1
|
Zhang W, Zeng M, Li Y, Yu L. Leveraging oncovirus-derived antigen against the viral malignancies in adoptive cell therapies. Biomark Res 2024; 12:71. [PMID: 39075601 PMCID: PMC11287861 DOI: 10.1186/s40364-024-00617-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/10/2024] [Indexed: 07/31/2024] Open
Abstract
Adoptive cell therapies (ACTs) have revolutionized cancer immunotherapy, prompting exploration into their application against oncoviruses. Oncoviruses such as human papillomavirus (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV), and Epstein-Barr virus (EBV) contribute significantly (12-25%) to human malignancies through direct or indirect oncogenic mechanisms. These viruses persistently or latently infect cells, disrupt cellular homeostasis and pathways, challenging current antiviral treatment paradigms. Moreover, viral infections pose additional risks in the setting of long-term cancer therapy and lead to morbidity and mortality. Virally encoded oncoproteins, which are tumor-restricted, immunologically foreign, and even uniformly expressed, represent promising targets for patient-tailored ACTs. This review elucidates the rationale for leveraging viral antigen-specific ACTs in combating viral-associated malignancies. On this basis, ongoing preclinical studies consolidate our understanding of harnessing ACTs against viral malignancies, underscoring their potential to eradicate viruses implicated in cancer progression. Furthermore, we scrutinize the current landscape of clinical trials focusing on virus-specific ACTs and discuss their implications for therapeutic advancement.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
| | - Miao Zeng
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
| | - Yisheng Li
- Shenzhen Haoshi Biotechnology Co., Ltd, No. 155 Hongtian Road, Xinqiao Street, Bao'an District, Shenzhen, Guangdong, 518125, China
- Haoshi Cell Therapy Institute, Shenzhen University, Shenzhen, China
| | - Li Yu
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China.
- Haoshi Cell Therapy Institute, Shenzhen University, Shenzhen, China.
| |
Collapse
|
2
|
Wang S, Chen CC, Hu MH, Cheng M, Tu HF, Tsai YC, Yang JM, Wu TC, Huang CH, Hung CF. Arginine-linked HPV-associated E7 displaying bacteria-derived outer membrane vesicles as a potent antigen-specific cancer vaccine. J Transl Med 2024; 22:378. [PMID: 38649894 PMCID: PMC11036690 DOI: 10.1186/s12967-024-05195-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Bacteria-based cancer therapy have demonstrated innovative strategies to combat tumors. Recent studies have focused on gram-negative bacterial outer membrane vesicles (OMVs) as a novel cancer immunotherapy strategy due to its intrinsic properties as a versatile carrier. METHOD Here, we developed an Human Papillomavirus (HPV)-associated E7 antigen displaying Salmonella-derived OMV vaccine, utilizing a Poly(L-arginine) cell penetrating peptide (CPP) to enhance HPV16 E7 (aa49-67) H-2 Db and OMV affinity, termed SOMV-9RE7. RESULTS Due to OMV's intrinsic immunogenic properties, SOMV-9RE7 effectively activates adaptive immunity through antigen-presenting cell uptake and antigen cross-presentation. Vaccination of engineered OMVs shows immediate tumor suppression and recruitment of infiltrating tumor-reactive immune cells. CONCLUSION The simplicity of the arginine coating strategy boasts the versatility of immuno-stimulating OMVs that can be broadly implemented to personalized bacterial immunotherapeutic applications.
Collapse
Affiliation(s)
- Suyang Wang
- Department of Pathology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II 307, Baltimore, MD, 21287, USA
| | - Chao-Cheng Chen
- Department of Pathology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II 307, Baltimore, MD, 21287, USA
| | - Ming-Hung Hu
- Department of Pathology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II 307, Baltimore, MD, 21287, USA
| | - Michelle Cheng
- Department of Pathology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II 307, Baltimore, MD, 21287, USA
| | - Hsin-Fang Tu
- Department of Pathology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II 307, Baltimore, MD, 21287, USA
| | - Ya-Chea Tsai
- Department of Pathology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II 307, Baltimore, MD, 21287, USA
| | - Jr-Ming Yang
- Department of Pathology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II 307, Baltimore, MD, 21287, USA
| | - T C Wu
- Department of Pathology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II 307, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II 307, Baltimore, MD, 21287, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Molecular Microbiology and Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chuan-Hsiang Huang
- Department of Pathology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II 307, Baltimore, MD, 21287, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II 307, Baltimore, MD, 21287, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, 1550 Orleans Street, CRB II 307, Baltimore, MD, 21287, USA.
| |
Collapse
|
3
|
Wang S, Cheng M, Chen CC, Chang CY, Tsai YC, Yang JM, Wu TC, Huang CH, Hung CF. Salmonella immunotherapy engineered with highly efficient tumor antigen coating establishes antigen-specific CD8+ T cell immunity and increases in antitumor efficacy with type I interferon combination therapy. Oncoimmunology 2023; 13:2298444. [PMID: 38170154 PMCID: PMC10761047 DOI: 10.1080/2162402x.2023.2298444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Bacteria-based cancer therapy employs various strategies to combat tumors, one of which is delivering tumor-associated antigen (TAA) to generate specific immunity. Here, we utilized a poly-arginine extended HPV E7 antigen (9RE7) for attachment on Salmonella SL7207 outer membrane to synthesize the bacterial vaccine Salmonella-9RE7 (Sal-9RE7), which yielded a significant improvement in the amount of antigen presentation compared to the previous lysine-extended antigen coating strategy. In TC-1 tumor mouse models, Sal-9RE7 monotherapy decreased tumor growth by inducing E7 antigen-specific immunity. In addition, pairing Sal-9RE7 with adjuvant Albumin-IFNβ (Alb-IFNβ), a protein cytokine fusion, the combination significantly increased the antitumor efficacy and enhanced immunogenicity in the tumor microenvironment (TME). Our study made a significant contribution to personalized bacterial immunotherapy via TAA delivery and demonstrated the advantage of combination therapy.
Collapse
Affiliation(s)
- Suyang Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michelle Cheng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chao-Cheng Chen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chia-Yu Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ya-Chea Tsai
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jr-Ming Yang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - TC Wu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Molecular Microbiology and Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chuan-Hsiang Huang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Abstract
The use of cancer vaccines is considered a promising therapeutic strategy in clinical oncology, which is achieved by stimulating antitumor immunity with tumor antigens delivered in the form of cells, peptides, viruses, and nucleic acids. The ideal cancer vaccine has many advantages, including low toxicity, specificity, and induction of persistent immune memory to overcome tumor heterogeneity and reverse the immunosuppressive microenvironment. Many therapeutic vaccines have entered clinical trials for a variety of cancers, including melanoma, breast cancer, lung cancer, and others. However, many challenges, including single antigen targeting, weak immunogenicity, off-target effects, and impaired immune response, have hindered their broad clinical translation. In this review, we introduce the principle of action, components (including antigens and adjuvants), and classification (according to applicable objects and preparation methods) of cancer vaccines, summarize the delivery methods of cancer vaccines, and review the clinical and theoretical research progress of cancer vaccines. We also present new insights into cancer vaccine technologies, platforms, and applications as well as an understanding of potential next-generation preventive and therapeutic vaccine technologies, providing a broader perspective for future vaccine design.
Collapse
Affiliation(s)
- Nian Liu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Xiangyu Xiao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Ziqiang Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| |
Collapse
|
5
|
Yadav A, Yadav S, Alam MA. Immunotherapies landscape and associated inhibitors for the treatment of cervical cancer. Med Oncol 2023; 40:328. [PMID: 37815596 DOI: 10.1007/s12032-023-02188-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/05/2023] [Indexed: 10/11/2023]
Abstract
Cervical cancer ranks as the fourth most common form of cancer worldwide. There is a large number of situations that may be examined in the developing world. The risk of contracting HPV (Human Papillomavirus) due to poor sanitation and sexual activity is mostly to blame for the disease's alarming rate of expansion. Immunotherapy is widely regarded as one of the most effective medicines available. The immunotherapy used to treat cervical cancer cells relies on inhibitors that block the immune checkpoint. The poly adenosine diphosphate ribose polymer inhibited cervical cancer cells by activating both the programmed death 1 (PD-1) and programmed death ligand 1 (CTLA-1) checkpoints, a strategy that has been shown to have impressive effects. Yet, immunotherapy directed towards tumors that have already been invaded by lymphocytes leaves a positive imprint on the healing process. Immunotherapy is used in conjunction with other treatments, including chemotherapy and radiation, to provide faster and more effective outcomes. In this combination therapy, several medications such as Pembrolizumab, Durvalumab, Atezolizumab, and so on are employed in clinical trials. Recent developments and future predictions suggest that vaccinations will soon be developed with the dual goal of reducing the patient's susceptibility to illness while simultaneously strengthening their immune system. Many clinical and preclinical studies are now investigating the effectiveness of immunotherapy in slowing the progression of cervical cancer. The field of immunotherapy is expected to witness more progress toward improving outcomes. Immunotherapies landscape and associated inhibitors for the treatment of Cervical Cancer.
Collapse
Affiliation(s)
- Agrima Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Shikha Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India.
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Plot No. 2, Sector 17-A, Yamuna Expressway, Gautam Buddh Nagar, Greater Noida, Uttar Pradesh, 201310, India.
| | - Md Aftab Alam
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| |
Collapse
|
6
|
Mo Y, Ma J, Zhang H, Shen J, Chen J, Hong J, Xu Y, Qian C. Prophylactic and Therapeutic HPV Vaccines: Current Scenario and Perspectives. Front Cell Infect Microbiol 2022; 12:909223. [PMID: 35860379 PMCID: PMC9289603 DOI: 10.3389/fcimb.2022.909223] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/06/2022] [Indexed: 12/20/2022] Open
Abstract
Persistent human papillomavirus (HPV) infection is recognized as the main cause of cervical cancer and other malignant cancers. Although early detection and treatment can be achieved by effective HPV screening methods and surgical procedures, the disease load has not been adequately mitigated yet, especially in the underdeveloped areas. Vaccine, being regarded as a more effective solution, is expected to prevent virus infection and the consequent diseases in the phases of both prevention and treatment. Currently, there are three licensed prophylactic vaccines for L1-VLPs, namely bivalent, quadrivalent and nonavalent vaccine. About 90% of HPV infections have been effectively prevented with the implementation of vaccines worldwide. However, no significant therapeutic effect has been observed on the already existed infections and lesions. Therapeutic vaccine designed for oncoprotein E6/E7 activates cellular immunity rather than focuses on neutralizing antibodies, which is considered as an ideal immune method to eliminate infection. In this review, we elaborate on the classification, mechanism, and clinical effects of HPV vaccines for disease prevention and treatment, in order to make improvements to the current situation of HPV vaccines by provoking new ideas.
