquiring HPV infections via HPV-specific antibodies. In order to treat established HPV-associated malignancies, however, new therapies are necessary. Multiple recombinant gorilla adenovirus HPV vaccine constructs were evaluated in NSG-β2m-/- peripheral blood mononuclear cell-humanized mice bearing SiHa, a human HPV16+ cervical tumor, and/or in the syngeneic HPV16+ TC-1 model. PRGN-2009 is a therapeutic gorilla adenovirus HPV vaccine containing multiple cytotoxic T cell epitopes of the viral oncoproteins HPV 16/18 E6 and E7, including T cell enhancer agonist epitopes. PRGN-2009 treatment reduced tumor volume and increased CD8+ and CD4+ T cells in the tumor microenvironment of humanized mice bearing the human cervical tumor SiHa. PRGN-2009 monotherapy in the syngeneic TC-1 model also reduced tumor volumes and weights, generated high levels of HPV16 E6-specific T cells, and increased multifunctional CD8+ and CD4+ T cells in the tumor microenvironment. These studies provide the first evaluation to our knowledge of a therapeutic gorilla adenovirus HPV vaccine, PRGN-2009, showing promising preclinical antitumor efficacy and induction of HPV-specific T cells, along with the rationale for its evaluation in clinical trials.
Collapse
Affiliation(s)
- Samuel T. Pellom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Claire Smalley Rumfield
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Y. Maurice Morillon
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Nicholas Roller
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Lisa K. Poppe
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | | | | | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Caroline Jochems
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Farmer E, Cheng MA, Hung CF, Wu TC. Vaccination Strategies for the Control and Treatment of HPV Infection and HPV-Associated Cancer. Recent Results Cancer Res 2021; 217:157-195. [PMID: 33200366 DOI: 10.1007/978-3-030-57362-1_8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human papillomavirus (HPV) is the most common sexually transmitted infection, currently affecting close to 80 million Americans. Importantly, HPV infection is recognized as the etiologic factor for numerous cancers, including cervical, vulval, vaginal, penile, anal, and a subset of oropharyngeal cancers. The prevalence of HPV infection and its associated diseases are a significant problem, affecting millions of individuals worldwide. Likewise, the incidence of HPV infection poses a significant burden on individuals and the broader healthcare system. Between 2011 and 2015, there were an estimated 42,700 new cases of HPV-associated cancers each year in the United States alone. Similarly, the global burden of HPV is high, with around 630,000 new cases of HPV-associated cancer occurring each year. In the last decade, a total of three preventive major capsid protein (L1) virus-like particle-based HPV vaccines have been licensed and brought to market as a means to prevent the spread of HPV infection. These prophylactic vaccines have been demonstrated to be safe and efficacious in preventing HPV infection. The most recent iteration of the preventive HPV vaccine, a nanovalent, L1-VLP vaccine, protects against a total of nine HPV types (seven high-risk and two low-risk HPV types), including the high-risk types HPV16 and HPV18, which are responsible for causing the majority of HPV-associated cancers. Although current prophylactic HPV vaccines have demonstrated huge success in preventing infection, existing barriers to vaccine acquisition have limited their widespread use, especially in low- and middle-income countries, where the burden of HPV-associated diseases is highest. Prophylactic vaccines are unable to provide protection to individuals with existing HPV infections or HPV-associated diseases. Instead, therapeutic HPV vaccines capable of generating T cell-mediated immunity against HPV infection and associated diseases are needed to ameliorate the burden of disease in individuals with existing HPV infection. To generate a cell-mediated immune response against HPV, most therapeutic vaccines target HPV oncoproteins E6 and E7. Several types of therapeutic HPV vaccine candidates have been developed including live-vector, protein, peptide, dendritic cell, and DNA-based vaccines. This chapter will review the commercially available prophylactic HPV vaccines and discuss the recent progress in the development of therapeutic HPV vaccines.
Collapse
Affiliation(s)
- Emily Farmer
- Department of Pathology, The Johns Hopkins School of Medicine, Cancer Research Building II, 1550 Orleans Street, Baltimore, MD, 21287, USA
| | - Max A Cheng
- Department of Pathology, The Johns Hopkins School of Medicine, Cancer Research Building II, 1550 Orleans Street, Baltimore, MD, 21287, USA
| | - Chien-Fu Hung
- Department of Pathology, The Johns Hopkins School of Medicine, Cancer Research Building II, 1550 Orleans Street, Baltimore, MD, 21287, USA.,Department of Oncology, The Johns Hopkins School of Medicine, Cancer Research Building II, 1550 Orleans Street, Baltimore, MD, 21287, USA
| | - T-C Wu
- Department of Pathology, The Johns Hopkins School of Medicine, Cancer Research Building II, 1550 Orleans Street, Baltimore, MD, 21287, USA. .,Department of Oncology, The Johns Hopkins School of Medicine, Cancer Research Building II, 1550 Orleans Street, Baltimore, MD, 21287, USA. .,Department of Obstetrics and Gynecology, The Johns Hopkins School of Medicine, Cancer Research Building II, 1550 Orleans Street, Baltimore, MD, 21287, USA. .,Department of Pathology, Oncology, Obstetrics and Gynecology, and Molecular Microbiology and Immunology, The Johns Hopkins Medical Institutions, Cancer Research Building II, Room 309, 1550 Orleans Street, Baltimore, MD, 21287, USA.
| |
Collapse
|
11
|
CCL25 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1302:99-111. [PMID: 34286444 DOI: 10.1007/978-3-030-62658-7_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Multiple checkpoint mechanisms are overridden by cancer cells in order to develop into a tumor. Neoplastic cells, while constantly changing during the course of cancer progression, also craft their surroundings to meet their growing needs. This crafting involves changing cell surface receptors, affecting response to extracellular signals and secretion of signals that affect the nearby cells and extracellular matrix architecture. This chapter briefly comprehends the non-cancer cells facilitating the cancer growth and elaborates on the notable role of the CCR9-CCL25 chemokine axis in shaping the tumor microenvironment (TME), directly and via immune cells. Association of increased CCR9 and CCL25 levels in various tumors has demonstrated the significance of this axis as a tool commonly used by cancer to flourish. It is involved in attracting immune cells in the tumor and determining their fate via various direct and indirect mechanisms and, leaning the TME toward immunosuppressive state. Besides, elevated CCR9-CCL25 signaling allows survival and rapid proliferation of cancer cells in an otherwise repressive environment. It modulates the intra- and extracellular protein matrix to instigate tumor dissemination and creates a supportive metastatic niche at the secondary sites. Lastly, this chapter abridges the latest research efforts and challenges in using the CCR9-CCL25 axis as a cancer-specific target.
