1
|
Shao Y, Shah PT, Su Q, Li S, Huang F, Wang J, Wang P, Wu C. Recombinant adenoviruses expressing HPV16/18 E7 upregulate the HDAC6 and DNMT3B genes in C33A cells. Front Cell Infect Microbiol 2024; 14:1459572. [PMID: 39411320 PMCID: PMC11473514 DOI: 10.3389/fcimb.2024.1459572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
Objective High-risk human papillomavirus (HPV) is a carcinogenic virus associated with nearly all cases of cervical cancer, as well as an increasing number of anal and oral cancers. The two carcinogenic proteins of HPV, E6 and E7, can immortalize keratinocytes and are essential for HPV-related cellular transformation. Currently, the global regulatory effects of these oncogenic proteins on the host proteome are not fully understood, and further exploration of the functions and carcinogenic mechanisms of E6 and E7 proteins is needed. Methods We used a previously established platform in our laboratory for constructing recombinant adenoviral plasmids expressing the HPV16 E7 gene to further construct recombinant virus particles expressing HPV16/18 E6, E7, and both E6 and E7 genes. These recombinant viruses were used to infect C33A cells to achieve sustained expression of the HPV16/18 E6/E7 genes. Subsequently, total RNA was extracted and RNA-Seq technology was employed for transcriptome sequencing to identify differentially expressed genes associated with HPV infection in cervical cancer. Results RNA-Seq analysis revealed that overexpression of the HPV16/18 E6/E7 genes upregulated GP6, CD36, HDAC6, ESPL1, and DNMT3B among the differentially expressed genes (DEGs) associated with cervical cancer. Spearman correlation analysis revealed a statistically significant correlation between the HDAC6 and DNMT3B genes and key pathways, including DNA replication, tumor proliferation signature, G2M checkpoint, p53 pathways, and PI3K/AKT/mTOR signaling pathways. Further, qRT-PCR and Western blot analyses indicated that both HPV16/18 E7 can upregulate the expression of HDAC6 and DNMT3B, genes associated with HPV infection-related cervical cancer. Conclusion The successful expression of HPV16/18 E6/E7 in cells indicates that the recombinant viruses retain the replication and infection capabilities of Ad4. Furthermore, the recombinant viruses expressing HPV16/18 E7 can upregulate the HDAC6 and DNMT3B genes involved in cervical cancer pathways, thereby influencing the cell cycle. Additionally, HDAC6 and DNMT3B are emerging as important therapeutic targets for cancer. This study lays the foundation for further exploration of the oncogenic mechanisms of HPV E6/E7 and may provide new directions for the treatment of HPV-related cancers.
Collapse
Affiliation(s)
- Yunting Shao
- Faculty of Medicine, School of Basic Medical Sciences, Dalian University of Technology, Dalian, China
| | - Pir Tariq Shah
- Faculty of Medicine, School of Basic Medical Sciences, Dalian University of Technology, Dalian, China
| | - Qisheng Su
- Faculty of Medicine, School of Basic Medical Sciences, Dalian University of Technology, Dalian, China
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Fang Huang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Jun Wang
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Dalian Medical University Mailing, Dalian, China
| | - Peng Wang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Chengjun Wu
- Faculty of Medicine, School of Basic Medical Sciences, Dalian University of Technology, Dalian, China
| |
Collapse
|
2
|
Goodarzi MM, Mosayebi G, Ganji A, Raoufi E, Sadelaji S, Babaei S, Abtahi H. HPV16 mutant E6/E7 construct is protective in mouse model. BMC Biotechnol 2024; 24:71. [PMID: 39350162 PMCID: PMC11443707 DOI: 10.1186/s12896-024-00893-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Human papillomavirus type 16 (HPV-16) infection is strongly associated with considerable parts of cervical, neck, and head cancers. Performed investigations have had moderate clinical success, so research to reach an efficient vaccine has been of great interest. In the present study, the immunization potential of a newly designed HPV-16 construct was evaluated in a mouse model. RESULTS Initially, a construct containing HPV-16 mutant (m) E6/E7 fusion gene was designed and antigen produced in two platforms (i.e., DNA vaccine and recombinant protein). Subsequently, the immunogenicity of these platforms was investigated in five mice) C57BL/6 (groups based on several administration strategies. Three mice groups were immunized recombinant protein, DNA vaccine, and a combination of them, and two other groups were negative controls. The peripheral blood mononuclear cells (PBMCs) proliferation, Interleukin-5 (IL-5) and interferon-γ (IFN-γ) cytokines, IgG1 and IgG2a antibody levels were measured. After two weeks, TC-1 tumor cells were injected into all mice groups, and subsequently further analysis of tumor growth and metastasis and mice survival were performed according to the schedule. Overall, the results obtained from in vitro immunology and tumor cells challenging assays indicated the potential of the mE6/E7 construct as an HPV16 therapeutic vaccine candidate. The results demonstrated a significant increase in IFN-γ cytokine (P value < 0.05) in the Protein/Protein (D) and DNA/Protein (E) groups. This finding was in agreement with in vivo assays. Control groups show a 10.5-fold increase (P value < 0.001) and (C) DNA/DNA group shows a 2.5-fold increase (P value < 0.01) in tumor growth compared to D and E groups. Also, a significant increase in survival of D and E (P value < 0.001) and C (P value < 0.01) groups were observed. CONCLUSIONS So, according to the findings, the recombinant protein could induce stronger protection compared to the DNA vaccine form. Protein/Protein and DNA/Protein are promising administration strategies for presenting this construct to develop an HPV-16 therapeutic vaccine candidate.
Collapse
Affiliation(s)
- Maryam Moazami Goodarzi
- Department of Medical Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ghasem Mosayebi
- Department of Microbiology and Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ali Ganji
- Department of Microbiology and Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ehsan Raoufi
- Vaccine research center, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Sadelaji
- Department of Microbiology and Immunology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Saeid Babaei
- Department of Anatomy, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Hamid Abtahi
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran.
| |
Collapse
|
3
|
Wang J, Lv F, Sun T, Zhao S, Chen H, Liu Y, Liu Z. Sorafenib Nanomicelles Effectively Shrink Tumors by Vaginal Administration for Preoperative Chemotherapy of Cervical Cancer. NANOMATERIALS 2021; 11:nano11123271. [PMID: 34947619 PMCID: PMC8705954 DOI: 10.3390/nano11123271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022]
Abstract
To investigate the potential of sorafenib (SF) in preoperative chemotherapy for cervical cancer to reduce tumor volume, sorafenib micelles (SF micelles) with good stability and high drug loading were designed. SF micelles were prepared by film hydration followed by the ultrasonic method. The results showed that the SF micelles were spherical with an average particle size of 67.18 ± 0.66 nm (PDI 0.17 ± 0.01), a considerable drug loading of 15.9 ± 0.46% (w/w%) and satisfactory stability in buffers containing plasma or not for at least 2 days. In vitro release showed that SF was gradually released from SF micelles and almost completely released on the third day. The results of in vitro cellular intake, cytotoxicity and proliferation of cervical cancer cell TC-1 showed that SF micelles were superior to sorafenib (Free SF). For intravaginal administration, SF micelles were dispersed in HPMC (SF micelles/HPMC), showed good viscosity sustained-release profiles in vitro and exhibited extended residence in intravaginal in vivo. Compared with SF micelles dispersed in N.S. (SF micelles/N.S.), SF micelles/HPMC significantly reduced tumor size with a tumor weight inhibition rate of 73%. The results suggested that SF micelles had good potential for preoperative tumor shrinkage and improving the quality life of patients.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China;
| | - Fengmei Lv
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (F.L.); (T.S.); (S.Z.); (H.C.)
| | - Tao Sun
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (F.L.); (T.S.); (S.Z.); (H.C.)
| | - Shoujin Zhao
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (F.L.); (T.S.); (S.Z.); (H.C.)
| | - Haini Chen
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (F.L.); (T.S.); (S.Z.); (H.C.)
| | - Yu Liu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China;
- Correspondence: (Y.L.); (Z.L.)
| | - Zhepeng Liu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (F.L.); (T.S.); (S.Z.); (H.C.)