Collapse
Affiliation(s)
- Yicheng Mo
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jiabing Ma
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Hongtao Zhang
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Junjie Shen
- IND Center, Chongqing Precision Biotech Co., Ltd., Chongqing, China
| | - Jun Chen
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Juan Hong
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Yanmin Xu
- IND Center, Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
- *Correspondence: Yanmin Xu, ; Cheng Qian,
| | - Cheng Qian
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
- *Correspondence: Yanmin Xu, ; Cheng Qian,
| |
Collapse
|
7
|
Peng S, Xing D, Ferrall L, Tsai YC, Roden RBS, Hung CF, Wu TC. Development of a Spontaneous HPV16 E6/E7-Expressing Head and Neck Squamous Cell Carcinoma in HLA-A2 Transgenic Mice. mBio 2022; 13:e0325221. [PMID: 35089069 PMCID: PMC8725581 DOI: 10.1128/mbio.03252-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 11/23/2021] [Indexed: 01/10/2023] Open
Abstract
Human papillomavirus (HPV)-associated head and neck squamous cell carcinoma (HNSCC) is a growing global health problem. HPV16 has been attributed to a majority of HPV-associated HNSCCs. In order to test candidate immunotherapies, we developed a spontaneous HPV16-driven HNSCC model in HLA-A2 (AAD) transgenic mice. We sought to eliminate the confounding effects of dominant HPV antigen presentation through murine major histocompatibility complex class I (MHC-I) via epitope mutagenesis (without compromising tumorigenicity). We generated HPV16 E6(R55K)(delK75) and E7(N53S) expression constructs with mutations in known dominant H-2Db epitopes and characterized their presentation through murine and human MHC-I molecules using in vitro and in vivo activation of HPV16 E6/E7 antigen-specific CD8+ T cells. In addition, we tested the ability of E6(R55K)(delK75) and E7(N53S) for oncogenicity. The mutated E7(N53S) abolished the presentation of murine H-2Db-restricted HPV16 E7 peptide (i.e., amino acids [aa] 49 to 57) cytotoxic T lymphocyte (CTL) epitope and resulted in HLA-A2-restricted presentation of the HPV16 E7 (aa 11 to 20)-specific CTL epitope. The mutated E6(R55K)(delK75) abolished the activation of murine MHC-I-restricted E6-specific CD8+ T cell-mediated immune responses in C57BL/6 mice. In addition, the vaccination led to the activation of human HLA-A2-restricted E6-specific CD8+ T cell-mediated immune responses in HLA-A2 (AAD) transgenic mice. Injection of DNA plasmids encoding LucE7(N53S)E6(R55K)(delK75), AKT, c-Myc, and SB100 followed by electroporation results in development of squamous cell carcinoma in the oral/pharyngeal cavity of all of the HLA-A2 (AAD) transgenic mice (5/5), with 2/5 tumor-bearing mice developing metastatic carcinoma in the neck lymph nodes. IMPORTANCE Our data indicate that mutated HPV16 E6(R55K)(delK75) and mutated HPV16 E7(N53S) DNA abolishes the presentation of HPV16 E6 and E7 through murine MHC-I and results in their presentation through human HLA-A2 molecules. Additionally, the mutated HPV16 E6 and E7 remain oncogenic. Our approach is potentially applicable to different human MHC-I transgenic mice for the identification of human MHC-I restricted HPV16 E6/E7-specific CTL epitopes as well as the generation of spontaneous HPV E6/E7-expressing oral/pharyngeal carcinoma.
Collapse
Affiliation(s)
- Shiwen Peng
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Deyin Xing
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Louise Ferrall
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Ya-Chea Tsai
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Richard B. S. Roden
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Chien-Fu Hung
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - T.-C. Wu
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Local radiotherapy and E7 RNA-LPX vaccination show enhanced therapeutic efficacy in preclinical models of HPV16 + cancer. Cancer Immunol Immunother 2021; 71:1975-1988. [PMID: 34971406 PMCID: PMC9293862 DOI: 10.1007/s00262-021-03134-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/15/2021] [Indexed: 10/25/2022]
Abstract
Human papilloma virus (HPV) infection is a causative agent for several cancers types (genital, anal and head and neck region). The HPV E6 and E7 proteins are oncogenic drivers and thus are ideal candidates for therapeutic vaccination. We recently reported that a novel ribonucleic acid lipoplex (RNA-LPX)-based HPV16 vaccine, E7 RNA-LPX, mediates regression of mouse HPV16+ tumors and establishes protective T cell memory. An HPV16 E6/E7 RNA-LPX vaccine is currently being investigated in two phase I and II clinical trials in various HPV-driven cancer types; however, it remains a high unmet medical need for treatments for patients with radiosensitive HPV16+ tumors. Therefore, we set out to investigate the therapeutic efficacy of E7 RNA-LPX vaccine combined with standard-of-care local radiotherapy (LRT). We demonstrate that E7 RNA-LPX synergizes with LRT in HPV16+ mouse tumors, with potent therapeutic effects exceeding those of either monotherapy. Mode of action studies revealed that the E7 RNA-LPX vaccine induced high numbers of intratumoral-E7-specific CD8+ T cells, rendering cold tumors immunologically hot, whereas LRT primarily acted as a cytotoxic therapy, reducing tumor mass and intratumor hypoxia by predisposing tumor cells to antigen-specific T cell-mediated killing. Overall, LRT enhanced the effector function of E7 RNA-LPX-primed T cell responses. The therapeutic synergy was dependent on total radiation dose, rather than radiation dose-fractionation. Together, these results show that LRT synergizes with E7 RNA-LPX and enhances its anti-tumor activity against HPV16+ cancer models. This work paves into a new translational therapy for HPV16+ cancer patients.
Collapse
|
9
|
Abstract
Owing to the presence of known tumor-specific viral antigens, human papillomavirus (HPV)-associated cancers are well suited for treatment with immunotherapy designed to unleash, amplify or replace the T cell arm of the adaptive immune system. Immune checkpoint blockade designed to unleash existing T cell immunity is currently Food and Drug Administration approved for certain HPV-associated cancers. More specific immunotherapies such as therapeutic vaccines and T cell receptor-engineered cellular therapy are currently in clinical development. Such therapies may offer more specific immune activation against viral tumor antigens and decrease the risk of immune-related adverse events. Current and planned clinical study of these treatments will determine their utility in the treatment of patients with newly diagnosed advanced stage or relapsed HPV-associated cancer.
Collapse
Affiliation(s)
- Maxwell Y Lee
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Clint T Allen
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
10
|
Ferrall L, Lin KY, Roden RBS, Hung CF, Wu TC. Cervical Cancer Immunotherapy: Facts and Hopes. Clin Cancer Res 2021; 27:4953-4973. [PMID: 33888488 DOI: 10.1158/1078-0432.ccr-20-2833] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/12/2021] [Accepted: 04/08/2021] [Indexed: 11/16/2022]
Abstract
It is a sad fact that despite being almost completely preventable through human papillomavirus (HPV) vaccination and screening, cervical cancer remains the fourth most common cancer to affect women worldwide. Persistent high-risk HPV (hrHPV) infection is the primary etiologic factor for cervical cancer. Upward of 70% of cases are driven by HPV types 16 and 18, with a dozen other hrHPVs associated with the remainder of cases. Current standard-of-care treatments include radiotherapy, chemotherapy, and/or surgical resection. However, they have significant side effects and limited efficacy against advanced disease. There are a few treatment options for recurrent or metastatic cases. Immunotherapy offers new hope, as demonstrated by the recent approval of programmed cell death protein 1-blocking antibody for recurrent or metastatic disease. This might be augmented by combination with antigen-specific immunotherapy approaches, such as vaccines or adoptive cell transfer, to enhance the host cellular immune response targeting HPV-positive cancer cells. As cervical cancer progresses, it can foster an immunosuppressive microenvironment and counteract host anticancer immunity. Thus, approaches to reverse suppressive immune environments and bolster effector T-cell functioning are likely to enhance the success of such cervical cancer immunotherapy. The success of nonspecific immunostimulants like imiquimod against genital warts also suggest the possibility of utilizing these immunotherapeutic strategies in cervical cancer prevention to treat precursor lesions (cervical intraepithelial neoplasia) and persistent hrHPV infections against which the licensed prophylactic HPV vaccines have no efficacy. Here, we review the progress and challenges in the development of immunotherapeutic approaches for the prevention and treatment of cervical cancer.
Collapse
Affiliation(s)
- Louise Ferrall
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland
| | - Ken Y Lin
- Department of Obstetrics and Gynecology and Women's Health, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Richard B S Roden
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland.,Department of Oncology, The Johns Hopkins University, Baltimore, Maryland.,Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland
| | - Chien-Fu Hung
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland.,Department of Oncology, The Johns Hopkins University, Baltimore, Maryland.,Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland
| | - T-C Wu
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland. .,Department of Oncology, The Johns Hopkins University, Baltimore, Maryland.,Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland.,Department of Molecular Microbiology and Immunology, The Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
11
|
Peng S, Ferrall L, Gaillard S, Wang C, Chi WY, Huang CH, Roden RBS, Wu TC, Chang YN, Hung CF. Development of DNA Vaccine Targeting E6 and E7 Proteins of Human Papillomavirus 16 (HPV16) and HPV18 for Immunotherapy in Combination with Recombinant Vaccinia Boost and PD-1 Antibody. mBio 2021; 12:e03224-20. [PMID: 33468698 PMCID: PMC7845631 DOI: 10.1128/mbio.03224-20] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy for cervical cancer should target high-risk human papillomavirus types 16 and 18, which cause 50% and 20% of cervical cancers, respectively. Here, we describe the construction and characterization of the pBI-11 DNA vaccine via the addition of codon-optimized human papillomavirus 18 (HPV18) E7 and HPV16 and 18 E6 genes to the HPV16 E7-targeted DNA vaccine pNGVL4a-SigE7(detox)HSP70 (DNA vaccine pBI-1). Codon optimization of the HPV16/18 E6/E7 genes in pBI-11 improved fusion protein expression compared to that in DNA vaccine pBI-10.1 that utilized the native viral sequences fused 3' to a signal sequence and 5' to the HSP70 gene of Mycobacterium tuberculosis Intramuscular vaccination of mice with pBI-11 DNA better induced HPV antigen-specific CD8+ T cell immune responses than pBI-10.1 DNA. Furthermore, intramuscular vaccination with pBI-11 DNA generated stronger therapeutic responses for C57BL/6 mice bearing HPV16 E6/E7-expressing TC-1 tumors. The HPV16/18 antigen-specific T cell-mediated immune responses generated by pBI-11 DNA vaccination were further enhanced by boosting with tissue-antigen HPV vaccine (TA-HPV). Combination of the pBI-11 DNA and TA-HPV boost vaccination with PD-1 antibody blockade significantly improved the control of TC-1 tumors and extended the survival of the mice. Finally, repeat vaccination with clinical-grade pBI-11 with or without clinical-grade TA-HPV was well tolerated in vaccinated mice. These preclinical studies suggest that the pBI-11 DNA vaccine may be used with TA-HPV in a heterologous prime-boost strategy to enhance HPV 16/18 E6/E7-specific CD8+ T cell responses, either alone or in combination with immune checkpoint blockade, to control HPV16/18-associated tumors. Our data serve as an important foundation for future clinical translation.IMPORTANCE Persistent expression of high-risk human papillomavirus (HPV) E6 and E7 is an obligate driver for several human malignancies, including cervical cancer, wherein HPV16 and HPV18 are the most common types. PD-1 antibody immunotherapy helps a subset of cervical cancer patients, and its efficacy might be improved by combination with active vaccination against E6 and/or E7. For patients with HPV16+ cervical intraepithelial neoplasia grade 2/3 (CIN2/3), the precursor of cervical cancer, intramuscular vaccination with a DNA vaccine targeting HPV16 E7 and then a recombinant vaccinia virus expressing HPV16/18 E6-E7 fusion proteins (TA-HPV) was safe, and half of the patients cleared their lesions in a small study (NCT00788164). Here, we sought to improve upon this therapeutic approach by developing a new DNA vaccine that targets E6 and E7 of HPV16 and HPV18 for administration prior to a TA-HPV booster vaccination and for application against cervical cancer in combination with a PD-1-blocking antibody.