Collapse
|
12
|
Bahreyni A, Luo H. Advances in Targeting Cancer-Associated Genes by Designed siRNA in Prostate Cancer. Cancers (Basel) 2020; 12:E3619. [PMID: 33287240 PMCID: PMC7761674 DOI: 10.3390/cancers12123619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 11/28/2020] [Accepted: 11/29/2020] [Indexed: 12/21/2022] Open
Abstract
Short interfering RNAs (siRNAs) have provided novel insights into the field of cancer treatment in light of their ability to specifically target and silence cancer-associated genes. In recent years, numerous studies focus on determining genes that actively participate in tumor formation, invasion, and metastasis in order to establish new targets for cancer treatment. In spite of great advances in designing various siRNAs with diverse targets, efficient delivery of siRNAs to cancer cells is still the main challenge in siRNA-mediated cancer treatment. Recent advancements in the field of nanotechnology and nanomedicine hold great promise to meet this challenge. This review focuses on recent findings in cancer-associated genes and the application of siRNAs to successfully silence them in prostate cancer, as well as recent progress for effectual delivery of siRNAs to cancer cells.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Centre for Heart Lung Innovation, St. Paul’s Hospital, 1081 Burrard St, Vancouver, BC V6Z 1Y6, Canada;
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St. Paul’s Hospital, 1081 Burrard St, Vancouver, BC V6Z 1Y6, Canada;
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|
13
|
Wang K, Du H, Chen Z, Lu H, Xu R, Xue D. ACTH 4-10 protects the ADR-injured podocytes by stimulating B lymphocytes to secrete interleukin-10. Int Immunopharmacol 2020; 87:106769. [PMID: 32682256 DOI: 10.1016/j.intimp.2020.106769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/10/2020] [Accepted: 06/29/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES In the present study, we aimed to assess whether adrenocorticotropic hormone (ACTH) could protect the podocytes from adriamycin (ADR)-induced injury by stimulating B lymphocytes to secrete the associated cytokines. METHODS Proliferation assay was used to assess the proliferation and activity of podocytes. Enzyme-linked immunosorbent assay was used to examine the secretion of IL-10 and IL-4. TUNEL apoptosis detection kit was used to detect the apoptosis of podocytes. Real-time PCR and Western blotting analysis were used to examine the expressions of nephrin and podocin at the mRNA and protein levels. RESULTS Compared with the normal control group, the podocyte proliferation of ADR group was significantly inhibited. However, compared with the ADR group, the podocyte proliferation of the supernatant (1 µg/L, 10 µg/L or 100 µg/L ACTH4-10) + ADR groups was generally increased, and the pro-proliferative effect of the supernatant containing 10 µg/L ACTH4-10 was the highest. Moreover, we found that after B lymphocytes were intervened by 10 µg/L ACTH4-10, the IL-10 level in the cell supernatant was significantly elevated (p < 0.05). When anti-IL-10R was added, the podocyte proliferation of the supernatant (10 µg/L ACTH4-10) + ADR group was significantly inhibited. Furthermore, the supernatant of B cells stimulated with 10 µg/L ACTH4-10 could better decrease the apoptosis rate of injured podocytes and increase the expressions of nephrin and podocin at the mRNA and protein levels by elevating the secretion of IL-10. CONCLUSION Compared with ACTH4-10, the supernatant of B cells stimulated with ACTH4-10 could better protect the podocytes from ADR-induced injury by elevating the secretion of IL-10.
Collapse
Affiliation(s)
- Kun Wang
- Department of Surgical Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Huaping Du
- Suzhou Ninth People's Hospital, Suzhou, Jiangsu, China
| | - Zhen Chen
- Department of Surgical Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Hao Lu
- Department of Surgical Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Renfang Xu
- Department of Surgical Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Dong Xue
- Department of Surgical Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| |
Collapse
|
14
|
Ahmed R, Sayegh N, Graciotti M, Kandalaft LE. Electroporation as a method of choice to generate genetically modified dendritic cell cancer vaccines. Curr Opin Biotechnol 2020; 65:142-155. [PMID: 32240923 DOI: 10.1016/j.copbio.2020.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/10/2019] [Accepted: 02/17/2020] [Indexed: 12/31/2022]
Abstract
In the last few decades, immunotherapy has emerged as an alternative therapeutic approach to treat cancer. Immunotherapy offers a plethora of different treatment possibilities. Among these, dendritic cell (DC)-based cancer vaccines constitute one of the most promising and valuable therapeutic options. DC-vaccines have been introduced into the clinics more than 15 years ago, and preclinical studies showed their general safety and low toxic effects on patients. However, their treatment efficacy is still rather limited, demanding for novel avenues to improve vaccine efficacy. One way to potentially achieve this is to focus on improving the DC-T cell interaction to further increase T cell priming and downstream activity. A successful DC-T cell interaction requires three different signals (Figure 1): (1) Major Histocompatibility Complex (MHC) and antigen complex interaction with T cell receptor (TCR) (2) interaction between co-stimulatory molecules and their cognate ligands at the cell surface and (3) secretion of cytokines to polarize the immune response toward a Type 1 helper (Th1) phenotype. In recent years, many studies attempted to improve the DC-T cell interaction and overall cancer vaccine therapeutic outcomes by increasing the expression of mediators of signal 1, 2 and/or 3, through genetic modifications of DCs. Transfection of genes of interest can be achieved through many different methods such as passive pulsing, lipofection, viral transfection, or electroporation (EP). However, EP is currently emerging as the method of choice thanks to its safety, versatility, and relatively easy clinical translation. In this review we will highlight the potential benefits of EP over other transfection methods as well as giving an overview of the available studies employing EP to gene-modify DCs in cancer vaccines. Crucial aspects such as safety, feasibility, and gene(s) of choice will be also discussed, together with future perspectives and opportunities for DC genetic engineering.
Collapse
Affiliation(s)
- Rita Ahmed
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland
| | - Naya Sayegh
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland
| | - Michele Graciotti
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland
| | - Lana E Kandalaft
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
| |
Collapse
|
15
|
Wang R, Pan W, Jin L, Huang W, Li Y, Wu D, Gao C, Ma D, Liao S. Human papillomavirus vaccine against cervical cancer: Opportunity and challenge. Cancer Lett 2020; 471:88-102. [DOI: 10.1016/j.canlet.2019.11.039] [Citation(s) in RCA: 258] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/28/2019] [Accepted: 11/30/2019] [Indexed: 12/20/2022]
|
16
|
Thepmalee C, Panya A, Sujjitjoon J, Sawasdee N, Poungvarin N, Junking M, Yenchitsomanus PT. Suppression of TGF-β and IL-10 receptors on self-differentiated dendritic cells by short-hairpin RNAs enhanced activation of effector T-cells against cholangiocarcinoma cells. Hum Vaccin Immunother 2020; 16:2318-2327. [PMID: 31976810 DOI: 10.1080/21645515.2019.1701913] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive tumor that is associated with high rates of recurrence and mortality. This is due, in part, to the fact that CCA cells and their microenvironment secrete immunosuppressive cytokines, transforming growth factor-β (TGF-β) and interleukin-10 (IL-10), that inhibit dendritic cell (DC) functions, which, in turn, results in the decreased anti-tumor activity of T-cells. We hypothesized that the TGF-β receptor and IL-10 blockade on dendritic cells would improve DC function, thereby allowing improved activation of T cells against CCA cells. To test our hypothesis, we generated self-differentiated DCs (SD-DCs) via transduction of human peripheral blood monocytes with lentivirus expressing IL-4 and GM-CSF. SD-DCs were transduced with a second lentivirus containing short-hairpin RNAs (shRNAs) to knock-down TGF-βRII and IL-10RA mRNAs. Immunoblot confirmed the reduced expression levels of TGF-β and IL-10 receptors in both SD-DCs that were transduced with a single and/or combination of lentiviruses containing shRNAs. SD-DCs were thereafter pulsed with tumor antigens extracted from CCA cell lines in an effort to activate DC function. MHC class II (HLA-DR) and co-stimulatory molecules (CD40 and CD86) on SD-DCs were upregulated to levels comparable to those on DCs generated by the conventional method. Suppression of TGF-β and IL-10 receptors on SD-DCs influenced the effector T-cells to produce IFN-γ, which enhanced their ability to kill CCA cells. The preparation of adoptive effector T-cells holds the potential of becoming a novel therapy for cellular immunotherapy in CCA.