- Correspondence: (Y.L.); (Z.L.)
| |
Collapse
|
4
|
Silva MO, Almeida BS, Sales NS, Diniz MO, Aps LRMM, Rodrigues KB, Silva JR, Moreno ACR, Porchia BFMM, Sulczewski FB, Boscardin SB, Ferreira LCS. Antigen Delivery to DEC205 + Dendritic Cells Induces Immunological Memory and Protective Therapeutic Effects against HPV-Associated Tumors at Different Anatomical Sites. Int J Biol Sci 2021; 17:2944-2956. [PMID: 34345218 PMCID: PMC8326119 DOI: 10.7150/ijbs.57038] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 05/02/2021] [Indexed: 12/27/2022] Open
Abstract
The generation of successful anticancer vaccines relies on the ability to induce efficient and long-lasting immune responses to tumor antigens. In this scenario, dendritic cells (DCs) are essential cellular components in the generation of antitumor immune responses. Thus, delivery of tumor antigens to specific DC populations represents a promising approach to enhance the efficiency of antitumor immunotherapies. In the present study, we employed antibody-antigen conjugates targeting a specific DC C-type lectin receptor. For that purpose, we genetically fused the anti-DEC205 monoclonal antibody to the type 16 human papillomavirus (HPV-16) E7 oncoprotein to create a therapeutic vaccine to treat HPV-associated tumors in syngeneic mouse tumor models. The therapeutic efficacy of the αDEC205-E7 mAb was investigated in three distinct anatomical tumor models (subcutaneous, lingual and intravaginal). The immunization regimen comprised two doses of the αDEC205-E7 mAb coadministered with a DC maturation stimulus (Polyinosinic:polycytidylic acid, poly (I:C)) as an adjuvant. The combined immunotherapy produced robust antitumor effects on both the subcutaneous and orthotopic tumor models, stimulating rapid tumor regression and long-term survival. These outcomes were related to the activation of tumor antigen-specific CD8+ T cells in both systemic compartments and lymphoid tissues. The αDEC205-E7 antibody plus poly (I:C) administration induced long-lasting immunity and controlled tumor relapses. Our results highlight that the delivery of HPV tumor antigens to DCs, particularly via the DEC205 surface receptor, is a promising therapeutic approach, providing new opportunities for the development of alternative immunotherapies for patients with HPV-associated tumors at different anatomical sites.
Collapse
Affiliation(s)
- Mariângela O Silva
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Bianca S Almeida
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Natiely S Sales
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mariana O Diniz
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Luana R M M Aps
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Karine B Rodrigues
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Jamile R Silva
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Ana C R Moreno
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Bruna F M M Porchia
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Fernando B Sulczewski
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Silvia B Boscardin
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Luís C S Ferreira
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
5
|
Zhang C, Chen H, Deng Z, Long D, Xu L, Liu Z. DGCR8/miR-106 Axis Enhances Radiosensitivity of Head and Neck Squamous Cell Carcinomas by Downregulating RUNX3. Front Med (Lausanne) 2020; 7:582097. [PMID: 33385002 PMCID: PMC7770216 DOI: 10.3389/fmed.2020.582097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Abstract
Purpose: Head and neck squamous cell carcinoma (HNSCC) is the sixth most prevalent malignant tumor worldwide, and the radiotherapy effect is strongly associated with human papillomavirus (HPV) infection. Therefore, the aim of our study was to analyze the mechanism of HPV E7 and its effects on radiosensitivity in HNSCC cells. Methods: The mRNA expression of DiGeorge syndrome critical region gene 8 (DGCR8), has-miR-106a, and Runt-related transcription factor 3 (RUNX3) was examined by quantitative real-time PCR (RT-qPCR). The protein expression of DGCR8, E7, RUNX3, caspase-3/cleaved caspase-3, poly(ADP-ribose) polymerase (PARP)/cleaved PARP, and γH2AX was measured by Western blot. The expression level of DGCR8 was measured by immunofluorescence assay. Starbase database (http://starbase.sysu.edu.cn/) was used to analyze the correlation between has-miR-106a-5p and DGCR8. TargetScan database (http://www.targetscan.org/vert_72/) was adopted to calculate the prediction of binding sites. Radiosensitivity was evaluated through clone formation assays and Cell Counting Kit-8 (CCK-8) assays. Results: In our study, we found that the mRNA and protein expression levels of HPV E7 and DGCR8 in HPV-positive HNSCC cells were higher than those in HPV-negative cells. The expression of DGCR8 was increased in FaDu and UM-SCC-4 with E7 overexpression, while the expression of DGCR8 was decreased in UM-SCC-47 and UPCI-SCC-090 with E7 silence. The miR-106a expression was increased after DGCR8 overexpression in FaDu and UM-SCC-4. However, the miR-106a expression was decreased in UM-SCC-47 and UPCI-SCC-090 with E7 silence. In radiation conditions, clone formation assays found that less clones formed in FaDu and UM-SCC-4 cells subsequent to silencing DGCR8 or miR-106a than that in the control group, and more clones were formed in UM-SCC-47 and UPCI-SCC-090 cells overexpressing DGCR8 or miR-106a than that in the control group. Luciferase reporter gene assays verified that miR-106a targeted the 3' untranslated region (UTR) of RUNX3 mRNA. MiR-106a overexpression resulted in a decrease in RUNX3 expression, and miR-106a silence increased RUNX3 expression. Rescue experiments conducted with miR-106a inhibitor restored radiation resistance and reduced DNA damage in radiation condition. Conclusions: Our study indicated that HPV E7 activated DGCR8/miR-106a/RUNX3 axis to enhance radiation sensitivity and provided directions for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Chunlin Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hangqi Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zeyi Deng
- Department of Otorhinolaryngology, Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Dan Long
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Li Xu
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhaohui Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
6
|
Li S, Zhu W, Ye C, Sun W, Xie H, Yang X, Zhang Q, Ma Y. Local mucosal immunization of self-assembled nanofibers elicits robust antitumor effects in an orthotopic model of mouse genital tumors. NANOSCALE 2020; 12:3076-3089. [PMID: 31965136 DOI: 10.1039/c9nr10334a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Human papillomavirus (HPV) is the identified causative agent of cervical cancer. Current therapeutic HPV vaccine candidates lack significant clinical efficacy, which can be attributed to insufficient activation of effector cells, lack of effective modification of the immunosuppressive tumor microenvironment, and the limitations of applied tumor models for preclinical vaccine evaluation. Here, a mouse model of orthotopic genital tumors was used to assess the effect of self-assembled nanofibers on eliciting a robust antitumor response via local mucosal immunization. A candidate vaccine was obtained by fusing HPV16 E744-62 to the self-assembling peptide Q11, which was assembled into nanofibers in a salt solution. Mice bearing an established genital TC-1 tumor were immunized with nanofibers through the intravaginal, intranasal, or subcutaneous route. Mucosal vaccination, especially via the intravaginal route, was more effective for suppressing tumor growth than subcutaneous immunization. The potential underlying mechanisms include promoting the systemic generation and tumor accumulation of antigen-specific cytotoxic T lymphocytes expressing high levels of interferon (IFN)-γ or granzyme-B, and reducing the tumor infiltration of immunosuppressive regulatory T cells and myeloid-derived suppressor cells. The levels of IFN-γ, the chemokines CXCL9 and CXCL10, and CXCR3+CD8+ T cells were significantly increased in tumor tissues, which may account for the improved recruitment of effector T cells into the tumor. Local mucosal immunization of nanofibers via the intravaginal route represents a new and promising vaccination strategy for the treatment of genital tumor lesions such as cervical cancer.
Collapse
Affiliation(s)
- Sijin Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, People's Republic of China. and Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease, Kunming, People's Republic of China
| | - Wenbing Zhu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, People's Republic of China. and Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease, Kunming, People's Republic of China
| | - Chao Ye
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, People's Republic of China. and Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease, Kunming, People's Republic of China
| | - Wenjia Sun
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, People's Republic of China. and Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease, Kunming, People's Republic of China
| | - Hanghang Xie
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, People's Republic of China. and Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease, Kunming, People's Republic of China
| | - Xu Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, People's Republic of China. and Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease, Kunming, People's Republic of China
| | - Qishu Zhang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, People's Republic of China. and Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease, Kunming, People's Republic of China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, People's Republic of China. and Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Disease, Kunming, People's Republic of China
| |
Collapse
|
7
|
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: 116] [Impact Index Per Article: 23.2] [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]
|
8
|
Qian Q, Shi L, Gao X, Ma Y, Yang J, Zhang Z, Qian J, Zhu X. A Paclitaxel-Based Mucoadhesive Nanogel with Multivalent Interactions for Cervical Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1903208. [PMID: 31617295 DOI: 10.1002/smll.201903208] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/24/2019] [Indexed: 06/10/2023]
Abstract
Cervical cancer treatment is subject to limited drug access to locally diseased targets and generally resistant to chemotherapy, thus it is essential to develop a local drug delivery system to overcome these problems, premised on guaranteeing drug efficacy. With this goal in mind, a multivalent interactions-based mucoadhesive nanogel for vaginal delivery is proposed. Briefly, the nanogel is constructed with mucoadhesive poly(acrylic acid) as the backbone and multiple inclusions between β-cyclodextrin and paclitaxel as the crosslinking points. The in vitro experiments demonstrate that nanogel exerts high cytotoxicity to cancer cells, reverses multidrug resistance effectively, and successfully promotes the permeation of drugs. More to the point, as proved in the in vivo experiments, the retention time in the vagina is prolonged and the tumor growth is effectively suppressed by the nanogel without any side effects in the orthotopic cervical cancer model. As mentioned above, this novel mucoadhesive nanogel is believed to be a useful tool toward designing drug delivery systems for cervical cancer treatment.