Collapse
Affiliation(s)
- Shiwen Peng
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Louise Ferrall
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Stephanie Gaillard
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Chenguang Wang
- Department of Oncology Biostatistics, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Wei-Yu Chi
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Chuan-Hsiang Huang
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Richard B S Roden
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - T-C Wu
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Chien-Fu Hung
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Rotavirus VP6 Adjuvant Effect on Norovirus GII.4 Virus-Like Particle Uptake and Presentation by Bone Marrow-Derived Dendritic Cells In Vitro and In Vivo. J Immunol Res 2020; 2020:3194704. [PMID: 32411793 PMCID: PMC7204108 DOI: 10.1155/2020/3194704] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 12/20/2019] [Indexed: 12/31/2022] Open
Abstract
We have previously shown that rotavirus (RV) inner capsid protein VP6 has an adjuvant effect on norovirus (NoV) virus-like particle- (VLP-) induced immune responses and studied the adjuvant mechanism in immortalized cell lines used as antigen-presenting cells (APCs). Here, we investigated the uptake and presentation of RV VP6 and NoV GII.4 VLPs by primary bone marrow-derived dendritic cells (BMDCs). The adjuvant effect of VP6 on GII.4 VLP presentation and NoV-specific immune response induction by BMDC in vivo was also studied. Intracellular staining demonstrated that BMDCs internalized both antigens, but VP6 more efficiently than NoV VLPs. Both antigens were processed and presented to antigen-primed T cells, which responded by robust interferon γ secretion. When GII.4 VLPs and VP6 were mixed in the same pulsing reaction, a subpopulation of the cells had uptaken both antigens. Furthermore, VP6 copulsing increased GII.4 VLP uptake by 37% and activated BMDCs to secrete 2-5-fold increased levels of interleukin 6 and tumor necrosis factor α compared to VLP pulsing alone. When in vitro-pulsed BMDCs were transferred to syngeneic BALB/c mice, VP6 improved NoV-specific antibody responses. The results of this study support the earlier findings of VP6 adjuvant effect in vitro and in vivo.
Collapse
|
13
|
Kang TH, Mao CP, Kim YS, Kim TW, Yang A, Lam B, Tseng SH, Farmer E, Park YM, Hung CF. TLR9 acts as a sensor for tumor-released DNA to modulate anti-tumor immunity after chemotherapy. J Immunother Cancer 2019; 7:260. [PMID: 31619293 PMCID: PMC6794732 DOI: 10.1186/s40425-019-0738-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/11/2019] [Indexed: 12/19/2022] Open
Abstract
The tumor microenvironment exists in a state of dynamic equilibrium, in which a balance of agonist and antagonist signals govern the anti-tumor immune responses. Previous studies have shown that chemotherapy could shift this balance in favor of agonistic signals for the anti-tumor immune responses mounted by CD8+ cytotoxic T lymphocytes (CTL), providing sufficiently high antigen density within the tumor. We undertook the current study to characterize the anti-tumor immune response following chemotherapy and its underlying mechanisms. We show that this 'adjuvant effect' of chemotherapy is, at least partially, mediated by the release of tumor DNA and acts through the Toll-like receptor 9 (TLR9) pathway. We found that tumor-released DNA causes accumulation, antigen uptake, and maturation of dendritic cells (DCs) in the tumor in a TLR9-dependent manner. These DCs subsequently migrate into the draining lymph nodes and prime tumor-specific CTLs. Our study provides novel insights to the molecular and cellular mechanisms by which chemotherapy converts the tumor microenvironment into a site permissive for the activation of a potent tumor-specific adaptive immune response.
Collapse
Affiliation(s)
- Tae Heung Kang
- Department of Immunology, College of Medicine, Konkuk University, 268, Chungju, South Korea
| | - Chih-Ping Mao
- MD-PhD Program, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Graduate Program in Immunology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Young Seob Kim
- Department of Immunology, College of Medicine, Konkuk University, 268, Chungju, South Korea
| | - Tae Woo Kim
- Division of Infection and Immunology, Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Andrew Yang
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- MD-PhD Program, Baylor College of Medicine, Houston, TX, USA
| | - Brandon Lam
- Graduate Program in Immunology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Ssu-Hsueh Tseng
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Emily Farmer
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Yeong-Min Park
- Department of Immunology, College of Medicine, Konkuk University, 268, Chungju, South Korea.
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
14
|
Sun NY, Chen YL, Wu WY, Lin HW, Chiang YC, Chang CF, Tai YJ, Hsu HC, Chen CA, Sun WZ, Cheng WF. Blockade of PD-L1 Enhances Cancer Immunotherapy by Regulating Dendritic Cell Maturation and Macrophage Polarization. Cancers (Basel) 2019; 11:cancers11091400. [PMID: 31546897 PMCID: PMC6769724 DOI: 10.3390/cancers11091400] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/06/2019] [Accepted: 09/16/2019] [Indexed: 01/16/2023] Open
Abstract
The immuno-inhibitory checkpoint PD-L1, regulated by tumor cells and antigen-presenting cells (APCs), dampened the activation of T cells from the PD-1/PD-L1 axis. PD-L1-expressing APCs rather than tumor cells demonstrated the essential anti-tumor effects of anti-PD-L1 monotherapy in preclinical tumor models. Using the murine tumor model, we investigated whether anti-PD-L1 antibody increased the antigen-specific immune response and anti-tumor effects induced by the antigen-specific protein vaccine, as well as the possible mechanisms regarding activation of APCs. Anti-PD-L1 antibody combined with the PEK protein vaccine generated more potent E7-specific immunity (including the number and cytotoxic activity of E7-specific cytotoxic CD8+ T lymphocytes) and anti-tumor effects than protein vaccine alone. Anti-PD-L1 antibody enhanced the maturation of dendritic cells and the proportion of M1-like macrophages in tumor-draining lymph nodes and tumors in tumor-bearing mice treated with combinatorial therapy. PD-L1 blockade overturned the immunosuppressive status of the tumor microenvironment and then enhanced the E7 tumor-specific antigen-specific immunity and anti-tumor effects generated by an E7-specific protein vaccine through modulation of APCs in an E7-expressing small tumor model. Tumor-specific antigen (like HPV E7 antigen)-specific immunotherapy combined with APC-targeting modality by PD-L1 blockade has a high translational potential in E7-specific cancer therapy.
Collapse
Affiliation(s)
- Nai-Yun Sun
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Yu-Li Chen
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Wen-Yih Wu
- Department of Obstetrics and Gynecology, Far Eastern Memorial Hospital, New Taipei 220, Taiwan.
| | - Han-Wei Lin
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Ying-Cheng Chiang
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Chi-Fang Chang
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Yi-Jou Tai
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Heng-Cheng Hsu
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
- Department of Obstetrics and Gynecology, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu City 300, Taiwan.
| | - Chi-An Chen
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Wei-Zen Sun
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
- Department of Anesthesiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Wen-Fang Cheng
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| |
Collapse
|
15
|
Liu Y, Li H, Pi R, Yang Y, Zhao X, Qi X. Current strategies against persistent human papillomavirus infection (Review). Int J Oncol 2019; 55:570-584. [PMID: 31364734 DOI: 10.3892/ijo.2019.4847] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/16/2019] [Indexed: 11/06/2022] Open
Abstract
Human papillomavirus (HPV) is the most common sexually transmitted infection, exhibiting a tropism for the epidermis and mucosae. The link between persistent HPV infection and malignancies involving the anogenital tract as well as the head and neck has been well‑established, and it is estimated that HPV‑related cancers involving various anatomical sites account for 4.5% of all human cancers. Current prophylactic vaccines against HPV have enabled the prevention of associated malignancies. However, the sizeable population base of current infection in whom prophylactic vaccines are not applicable, certain high‑risk HPV types not included in vaccines, and the vast susceptible population in developing countries who do not have access to the costly prophylactic vaccines, put forward an imperative need for effective therapies targeting persistent infection. In this article, the life cycle of HPV, the mechanisms facilitating HPV evasion of recognition and clearance by the host immune system, and the promising therapeutic strategies currently under investigation, particularly antiviral drugs and therapeutic vaccines, are reviewed.
Collapse
Affiliation(s)
- Yu Liu
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hongyi Li
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ruyu Pi
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yang Yang
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaorong Qi
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
16
|
Grunwitz C, Salomon N, Vascotto F, Selmi A, Bukur T, Diken M, Kreiter S, Türeci Ö, Sahin U. HPV16 RNA-LPX vaccine mediates complete regression of aggressively growing HPV-positive mouse tumors and establishes protective T cell memory. Oncoimmunology 2019; 8:e1629259. [PMID: 31428528 PMCID: PMC6685602 DOI: 10.1080/2162402x.2019.1629259] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 10/27/2022] Open
Abstract
HPV16 infections are associated with a variety of cancers and there is compelling evidence that the transforming activity of HPV16 critically depends on the expression of the viral oncoproteins E6 and E7. Therapeutic cancer vaccines capable of generating durable and specific immunity against these HPV16 antigens hold great promise to achieve long-term disease control. Here we show in mice that HPV16 E7 RNA-LPX, an intravenously administered cancer vaccine based on immuno-pharmacologically optimized antigen-encoding mRNA, efficiently primes and expands antigen-specific effector and memory CD8+ T cells. HPV-positive TC-1 and C3 tumors of immunized mice are heavily infiltrated with activated immune cells and HPV16-specific T cells and are polarized towards a proinflammatory, cytotoxic and less immune-suppressive contexture. E7 RNA-LPX immunization mediates complete and durable remission of progressing tumors. Circulating memory T cells are highly cytotoxic and protect from tumor rechallenge. Moreover, E7 RNA-LPX immunization sensitizes anti-PD-L1 refractory tumors to checkpoint blockade. In conclusion, our data highlight the potential of HPV16 RNA-LPX for the treatment of HPV-driven cancers.