Collapse
Affiliation(s)
- Chutamas Thepmalee
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand.,Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand.,Division of Biochemistry, School of Medical Sciences, University of Phayao , Phayao, Thailand
| | - Aussara Panya
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand.,Department of Biology, Faculty of Science, Chiang Mai University , Chiang Mai, Thailand.,Center of Excellence in Bioresources for Agriculture, Industry and Medicine, Faculty of Science, Chiang Mai University , Chiang Mai, Thailand
| | - Jatuporn Sujjitjoon
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| | - Nunghathai Sawasdee
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| | - Naravat Poungvarin
- Clinical Molecular Pathology Laboratory, Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| | - Mutita Junking
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| |
Collapse
|
17
|
Sioud M. Unleashing the Therapeutic Potential of Dendritic and T Cell Therapies Using RNA Interference. Methods Mol Biol 2020; 2115:259-280. [PMID: 32006406 DOI: 10.1007/978-1-0716-0290-4_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Therapeutic dendritic cell (DC) cancer vaccines work to boost the body's immune system to fight a cancer. Although this type of immunotherapy often leads to the activation of tumor-specfic T cells, clinical responses are fairly low, arguing for the need to improve the design of DC-based vaccines. Recent studies revealed a promising strategy of combining DC vaccines with small interfering RNAs (siRNAs) targeting immunosuppressive signals such as checkpoint receptors. Similarly, incorporating checkpoint siRNA blockers in adoptive T-cell therapy to amplify cytotoxic T lymphocyte responses is now being tested in the clinic. The development of the next generation of cancer immunotherapies using siRNA technology will hopefuly benefit patients with various cancer types including those who did not respond to current therapies. This review highlights the latest advances in RNA interference technology to improve the therapeutic efficacy of DC cancer vaccines and T cell therapy.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Ullernchausseen 70, Oslo, Norway.
| |
Collapse
|
18
|
Vonsky MS, Runov AL, Gordeychuk IV, Isaguliants MG. Therapeutic Vaccines Against Human Papilloma Viruses: Achievements and Prospects. BIOCHEMISTRY (MOSCOW) 2019; 84:800-816. [PMID: 31509730 DOI: 10.1134/s0006297919070101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Human papillomaviruses of high carcinogenic risk (HR HPVs) are major etiological agents of malignant diseases of the cervix, vulva, penis, anal canal, larynx, head, and neck. Prophylactic vaccination against HPV, which mainly covers girls and women under 25, does not prevent vertical and horizontal HPV transmission in infants and children and does not have a therapeutic effect. As a result, a significant proportion of the population is not protected from the HPV infection and development of HPV-associated neoplastic transformation and cancer, which indicates the need for development and introduction of therapeutic HPV vaccines. Unlike prophylactic vaccines aimed at the formation of virus-neutralizing antibodies, therapeutic vaccines elicit cellular immune response leading to the elimination of infected and malignant cells expressing viral proteins. The ideal targets for vaccine immunotherapy are highly conserved HR HPV oncoproteins E6 and E7 expressed in precancerous and tumor tissues. Here, we describe expression of these proteins during different stages of HPV infection, their antigenic and immunogenic properties, and T-cell epitopes, the response to which correlates with natural regression of HPV-induced neoplastic changes. The review describes patterns of E6 and E7 oncoproteins presentation to the immune system as components of candidate vaccines along with the results of the most promising preclinical trials and animal models used in these trials. Special attention is paid to vaccine candidates which have shown efficacy in clinical trials in patients with HPV-associated neoplastic changes.
Collapse
Affiliation(s)
- M S Vonsky
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia. .,Almazov National Medical Research Centre, Ministry of Health of Russian Federation, St. Petersburg, 197341, Russia
| | - A L Runov
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia.,Almazov National Medical Research Centre, Ministry of Health of Russian Federation, St. Petersburg, 197341, Russia.,Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Health of Russian Federation, Moscow, 123098, Russia
| | - I V Gordeychuk
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Health of Russian Federation, Moscow, 123098, Russia. .,Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences, Moscow, 108819, Russia.,Sechenov First Moscow State Medical University, Ministry of Health of Russian Federation, Moscow, 119991, Russia
| | - M G Isaguliants
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Health of Russian Federation, Moscow, 123098, Russia. .,Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences, Moscow, 108819, Russia.,Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, Stockholm, SE-171 77, Sweden.,Riga Stradins University, Department of Pathology, Riga, LV-1007, Latvia
| |
Collapse
|
19
|
King Thomas J, Mir H, Kapur N, Singh S. Racial Differences in Immunological Landscape Modifiers Contributing to Disparity in Prostate Cancer. Cancers (Basel) 2019; 11:cancers11121857. [PMID: 31769418 PMCID: PMC6966521 DOI: 10.3390/cancers11121857] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022] Open
Abstract
Prostate cancer affects African Americans disproportionately by exhibiting greater incidence, rapid disease progression, and higher mortality when compared to their Caucasian counterparts. Additionally, standard treatment interventions do not achieve similar outcome in African Americans compared to Caucasian Americans, indicating differences in host factors contributing to racial disparity. African Americans have allelic variants and hyper-expression of genes that often lead to an immunosuppressive tumor microenvironment, possibly contributing to more aggressive tumors and poorer disease and therapeutic outcomes than Caucasians. In this review, we have discussed race-specific differences in external factors impacting internal milieu, which modify immunological topography as well as contribute to disparity in prostate cancer.
Collapse
Affiliation(s)
- Jeronay King Thomas
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (J.K.T.); (H.M.); (N.K.)
- Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Hina Mir
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (J.K.T.); (H.M.); (N.K.)
- Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Neeraj Kapur
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (J.K.T.); (H.M.); (N.K.)
- Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Shailesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (J.K.T.); (H.M.); (N.K.)
- Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
- Correspondence: ; Tel.: +1-404-756-5718; Fax: +1-404-752-1179
| |
Collapse
|
20
|
Liu Y, Li H, Pi R, Yang Y, Zhao X, Qi X. Current strategies against persistent human papillomavirus infection (Review). Int J Oncol 2019; 55:570-584. [PMID: 31364734 DOI: 10.3892/ijo.2019.4847] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/16/2019] [Indexed: 11/06/2022] Open
Abstract
Human papillomavirus (HPV) is the most common sexually transmitted infection, exhibiting a tropism for the epidermis and mucosae. The link between persistent HPV infection and malignancies involving the anogenital tract as well as the head and neck has been well‑established, and it is estimated that HPV‑related cancers involving various anatomical sites account for 4.5% of all human cancers. Current prophylactic vaccines against HPV have enabled the prevention of associated malignancies. However, the sizeable population base of current infection in whom prophylactic vaccines are not applicable, certain high‑risk HPV types not included in vaccines, and the vast susceptible population in developing countries who do not have access to the costly prophylactic vaccines, put forward an imperative need for effective therapies targeting persistent infection. In this article, the life cycle of HPV, the mechanisms facilitating HPV evasion of recognition and clearance by the host immune system, and the promising therapeutic strategies currently under investigation, particularly antiviral drugs and therapeutic vaccines, are reviewed.
Collapse
Affiliation(s)
- Yu Liu
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hongyi Li
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ruyu Pi
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yang Yang
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaorong Qi
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
21
|
Silva JR, Sales NS, Silva MO, Aps LRMM, Moreno ACR, Rodrigues EG, Ferreira LCS, Diniz MO. Expression of a soluble IL-10 receptor enhances the therapeutic effects of a papillomavirus-associated antitumor vaccine in a murine model. Cancer Immunol Immunother 2019; 68:753-763. [PMID: 30806747 PMCID: PMC11028134 DOI: 10.1007/s00262-018-02297-2] [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: 05/22/2018] [Accepted: 12/28/2018] [Indexed: 12/25/2022]
Abstract
The presence of IL-10, produced either by tumor cells or immunosuppressive cells, is frequently associated with a poor prognosis for cancer progression. It may also negatively impact anticancer treatments, such as immunotherapies, that otherwise would promote the activation of cytotoxic T cells capable of detecting and destroying malignant cells. In the present study, we evaluated a new adjuvant approach for anticancer immunotherapy using a plasmid vector encoding a soluble form of the IL-10 receptor (pIL-10R). pIL-10R was coadministered to mice with a DNA vaccine encoding the type 16 human papillomavirus (HPV-16) E7 oncoprotein genetically fused with glycoprotein D of herpes simplex virus (HSV) (pgDE7h). Immunization regimens based on the coadministration of pIL-10R and pgDE7h enhanced the antitumor immunity elicited in mice injected with TC-1 cells, which express HPV-16 oncoproteins. The administration of the DNA vaccines by in vivo electroporation further enhanced the anticancer effects of the vaccines, leading to the activation of tumor-infiltrating polyfunctional E7-specific cytotoxic CD8+ T cells and control of the expansion of immunosuppressive cells. In addition, the combination of immunotherapy and pIL-10R allowed the control of tumors in more advanced growth stages that otherwise would not be treatable by the pgDE7h vaccine. In conclusion, the proposed treatment involving the expression of IL-10R enhanced the antitumor protective immunity induced by pgDE7h administration and may contribute to the development of more efficient clinical interventions against HPV-induced tumors.
Collapse
Affiliation(s)
- Jamile R Silva
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil
| | - Natiely S Sales
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil
| | - Mariângela O Silva
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil
| | - Luana R M M Aps
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil
| | - Ana C R Moreno
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil
| | - Elaine G Rodrigues
- Tumor Immunobiology Laboratory, Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - Luís C S Ferreira
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil.
| | - Mariana O Diniz
- Vaccine Development Laboratory, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-000, Brazil
- Division of Infection and Immunity, University College London, 5 University St, Bloomsbury, London, WC1E 6JF, UK
| |
Collapse
|
22
|
Sioud M. Releasing the Immune System Brakes Using siRNAs Enhances Cancer Immunotherapy. Cancers (Basel) 2019; 11:cancers11020176. [PMID: 30717461 PMCID: PMC6406640 DOI: 10.3390/cancers11020176] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/29/2019] [Accepted: 01/31/2019] [Indexed: 12/12/2022] Open
Abstract
Therapeutic dendritic cell (DC) cancer vaccines rely on the immune system to eradicate tumour cells. Although tumour antigen-specific T cell responses have been observed in most studies, clinical responses are fairly low, arguing for the need to improve the design of DC-based vaccines. The incorporation of small interfering RNAs (siRNAs) against immunosuppressive factors in the manufacturing process of DCs can turn the vaccine into potent immune stimulators. Additionally, siRNA modification of ex vivo-expanded T cells for adoptive immunotherapy enhanced their killing potency. Most of the siRNA-targeted immune inhibitory factors have been successful in that their blockade produced the strongest cytotoxic T cell responses in preclinical and clinical studies. Cancer patients treated with the siRNA-modified DC vaccines showed promising clinical benefits providing a strong rationale for further development of these immunogenic vaccine formulations. This review covers the progress in combining siRNAs with DC vaccines or T cell therapy to boost anti-tumour immunity.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Montebello, N-0310 Oslo, Norway.
| |
Collapse
|
23
|
Llopiz D, Ruiz M, Silva L, Sarobe P. Enhancement of Antitumor Vaccination by Targeting Dendritic Cell-Related IL-10. Front Immunol 2018; 9:1923. [PMID: 30233565 PMCID: PMC6129595 DOI: 10.3389/fimmu.2018.01923] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/06/2018] [Indexed: 12/24/2022] Open
Abstract
Understanding mechanisms associated to dendritic cell (DC) functions has allowed developing new antitumor therapeutic vaccination strategies. However, these vaccines have demonstrated limited clinical results. Although the low immunogenicity of tumor antigens used and the presence of tumor-associated suppressive factors may in part account for these results, intrinsic vaccine-related factors may also be involved. Vaccines modulate DC functions by inducing activating and inhibitory signals that determine ensuing T cell responses. In this mini review, we focus on IL-10, inhibitory cytokine induced in DC upon vaccination, which defines a suppressive cell subset, discussing its implications as a potential target in combined vaccination immunotherapies.
Collapse
Affiliation(s)
- Diana Llopiz
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Marta Ruiz
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Leyre Silva
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Pablo Sarobe
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
24
|
Description of CD8 + Regulatory T Lymphocytes and Their Specific Intervention in Graft-versus-Host and Infectious Diseases, Autoimmunity, and Cancer. J Immunol Res 2018; 2018:3758713. [PMID: 30155493 PMCID: PMC6098849 DOI: 10.1155/2018/3758713] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/09/2018] [Accepted: 06/06/2018] [Indexed: 12/13/2022] Open
Abstract
Gershon and Kondo described CD8+ Treg lymphocytes as the first ones with regulating activity due to their tolerance ability to foreign antigens and their capacity to inhibit the proliferation of other lymphocytes. Regardless, CD8+ Treg lymphocytes have not been fully described-unlike CD4+ Treg lymphocytes-because of their low numbers in blood and the lack of specific and accurate population markers. Still, these lymphocytes have been studied for the past 30 years, even after finding difficulties during investigations. As a result, studies have identified markers that define their subpopulations. This review is focused on the expression of cell membrane markers as CD25, CD122, CD103, CTLA-4, CD39, CD73, LAG-3, and FasL as well as soluble molecules such as FoxP3, IFN-γ, IL-10, TGF-β, IL-34, and IL-35, in addition to the lack of expression of cell activation markers such as CD28, CD127 CD45RC, and CD49d. This work also underlines the importance of identifying some of these markers in infections with several pathogens, autoimmunity, cancer, and graft-versus-host disease as a strategy in their prevention, monitoring, and cure.