Collapse
Affiliation(s)
- Qiuhui Qian
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Leilei Shi
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Xihui Gao
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Yuan Ma
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Jiapei Yang
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Zhihao Zhang
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Jiwen Qian
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| |
Collapse
|
9
|
Abstract
Infectious diseases caused by germs, parasites, fungi, virus and bacteria are one of the leading causes of death worldwide. Polymeric therapeutics are nanomedicines that offer several advantages making them useful for the treatment of infectious diseases such as targeted drug release mechanism, ability to maintain the drug concentration within a therapeutic window for a desired duration, biocompatibility with low immunogenicity and reduced drug toxicity resulting in enhanced therapeutic efficacy of the incorporated drug. Although polymeric therapeutics have been evaluated for the treatment of infectious diseases in vitro and in vivo with improved therapeutic efficacy, most treatments for infectious disease have not been evaluated using polymeric therapeutics. This review will focus on the applications of polymeric therapeutics for the treatment of infectious diseases (preclinical studies and clinical trials), with particular focus on parasitic and viral infections.
Collapse
|
10
|
Gadkaree SK, Fu J, Sen R, Korrer MJ, Allen C, Kim YJ. Induction of tumor regression by intratumoral STING agonists combined with anti-programmed death-L1 blocking antibody in a preclinical squamous cell carcinoma model. Head Neck 2017; 39:1086-1094. [PMID: 28323387 DOI: 10.1002/hed.24704] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 10/31/2016] [Accepted: 12/09/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cyclic dinucleotides (CDNs) are bacterial intracellular messengers that have demonstrated antitumor activity in melanoma and breast tumors, although their role in immunotherapy of head and neck squamous cell cancers (HNSCCs) has not been well investigated. METHODS We measured primary tumor growth rates, mechanism of antitumor activity, and efficacy of programmed death-L1 blockade combinatorial therapy in SCCFVII tumor-bearing C3H/HeOUJ mice undergoing intratumoral injections with RR-cyclic-di-guanine (synthetic CDG), CDG (natural cyclic-di-guanine), R848 (TLR 7/8 agonist), or phosphate buffered saline (PBS, control). RESULTS Intratumoral CDN treatment groups showed decreased tumor size and enhanced splenocyte Th1 response when compared to the PBS treatment control group (p < .05). The RR-CDG tumor microenvironment showed upregulated interferon (IFN)-γ+CD8+ and programmed death-L1. Combining programmed death-L1 blocking antibody with RR-CDG induced regression of established tumors. CONCLUSION This study demonstrates the antitumor effects of CDNs in a HNSCC cell line. These preclinical data strongly support the future clinical development of intratumoral CDN in patients with HNSCC. © 2017 Wiley Periodicals, Inc. Head Neck 39: 1086-1094, 2017.
Collapse
Affiliation(s)
- Shekhar K Gadkaree
- Department of Otolaryngology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Juan Fu
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rupashree Sen
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael J Korrer
- Department of Otolaryngology - Head and Neck Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Clint Allen
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Young J Kim
- Department of Otolaryngology - Head and Neck Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
11
|
Hussein WM, Liu TY, Jia Z, McMillan NA, Monteiro MJ, Toth I, Skwarczynski M. Multiantigenic peptide–polymer conjugates as therapeutic vaccines against cervical cancer. Bioorg Med Chem 2016; 24:4372-4380. [DOI: 10.1016/j.bmc.2016.07.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/14/2016] [Accepted: 07/16/2016] [Indexed: 01/10/2023]
|
12
|
Bagley K, Xu R, Ota-Setlik A, Egan M, Schwartz J, Fouts T. The catalytic A1 domains of cholera toxin and heat-labile enterotoxin are potent DNA adjuvants that evoke mixed Th1/Th17 cellular immune responses. Hum Vaccin Immunother 2016; 11:2228-40. [PMID: 26042527 PMCID: PMC4635876 DOI: 10.1080/21645515.2015.1026498] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
DNA encoded adjuvants are well known for increasing the magnitude of cellular and/or humoral immune responses directed against vaccine antigens. DNA adjuvants can also tune immune responses directed against vaccine antigens to better protect against infection of the target organism. Two potent DNA adjuvants that have unique abilities to tune immune responses are the catalytic A1 domains of Cholera Toxin (CTA1) and Heat-Labile Enterotoxin (LTA1). Here, we have characterized the adjuvant activities of CTA1 and LTA1 using HIV and SIV genes as model antigens. Both of these adjuvants enhanced the magnitude of antigen-specific cellular immune responses on par with those induced by the well-characterized cytokine adjuvants IL-12 and GM-CSF. CTA1 and LTA1 preferentially enhanced cellular responses to the intracellular antigen SIVmac239-gag over those for the secreted HIVBaL-gp120 antigen. IL-12, GM-CSF and electroporation did the opposite suggesting differences in the mechanisms of actions of these diverse adjuvants. Combinations of CTA1 or LTA1 with IL-12 or GM-CSF generated additive and better balanced cellular responses to both of these antigens. Consistent with observations made with the holotoxin and the CTA1-DD adjuvant, CTA1 and LTA1 evoked mixed Th1/Th17 cellular immune responses. Together, these results show that CTA1 and LTA1 are potent DNA vaccine adjuvants that favor the intracellular antigen gag over the secreted antigen gp120 and evoke mixed Th1/Th17 responses against both of these antigens. The results also indicate that achieving a balanced immune response to multiple intracellular and extracellular antigens delivered via DNA vaccination may require combining adjuvants that have different and complementary mechanisms of action.
Collapse
|
13
|
Vici P, Pizzuti L, Mariani L, Zampa G, Santini D, Di Lauro L, Gamucci T, Natoli C, Marchetti P, Barba M, Maugeri-Saccà M, Sergi D, Tomao F, Vizza E, Di Filippo S, Paolini F, Curzio G, Corrado G, Michelotti A, Sanguineti G, Giordano A, De Maria R, Venuti A. Targeting immune response with therapeutic vaccines in premalignant lesions and cervical cancer: hope or reality from clinical studies. Expert Rev Vaccines 2016; 15:1327-36. [PMID: 27063030 PMCID: PMC5152541 DOI: 10.1080/14760584.2016.1176533] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Human papillomavirus (HPV) is widely known as a cause of cervical cancer (CC) and cervical intraepithelial neoplasia (CIN). HPVs related to cancer express two main oncogenes, i.e. E6 and E7, considered as tumorigenic genes; their integration into the host genome results in the abnormal regulation of cell cycle control. Due to their peculiarities, these oncogenes represent an excellent target for cancer immunotherapy. In this work the authors highlight the potential use of therapeutic vaccines as safe and effective pharmacological tools in cervical disease, focusing on vaccines that have reached the clinical trial phase. Many therapeutic HPV vaccines have been tested in clinical trials with promising results. Adoptive T-cell therapy showed clinical activity in a phase II trial involving advanced CC patients. A phase II randomized trial showed clinical activity of a nucleic acid-based vaccine in HPV16 or HPV18 positive CIN. Several trials involving peptide-protein-based vaccines and live-vector based vaccines demonstrated that these approaches are effective in CIN as well as in advanced CC patients. HPV therapeutic vaccines must be regarded as a therapeutic option in cervical disease. The synergic combination of HPV therapeutic vaccines with radiotherapy, chemotherapy, immunomodulators or immune checkpoint inhibitors opens a new and interesting scenario in this disease.