Collapse
Affiliation(s)
- Christian Grunwitz
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Center Mainz, Mainz, Germany
| | - Nadja Salomon
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Fulvia Vascotto
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Abderaouf Selmi
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Thomas Bukur
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Mustafa Diken
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany.,TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Sebastian Kreiter
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany.,TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Özlem Türeci
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Ugur Sahin
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany.,TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| |
Collapse
|
17
|
Sun NY, Chen YL, Lin HW, Chiang YC, Chang CF, Tai YJ, Chen CA, Sun WZ, Chien CL, Cheng WF. Immune checkpoint Ab enhances the antigen-specific anti-tumor effects by modulating both dendritic cells and regulatory T lymphocytes. Cancer Lett 2018; 444:20-34. [PMID: 30543813 DOI: 10.1016/j.canlet.2018.11.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 11/29/2018] [Accepted: 11/29/2018] [Indexed: 01/03/2023]
Abstract
We determined the anti-tumor effects and possible mechanisms of an antigen-specific DNA vaccine combined with PD-1 or CTLA-4 blockade. Using the HPV16 E6/E7+ syngeneic mouse tumor model, we investigated whether anti-CTLA-4 antibody (Ab) or anti-PD-1 Ab increases the antigen-specific anti-tumor effects and immune response induced by CTGF/E7 chimeric DNA vaccine and the possible mechanisms. Anti-PD-1 Ab or anti-CTLA-4 Ab combined with E7-specific DNA vaccine generated more potent antigen-specific immunity, including anti-E7 Abs and the number and cytotoxic activity of E7-specific cytotoxic CD8+ T lymphocytes, and anti-tumor effects than E7-specific DNA vaccine alone. In addition, the number of systemic and intratumoral Tregs was lower with the anti-PD-1 or anti-CTLA-4 Ab and E7-specific DNA vaccine. Furthermore, anti-PD-1 and anti-CTLA-4 Abs could enhance the maturation and abilities of intratumoral DCs to activate E7-specific cytotoxic CD8+ T cells. Immune checkpoint blockade overcomes the immunosuppressive status of the tumor-microenvironment to enhance the antigen-specific immunity and anti-tumor effects generated by an antigen-specific DNA vaccine. Antigen-specific immunotherapy combined with immune checkpoint blockade can be a novel strategy in clinical cancer therapy.
Collapse
Affiliation(s)
- Nai-Yun Sun
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Li Chen
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Han-Wei Lin
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ying-Cheng Chiang
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-Fang Chang
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Jou Tai
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-An Chen
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wei-Zen Sun
- Department of Anesthesiology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chung-Liang Chien
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Fang Cheng
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
18
|
Barros MR, de Melo CML, Barros MLCMGR, de Cássia Pereira de Lima R, de Freitas AC, Venuti A. Activities of stromal and immune cells in HPV-related cancers. J Exp Clin Cancer Res 2018; 37:137. [PMID: 29976244 PMCID: PMC6034319 DOI: 10.1186/s13046-018-0802-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023] Open
Abstract
The immune system is composed of immune as well as non-immune cells. As this system is a well-established component of human papillomavirus- (HPV)-related carcinogenesis, high risk human papillomavirus (hrHPV) prevents its routes and mechanisms in order to cause the persistence of infection. Among these mechanisms are those originated from stromal cells, which include the cancer-associated fibroblasts (CAFs), the myeloid-derived suppressor cells (MDSCs) and the host infected cells themselves, i.e. the keratinocytes. These types of cells play central role since they modulate immune cells activities to create a prosperous milieu for cancer development, and the knowledge how such interactions occur are essential for prognostic assessment and development of preventive and therapeutic approaches. Nevertheless, the precise mechanisms are not completely understood, and this lack of knowledge precluded the development of entirely efficient immunotherapeutic strategies for HPV-associated tumors. As a result, an intense work for attaining how host immune response works, and developing of effective therapies has been applied in the last decade. Based on this, this review aims to discuss the major mechanisms of immune and non-immune cells modulated by hrHPV and the potential and existing immunotherapies involving such mechanisms in HPV-related cancers. It is noticed that the combination of immunotherapies has been demonstrated to be essential for obtaining better results, especially because the possibility of increasing the modulating capacity of the HPV-tumor microenvironment has been shown to be central in strengthening the host immune system.
Collapse
Affiliation(s)
- Marconi Rego Barros
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Center of Biological Sciences, Federal University of Pernambuco, Cidade Universitária, Av. Prof Moraes Rego, 1235, Recife, PE CEP-50670-901 Brazil
| | - Cristiane Moutinho Lagos de Melo
- Laboratory of Immunological and Antitumor Analysis (LAIA), Department of Antibiotics, Center of Biological Sciences, Federal University of Pernambuco, Cidade Universitária, Av. Prof Artur de Sá, s/n, Recife, PE CEP-50740-525 Brazil
| | | | - Rita de Cássia Pereira de Lima
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Center of Biological Sciences, Federal University of Pernambuco, Cidade Universitária, Av. Prof Moraes Rego, 1235, Recife, PE CEP-50670-901 Brazil
| | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Center of Biological Sciences, Federal University of Pernambuco, Cidade Universitária, Av. Prof Moraes Rego, 1235, Recife, PE CEP-50670-901 Brazil
| | - Aldo Venuti
- HPV-Unit, Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| |
Collapse
|
19
|
Enhanced anti-tumor therapeutic efficacy of DNA vaccine by fusing the E7 gene to BAFF in treating human papillomavirus-associated cancer. Oncotarget 2018; 8:33024-33036. [PMID: 28423693 PMCID: PMC5464847 DOI: 10.18632/oncotarget.16032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 02/13/2017] [Indexed: 12/17/2022] Open
Abstract
B-cell-activating factor (BAFF) belongs to the tumor necrosis factor family that not only stimulates B and T cells but also counteracts immune tolerance. BAFF is also a type II membrane protein, which is secreted through the endoplasmic reticulum (ER)–Golgi apparatus pathway. Fusing an antigen to BAFF might enhance the presentation of major histocompatibility complex class I molecules. These characteristics represent an opportunity to enhance the antitumor effects of DNA vaccines. Therefore, we fused BAFF to human papillomavirus type 16 E7 as a DNA vaccine and evaluated its antitumor effects. We found that this vaccine increased E7-specific CD8+ T-cell immune responses, engendered major antitumor effects against E7-expressing tumors, and prolonged the survival of the immunized mice. Interestingly, vaccinating B-cell-deficient mice with BAFF–E7 revealed considerable E7-specific CD8+ T-cell immune responses, suggesting that B cells do not contribute to this immune response. Image analysis through confocal fluorescence microscopy revealed that fusing BAFF to E7 targeted the protein to the ER, but not BAFF lacking 128 N-terminal residues that generated a lower number of E7-specific CD8+ T cells in the vaccinated mice. Our data indicated that the ER-targeting characteristic of BAFF is the main factor improving the potency of DNA vaccines.
Collapse
|
20
|
Poly(I:C) enhanced anti-cervical cancer immunities induced by dendritic cells-derived exosomes. Int J Biol Macromol 2018; 113:1182-1187. [PMID: 29427678 DOI: 10.1016/j.ijbiomac.2018.02.034] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/06/2018] [Indexed: 12/12/2022]
Abstract
Dendritic cell (DC)-derived exosomes (Dexo) has been confirmed to be able to induce the specific anti-tumor immune response ex vivo and in vivo. Here, the aim of this study was to evaluate the application of the antigen-pulsed Dexo as a new vaccination platform in immunotherapy for cervical cancer. The immunogenic profile of the different Dexo was assessed by the cell proliferation, cytokines secretion and effector functions of CD8+ T cells and the splenocytes from Dexo-vaccinated mice. Furthermore, the anti-tumor immunity elicited by Dexo was further compared in cervical cancer-bearing mice. Dexo from DCs loaded with E749-57 peptide could efficiently induce the cytotoxic activity of CD8+ T cells on TC-1 tumor cells ex vivo, the proliferation and IFN-γ excretion of CD8+ T cells. Moreover, Dexo vaccine promoted the immune responses of vaccinated mice splenocytes induced by antigen E7 in vitro restimulation. Of note, poly(I:C) was significantly more potent inducer of the antigen-loaded Dexo mediated protective immunity responses for cervical cancer and further evidenced by that Dexo(E7+pIC) markedly inhibited the tumor growth and improved the survival rate of the tumor-bearing mice. We provided evidence that poly(I:C) dramatically increased the potent antitumoral immunity induced by antigen-pulsed Dexo for ameliorating cervical cancer.
Collapse
|
21
|
Yang A, Peng S, Farmer E, Zeng Q, Cheng MA, Pang X, Wu TC, Hung CF. Enhancing antitumor immunogenicity of HPV16-E7 DNA vaccine by fusing DNA encoding E7-antigenic peptide to DNA encoding capsid protein L1 of Bovine papillomavirus. Cell Biosci 2017; 7:46. [PMID: 28852471 PMCID: PMC5569540 DOI: 10.1186/s13578-017-0171-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/12/2017] [Indexed: 01/10/2023] Open
Abstract
Background Human papillomavirus (HPV) has been identified as the primary etiologic factor of cervical cancer, the fourth leading cause of cancer death in females worldwide. We have previously shown that coadministration of DNA encoding L1 capsid protein of Bovine papillomavirus (BPV) can enhance the antigen-specific immune response elicited by a therapeutic HPV16-E7 DNA vaccination. In this study, we sought to generate and evaluate the immunogenicity of a therapeutic HPV16-E7 DNA vaccine that encodes the fusion construct of HPV16-E7 and BPV-L1. Results We generated a therapeutic HPV16-E7 DNA vaccine construct, pcDNA3-BPVL1-E7(49-57), encoding the fusion sequence of full-length BPVL1 protein and a murine E7 antigenic epitope, aa49-57. Transfecting 293-Db cells with pcDNA3-BPVL1-E7(49-57) demonstrated that this DNA construct can effectively lead to the presentation of E7 epitope for the activation of E7-specific CD8+ T cells in vitro. Intramuscular vaccination of pcDNA3-BPVL1-E7(49-57) with electroporation generated a stronger E7-specific CD8+ T cell-mediated immune response than coadministration of pcDNA3-BPVL1 and pcDNA3-E7(49-57) in C57BL/6 mice. Furthermore, we observed that the strong E7-specific CD8+ T cell response elicited by pcDNA3-BPVL1-E7(49-57) vaccination translated into potent protective and therapeutic antitumor effects in C57BL/6 mice against HPV16-E7 expressing TC-1 tumor cells. Finally, using antibody depletion experiment, we showed that the antitumor immune response generated by pcDNA3-BPVL1-E7(49-57) is CD8+ T cell dependent, and CD4+ T cell and NK cell independent. Conclusion Treatment with fusion construct of BPV-L1 and HPV16-E7 epitope can elicit effective E7-specific antitumor immune response in mice. Due to the potential ability of the fusion DNA construct to also trigger immune responses specific to the L1 protein, the current study serves to support future design of HPV DNA vaccines encoding fusion HPVL1-E6/E7 constructs for the generation of both T cell and B cell mediated immune responses against HPV infections and associated diseases.