Collapse
|
25
|
Mainini F, Larsen DS, Webster GA, Young SL, Eccles MR. MIS416 as a siRNA Delivery System with the Ability to Target Antigen-Presenting Cells. Nucleic Acid Ther 2018; 28:225-232. [PMID: 29893623 PMCID: PMC6080116 DOI: 10.1089/nat.2017.0695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
MIS416 is a microparticulate formulation derived from propionibacterium acnes cell wall skeletons with intrinsic adjuvant activity. Conjugates of MIS416-SS-peptide containing a disulfide linkage facilitate the cytoplasmic delivery and release of peptides in antigen-presenting cells (APCs). We hypothesized that MIS416-siRNA (small interfering RNA) conjugates, containing a disulfide linkage between MIS416 and the siRNA, would allow cytoplasmic release of siRNA in APCs. MIS416-SS-siStat3 conjugates added to cell culture medium of monolayers of DCs in culture flasks successfully targeted Stat3 mRNA in DCs in vitro without transfection, downregulating Stat3 mRNA and protein levels. These results suggest that MIS416-SS-siRNA conjugates can be used as a novel siRNA delivery system for the knockdown of mRNA levels in APCs.
Collapse
Affiliation(s)
- Francesco Mainini
- 1 Department of Pathology, University of Otago , Dunedin, New Zealand
| | - David S Larsen
- 2 Department of Chemistry, University of Otago , Dunedin, New Zealand
| | | | - Sarah L Young
- 1 Department of Pathology, University of Otago , Dunedin, New Zealand .,4 Maurice Wilkins Center for Molecular Biodiscovery , Auckland, New Zealand
| | - Michael R Eccles
- 1 Department of Pathology, University of Otago , Dunedin, New Zealand .,4 Maurice Wilkins Center for Molecular Biodiscovery , Auckland, New Zealand
| |
Collapse
|
26
|
Chabeda A, Yanez RJR, Lamprecht R, Meyers AE, Rybicki EP, Hitzeroth II. Therapeutic vaccines for high-risk HPV-associated diseases. PAPILLOMAVIRUS RESEARCH (AMSTERDAM, NETHERLANDS) 2018; 5:46-58. [PMID: 29277575 PMCID: PMC5887015 DOI: 10.1016/j.pvr.2017.12.006] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 12/13/2017] [Accepted: 12/17/2017] [Indexed: 12/16/2022]
Abstract
Cancer is the second leading cause of death worldwide, and it is estimated that Human papillomavirus (HPV) related cancers account for 5% of all human cancers. Current HPV vaccines are extremely effective at preventing infection and neoplastic disease; however, they are prophylactic and do not clear established infections. Therapeutic vaccines which trigger cell-mediated immune responses for the treatment of established infections and malignancies are therefore required. The E6 and E7 early genes are ideal targets for vaccine therapy due to their role in disruption of the cell cycle and their constitutive expression in premalignant and malignant tissues. Several strategies have been investigated for the development of therapeutic vaccines, including live-vector, nucleic acid, peptide, protein-based and cell-based vaccines as well as combinatorial approaches, with several vaccine candidates progressing to clinical trials. With the current understanding of the HPV life cycle, molecular mechanisms of infection, carcinogenesis, tumour biology, the tumour microenvironment and immune response mechanisms, an approved HPV therapeutic vaccine seems to be a goal not far from being achieved. In this article, the status of therapeutic HPV vaccines in clinical trials are reviewed, and the potential for plant-based vaccine production platforms described.
Collapse
Affiliation(s)
- Aleyo Chabeda
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa
| | - Romana J R Yanez
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa
| | - Renate Lamprecht
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa
| | - Ann E Meyers
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa
| | - Edward P Rybicki
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Inga I Hitzeroth
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa.
| |
Collapse
|
27
|
Cornel AM, van Til NP, Boelens JJ, Nierkens S. Strategies to Genetically Modulate Dendritic Cells to Potentiate Anti-Tumor Responses in Hematologic Malignancies. Front Immunol 2018; 9:982. [PMID: 29867960 PMCID: PMC5968097 DOI: 10.3389/fimmu.2018.00982] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/20/2018] [Indexed: 12/12/2022] Open
Abstract
Dendritic cell (DC) vaccination has been investigated as a potential strategy to target hematologic malignancies, while generating sustained immunological responses to control potential future relapse. Nonetheless, few clinical trials have shown robust long-term efficacy. It has been suggested that a combination of surmountable shortcomings, such as selection of utilized DC subsets, DC loading and maturation strategies, as well as tumor-induced immunosuppression may be targeted to maximize anti-tumor responses of DC vaccines. Generation of DC from CD34+ hematopoietic stem and progenitor cells (HSPCs) may provide potential in patients undergoing allogeneic HSPC transplantations for hematologic malignancies. CD34+ HSPC from the graft can be genetically modified to optimize antigen presentation and to provide sufficient T cell stimulatory signals. We here describe beneficial (gene)-modifications that can be implemented in various processes in T cell activation by DC, among which major histocompatibility complex (MHC) class I and MHC class II presentation, DC maturation and migration, cross-presentation, co-stimulation, and immunosuppression to improve anti-tumor responses.
Collapse
Affiliation(s)
- Annelisa M Cornel
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Niek P van Til
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jaap Jan Boelens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Pediatric Blood and Marrow Transplantation Program, University Medical Center Utrecht, Utrecht, Netherlands.,Blood and Marrow Transplantation Program, Princess Maxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Stefan Nierkens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
28
|
Thepmalee C, Panya A, Junking M, Chieochansin T, Yenchitsomanus PT. Inhibition of IL-10 and TGF-β receptors on dendritic cells enhances activation of effector T-cells to kill cholangiocarcinoma cells. Hum Vaccin Immunother 2018; 14:1423-1431. [PMID: 29420117 DOI: 10.1080/21645515.2018.1431598] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Tumor escapes host immune responses by producing immunosuppressive cytokines, such as IL-10 and TGF-β, secreted into the tumor microenvironment. These cytokines play important roles in the suppression of dendritic cell (DC) function, leading to decreased immune responses of the effector CD4+ and CD8+ T cells. To improve DC functions and enhance cytolytic activity of activated effector T-cells, we suppressed the effect of these cytokines on DCs by using specific neutralizing antibodies that inhibit IL-10 and TGF-β receptors. Monocyte-derived DCs generated in vitro showed up-regulation of MHC (HLA-DR) and co-stimulatory molecules (CD40 and CD86). The IL-10 and TGF-β receptors were expressed and localized on cell membrane of DCs, as shown by Western blot analysis and immunofluorescence staining, whereas the IL-10 and TGF-β ligands were detected in the culture supernatants of DCs and cholangiocarcinoma (CCA) cell line, respectively. Inhibition of the IL-10 and TGF-β receptors on DCs by specific neutralizing antibodies significantly increased level of IFN-γ and enhanced cytolytic activity of the DC-activated effector T-cells against CCA cell line. These results indicate that the IL-10 and TGF-β receptors are the targets for inhibition to increase DC functions and enhance cytolytic activity of the DC-activated effector T-cells against CCA cells. Thus, inhibition of the IL-10 and TGF-β receptors on DCs is crucial in the preparation of DC-activated effector T cells for adoptive T-cell therapy.