Collapse
Affiliation(s)
- P Vici
- a Division of Medical Oncology 2 , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - L Pizzuti
- a Division of Medical Oncology 2 , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - L Mariani
- b HPV-UNIT Laboratory of Virology , 'Regina Elena' National Cancer Institute , Rome , Italy.,c Department of Gynecologic Oncology , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - G Zampa
- d Oncology Unit , Nuovo Regina Margherita Hospital , Rome , Italy
| | - D Santini
- e Department of Medical Oncology , University Campus Bio-Medico , Rome , Italy
| | - L Di Lauro
- a Division of Medical Oncology 2 , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - T Gamucci
- f Medical Oncology Unit, ASL Frosinone , Frosinone , Italy
| | - C Natoli
- g Department of Medical, Oral and Biotechnological Sciences, Experimental and Clinical Sciences , University 'G. d'Annunzio' , Chieti , Italy
| | - P Marchetti
- h Oncology Unit, Sant'Andrea Hospital , 'Sapienza' University of Rome , Rome , Italy
| | - M Barba
- a Division of Medical Oncology 2 , 'Regina Elena' National Cancer Institute , Rome , Italy.,i Scientific Direction , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - M Maugeri-Saccà
- a Division of Medical Oncology 2 , 'Regina Elena' National Cancer Institute , Rome , Italy.,i Scientific Direction , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - D Sergi
- a Division of Medical Oncology 2 , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - F Tomao
- j Department of Gynecologic and Obstetric Sciences , La Sapienza University of Rome , Rome , Italy
| | - E Vizza
- b HPV-UNIT Laboratory of Virology , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - S Di Filippo
- k Emergency Department , Santa Maria Goretti Hospital , Latina , Italy
| | - F Paolini
- b HPV-UNIT Laboratory of Virology , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - G Curzio
- b HPV-UNIT Laboratory of Virology , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - G Corrado
- c Department of Gynecologic Oncology , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - A Michelotti
- l Oncology Unit I , Azienda Ospedaliera Universitaria Pisana , Pisa , Italy
| | - G Sanguineti
- m Radiotherapy , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - A Giordano
- n Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology , Temple University , Philadelphia , PA , USA.,o Department of Human Pathology and Oncology , University of Siena , Siena , Italy
| | - R De Maria
- i Scientific Direction , 'Regina Elena' National Cancer Institute , Rome , Italy
| | - A Venuti
- b HPV-UNIT Laboratory of Virology , 'Regina Elena' National Cancer Institute , Rome , Italy
| |
Collapse
|
14
|
Liu Z, Zhou H, Wang W, Fu YX, Zhu M. A novel dendritic cell targeting HPV16 E7 synthetic vaccine in combination with PD-L1 blockade elicits therapeutic antitumor immunity in mice. Oncoimmunology 2016; 5:e1147641. [PMID: 27471615 DOI: 10.1080/2162402x.2016.1147641] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/18/2016] [Accepted: 01/24/2016] [Indexed: 10/22/2022] Open
Abstract
Human papilliomavirus (HPV) oncogene E7, essential for the transformation and maintenance of the malignancy of cervical cancer cells, represents an ideal tumor-specific antigen for vaccine development. However, due to the poor immunogenicity of E7 protein, an effective therapeutic E7 vaccine is still lacking. Dendritic cells (DCs) are probably the most potent antigen presenting cells for the induction of cytotoxic T lymphocyte (CTL) response, which is crucial for tumor control. In this study, we tested whether targeting the E7 antigen to DCs in vivo would elicit therapeutic antitumor CTL response. We generated the DEC205-specific single-chain variable fragment (scFv) and E7 long peptide fusion protein [scFv(DEC205)-E7] based on the novel method of protein assembly we recently developed. This fusion protein vaccine demonstrated highly efficient DC-targeting in vivo and elicited much stronger protective CTL response than non-DC-targeting control vaccine in naive mice. Furthermore, the scFv(DEC205)-E7 vaccine showed significant therapeutic antitumor response in TC-1 tumor bearing mice. Importantly, PD-L1 blockade further improved the therapeutic effect of the scFv(DEC205)-E7 vaccine. Thus, the current study suggests an efficient strategy for cervical cancer immunotherapy by combining the DC(DEC205)-targeting E7 vaccine and PD-L1 blockade.
Collapse
Affiliation(s)
- Zhida Liu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China; Department of Pathology, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - Hang Zhou
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Wenjun Wang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yang-Xin Fu
- Department of Pathology, University of Texas, Southwestern Medical Center , Dallas, TX, USA
| | - Mingzhao Zhu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences , Beijing, China
| |
Collapse
|
15
|
Hussein WM, Liu TY, Maruthayanar P, Mukaida S, Moyle PM, Wells JW, Toth I, Skwarczynski M. Double conjugation strategy to incorporate lipid adjuvants into multiantigenic vaccines. Chem Sci 2016; 7:2308-2321. [PMID: 29910921 PMCID: PMC5977935 DOI: 10.1039/c5sc03859f] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/28/2015] [Indexed: 11/30/2022] Open
Abstract
Conjugation of multiple peptides by their N-termini is a promising technique to produce branched multiantigenic vaccines.
Conjugation of multiple peptides by their N-termini is a promising technique to produce branched multiantigenic vaccines. We established a double conjugation strategy that combines a mercapto-acryloyl Michael addition and a copper-catalysed alkyne-azide 1,3-dipolar cycloaddition (CuAAC) reaction to synthesise self-adjuvanting branched multiantigenic vaccine candidates. These vaccine candidates aim to treat cervical cancer and include two HPV-16 derived epitopes and a novel self-adjuvanting moiety. This is the first report of mercapto-acryloyl conjugation applied to the hetero conjugation of two unprotected peptides by their N-termini followed by a CuAAC reaction to conjugate a novel synthetic lipoalkyne self-adjuvanting moiety. In vivo experiments showed that the most promising vaccine candidate completely eradicated tumours in 46% of the mice (6 out of 13 mice).
Collapse
Affiliation(s)
- Waleed M Hussein
- School of Chemistry and Molecular Biosciences , The University of Queensland , Brisbane , QLD 4072 , Australia . ;
| | - Tzu-Yu Liu
- School of Chemistry and Molecular Biosciences , The University of Queensland , Brisbane , QLD 4072 , Australia . ;
| | - Pirashanthini Maruthayanar
- The University of Queensland Diamantina Institute , The University of Queensland , Translational Research Institute , Brisbane , Australia
| | - Saori Mukaida
- School of Chemistry and Molecular Biosciences , The University of Queensland , Brisbane , QLD 4072 , Australia . ;
| | - Peter M Moyle
- School of Pharmacy , The University of Queensland , Brisbane , QLD 4102 , Australia
| | - James W Wells
- The University of Queensland Diamantina Institute , The University of Queensland , Translational Research Institute , Brisbane , Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences , The University of Queensland , Brisbane , QLD 4072 , Australia . ; .,School of Pharmacy , The University of Queensland , Brisbane , QLD 4102 , Australia.,Institute for Molecular Bioscience , The University of Queensland , Brisbane , QLD 4072 , Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences , The University of Queensland , Brisbane , QLD 4072 , Australia . ;
| |
Collapse
|
16
|
Sun Y, Peng S, Qiu J, Miao J, Yang B, Jeang J, Hung CF, Wu TC. Intravaginal HPV DNA vaccination with electroporation induces local CD8+ T-cell immune responses and antitumor effects against cervicovaginal tumors. Gene Ther 2015; 22:528-35. [PMID: 25786869 PMCID: PMC4490060 DOI: 10.1038/gt.2015.17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 01/14/2015] [Accepted: 01/29/2015] [Indexed: 12/24/2022]
Abstract
Therapeutic human papillomavirus (HPV) vaccines have the potential to inhibit the progression of an established HPV infection to precancer and cancer lesions by targeting HPV oncoproteins. We have previously developed a therapeutic DNA vaccine encoding calreticulin (CRT) linked to E7, CRT/E7 DNA vaccine, for use in the treatment of HPV-associated lesions. Since the transfection efficiency of DNA vaccines administered in vivo is typically low, we examined the use of electroporation as well as different routes of administration to enhance antigen-specific tumor control. We tested the effects of the CRT/E7 DNA vaccine administered intramuscularly or intravaginally, with or without electroporation, on the generation of CD8+ T-cell immunity and therapeutic antitumor effects in HPV16 E7-expressing cervicovaginal tumor-bearing mice. We found that intravaginal vaccination of CRT/E7 DNA followed by electroporation-induced potent E7-specific CD8(+) T-cell responses in the cervicovaginal tract, compared with intramuscular injection followed by electroporation. Furthermore, tumor-bearing mice vaccinated intravaginally followed by electroporation had an enhanced survival, antitumor effects and local production of IFN-γ+CD8+ T cells compared with those vaccinated intramuscularly with electroporation. Thus, we show that intravaginal CRT/E7 DNA vaccination followed by electroporation generates the most potent therapeutic antitumor effects against an orthotopic E7-expressing tumor model. The current study will have significant clinical implications once a clinically applicable electroporation device for intravaginal use becomes available.