Collapse
Affiliation(s)
- Andrew Yang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD USA
| | - Shiwen Peng
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD USA
| | - Emily Farmer
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD USA
| | - Qi Zeng
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD USA
| | - Max A Cheng
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD USA
| | - Xiaowu Pang
- Department of Oral Pathology, Howard University College of Dentistry, Washington, DC USA
| | - T-C Wu
- Department of Pathology, 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.,Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD USA.,The Johns Hopkins University School of Medicine, CRB II Room 309, 1550 Orleans Street, Baltimore, MD 21231 USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD USA.,Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD USA.,The Johns Hopkins University School of Medicine, CRB II Room 307, 1550 Orleans Street, Baltimore, MD 21231 USA
| |
Collapse
|
22
|
Ahn SB, Lee SB, Singh TD, Cho SJ, Kim SK, Lee IK, Jeong SY, Ahn BC, Lee J, Lee SW, Jeon YH. Multimodality Imaging of Bone Marrow-Derived Dendritic Cell Migration and Antitumor Immunity. Transl Oncol 2017; 10:262-270. [PMID: 28214774 PMCID: PMC5314440 DOI: 10.1016/j.tranon.2017.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/11/2017] [Accepted: 01/13/2017] [Indexed: 11/17/2022] Open
Abstract
Here, we sought to monitor bone marrow-derived dendritic cell (BMDC) migration and antitumor effects using a multimodal reporter imaging strategy in living mice. BMDCs were transduced with retroviral vector harboring human sodium iodide symporter (hNIS, nuclear imaging reporter), firefly luc2 (optical imaging reporter), and thy1.1 (surrogate marker of NIS and luc2) genes (BMDC/NF cells). No significant differences in biological functions, including cell proliferation, antigen uptake, phenotype expression, and migration ability, were observed between BMDC and BMDC/NF cells. Combined bioluminescence imaging and I-124 positron emission tomography/computed tomography clearly revealed the migration of BMDC/NF cells to draining popliteal lymph nodes at day 7 postinjection. Interestingly, marked tumor protection was observed in mice immunized with TC-1 lysate-pulsed BMDC/NF cells. Our findings suggested that multimodal reporter gene imaging of NIS and luciferase could provide insights into the biological behaviors of dendritic cells in living organisms and could be a useful tool for the optimization of DC-based immunotherapy protocols.
Collapse
Affiliation(s)
- Su-Bi Ahn
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Sang Bong Lee
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea
| | - Thoudam Debraj Singh
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Sung Jin Cho
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea; New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, South Korea
| | - Sang Kyoon Kim
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - In-Kyu Lee
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea; Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea.
| | - Yong Hyun Jeon
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea; Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea.
| |
Collapse
|
23
|
Yang A, Farmer E, Lin J, Wu TC, Hung CF. The current state of therapeutic and T cell-based vaccines against human papillomaviruses. Virus Res 2016; 231:148-165. [PMID: 27932207 DOI: 10.1016/j.virusres.2016.12.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 12/01/2016] [Accepted: 12/02/2016] [Indexed: 12/12/2022]
Abstract
Human papillomavirus (HPV) is known to be a necessary factor for many gynecologic malignancies and is also associated with a subset of head and neck malignancies. This knowledge has created the opportunity to control these HPV-associated cancers through vaccination. However, despite the availability of prophylactic HPV vaccines, HPV infections remain extremely common worldwide. In addition, while prophylactic HPV vaccines have been effective in preventing infection, they are ineffective at clearing pre-existing HPV infections. Thus, there is an urgent need for therapeutic and T cell-based vaccines to treat existing HPV infections and HPV-associated lesions and cancers. Unlike prophylactic vaccines, which generate neutralizing antibodies, therapeutic, and T cell-based vaccines enhance cell-mediated immunity against HPV antigens. Our review will cover various therapeutic and T cell-based vaccines in development for the treatment of HPV-associated diseases. Furthermore, we review the strategies to enhance the efficacy of therapeutic vaccines and the latest clinical trials on therapeutic and T cell-based HPV vaccines.
Collapse
Affiliation(s)
- Andrew Yang
- Department of Pathology, Johns Hopkins University, Baltimore, MD USA
| | - Emily Farmer
- Department of Pathology, Johns Hopkins University, Baltimore, MD USA
| | - John Lin
- Department of Pathology, Johns Hopkins University, Baltimore, MD USA
| | - T-C Wu
- Department of Pathology, Johns Hopkins University, Baltimore, MD USA; Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, MD USA; Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD USA; Department of Oncology, Johns Hopkins University, Baltimore, MD USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, Baltimore, MD USA; Department of Oncology, Johns Hopkins University, Baltimore, MD USA.
| |
Collapse
|
24
|
Yang A, Farmer E, Wu TC, Hung CF. Perspectives for therapeutic HPV vaccine development. J Biomed Sci 2016; 23:75. [PMID: 27809842 PMCID: PMC5096309 DOI: 10.1186/s12929-016-0293-9] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 10/26/2016] [Indexed: 12/24/2022] Open
Abstract
Background Human papillomavirus (HPV) infections and associated diseases remain a serious burden worldwide. It is now clear that HPV serves as the etiological factor and biologic carcinogen for HPV-associated lesions and cancers. Although preventative HPV vaccines are available, these vaccines do not induce strong therapeutic effects against established HPV infections and lesions. These concerns create a critical need for the development of therapeutic strategies, such as vaccines, to treat these existing infections and diseases. Main Body Unlike preventative vaccines, therapeutic vaccines aim to generate cell-mediated immunity. HPV oncoproteins E6 and E7 are responsible for the malignant progression of HPV-associated diseases and are consistently expressed in HPV-associated diseases and cancer lesions; therefore, they serve as ideal targets for the development of therapeutic HPV vaccines. In this review we revisit therapeutic HPV vaccines that utilize this knowledge to treat HPV-associated lesions and cancers, with a focus on the findings of recent therapeutic HPV vaccine clinical trials. Conclusion Great progress has been made to develop and improve novel therapeutic HPV vaccines to treat existing HPV infections and diseases; however, there is still much work to be done. We believe that therapeutic HPV vaccines have the potential to become a widely available and successful therapy to treat HPV and HPV-associated diseases in the near future.
Collapse
Affiliation(s)
- Andrew Yang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Emily Farmer
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - T C Wu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.,Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, MD, USA.,Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA. .,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA. .,The Johns Hopkins University School of Medicine, CRB II Room 307, 1550 Orleans Street, Baltimore, MD, 21231, USA.
| |
Collapse
|
25
|
Affiliation(s)
- Michael A. Steller
- Program in Women's Oncology, Women and Infants' Hospital, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Brown University School of Medicine, Providence, Rhode Island; St. Elizabeth's Medical Center, Division of Gynecologic Oncology, 736 Cambridge Street, Boston, MA 02135-2997
| |
Collapse
|
26
|
Gu L, Icyuz M, Krendelchtchikova V, Krendelchtchikov A, Johnston AE, Matthews QL. Development of an Ad5H3 Chimera Using the "Antigen Capsid-Incorporation" Strategy for an Alternative Vaccination Approach. Open Virol J 2016; 10:10-20. [PMID: 27335626 PMCID: PMC4892130 DOI: 10.2174/1874357901610010010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 02/10/2016] [Accepted: 02/23/2016] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Adenovirus type 5 (Ad5) achieved success as a conventional transgene vaccine vector in preclinical trials, however; achieved poor efficiency in some of the clinical trials, due to the major hurdle associated with Ad5 pre-existing immunity (PEI) in the majority of the human population. OBJECTIVE We sought to generate Ad5-based chimeras to assess their capabilities to bypass this bottleneck and to induce antigen-specific humoral immune response. METHODS A His6 tag was incorporated into the hypervariable region 2 (HVR2) of hexon3 (H3) capsid protein using the "Antigen Capsid-Incorporation" strategy. This lead to the construction of a viral chimera, Ad5H3-HVR2-His. Ad5H3 was generated previously by substituting the hexon of Ad5 (hexon5) with the hexon from adenovirus type 3 (Ad3). RESULTS His6 was presented on the viral capsid surface and recognized by a His6 antibody. An in vitro neutralization assay with Ad5 sera indicated the ability of Ad5 chimeras to partially escape Ad5 immunity. Immunization with Ad5H3-HVR2-His generated significant humoral response to the incorporated tagged peptide, when compared to the immunizations with controls. CONCLUSION Based on our in vitro studies the data suggested that Ad5H3 as a novel chimeric vaccine platform yields the possibility to escape Ad5 neutralization, and the potential to generate robust humoral immunity against incorporated antigens using the "Antigen Capsid-Incorporation" strategy.