Collapse
Affiliation(s)
- Chutamas Thepmalee
- a Division of Molecular Medicine , Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok , Thailand.,b Graduate Program in Immunology , Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok , Thailand.,c Division of Biochemistry and Nutrition, School of Medical Science, University of Phayao , Phayao , Thailand
| | - Aussara Panya
- a Division of Molecular Medicine , Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok , Thailand.,d Department of Biology , Faculty of Science, Chiang Mai University , Chiang Mai , Thailand
| | - Mutita Junking
- a Division of Molecular Medicine , Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok , Thailand
| | - Thaweesak Chieochansin
- a Division of Molecular Medicine , Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok , Thailand
| | - Pa-Thai Yenchitsomanus
- a Division of Molecular Medicine , Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok , Thailand
| |
Collapse
|
29
|
Meng Q, Pan J, Liu Y, Chen L, Ren Y. Anti-tumour effects of polysaccharide extracted from Acanthopanax senticosus and cell-mediated immunity. Exp Ther Med 2018; 15:1694-1701. [PMID: 29434755 PMCID: PMC5774378 DOI: 10.3892/etm.2017.5568] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 09/22/2017] [Indexed: 12/30/2022] Open
Abstract
Acanthopanax senticosus, also known as Siberian ginseng, is widely distributed throughout northern Asia and used in traditional Chinese medicine; it has been reported to prevent a number of diseases. However, the association between the antitumour and immunostimulatory activities of polysaccharide extracted from A. senticosus (ASPS) remains to be elucidated. The aim of the present study was to investigate the anti-tumour and immunomodulatory effects of polysaccharide extracted from ASPS on Crocker sarcoma S180, hepatic carcinoma H22 and uterine cervical carcinoma U14 tumour cell lines implanted in mice. High performance liquid chromatography, gas chromatography and infrared spectroscopy were used to analyse the monosaccharide composition of ASPS. The monosaccharide composition of ASPS (Arabic candy: Xylose: Glucose: Mannose) was 7.1:22.3:7.6:1.0. On day 0, female Kunming mice, were injected subcutaneously with 1×108 tumour cells in 0.2 ml. The inoculated mice were subsequently divided into five groups (10 mice/group) as follows: Model group, treated with normal saline; positive control group, treated with 30 mg/kg cyclophosphamide (CTX); and three treatment groups, treated with 200, 100 or 50 mg/kg ASPS. Non-inoculated mice were divided into the normal group, which was treated with normal saline, and the negative control group, which was treated with 200 mg/kg ASPS (n=10/group). CTX and ASPS were administered intragastrically once daily for 10 days. All mice were sacrificed on day 11. ASPS was observed to have an inhibitory effect on the growth of S180, H22 and U14 cells in solid and ascites tumour-bearing mice. Serum interleukin (IL)-2 and IL-12 levels were significantly increased in S180 solid tumour-bearing mice treated with 200 or 100 mg/kg ASPS compared with mice in the normal, control and model groups (P<0.05), whereas serum IL-2 and IL-12 levels were significantly decreased in the cyclophosphamide treatment group compared with the normal, control and model groups (P<0.05). No significant difference in serum levels of tumour necrosis factor-α level was observed between any groups. In S180 and U14 solid tumour-bearing mice, no significant differences in serum levels of interferon (INF)-γ level in were observed between groups; however, in H22 solid tumour-bearing mice, treatment with ASPS significantly increased serum INF-γ compared with the positive control group (P<0.05). The results may provide a basis for the potential application of ASPS in clinical treatment for cancer.
Collapse
Affiliation(s)
- Qinglong Meng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin 130118, P.R. China
| | - Jingzhi Pan
- Tuberculous Meningitis Research Center, Infectious Disease Hospital, Changchun, Jilin 130123, P.R. China
| | - Yajing Liu
- College of Plant Science, Jilin University, Changchun, Jilin 130062, P.R. China
| | - Li Chen
- Innovation and Development Centre of Small and Medium Enterprises, Siping, Jilin 136000, P.R. China
| | - Yueying Ren
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin 130118, P.R. China
| |
Collapse
|
30
|
Qin S, Gao Z, Liu Y, Liu C, Wang J, Zou LL. Silencing of suppressor of cytokine signaling 1 enhances the immunological effect of mucin 1-calreticulin-primed 4T1 cell-treated dendritic cells in breast cancer treatment. Oncol Lett 2017; 15:1630-1638. [PMID: 29434859 PMCID: PMC5774391 DOI: 10.3892/ol.2017.7477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 09/28/2017] [Indexed: 11/06/2022] Open
Abstract
In cancer immunotherapy, dendritic cell (DC)-based vaccines represent a promising, yet challenging, treatment method. In addition to overcoming the low expression levels of antigenic epitopes on cancer cells, it is also necessary to overcome the inhibitory effect of suppressor of cytokine signaling 1 (SOCS1) on DC self-antigen presentation. Our group previously demonstrated that calreticulin (CRT) translocated type I transmembrane glycoprotein mucin 1 (MUC1), a breast cancer antigen, to the surface of 4T1 cells, and that treatment with MUC1-CRT-primed 4T1 cell-treated DCs induced apoptosis in a breast cancer cell line. In the present study, cell penetrate peptide, hpp10-DRBD was successfully used to deliver siRNAs into bone marrow-derived (BM) DCs to construct SOCS1-silenced DCs, which were incubated with MUC1-CRT-primed 4T1 cells, and antigen-specific antitumor immunity was markedly enhanced in vitro and in vivo. These results demonstrated that SOCS1-silencing, combined with MUC1-CRT-primed 4T1 cell treatment, may induce increased cytokine production and T cell proliferation by DCs. Furthermore, the in vivo experimental data demonstrated that the silencing of SOCS1 combined with MUC1-CRT-primed 4T1 treatment of BMDCs may induce enhanced immunological effects. The results of the present study have implications for the development of more effective DC-based tumor vaccines, suggesting that the combination of high tumor-associated antigen expression levels on cancer cells with the silencing of a critical inhibitor of DC antigen presentation may be beneficial.