Collapse
Affiliation(s)
- Y Sun
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - S Peng
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - J Qiu
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital of Tongji University Shanghai, Shanghai, China
| | - J Miao
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - B Yang
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - J Jeang
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - C-F Hung
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - T-C Wu
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|
17
|
Wu XM, Liu X, Jiao QF, Fu SY, Bu YQ, Song FZ, Yi FP. Cytotoxic T lymphocytes elicited by dendritic cell-targeted delivery of human papillomavirus type-16 E6/E7 fusion gene exert lethal effects on CaSki cells. Asian Pac J Cancer Prev 2015; 15:2447-51. [PMID: 24761845 DOI: 10.7314/apjcp.2014.15.6.2447] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Human papillomavirus (HPV) is the primary etiologic agent of cervical cancer. Consideration of safety and non human leukocyte antigen restriction, protein vaccine has become the most likely form of HPV therapeutic vaccine, although none have so far been reported as effective. Since tumor cells consistently express the two proteins E6 and E7, most therapeutic vaccines target one or both of them. In this study, we fabricated DC vaccines by transducing replication-defective recombinant adenoviruses expressing E6/E7 fusion gene of HPV-16, to investigate the lethal effects of specific cytotoxic T lymphocytes (CTL) against CaSki cells in vitro. Mouse immature dendritic cells (DC) were generated from bone marrow, and transfected with pAd-E6/E7 to prepare a DC vaccine and to induce specific CTL. The surface expression of CD40, CD68, MHC II and CD11c was assessed by flow cytometry (FCM), and the lethal effects of CTL against CaSki cells were determined by DAPI, FCM and CCK-8 methods. Immature mouse DC was successfully transfected by pAd-E6/E7 in vitro, and the transfecting efficiency was 40%-50%. A DC vaccine was successfully prepared and was used to induce specific CTL. Experimental results showed that the percentage of apoptosis and killing rate of CaSki cells were significantly increased by coculturing with the specific CTL (p <0.05). These results illustrated that a DC vaccine modified by HPV-16 E6/E7 gene can induce apoptosis of CaSki cells by inducing CTL, which may be used as a new strategy for biological treatment of cervical cancer.
Collapse
Affiliation(s)
- Xiang-Mei Wu
- Chongqing Molecular Medicine and Cancer Research Center, Chongqing, China E-mail :
| | | | | | | | | | | | | |
Collapse
|
18
|
Liu TY, Hussein WM, Giddam AK, Jia Z, Reiman JM, Zaman M, McMillan NAJ, Good MF, Monteiro MJ, Toth I, Skwarczynski M. Polyacrylate-based delivery system for self-adjuvanting anticancer peptide vaccine. J Med Chem 2014; 58:888-96. [PMID: 25489968 DOI: 10.1021/jm501514h] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vaccination can provide a safe alternative to chemotherapy by using the body's natural defense mechanisms to create a potent immune response against tumor cells. Peptide-based therapeutic vaccines against human papillomavirus (HPV)-related cancers are usually designed to elicit cytotoxic T cell responses by targeting the HPV-16 E7 oncoprotein. However, peptides alone lack immunogenicity, and an additional adjuvant or external delivery system is required. In this study, we developed new polymer-peptide conjugates to create an efficient self-adjuvanting system for peptide-based therapeutic vaccines. These conjugates reduced tumor growth and eradicated E7-positive TC-1 tumors in mice after a "single shot" immunization, without the help from an external adjuvant. The new conjugates had a significantly higher anticancer efficacy than the antigen formulated with a commercial adjuvant. Furthermore, the polymer-peptide conjugates were promptly taken up by antigen presenting cells, including dendritic cells and macrophages, and efficiently activated CD4(+) T-helper cells and CD8(+) cytotoxic T lymphocyte cells.
Collapse
Affiliation(s)
- Tzu-Yu Liu
- School of Chemistry and Molecular Biosciences, ‡Australian Institute for Bioengineering and Nanotechnology, and §School of Pharmacy, The University of Queensland , Brisbane, QLD 4072, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Soong RS, Song L, Trieu J, Knoff J, He L, Tsai YC, Huh W, Chang YN, Cheng WF, Roden RBS, Wu TC, Trimble CL, Hung CF. Toll-like receptor agonist imiquimod facilitates antigen-specific CD8+ T-cell accumulation in the genital tract leading to tumor control through IFNγ. Clin Cancer Res 2014; 20:5456-67. [PMID: 24893628 PMCID: PMC4216740 DOI: 10.1158/1078-0432.ccr-14-0344] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Imiquimod is a Toll-like receptor 7 agonist used topically to treat external genital warts and basal cell carcinoma. We examined the combination of topical imiquimod with intramuscular administration of CRT/E7, a therapeutic human papillomavirus (HPV) vaccine comprised of a naked DNA vector expressing calreticulin fused to HPV16 E7. EXPERIMENTAL DESIGN Using an orthotopic HPV16 E6/E7(+) syngeneic tumor, TC-1, as a model of high-grade cervical/vaginal/vulvar intraepithelial neoplasia, we assessed if combining CRT/E7 vaccination with cervicovaginal deposition of imiquimod could result in synergistic activities promoting immune-mediated tumor clearance. RESULTS Imiquimod induced cervicovaginal accumulation of activated E7-specific CD8(+) T cells elicited by CRT/E7 vaccination. Recruitment was not dependent upon the specificity of the activated CD8(+) T cells, but was significantly reduced in mice lacking the IFNγ receptor. Intravaginal imiquimod deposition induced upregulation of CXCL9 and CXCL10 mRNA expression in the genital tract, which are produced in response to IFNγ receptor signaling and attract cells expressing their ligand, CXCR3. The T cells attracted by imiquimod to the cervicovaginal tract expressed CXCR3 as well as CD49a, an integrin involved in homing and retention of CD8(+) T cells at mucosal sites. Our results indicate that intramuscular CRT/E7 vaccination in conjunction with intravaginal imiquimod deposition recruits antigen-specific CXCR3(+) CD8(+) T cells to the genital tract. CONCLUSIONS Several therapeutic HPV vaccination clinical trials using a spectrum of DNA vaccines, including vaccination in concert with cervical imiquimod, are ongoing. Our study identifies a mechanism by which these strategies could provide therapeutic benefit. Our findings support accumulating evidence that manipulation of the tumor microenvironment can enhance the therapeutic efficacy of strategies that induce tumor-specific T cells.
Collapse
Affiliation(s)
- Ruey-Shyang Soong
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of General Surgery, Chang Gung Memorial Hospital at Keelung, Keelung City, Taiwan. College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Liwen Song
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland. School of Pharmacy, Fudan University, Shanghai, China. Department of Pharmacology and Toxicology, Shanghai Institute of Planned Parenthood Research, Shanghai, China. Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | | | - Jayne Knoff
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Liangmei He
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Ya-Chea Tsai
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Warner Huh
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Wen-Fang Cheng
- Department of Obstetrics and Gynecology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Richard B S Roden
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - T-C Wu
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions, Baltimore, Maryland.
| | - Cornelia L Trimble
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland. Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland.
| |
Collapse
|
20
|
Yang M, Lai SK, Yu T, Wang YY, Happe C, Zhong W, Zhang M, Anonuevo A, Fridley C, Hung A, Fu J, Hanes J. Nanoparticle penetration of human cervicovaginal mucus: the effect of polyvinyl alcohol. J Control Release 2014; 192:202-8. [PMID: 25090196 DOI: 10.1016/j.jconrel.2014.07.045] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/10/2014] [Accepted: 07/20/2014] [Indexed: 11/17/2022]
Abstract
Therapeutic nanoparticles must rapidly penetrate the mucus secretions lining the surfaces of the respiratory, gastrointestinal and cervicovaginal tracts to efficiently reach the underlying tissues. Whereas most polymeric nanoparticles are highly mucoadhesive, we previously discovered that a dense layer of low MW polyethylene glycol (PEG) conferred a sufficiently hydrophilic and uncharged surface to effectively minimize mucin-nanoparticle adhesive interactions, allowing well-coated particles to rapidly diffuse through human mucus. Here, we sought to investigate the influence of surface coating by polyvinyl alcohol (PVA), a relatively hydrophilic and uncharged polymer routinely used as a surfactant to formulate drug carriers, on the transport of nanoparticles in fresh human cervicovaginal mucus. We found that PVA-coated polystyrene (PS) particles were immobilized, with speeds at least 4000-fold lower in mucus than in water, regardless of the PVA molecular weight or incubation concentration tested. Nanoparticles composed of poly(lactide-co-glycolide) (PLGA) or diblock copolymers of PEG-PLGA were similarly immobilized when coated with PVA (slowed 29,000- and 2500-fold, respectively). PVA coatings could not be adequately removed upon washing, and the residual PVA prevented sufficient coating with Pluronic F127 capable of reducing particle mucoadhesion. In contrast to PVA-coated particles, the similar sized PEG-coated formulations were slowed only ~6- to 10-fold in mucus compared to in water. Our results suggest that incorporating PVA in the particle formulation process may lead to the formation of mucoadhesive particles for many nanoparticulate systems. Thus, alternative methods for particle formulation, based on novel surfactants or changes in the formulation process, should be identified and developed in order to produce mucus-penetrating particles for mucosal applications.