Collapse
Affiliation(s)
- Linlin Gu
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, 845 19 street south, Birmingham, AL,35294, USA
| | - Mert Icyuz
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Valentina Krendelchtchikova
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, 845 19 street south, Birmingham, AL,35294, USA
| | - Alexandre Krendelchtchikov
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, 845 19 street south, Birmingham, AL,35294, USA
| | - Alison E Johnston
- Division of Natural Sciences and Math, Miles College, Fairfield, AL, 35064, USA
| | - Qiana L Matthews
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, 845 19 street south, Birmingham, AL,35294, USA; Center for AIDS Research, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| |
Collapse
|
27
|
Yang A, Jeang J, Cheng K, Cheng T, Yang B, Wu TC, Hung CF. Current state in the development of candidate therapeutic HPV vaccines. Expert Rev Vaccines 2016; 15:989-1007. [PMID: 26901118 DOI: 10.1586/14760584.2016.1157477] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The identification of human papillomavirus (HPV) as an etiological factor for HPV-associated malignancies creates the opportunity to control these cancers through vaccination. Currently, available preventive HPV vaccines have not yet demonstrated strong evidences for therapeutic effects against established HPV infections and lesions. Furthermore, HPV infections remain extremely common. Thus, there is urgent need for therapeutic vaccines to treat existing HPV infections and HPV-associated diseases. Therapeutic vaccines differ from preventive vaccines in that they are aimed at generating cell-mediated immunity rather than neutralizing antibodies. The HPV-encoded early proteins, especially oncoproteins E6 and E7, form ideal targets for therapeutic HPV vaccines since they are consistently expressed in HPV-associated malignancies and precancerous lesions, playing crucial roles in the generation and maintenance of HPV-associated disease. Our review will cover various therapeutic vaccines in development for the treatment of HPV-associated lesions and cancers. Furthermore, we review strategies to enhance vaccine efficacy and the latest clinical trials on therapeutic HPV vaccines.
Collapse
Affiliation(s)
- Andrew Yang
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Jessica Jeang
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Kevin Cheng
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Ting Cheng
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Benjamin Yang
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - T-C Wu
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA.,b Department of Obstetrics and Gynecology , Johns Hopkins University , Baltimore , MD , USA.,c Department of Molecular Microbiology and Immunology , Johns Hopkins University , Baltimore , MD , USA.,d Department of Oncology , Johns Hopkins University , Baltimore , MD , USA
| | - Chien-Fu Hung
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA.,d Department of Oncology , Johns Hopkins University , Baltimore , MD , USA
| |
Collapse
|
28
|
Lee HW, Yoon SY, Singh TD, Choi YJ, Lee HJ, Park JY, Jeong SY, Lee SW, Ha JH, Ahn BC, Jeon YH, Lee J. Tracking of dendritic cell migration into lymph nodes using molecular imaging with sodium iodide symporter and enhanced firefly luciferase genes. Sci Rep 2015; 5:9865. [PMID: 25974752 PMCID: PMC4431315 DOI: 10.1038/srep09865] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 02/26/2015] [Indexed: 12/14/2022] Open
Abstract
We sought to evaluate the feasibility of molecular imaging using the human sodium iodide symporter (hNIS) gene as a reporter, in addition to the enhanced firefly luciferase (effluc) gene, for tracking dendritic cell (DCs) migration in living mice. A murine dendritic cell line (DC2.4) co-expressing hNIS and effluc genes (DC/NF) was established. For the DC-tracking study, mice received either parental DCs or DC/NF cells in the left or right footpad, respectively, and combined I-124 PET/CT and bioluminescence imaging (BLI) were performed. In vivo PET/CT imaging with I-124 revealed higher activity of the radiotracer in the draining popliteal lymph nodes (DPLN) of the DC/NF injection site at day 1 than DC injection site (p < 0.05). The uptake value further increased at day 4 (p < 0.005). BLI also demonstrated migration of DC/NF cells to the DPLNs at day 1 post-injection, and signals at the DPLNs were much higher at day 4. These data support the feasibility of hNIS reporter gene imaging in the tracking of DC migration to lymphoid organs in living mice. DCs expressing the NIS reporter gene could be a useful tool to optimize various strategies of cell-based immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Hong Je Lee
- Department of Nuclear Medicine, Dongnam Institution of Radiological &Medical SciencesBusan
| | - Ji Young Park
- Department of Pathology, School of Medicine, Kyungpook National UniversityDaegu
| | | | - Sang-Woo Lee
- 1] Department of Nuclear Medicine [2] Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, 807 Hogukro, Bukgu, Daegu
| | | | | | - Yong Hyun Jeon
- 1] Department of Nuclear Medicine [2] Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, 807 Hogukro, Bukgu, Daegu
| | - Jaetae Lee
- 1] Department of Nuclear Medicine [2] Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu, 701-310, Republic of Korea
| |
Collapse
|
29
|
Local administration of granulocyte macrophage colony-stimulating factor induces local accumulation of dendritic cells and antigen-specific CD8+ T cells and enhances dendritic cell cross-presentation. Vaccine 2015; 33:1549-55. [PMID: 25701675 DOI: 10.1016/j.vaccine.2015.02.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/26/2015] [Accepted: 02/07/2015] [Indexed: 11/20/2022]
Abstract
Immunotherapy has emerged as a promising treatment strategy for the control of HPV-associated malignancies. Various therapeutic HPV vaccines have elicited potent antigen-specific CD8+ T cell mediated antitumor immune responses in preclinical models and are currently being tested in several clinical trials. Recent evidence indicates the importance of local immune activation, and higher number of immune cells in the site of lesion correlates with positive prognosis. Granulocyte macrophage colony-stimulating factor (GMCSF) has been reported to posses the ability to induce migration of antigen presentation cells and CD8+ T cells. Therefore, in the current study, we employ a combination of systemic therapeutic HPV DNA vaccination with local GMCSF application in the TC-1 tumor model. We show that intramuscular vaccination with CRT/E7 DNA followed by GMCSF intravaginal administration effectively controls cervicovaginal TC-1 tumors in mice. Furthermore, we observe an increase in the accumulation of E7-specific CD8+ T cells and dendritic cells in vaginal tumors following the combination treatment. In addition, we show that GMCSF induces activation and maturation in dendritic cells and promotes antigen cross-presentation. Our results support the clinical translation of the combination treatment of systemic therapeutic vaccination followed by local GMCSF administration as an effective strategy for tumor treatment.
Collapse
|
30
|
Huang Z, Peng S, Knoff J, Lee SY, Yang B, Wu TC, Hung CF. Combination of proteasome and HDAC inhibitor enhances HPV16 E7-specific CD8+ T cell immune response and antitumor effects in a preclinical cervical cancer model. J Biomed Sci 2015; 22:7. [PMID: 25591912 PMCID: PMC4298946 DOI: 10.1186/s12929-014-0111-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 12/31/2014] [Indexed: 11/15/2022] Open
Abstract
Background Bortezomib, a proteasome inhibitor and suberoylanilide hydroxamic acid (SAHA, also known as Vorinostat), a histone deacetylase inhibitor, have been recognized as potent chemotherapeutic drugs. Bortezomib and SAHA are FDA-approved for the treatment of cutaneous T cell lymphoma and multiple myeloma/mantle cell lymphoma, respectively. Furthermore, the combination of the bortezomib and SAHA has been tested in a variety of preclinical models and in clinical trials and may be ideal for the treatment of cancer. However, it remains unclear how this treatment strategy affects the host immune response against tumors. Results Here, we used a well-defined E6/E7-expressing tumor model to examine how the immune system can be motivated to act against tumor cells expressing tumor antigens. We demonstrate that the combination of bortezomib and SAHA elicits potent antitumor effects in TC-1 tumor-bearing mice. Additionally, we are the first to show that treatment with bortezomib and SAHA leads to tumor-specific immunity by rendering tumor cells more susceptible to killing by antigen-specific CD8+ T cells than treatment with either drug alone. Conclusions The current study serves an important foundation for the future clinical application of both drugs for the treatment of cervical cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12929-014-0111-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhuomin Huang
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA. .,Department of Gynecology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China.
| | - Shiwen Peng
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA.
| | - Jayne Knoff
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA.
| | - Sung Yong Lee
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA. .,Department of Internal Medicine, Korea University Medical Center, Seoul, South Korea.
| | - Benjamin Yang
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA.
| | - Tzyy-Choou Wu
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA. .,Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, MD, USA. .,Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions, Baltimore, MD, USA. .,Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA.
| |
Collapse
|
31
|
Huang CT, Chang MC, Chen YL, Chen TC, Chen CA, Cheng WF. Insulin-like growth factors inhibit dendritic cell-mediated anti-tumor immunity through regulating ERK1/2 phosphorylation and p38 dephosphorylation. Cancer Lett 2015; 359:117-26. [PMID: 25592043 DOI: 10.1016/j.canlet.2015.01.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/26/2014] [Accepted: 01/06/2015] [Indexed: 01/21/2023]
Abstract
Insulin-like growth factors (IGFs) can promote tumorigenesis via inhibiting the apoptosis of cancer cells. The relationship between IGFs and dendritic cell (DC)-mediated immunity were investigated. Advanced-stage ovarian carcinoma patients were first evaluated to show higher IGF-1 and IGF-2 concentrations in their ascites than early-stage patients. IGFs could suppress DCs' maturation, antigen presenting abilities, and the ability to activate antigen-specific CD8(+) T cell. IGF-treated DCs also secreted higher concentrations of IL-10 and TNF-α. IGF-treated DCs showed decreased ERK1/2 phosphorylation and reduced p38 dephosphorylation. The percentages of matured DCs in the ascites were significantly lower in the IGF-1 or IGF-2 highly-expressing WF-3 tumor-bearing mice. The IGF1R inhibitor - NVP-AEW541, could block the effects of IGFs to rescue DCs' maturation and to restore DC-mediated antigen-specific immunity through enhancing ERK1/2 phosphorylation and p38 dephosphorylation. IGFs can inhibit DC-mediated anti-tumor immunity through suppressing maturation and function and the IGF1R inhibitor could restore the DC-mediated anti-tumor immunity. Blockade of IGFs could be a potential strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Ching-Ting Huang
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Cheng Chang
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Li Chen
- Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei, Taiwan
| | - Tsung-Ching Chen
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-An Chen
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Fang Cheng
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
32
|
Kang TH, Knoff J, Yeh WH, Yang B, Wang C, Kim YS, Kim TW, Wu TC, Hung CF. Treatment of tumors with vitamin E suppresses myeloid derived suppressor cells and enhances CD8+ T cell-mediated antitumor effects. PLoS One 2014; 9:e103562. [PMID: 25072795 PMCID: PMC4114790 DOI: 10.1371/journal.pone.0103562] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 07/03/2014] [Indexed: 02/04/2023] Open
Abstract
Vitamin E has been shown to have strong anticarcinogenic properties, including antioxidant characteristics, making it an ideal candidate for use in combination with immunotherapies that modify the tumor microenvironment. The tumor microenvironment contains immunosuppressive components, which can be diminished, and immunogenic components, which can be augmented by immunotherapies in order to generate a productive immune response. In the current study, we employ the α-tocopherol succinate isomer of vitamin E to reduce immunosuppression by myeloid derived suppressor cells (MDSCs) as well as adoptive transfer of antigen-specific CD8+ T cells to generate potent antitumor effects against the HPV16 E7-expressing TC-1 tumor model. We show that vitamin E alone induces necrosis of TC-1 cells and elicits antitumor effects in TC-1 tumor-bearing mice. We further demonstrate that vitamin E reverses the suppression of T cell activation by MDSCs and that this effect is mediated in part by a nitric oxide-dependent mechanism. Additionally, treatment with vitamin E reduces the percentage of MDSCs in tumor loci, and induces a higher percentage of T cells, following T cell adoptive transfer. Finally, we demonstrate that treatment with vitamin E followed by E7-specific T cell adoptive transfer experience elicits potent antitumor effects in tumor-bearing mice. Our data provide additional evidence that vitamin E has anticancer properties and that it has promise for use as an adjuvant in combination with a variety of cancer therapies.