Collapse
Affiliation(s)
- Song Qin
- Translational Neuroscience and Neural Regeneration and Repair Institute/Institute of Cell Therapy, The First Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Zhipeng Gao
- Translational Neuroscience and Neural Regeneration and Repair Institute/Institute of Cell Therapy, The First Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Yu Liu
- Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Department of Oncology, Gong'an County Hospital, Yichang, Hubei 434300, P.R. China
| | - Changbai Liu
- Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Jun Wang
- Translational Neuroscience and Neural Regeneration and Repair Institute/Institute of Cell Therapy, The First Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Li Li Zou
- Translational Neuroscience and Neural Regeneration and Repair Institute/Institute of Cell Therapy, The First Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443002, P.R. China.,Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| |
Collapse
|
31
|
Wang J, Gao ZP, Qin S, Liu CB, Zou LL. Calreticulin is an effective immunologic adjuvant to tumor-associated antigens. Exp Ther Med 2017; 14:3399-3406. [PMID: 29042925 DOI: 10.3892/etm.2017.4989] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/24/2017] [Indexed: 12/18/2022] Open
Abstract
As a key molecule involved in cell recognition, calreticulin (CRT) may be expressed on the surface of (pre-) apoptotic cells and provide the signal that is recognized by dendritic cells (DCs) or other antigen presenting cells (APCs), which results in phagocytosis. Within the APCs, tumor-associated antigens (TAAs) may be subsequently presented to T lymphocytes, which triggers a specific antitumor immune response. It has been hypothesized that CRT is able to act as the immunologic adjuvant and translocate itself and TAAs to the cell surface and induce a specific antitumor immune response. In the present study, CRT was demonstrated to translocate itself and mucin 1 (MUC1), a breast cancer antigen, to the surface of 4T1 cells and the MUC1-CRT-coated cells were able to induce apoptosis in a time-dependent manner. When DCs were infected with adenovirus containing MUC1-CRT, an increase in T cell proliferation and cytokine production was exhibited. These results suggest that CRT may act as an immunologic adjuvant with MUC1 and induce a strong immune response.
Collapse
Affiliation(s)
- Jun Wang
- Translational Neuroscience and Neural Regeneration and Repair Institute/Institute of Cell Therapy, The First People's Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Zhi Peng Gao
- Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Song Qin
- Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Chang Bai Liu
- Translational Neuroscience and Neural Regeneration and Repair Institute/Institute of Cell Therapy, The First People's Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Li Li Zou
- Translational Neuroscience and Neural Regeneration and Repair Institute/Institute of Cell Therapy, The First People's Hospital of Yichang, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| |
Collapse
|
32
|
Ramachandran M, Dimberg A, Essand M. The cancer-immunity cycle as rational design for synthetic cancer drugs: Novel DC vaccines and CAR T-cells. Semin Cancer Biol 2017; 45:23-35. [DOI: 10.1016/j.semcancer.2017.02.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/26/2017] [Indexed: 01/18/2023]
|
33
|
Abraham RS, Mitchell DA. Gene-modified dendritic cell vaccines for cancer. Cytotherapy 2017; 18:1446-1455. [PMID: 27745604 DOI: 10.1016/j.jcyt.2016.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/16/2016] [Indexed: 12/13/2022]
Abstract
Dendritic cell (DC) vaccines are an immunotherapeutic approach to cancer treatment that use the antigen-presentation machinery of DCs to activate an endogenous anti-tumor response. In this treatment strategy, DCs are cultured ex vivo, exposed to tumor antigens and administered to the patient. The ex vivo culturing provides a unique and powerful opportunity to modify and enhance the DCs. As such, a variety of genetic engineering approaches have been employed to optimize DC vaccines, including the introduction of messenger RNA and small interfering RNA, viral gene transduction, and even fusion with whole tumor cells. In general, these modifications aim to improve targeting, enhance immunogenicity, and reduce susceptibility to the immunosuppressive tumor microenvironment. It has been demonstrated that several of these modifications can be employed in tandem, allowing for fine-tuning and optimization of the DC vaccine across multiple metrics. Thus, the application of genetic engineering techniques to the dendritic cell vaccine platform has the potential to greatly enhance its efficacy in the clinic.
Collapse
Affiliation(s)
- Rebecca S Abraham
- UF Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Department of Neurosurgery, University of Florida, Gainesville, FL 32605
| | - Duane A Mitchell
- UF Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Department of Neurosurgery, University of Florida, Gainesville, FL 32605.
| |
Collapse
|
34
|
Ita K. Dermal/transdermal delivery of small interfering RNA and antisense oligonucleotides- advances and hurdles. Biomed Pharmacother 2017; 87:311-320. [PMID: 28064104 DOI: 10.1016/j.biopha.2016.12.118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/26/2016] [Accepted: 12/27/2016] [Indexed: 10/20/2022] Open
Abstract
A diverse array of nucleic acids has been studied by several researchers for the management of several diseases. Among these compounds, small interfering RNA and antisense oligonucleotides have attracted considerable attention. Antisense oligonucleotides are synthetic single stranded strings of nucleic acids that bind to RNA and thereby alter or reduce expression of the target RNA while siRNAs, on the other hand, are double-stranded RNA molecules which can hybridize with a specific mRNA sequence and block the translation of numerous genes. One of the main obstacles in the dermal or transdermal delivery of these compounds is their low skin permeability. In this review, various techniques used to enhance the delivery of these molecules into or across the skin are described and in some cases, the correlation between enhanced dermal/transdermal delivery and therapeutic efficacy is highlighted.
Collapse
Affiliation(s)
- Kevin Ita
- College of Pharmacy, Touro University, Mare Island-Vallejo, CA 94592, USA.
| |
Collapse
|
35
|
Yang A, Farmer E, Wu TC, Hung CF. Perspectives for therapeutic HPV vaccine development. J Biomed Sci 2016; 23:75. [PMID: 27809842 PMCID: PMC5096309 DOI: 10.1186/s12929-016-0293-9] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 10/26/2016] [Indexed: 12/24/2022] Open
Abstract
Background Human papillomavirus (HPV) infections and associated diseases remain a serious burden worldwide. It is now clear that HPV serves as the etiological factor and biologic carcinogen for HPV-associated lesions and cancers. Although preventative HPV vaccines are available, these vaccines do not induce strong therapeutic effects against established HPV infections and lesions. These concerns create a critical need for the development of therapeutic strategies, such as vaccines, to treat these existing infections and diseases. Main Body Unlike preventative vaccines, therapeutic vaccines aim to generate cell-mediated immunity. HPV oncoproteins E6 and E7 are responsible for the malignant progression of HPV-associated diseases and are consistently expressed in HPV-associated diseases and cancer lesions; therefore, they serve as ideal targets for the development of therapeutic HPV vaccines. In this review we revisit therapeutic HPV vaccines that utilize this knowledge to treat HPV-associated lesions and cancers, with a focus on the findings of recent therapeutic HPV vaccine clinical trials. Conclusion Great progress has been made to develop and improve novel therapeutic HPV vaccines to treat existing HPV infections and diseases; however, there is still much work to be done. We believe that therapeutic HPV vaccines have the potential to become a widely available and successful therapy to treat HPV and HPV-associated diseases in the near future.