Collapse
Affiliation(s)
- Ming Yang
- Center for Nanomedicine, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21287, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Samuel K Lai
- Center for Nanomedicine, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21287, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA; Center for Cancer Nanotechnology Excellence, Institute for NanoBioTechnology, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA
| | - Tao Yu
- Center for Nanomedicine, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21287, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Ying-Ying Wang
- Center for Nanomedicine, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21287, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Christina Happe
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA
| | - Weixi Zhong
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Michael Zhang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA
| | - Abraham Anonuevo
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA
| | - Colleen Fridley
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA
| | - Amy Hung
- Center for Nanomedicine, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21287, USA
| | - Jie Fu
- Center for Nanomedicine, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21287, USA; Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21287, USA
| | - Justin Hanes
- Center for Nanomedicine, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21287, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 600 N Wolfe Street, Baltimore, MD 21287, USA; Center for Cancer Nanotechnology Excellence, Institute for NanoBioTechnology, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA; Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21287, USA.
| |
Collapse
|
21
|
Yang M, Yu T, Wang YY, Lai SK, Zeng Q, Miao B, Tang BC, Simons BW, Ensign LM, Liu G, Chan KW, Juang CY, Mert O, Wood J, Fu J, McMahon MT, Wu TC, Hung CF, Hanes J. Vaginal delivery of paclitaxel via nanoparticles with non-mucoadhesive surfaces suppresses cervical tumor growth. Adv Healthc Mater 2014; 3:1044-52. [PMID: 24339398 DOI: 10.1002/adhm.201300519] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/21/2013] [Indexed: 12/29/2022]
Abstract
Local delivery of chemotherapeutics in the cervicovaginal tract using nanoparticles may reduce adverse side effects associated with systemic chemotherapy, while improving outcomes for early-stage cervical cancer. It is hypothesized here that drug-loaded nanoparticles that rapidly penetrate cervicovaginal mucus (CVM) lining the female reproductive tract will more effectively deliver their payload to underlying diseased tissues in a uniform and sustained manner compared with nanoparticles that do not efficiently penetrate CVM. Paclitaxel-loaded nanoparticles are developed, composed entirely of polymers used in FDA-approved products, which rapidly penetrate human CVM and provide sustained drug release with minimal burst effect. A mouse model is further employed with aggressive cervical tumors established in the cervicovaginal tract to compare paclitaxel-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (conventional particles, or CP) and similar particles coated with Pluronic F127 (mucus-penetrating particles, or MPP). CP are mucoadhesive and, thus, aggregated in mucus, while MPP achieve more uniform distribution and close proximity to cervical tumors. Paclitaxel-MPP suppress tumor growth more effectively and prolong median survival of mice compared with unencapsulated paclitaxel or paclitaxel-CP. Histopathological studies demonstrate minimal toxicity to the cervicovaginal epithelia, suggesting paclitaxel-MPP may be safe for intravaginal use. These results demonstrate the in vivo advantages of polymer-based MPP for treatment of tumors localized to a mucosal surface.
Collapse
Affiliation(s)
- Ming Yang
- Center for Nanomedicine, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
- Department of Biomedical Engineering; Johns Hopkins University School of Medicine; 720 Rutland Avenue Baltimore MD 21205 USA
| | - Tao Yu
- Center for Nanomedicine, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
- Department of Biomedical Engineering; Johns Hopkins University School of Medicine; 720 Rutland Avenue Baltimore MD 21205 USA
| | - Ying-Ying Wang
- Center for Nanomedicine, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
- Department of Biomedical Engineering; Johns Hopkins University School of Medicine; 720 Rutland Avenue Baltimore MD 21205 USA
| | - Samuel K. Lai
- Center for Nanomedicine, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; 3400 N Charles Street Baltimore MD 21218 USA
- Eshelman School of Pharmacy; University of North Carolina at Chapel; Hill, 120 Mason Farm Road Chapel Hill NC 27599 USA
| | - Qi Zeng
- Department of Pathology; Johns Hopkins University School of Medicine; 600 N Wolfe Street Baltimore MD 21287 USA
| | - Bolong Miao
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; 3400 N Charles Street Baltimore MD 21218 USA
| | - Benjamin C. Tang
- Center for Nanomedicine, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; 3400 N Charles Street Baltimore MD 21218 USA
- Koch Institute for Integrated Cancer Research; Massachusetts Institute of Technology; 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - Brian W. Simons
- Department of Molecular and Comparative Pathobiology; Johns Hopkins University School of Medicine; 1550 Orleans Street Baltimore MD 21231 USA
| | - Laura M. Ensign
- Center for Nanomedicine, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; 3400 N Charles Street Baltimore MD 21218 USA
- Department of Ophthalmology; The Wilmer Eye Institute, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
| | - Guanshu Liu
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute; 707 N Broadway Baltimore MD 21205 USA
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine; 600 N Wolfe Street Baltimore MD 21287 USA
| | - Kannie W.Y. Chan
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute; 707 N Broadway Baltimore MD 21205 USA
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine; 600 N Wolfe Street Baltimore MD 21287 USA
| | - Chih-Yin Juang
- Center for Nanomedicine, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
| | - Olcay Mert
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; 3400 N Charles Street Baltimore MD 21218 USA
| | - Joseph Wood
- Department of Biomedical Engineering; Johns Hopkins University School of Medicine; 720 Rutland Avenue Baltimore MD 21205 USA
| | - Jie Fu
- Center for Nanomedicine, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
- Department of Ophthalmology; The Wilmer Eye Institute, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
| | - Michael T. McMahon
- Center for Nanomedicine, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
| | - T.-C. Wu
- Center for Nanomedicine, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
- Department of Pathology; Johns Hopkins University School of Medicine; 600 N Wolfe Street Baltimore MD 21287 USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center; Johns Hopkins University School of Medicine; 600 N Wolfe Street Baltimore MD 21287 USA
| | - Chien-Fu Hung
- Center for Nanomedicine, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
- Department of Pathology; Johns Hopkins University School of Medicine; 600 N Wolfe Street Baltimore MD 21287 USA
- Department of Obstetrics and Gynecology; Johns Hopkins University School of Medicine; 600 N Wolfe Street Baltimore MD 21287 USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center; Johns Hopkins University School of Medicine; 600 N Wolfe Street Baltimore MD 21287 USA
| | - Justin Hanes
- Center for Nanomedicine, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
- Department of Biomedical Engineering; Johns Hopkins University School of Medicine; 720 Rutland Avenue Baltimore MD 21205 USA
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; 3400 N Charles Street Baltimore MD 21218 USA
- Department of Ophthalmology; The Wilmer Eye Institute, Johns Hopkins University School of Medicine; 400 N Broadway Baltimore MD 21231 USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center; Johns Hopkins University School of Medicine; 600 N Wolfe Street Baltimore MD 21287 USA. Center for Cancer Nanotechnology Excellence; Institute for NanoBioTechnology, Johns Hopkins University; 3400 N Charles Street Baltimore MD 21218 USA
| |
Collapse
|
22
|
Vici P, Mariani L, Pizzuti L, Sergi D, Di Lauro L, Vizza E, Tomao F, Tomao S, Cavallotti C, Paolini F, Venuti A. Immunologic treatments for precancerous lesions and uterine cervical cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:29. [PMID: 24667138 PMCID: PMC3986944 DOI: 10.1186/1756-9966-33-29] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 03/18/2014] [Indexed: 01/24/2023]
Abstract
Development of HPV-associated cancers not only depends on efficient negative regulation of cell cycle control that supports the accumulation of genetic damage, but also relies on immune evasion that enable the virus to go undetected for long periods of time. In this way, HPV-related tumors usually present MHC class I down-regulation, impaired antigen-processing ability, avoidance of T-cell mediated killing, increased immunosuppression due to Treg infiltration and secrete immunosuppressive cytokines. Thus, these are the main obstacles that immunotherapy has to face in the treatment of HPV-related pathologies where a number of different strategies have been developed to overcome them including new adjuvants. Although antigen-specific immunotherapy induced by therapeutic HPV vaccines was proved extremely efficacious in pre-clinical models, its progression through clinical trials suffered poor responses in the initial trials. Later attempts seem to have been more promising, particularly against the well-defined precursors of cervical, anal or vulvar cancer, where the local immunosuppressive milieu is less active. This review focuses on the advances made in these fields, highlighting several new technologies (such as mRNA vaccine, plant-derived vaccine). The most promising immunotherapies used in clinical trials are also summarized, along with integrated strategies, particularly promising in controlling tumor metastasis and in eliminating cancer cells altogether. After the early promising clinical results, the development of therapeutic HPV vaccines need to be implemented and applied to the users in order to eradicate HPV-associated malignancies, eradicating existing perception (after the effectiveness of commercial preventive vaccines) that we have already solved the problem.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Aldo Venuti
- HPV Unit, Regina Elena National Cancer Institute, V Elio Chianesi 53, Rome 00144, Italy.