Collapse
Affiliation(s)
- Tae Heung Kang
- Seoul Department of Immunology, College of Medicine, Konkuk University, Chungju, South Korea
- * E-mail: (THK); (CFH)
| | - Jayne Knoff
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Wei-Hsi Yeh
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Benjamin Yang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Chenguang Wang
- Department of Biostatistics and Bioinformatics, Sidney Kimmel Cancer Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Young Seob Kim
- Seoul Department of Immunology, College of Medicine, Konkuk University, Chungju, South Korea
| | - Tae Woo Kim
- Division of Infection and Immunology, Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Tzyy-Choou Wu
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
- Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
- Department of Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
- * E-mail: (THK); (CFH)
| |
Collapse
|
33
|
Wu CY, Yang LH, Yang HY, Knoff J, Peng S, Lin YH, Wang C, Alvarez RD, Pai SI, Roden RBS, Hung CF, Wu TC. Enhanced cancer radiotherapy through immunosuppressive stromal cell destruction in tumors. Clin Cancer Res 2013; 20:644-57. [PMID: 24300786 DOI: 10.1158/1078-0432.ccr-13-1334] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE Radiotherapy kills cancer cells by causing DNA damage, and stimulates a systemic antitumor immune response by releasing tumor antigen and endogenous adjuvant within the tumor microenvironment. However, radiotherapy also induces the recruitment of immunosuppressive myeloid cells, which can interfere with the antitumor immune responses elicited by apoptotic tumor cells. We hypothesized that local delivery of vaccine following radiotherapy will lead to the priming of antigen-specific CTL immune responses and render immunosuppressive myeloid cells susceptible to killing by the activated CTLs. EXPERIMENTAL DESIGN Using several antigenic systems, we tested whether intratumoral injection of antigenic peptide/protein in irradiated tumors would be able to prime CTLs as well as load myeloid cells with antigen, rendering them susceptible to antigen-specific CTL killing. RESULTS We show that by combining radiotherapy and targeted antigenic peptide delivery to the tumor, the adjuvant effect generated by radiotherapy itself was sufficient to elicit the priming and expansion of antigen-specific CTLs, through the type I IFN-dependent pathway, leading to synergistic therapeutic antitumor effects compared with either treatment alone. In addition, using two different types of transgenic mice, we demonstrated that CTL-mediated killing of stromal cells in tumors by our approach is important for tumor control. Finally, we confirmed the efficacy of this approach in our preclinical model using two clinically tested therapeutic human papilloma virus (HPV) vaccines. CONCLUSIONS These data serve as an important foundation for the future clinical translation of radiotherapy combined with a clinically tested therapeutic HPV vaccine for the control of HPV-associated cancers.
Collapse
Affiliation(s)
- Chao-Yi Wu
- Authors' Affiliations: Departments of Pathology, Obstetrics and Gynecology, Molecular Microbiology and Immunology, Oncology, and Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions; Department of Biostatistics and Bioinformatics, Sidney Kimmel Cancer Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland; Departments of Obstetrics and Gynecology, Kunming Medical University, Yunnan, China; Nephrology, Chang-Gung Memorial Hospital, Gueishan; Department of Obstetrics and Gynecology, Tri-Service General Hospital, Taipei, Taiwan; and Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Lee SY, Huang Z, Kang TH, Soong RS, Knoff J, Axenfeld E, Wang C, Alvarez RD, Chen CS, Hung CF, Wu TC. Histone deacetylase inhibitor AR-42 enhances E7-specific CD8⁺ T cell-mediated antitumor immunity induced by therapeutic HPV DNA vaccination. J Mol Med (Berl) 2013; 91:1221-31. [PMID: 23715898 PMCID: PMC3783646 DOI: 10.1007/s00109-013-1054-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED We have previously created a potent DNA vaccine encoding calreticulin linked to the human papillomavirus (HPV) oncogenic protein E7 (CRT/E7). While treatment with the CRT/E7 DNA vaccine generates significant tumor-specific immune responses in vaccinated mice, the potency with the DNA vaccine could potentially be improved by co-administration of a histone deacetylase inhibitor (HDACi) as HDACi has been shown to increase the expression of MHC class I and II molecules. Thus, we aimed to determine whether co-administration of a novel HDACi, AR-42, with therapeutic HPV DNA vaccines could improve the activation of HPV antigen-specific CD8(+) T cells, resulting in potent therapeutic antitumor effects. To do so, HPV-16 E7-expressing murine TC-1 tumor-bearing mice were treated orally with AR-42 and/or CRT/E7 DNA vaccine via gene gun. Mice were monitored for E7-specific CD8(+) T cell immune responses and antitumor effects. TC-1 tumor-bearing mice treated with AR-42 and CRT/E7 DNA vaccine experienced longer survival, decreased tumor growth, and enhanced E7-specific immune response compared to mice treated with AR-42 or CRT/E7 DNA vaccine alone. Additionally, treatment of TC-1 cells with AR-42 increased the surface expression of MHC class I molecules and increased the susceptibility of tumor cells to the cytotoxicity of E7-specific T cells. This study indicates the ability of AR-42 to significantly enhance the potency of the CRT/E7 DNA vaccine by improving tumor-specific immune responses and antitumor effects. Both AR-42 and CRT/E7 DNA vaccines have been used in independent clinical trials; the current study serves as foundation for future clinical trials combining both treatments in cervical cancer therapy. KEY MESSAGE AR-42, a novel HDAC inhibitor, enhances potency of therapeutic HPV DNA vaccines AR-42 treatment leads to strong E7-specific CD8+ T cell immune responses AR-42 improves tumor-specific immunity and antitumor effects elicited by HPV DNA vaccine AR-42 is more potent than clinically available HDACi in combination with HPV DNA vaccine.
Collapse
Affiliation(s)
- Sung Yong Lee
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Internal Medicine, Korea University Medical Center, Seoul, South Korea
| | - Zhuomin Huang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Gynecology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Tae Heung Kang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Ruey-Shyang Soong
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of General Surgery, Chang Gung Memorial Hospital at Keelung, Taiwan
| | - Jayne Knoff
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Ellen Axenfeld
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Chenguang Wang
- Department of Biostatistics and Bioinformatics, Sidney Kimmel Cancer Comprehensive Cancer Center Johns Hopkins University, Baltimore, MD, USA
| | - Ronald D. Alvarez
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham
| | - Ching-Shih Chen
- Division of Medical Chemistry, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - T.-C. Wu
- Department of Pathology, 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
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|
35
|
Lee SY, Kang TH, Knoff J, Huang Z, Soong RS, Alvarez RD, Hung CF, Wu TC. Intratumoral injection of therapeutic HPV vaccinia vaccine following cisplatin enhances HPV-specific antitumor effects. Cancer Immunol Immunother 2013; 62:1175-85. [PMID: 23615841 DOI: 10.1007/s00262-013-1421-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 03/25/2013] [Indexed: 12/20/2022]
Abstract
Despite the conventional treatments of radiation therapy and chemotherapy, the 5-year survival rates for patients with advanced-stage cervical cancers remain low. Cancer immunotherapy has emerged as an alternative, innovative therapy that may improve survival. Here, we utilize a preclinical HPV-16 E6/E7-expressing tumor model, TC-1, and employ the chemotherapeutic agent cisplatin to generate an accumulation of CD11c+ dendritic cells in tumor loci making it an ideal location for the administration of therapeutic vaccines. Following cisplatin treatment, we tested different routes of administration of a therapeutic HPV vaccinia vaccine encoding HPV-16 E7 antigen (CRT/E7-VV). We found that TC-1 tumor-bearing C57BL/6 mice treated with cisplatin and intratumoral injection of CRT/E7-VV significantly increased E7-specific CD8+ T cells in the blood and generated potent local and systemic antitumor immune responses compared to mice receiving cisplatin and CRT/E7-VV intraperitoneally or mice treated with cisplatin alone. We further extended our study using a clinical grade recombinant vaccinia vaccine encoding HPV-16/18 E6/E7 antigens (TA-HPV). We found that intratumoral injection with TA-HPV following cisplatin treatment also led to increased E7-specific CD8+ T cells in the blood as well as significantly decreased tumor size compared to intratumoral injection with wild type vaccinia virus. Our study has strong implications for future clinical translation using intratumoral injection of TA-HPV in conjunction with the current treatment strategies for patients with advanced cervical cancer.
Collapse
Affiliation(s)
- Sung Yong Lee
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 309, 1550 Orleans Street, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Kang TH, Mao CP, La V, Chen A, Hung CF, Wu TC. Innovative DNA vaccine to break immune tolerance against tumor self-antigen. Hum Gene Ther 2013; 24:181-8. [PMID: 23268564 DOI: 10.1089/hum.2012.141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Vaccination is, in theory, a safe and effective approach for controlling disseminated or metastatic cancer due to the specificity of the mammalian immune system, yet its success in the clinic has been hampered thus far by the problem of immune tolerance to tumor self-antigen. Here we describe a DNA vaccination strategy that is able to control cancer by overcoming immune tolerance to tumor self-antigen. We engineered a DNA construct encoding a dimeric form of a secreted single-chain trimer of major histocompatibility complex class I heavy chain, β2-microglobulin, and peptide antigen linked to immunoglobulin G (SCT-Ag/IgG). The chimeric protein was able to bind to antigen-specific CD8(+) T cells with nearly 100% efficiency and strongly induce their activation and proliferation. In addition, the chimeric protein was able to coat professional antigen-presenting cells through the F(c) receptor to activate antigen-specific CD8(+) T cells. Furthermore, intradermal vaccination with DNA-encoding SCT-Ag/IgG could generate significant numbers of cytotoxic effector T cells against tumor self-antigen and leads to successful therapeutic outcomes in a preclinical model of metastatic melanoma. Our data suggest that the DNA vaccine strategy described in the current study is able to break immune tolerance against endogenous antigen from melanoma and result in potent therapeutic antitumor effects. Such strategy may be used in other antigenic systems for the control of infections and/or cancers.