Collapse
Affiliation(s)
- Andrew Yang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Emily Farmer
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - T C Wu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.,Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, MD, USA.,Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA. .,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA. .,The Johns Hopkins University School of Medicine, CRB II Room 307, 1550 Orleans Street, Baltimore, MD, 21231, USA.
| |
Collapse
|
36
|
Dysregulation of TGFβ1 Activity in Cancer and Its Influence on the Quality of Anti-Tumor Immunity. J Clin Med 2016; 5:jcm5090076. [PMID: 27589814 PMCID: PMC5039479 DOI: 10.3390/jcm5090076] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/26/2016] [Accepted: 08/29/2016] [Indexed: 01/01/2023] Open
Abstract
TGFβ1 is a pleiotropic cytokine that exhibits a variety of physiologic and immune regulatory functions. Although its influence on multiple cell types is critical for the regulation of numerous biologic processes in the host, dysregulation of both TGFβ1 expression and activity is frequently observed in cancer and contributes to various aspects of cancer progression. This review focuses on TGFβ1’s contribution to tumor immune suppression and escape, with emphasis on the influence of this regulatory cytokine on the differentiation and function of dendritic cells and T cells. Clinical trials targeting TGFβ1 in cancer patients are also reviewed, and strategies for future therapeutic interventions that build on our current understanding of immune regulation by TGFβ1 are discussed.
Collapse
|
37
|
Yang A, Jeang J, Cheng K, Cheng T, Yang B, Wu TC, Hung CF. Current state in the development of candidate therapeutic HPV vaccines. Expert Rev Vaccines 2016; 15:989-1007. [PMID: 26901118 DOI: 10.1586/14760584.2016.1157477] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The identification of human papillomavirus (HPV) as an etiological factor for HPV-associated malignancies creates the opportunity to control these cancers through vaccination. Currently, available preventive HPV vaccines have not yet demonstrated strong evidences for therapeutic effects against established HPV infections and lesions. Furthermore, HPV infections remain extremely common. Thus, there is urgent need for therapeutic vaccines to treat existing HPV infections and HPV-associated diseases. Therapeutic vaccines differ from preventive vaccines in that they are aimed at generating cell-mediated immunity rather than neutralizing antibodies. The HPV-encoded early proteins, especially oncoproteins E6 and E7, form ideal targets for therapeutic HPV vaccines since they are consistently expressed in HPV-associated malignancies and precancerous lesions, playing crucial roles in the generation and maintenance of HPV-associated disease. Our review will cover various therapeutic vaccines in development for the treatment of HPV-associated lesions and cancers. Furthermore, we review strategies to enhance vaccine efficacy and the latest clinical trials on therapeutic HPV vaccines.
Collapse
Affiliation(s)
- Andrew Yang
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Jessica Jeang
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Kevin Cheng
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Ting Cheng
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Benjamin Yang
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - T-C Wu
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA.,b Department of Obstetrics and Gynecology , Johns Hopkins University , Baltimore , MD , USA.,c Department of Molecular Microbiology and Immunology , Johns Hopkins University , Baltimore , MD , USA.,d Department of Oncology , Johns Hopkins University , Baltimore , MD , USA
| | - Chien-Fu Hung
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA.,d Department of Oncology , Johns Hopkins University , Baltimore , MD , USA
| |
Collapse
|
38
|
Endostatin gene therapy inhibits intratumoral macrophage M2 polarization. Biomed Pharmacother 2016; 79:102-11. [PMID: 27044818 DOI: 10.1016/j.biopha.2016.01.035] [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: 10/28/2015] [Revised: 01/27/2016] [Accepted: 01/27/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) is a highly vascularized cancer resistant to chemotherapy and radiotherapy. RCC is frequently infiltrated with immune cells, with macrophages being the most abundant cell type. Alternatively activated M2 macrophages are known to contribute to tumor progression. Endostatin (ES) is a fragment of collagen XVIII that possesses antiangiogenic activity. In this study, we investigated the impact of ES gene therapy on the polarization of tumor-associated macrophages (TAMs) in lung metastases from tumor-bearing mice. METHODS BALB/c mice divided into three groups: Normal, Control and ES-treated. Tumor-bearing mice were treated with ES-transduced cells or control cells over ten days. At the end of the study, plasma was collected, and pulmonary macrophages were isolated and used for FACS or RT-PCR. ELISA tests were used to analyze plasma and cell culture supernatant cytokines. RESULTS ES treatment significantly reduced the levels of anti-inflammatory and pro-angiogenic cytokines, including IL4, IL-10, IL-13 and VEGF. Gene expression of M2 markers, such as IL-10, Arg-1, VEGF and YM-1, declined significantly. Flow cytometry showed a reduction in the number of M2 F4/80+CD36+CD206+CD209+ macrophages and in IL-10 secretion by these cells. Reduced levels of IL-10 were also found in the culture supernatants of the ES-treated group. CONCLUSIONS Our research corroborates previous observations that ES has an important anti-tumoral role. However, aside from promoting interferon-ɤ secretion and an effective T cell response, we show here that this switch is extended to TAMs, complicating the maintenance of pro-tumorigenic M2 macrophages and thus favoring tumor elimination.
Collapse
|
39
|
Beavis PA, Slaney CY, Kershaw MH, Gyorki D, Neeson PJ, Darcy PK. Reprogramming the tumor microenvironment to enhance adoptive cellular therapy. Semin Immunol 2016; 28:64-72. [DOI: 10.1016/j.smim.2015.11.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/29/2015] [Accepted: 11/04/2015] [Indexed: 12/22/2022]
|
40
|
de Aquino MTP, Malhotra A, Mishra MK, Shanker A. Challenges and future perspectives of T cell immunotherapy in cancer. Immunol Lett 2015; 166:117-33. [PMID: 26096822 PMCID: PMC4499494 DOI: 10.1016/j.imlet.2015.05.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 05/10/2015] [Accepted: 05/27/2015] [Indexed: 12/15/2022]
Abstract
Since the formulation of the tumour immunosurveillance theory, considerable focus has been on enhancing the effectiveness of host antitumour immunity, particularly with respect to T cells. A cancer evades or alters the host immune response by various ways to ensure its development and survival. These include modifications of the immune cell metabolism and T cell signalling. An inhibitory cytokine milieu in the tumour microenvironment also leads to immune suppression and tumour progression within a host. This review traces the development in the field and attempts to summarize the hurdles that the approach of adoptive T cell immunotherapy against cancer faces, and discusses the conditions that must be improved to allow effective eradication of cancer.
Collapse
Affiliation(s)
- Maria Teresa P de Aquino
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Anshu Malhotra
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Manoj K Mishra
- Department of Biological Sciences, Alabama State University, Montgomery, AL 36101, USA
| | - Anil Shanker
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; Tumor-Host Interactions Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
41
|
Panatto D, Amicizia D, Bragazzi NL, Rizzitelli E, Tramalloni D, Valle I, Gasparini R. Human Papillomavirus Vaccine. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015:231-322. [DOI: 10.1016/bs.apcsb.2015.08.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|