| |
Collapse
|
23
|
Knoff J, Yang B, Hung CF, Wu TC. Cervical Cancer: Development of Targeted Therapies Beyond Molecular Pathogenesis. CURRENT OBSTETRICS AND GYNECOLOGY REPORTS 2014; 3:18-32. [PMID: 24533233 PMCID: PMC3921905 DOI: 10.1007/s13669-013-0068-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
It is well known that human papillomavirus (HPV) is the causative agent of cervical cancer. The integration of HPV genes into the host genome causes the upregulation of E6 and E7 oncogenes. E6 and E7 proteins inactivate and degrade tumor suppressors p53 and retinoblastoma, respectively, leading to malignant progression. HPV E6 and E7 antigens are ideal targets for the development of therapies for cervical cancer and precursor lesions because they are constitutively expressed in infected cells and malignant tumors but not in normal cells and they are essential for cell immortalization and transformation. Immunotherapies are being developed to target E6/E7 by eliciting antigen-specific immune responses. siRNA technologies target E6/E7 by modulating the expression of the oncoproteins. Proteasome inhibitors and histone deacetylase inhibitors are being developed to indirectly target E6/E7 by interfering with their oncogenic activities. The ultimate goal for HPV-targeted therapies is the progression through clinical trials to commercialization.
Collapse
Affiliation(s)
- Jayne Knoff
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Benjamin Yang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - T.-C. Wu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
24
|
Liu TY, Hussein WM, Jia Z, Ziora ZM, McMillan NAJ, Monteiro MJ, Toth I, Skwarczynski M. Self-Adjuvanting Polymer–Peptide Conjugates As Therapeutic Vaccine Candidates against Cervical Cancer. Biomacromolecules 2013; 14:2798-806. [DOI: 10.1021/bm400626w] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Tzu-Yu Liu
- School of Chemistry and Molecular
Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Waleed M. Hussein
- School of Chemistry and Molecular
Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zhongfan Jia
- Australian Institute for Bioengineering
and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zyta M. Ziora
- School of Chemistry and Molecular
Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nigel A. J. McMillan
- Cancer Research Centre, Griffith
Health Institute and School of Medical Science, Griffith University, Gold Coast, QLD 4222, Australia
| | - Michael J. Monteiro
- Australian Institute for Bioengineering
and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Istvan Toth
- School of Chemistry and Molecular
Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular
Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
25
|
Fassolari M, Chemes LB, Gallo M, Smal C, Sánchez IE, de Prat-Gay G. Minute time scale prolyl isomerization governs antibody recognition of an intrinsically disordered immunodominant epitope. J Biol Chem 2013; 288:13110-23. [PMID: 23504368 DOI: 10.1074/jbc.m112.444554] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Conformational rearrangements in antibody·antigen recognition are essential events where kinetic discrimination of isomers expands the universe of combinations. We investigated the interaction mechanism of a monoclonal antibody, M1, raised against E7 from human papillomavirus, a prototypic viral oncoprotein and a model intrinsically disordered protein. The mapped 12-amino acid immunodominant epitope lies within a "hinge" region between the N-terminal intrinsically disordered and the C-terminal globular domains. Kinetic experiments show that despite being within an intrinsically disordered region, the hinge E7 epitope has at least two populations separated by a high energy barrier. Nuclear magnetic resonance traced the origin of this barrier to a very slow (t(1/2)∼4 min) trans-cis prolyl isomerization event involving changes in secondary structure. The less populated (10%) cis isomer is the binding-competent species, thus requiring the 90% of molecules in the trans configuration to isomerize before binding. The association rate for the cis isomer approaches 6 × 10(7) M(-1) s(-1), a ceiling for antigen-antibody interactions. Mutagenesis experiments showed that Pro-41 in E7Ep was required for both binding and isomerization. After a slow postbinding unimolecular rearrangement, a consolidated complex with K(D) = 1.2 × 10(-7) M is reached. Our results suggest that presentation of this viral epitope by the antigen-presenting cells would have to be "locked" in the cis conformation, in opposition to the most populated trans isomer, in order to select the specific antibody clone that goes through affinity and kinetic maturation.
Collapse
Affiliation(s)
- Marisol Fassolari
- Protein Structure-Function and Engineering Laboratory, Fundación Instituto Leloir, IIBBA-CONICET, Patricias Argentinas 435, 1405 Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
26
|
Fridlender ZG, Jassar A, Mishalian I, Wang LC, Kapoor V, Cheng G, Sun J, Singhal S, Levy L, Albelda SM. Using macrophage activation to augment immunotherapy of established tumours. Br J Cancer 2013; 108:1288-97. [PMID: 23481183 PMCID: PMC3619255 DOI: 10.1038/bjc.2013.93] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background: Successful immunotherapy will require alteration of the tumour microenvironment and/or decreased immune suppression. Tumour-associated macrophages (TAMs) are one major factor affecting tumour microenvironment. We hypothesised that altering TAM phenotype would augment the efficacy of immunotherapy. Methods: We and others have reported that 5,6-Dimethylxanthenone-4-acetic-acid (DMXAA, Vadimezan) has the ability to change TAM phenotypes, inducing a tumour microenvironment conducive to antitumour immune responses. We therefore combined DMXAA with active immunotherapies, and evaluated anti-tumour efficacy, immune cell phenotypes (flow cytometry), and tumour microenvironment (RT–PCR). Results: In several different murine models of immunotherapy for lung cancer, DMXAA-induced macrophage activation significantly augmented the therapeutic effects of immunotherapy. By increasing influx of neutrophils and anti-tumour (M1) macrophages to the tumour, DMXAA altered myeloid cell phenotypes, thus changing the intratumoural M2/non-M2 TAM immunoinhibitory ratio. It also altered the tumour microenvironment to be more pro-inflammatory. Modulating macrophages during immunotherapy resulted in increased numbers, activity, and antigen-specificity of intratumoural CD8+ T cells. Macrophage depletion reduced the effect of combining immunotherapy with macrophage activation, supporting the importance of TAMs in the combined effect. Conclusion: Modulating intratumoural macrophages dramatically augmented the effect of immunotherapy. Our observations suggest that addition of agents that activate TAMs to immunotherapy should be considered in future trials.
Collapse
Affiliation(s)
- Z G Fridlender
- Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, POB 12000, Jerusalem 91120, Israel.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ma B, Maraj B, Tran NP, Knoff J, Chen A, Alvarez RD, Hung CF, Wu TC. Emerging human papillomavirus vaccines. Expert Opin Emerg Drugs 2012; 17:469-92. [PMID: 23163511 PMCID: PMC3786409 DOI: 10.1517/14728214.2012.744393] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Identification of human papillomavirus (HPV) as the etiologic factor of cervical, anogenital, and a subset of head and neck cancers has stimulated the development of preventive and therapeutic HPV vaccines to control HPV-associated malignancies. Excitement has been generated by the commercialization of two preventive L1-based vaccines, which use HPV virus-like particles (VLPs) to generate capsid-specific neutralizing antibodies. However, factors such as high cost and requirement for cold chain have prevented widespread implementation where they are needed most. AREAS COVERED Next generation preventive HPV vaccine candidates have focused on cost-effective stable alternatives and generating broader protection via targeting multivalent L1 VLPs, L2 capsid protein, and chimeric L1/L2 VLPs. Therapeutic HPV vaccine candidates have focused on enhancing T cell-mediated killing of HPV-transformed tumor cells, which constitutively express HPV-encoded proteins, E6 and E7. Several therapeutic HPV vaccines are in clinical trials. EXPERT OPINION Although progress is being made, cost remains an issue inhibiting the use of preventive HPV vaccines in countries that carry the majority of the cervical cancer burden. In addition, progression of therapeutic HPV vaccines through clinical trials may require combination strategies employing different therapeutic modalities. As research in the development of HPV vaccines continues, we may generate effective strategies to control HPV-associated malignancies.
Collapse
Affiliation(s)
- Barbara Ma
- The Johns Hopkins Medical Institutions, Departments of Pathology, Baltimore, MD, USA
| | - Bharat Maraj
- The Johns Hopkins Medical Institutions, Departments of Pathology, Baltimore, MD, USA
| | - Nam Phuong Tran
- The Johns Hopkins Medical Institutions, Departments of Pathology, Baltimore, MD, USA
| | - Jayne Knoff
- The Johns Hopkins Medical Institutions, Departments of Pathology, Baltimore, MD, USA
| | - Alexander Chen
- The Johns Hopkins Medical Institutions, Departments of Pathology, Baltimore, MD, USA
| | - Ronald D Alvarez
- University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, MD, USA
| | - Chien-Fu Hung
- The Johns Hopkins Medical Institutions, Departments of Pathology, Baltimore, MD, USA
- The Johns Hopkins Medical Institutions, Departments of Oncology, Baltimore, MD, USA
| | - T.-C. Wu
- The Johns Hopkins Medical Institutions, Departments of Pathology, Baltimore, MD, USA
- The Johns Hopkins Medical Institutions, Departments of Oncology, Baltimore, MD, USA
- The Johns Hopkins Medical Institutions, Departments of Obstetrics and Gynecology, Baltimore, MD, USA
- The Johns Hopkins Medical Institutions, Departments of Molecular Microbiology and Immunology, Baltimore, MD, USA
| |
Collapse
|
28
|
Jia Y, Yin Y, Duan F, Fu H, Hu M, Gao Y, Pan Z, Jiao X. Prophylactic and therapeutic efficacy of an attenuated Listeria monocytogenes-based vaccine delivering HPV16 E7 in a mouse model. Int J Mol Med 2012; 30:1335-42. [PMID: 23027427 DOI: 10.3892/ijmm.2012.1136] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 08/02/2012] [Indexed: 11/06/2022] Open
Abstract
Listeria monocytogenes (L. monocytogenes) has been developed as a cancer vaccine vector due to its ability to elicit strong innate and adaptive immune responses. For clinical application, it is necessary to exploit a Listeria platform strain that is safe and that also retains its immunogenicity to develop vaccine candidates against cancer. In this study, a highly attenuated strain with a deletion of actA/plcB was employed as a vector to deliver the human papillomavirus type 16 (HPV16) E7 antigen, which was stably inserted into the chromosome of L. monocytogenes. The prophylactic and therapeutic efficacy of the recombinant L. monocytogenes strain expressing E7 (LM1-2-E7) were evaluated in C57BL/6 mice. In prophylactic tumor challenge assays, immunization with the recombinant strain LM1-2-E7 was able to protect against tumor formation in 87.5% of the mice, even after a second challenge, suggesting that this prophylactic immunization can provide long-lasting immunity. In the therapeutic setting, immunization with LM1-2-E7 led to tumor regression in 50% of the mice and suppressed tumor growth in the remaining mice. The results showed that the recombinant strain was cleared by the immune system within 5 days after immunization and induced a Th1 immune response against E7 peptide and E7-specific cytotoxic T-lymphocyte (CTL) killing activity without severe inflammatory responses in the spleen and liver. Markedly, recombinant Listeria strain resulted in preferential accumulation within tumor tissues and induced higher numbers of CD8+ T cells that infiltrated into the tumor, which were associated with retardation of tumor growth. Collectively, these data indicate that LM1-2-E7 is a possible vaccine candidate against cervical cancer.
Collapse
Affiliation(s)
- Yanyan Jia
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Lin J, Xu J, Albers AE, Kaufmann AM. New Developments in Therapeutic HPV Vaccines. CURRENT OBSTETRICS AND GYNECOLOGY REPORTS 2012. [DOI: 10.1007/s13669-012-0015-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
30
|
TriVax-HPV: an improved peptide-based therapeutic vaccination strategy against human papillomavirus-induced cancers. Cancer Immunol Immunother 2012; 61:1307-17. [PMID: 22527249 DOI: 10.1007/s00262-012-1259-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 03/29/2012] [Indexed: 12/15/2022]
Abstract
BACKGROUND Therapeutic vaccines for cancer are an attractive alternative to conventional therapies, since the later result in serious adverse effects and in most cases are not effective against advanced disease. Human papillomavirus (HPV) is responsible for several malignancies such as cervical carcinoma. Vaccines targeting oncogenic viral proteins like HPV16-E6 and HPV16-E7 are ideal candidates to elicit strong immune responses without generating autoimmunity because: (1) these products are not expressed in normal cells and (2) their expression is required to maintain the malignant phenotype. Our group has developed peptide vaccination strategy called TriVax, which is effective in generating vast numbers of antigen-specific T cells in mice capable of persisting for long time periods. MATERIALS AND METHODS We have used two HPV-induced mouse cancer models (TC-1 and C3.43) to evaluate the immunogenicity and therapeutic efficacy of TriVax prepared with the immunodominant CD8 T-cell epitope HPV16-E7(49-57), mixed with poly-IC adjuvant and costimulatory anti-CD40 antibodies. RESULTS TriVax using HPV16-E7(49-57) induced large and persistent T-cell responses that were therapeutically effective against established HPV16-E7 expressing tumors. In most cases, TriVax was successful in attaining complete rejections of 6-11-day established tumors. In addition, TriVax induced long-term immunological memory, which prevented tumor recurrences. The anti-tumor effects of TriVax were independent of NK and CD4 T cells and, surprisingly, did not rely to a great extent on type-I or type-II interferon. CONCLUSIONS These findings indicate that the TriVax strategy is an appealing immunotherapeutic approach for the treatment of established viral-induced tumors. We believe that these studies may help to launch more effective and less invasive therapeutic vaccines for HPV-mediated malignancies.
Collapse
|
31
|
Peng S, Monie A, Pang X, Hung CF, Wu TC. Vascular disrupting agent DMXAA enhances the antitumor effects generated by therapeutic HPV DNA vaccines. J Biomed Sci 2011; 18:21. [PMID: 21385449 PMCID: PMC3062584 DOI: 10.1186/1423-0127-18-21] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 03/08/2011] [Indexed: 12/12/2022] Open
Abstract
Antigen-specific immunotherapy using DNA vaccines has emerged as an attractive approach for the control of tumors. Another novel cancer therapy involves the employment of the vascular disrupting agent, 5,6-dimethylxanthenone-4-acetic acid (DMXAA). In the current study, we aimed to test the combination of DMXAA treatment with human papillomavirus type 16 (HPV-16) E7 DNA vaccination to enhance the antitumor effects and E7-specific CD8+ T cell immune responses in treated mice. We determined that treatment with DMXAA generates significant therapeutic effects against TC-1 tumors but does not enhance the antigen-specific immune responses in tumor bearing mice. We then found that combination of DMXAA treatment with E7 DNA vaccination generates potent antitumor effects and E7-specific CD8+ T cell immune responses in the splenocytes of tumor bearing mice. Furthermore, the DMXAA-mediated enhancement or suppression of E7-specific CD8+ T cell immune responses generated by CRT/E7 DNA vaccination was found to be dependent on the time of administration of DMXAA and was also applicable to other antigen-specific vaccines. In addition, we determined that inducible nitric oxide synthase (iNOS) plays a role in the immune suppression caused by DMXAA administration before DNA vaccination. Our study has significant implications for future clinical translation.
Collapse
Affiliation(s)
- Shiwen Peng
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|