Collapse
Affiliation(s)
- Tae Heung Kang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | | | | | | | | | | |
Collapse
|
37
|
Targeted coating with antigenic peptide renders tumor cells susceptible to CD8(+) T cell-mediated killing. Mol Ther 2012. [PMID: 23183537 DOI: 10.1038/mt.2012.233] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The potency of immunotherapies targeting endogenous tumor antigens is hindered by immune tolerance. We created a therapeutic agent comprised of a tumor-homing module fused to a functional domain capable of selectively rendering tumor cells sensitive to foreign antigen-specific CD8(+) T cell-mediated immune attack, and thereby, circumventing concerns for immune tolerance. The tumor-homing module is comprised of a single-chain variable fragment (scFv) that specifically binds to mesothelin (Meso), which is commonly overexpressed in human cancers, including ovarian tumors. The functional domain is comprised of the Fc portion of IgG2a protein and foreign immunogenic CD8(+) T cell epitope flanked by furin cleavage sites (R), which can be recognized and cleaved by furin that is highly expressed in the tumor microenvironment. We show that our therapeutic protein specifically loaded antigenic epitope onto the surface of mesothelin-expressing tumor cells, rendering tumors susceptible to antigen-specific cytotoxic CD8(+) T lymphocytes (CTL)-mediated killing in vitro and in vivo. Our findings have important implications for bypassing immune tolerance to enhance cancer immunotherapy.
Collapse
|
38
|
Su JH, Wu A, Scotney E, Ma B, Monie A, Hung CF, Wu TC. Immunotherapy for cervical cancer: Research status and clinical potential. BioDrugs 2010; 24:109-29. [PMID: 20199126 DOI: 10.2165/11532810-000000000-00000] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The high-risk types of human papillomavirus (HPV) have been found to be associated with most cervical cancers and play an essential role in the pathogenesis of the disease. Despite recent advances in preventive HPV vaccine development, such preventive vaccines are unlikely to reduce the prevalence of HPV infections within the next few years, due to their cost and limited availability in developing countries. Furthermore, preventive HPV vaccines may not be capable of treating established HPV infections and HPV-associated lesions, which account for high morbidity and mortality worldwide. Thus, it is important to develop therapeutic HPV vaccines for the control of existing HPV infection and associated malignancies. Therapeutic vaccines are quite different from preventive vaccines in that they require the generation of cell-mediated immunity, particularly T cell-mediated immunity, instead of the generation of neutralizing antibodies. The HPV-encoded early proteins, the E6 and E7 oncoproteins, form ideal targets for therapeutic HPV vaccines, since they are consistently expressed in HPV-associated cervical cancer and its precursor lesions and thus play crucial roles in the generation and maintenance of HPV-associated disease. Our review covers the various therapeutic HPV vaccines for cervical cancer, including live vector-based, peptide or protein-based, nucleic acid-based, and cell-based vaccines targeting the HPV E6 and/or E7 antigens. Furthermore, we review the studies using therapeutic HPV vaccines in combination with other therapeutic modalities and review the latest clinical trials on therapeutic HPV vaccines.
Collapse
Affiliation(s)
- Jun-Han Su
- National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Human Papillomavirus (HPV) has been associated with several human cancers, including cervical cancer, vulvar cancer, vaginal and anal cancer, and a subset of head and neck cancers. Thus effective vaccination against HPV provides an opportunity to reduce the morbidity and mortality associated with HPV. The Food and Drug Administration of the United States has approved two preventive vaccines to limit the spread of HPV. However, these are unlikely to impact upon HPV prevalence and cervical cancer rates for many years. Furthermore, preventive vaccines do not exert therapeutic effects on pre-existing HPV infections and HPV-associated lesions. In order to further impact upon the burden of HPV infections worldwide, therapeutic vaccines are being developed. These vaccines aim to generate a cell-mediated immune response to infected cells. This review discusses current preventive and therapeutic HPV vaccines and their future directions.
Collapse
Affiliation(s)
- Ken Lin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | | | - Chien-Fu Hung
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - T-C Wu
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Molecular Microbiology and Immunology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|
40
|
Smith KA, Meisenburg BL, Tam VL, Pagarigan RR, Wong R, Joea DK, Lantzy L, Carrillo MA, Gross TM, Malyankar UM, Chiang CS, Da Silva DM, Kündig TM, Kast WM, Qiu Z, Bot A. Lymph node-targeted immunotherapy mediates potent immunity resulting in regression of isolated or metastatic human papillomavirus-transformed tumors. Clin Cancer Res 2009; 15:6167-76. [PMID: 19789304 DOI: 10.1158/1078-0432.ccr-09-0645] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE The goal of this study was to investigate the therapeutic potential of a novel immunotherapy strategy resulting in immunity to localized or metastatic human papillomavirus 16-transformed murine tumors. EXPERIMENTAL DESIGN Animals bearing E7-expressing tumors were coimmunized by lymph node injection with E7 49-57 antigen and TLR3-ligand (synthetic dsRNA). Immune responses were measured by flow cytometry and antitumor efficacy was evaluated by tumor size and survival. In situ cytotoxicity assays and identification of tumor-infiltrating lymphocytes and T regulatory cells were used to assess the mechanisms of treatment resistance in bulky disease. Chemotherapy with cyclophosphamide was explored to augment immunotherapy in late-stage disease. RESULTS In therapeutic and prophylactic settings, immunization resulted in a considerable expansion of E7 49-57 antigen-specific T lymphocytes in the range of 1/10 CD8(+) T cells. The resulting immunity was effective in suppressing disease progression and mortality in a pulmonary metastatic disease model. Therapeutic immunization resulted in control of isolated tumors up to a certain volume, and correlated with antitumor immune responses measured in blood. In situ analysis showed that within bulky tumors, T-cell function was affected by negative regulatory mechanisms linked to an increase in T regulatory cells and could be overcome by cyclophosphamide treatment in conjunction with immunization. CONCLUSIONS This study highlights a novel cancer immunotherapy platform with potential for translatability to the clinic and suggests its potential usefulness for controlling metastatic disease, solid tumors of limited size, or larger tumors when combined with cytotoxic agents that reduce the number of tumor-infiltrating T regulatory cells.
Collapse
Affiliation(s)
- Kent A Smith
- Department of Research and Development, MannKind Corporation, Valencia, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Chuang CM, Monie A, Wu A, Hung CF. Combination of apigenin treatment with therapeutic HPV DNA vaccination generates enhanced therapeutic antitumor effects. J Biomed Sci 2009; 16:49. [PMID: 19473507 PMCID: PMC2705346 DOI: 10.1186/1423-0127-16-49] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Accepted: 05/27/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND It is important to develop innovative therapies for advanced stage cancers in addition to the conventional therapies including chemotherapy, radiation and surgery. Antigen-specific immunotherapy has emerged as a novel alternate therapy for advanced stage cancers, which may be employed in conjunction with conventional therapies. METHODS In the current study, we tested the effect of treatment with the chemotherapeutic agent, apigenin in combination with DNA vaccines encoding the HPV-16 E7 antigen linked to heat shock protein 70 (HSP70) in the control of the E7-expressing tumor, TC-1. RESULTS We observed that treatment with apigenin rendered the TC-1 tumor cells more susceptible to lysis by E7-specific cytotoxic CD8+ T cells. Furthermore, treatment of TC-1 tumor cells with apigenin was found to enhance apoptotic tumor cell death in vitro in a dose-dependant manner. We showed that TC-1 tumor-bearing mice treated with apigenin combined with E7-HSP70 DNA generate highest frequency of primary and memory E7-specific CD8+ T cells, leading to potent therapeutic anti-tumor effects against E7-expressing tumors. CONCLUSION Thus, apigenin represents a promising chemotherapeutic agent, which may be used in combination with immunotherapy for the treatment of advanced stage cancers. The clinical implications of the current strategy are discussed.
Collapse
Affiliation(s)
- Chi-Mu Chuang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA.
| | | | | | | |
Collapse
|
42
|
Kim JH, Kang TH, Noh KH, Bae HC, Kim SH, Yoo YD, Seong SY, Kim TW. Enhancement of dendritic cell-based vaccine potency by anti-apoptotic siRNAs targeting key pro-apoptotic proteins in cytotoxic CD8(+) T cell-mediated cell death. Immunol Lett 2009; 122:58-67. [PMID: 19135479 DOI: 10.1016/j.imlet.2008.12.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2008] [Revised: 11/14/2008] [Accepted: 12/01/2008] [Indexed: 12/31/2022]
Abstract
Dendritic cells (DCs) have become an important measure for the treatment of malignancies. Current DC preparations, however, generate short-lived DCs because they are subject to cell death from various apoptotic pressures. Antigen-specific CD8(+) cytotoxic T lymphocytes (CTLs) is one of the main obstacles to limit the DC-mediated immune priming since CTLs can recognize the target antigen expressing DCs as target cells and kill the DCs. CTLs secret perforin and serine protease granzymes during CTL killing. Perforin and serine protease granzymes induce the release of a number of mitochondrial pro-apoptotic factors, which are controlled by members of the BCL-2 family, such as BAK, BAX and BIM. FasL linking to Fas on DCs triggers the activation of caspase-8, which eventually leads to mitochondria-mediated apoptosis via truncation of BID. In this study, we tried to enhance the DC priming capacity by prolonging DC survival using anti-apoptotic siRNA targeting these key pro-apoptotic molecules in CTL killing. Human papillomavirus (HPV)-16 E7 antigen presenting DCs that were transfected with these anti-apoptotic siRNAs showed increased resistance to T cell-mediated death, leading to enhanced E7-specific CD8(+) T cell activation in vitro and in vivo. Among them, siRNA targeting BIM (siBIM) generated strongest E7-specific E7-specific CD8(+) T cell immunity. More importantly, vaccination with E7 presenting DCs transfected with siBIM was capable of generating a marked therapeutic effect in vaccinated mice. Our data indicate that ex vivo manipulation of DCs with siBIM may represent a plausible strategy for enhancing dendritic cell-based vaccine potency.
Collapse
Affiliation(s)
- Jin Hee Kim
- Laboratory Of Infection and Immunology, Graduate School of Medicine, Korea University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Immune evasion is an important reason why the immune system cannot control tumor growth. To elucidate the mechanism for tumor immune evasion, we generated an immune-resistant human papillomavirus type 16 (HPV-16) E7-expressing tumor cell line by subjecting a susceptible tumor cell line to multiple rounds of in vivo immune selection with an E7-specific vaccine. Comparison of parental and immune-resistant tumors revealed that Akt is highly activated in the immune-resistant tumors. Retroviral transfer of a constitutively active form of Akt into the parental tumor significantly increased its resistance against E7-specific CD8(+) T-cell mediated apoptosis. The observed resistance against apoptosis was found to be associated with the upregulation of antiapoptotic molecules. We also observed that intratumoral injection of an Akt inhibitor enhanced the therapeutic efficacy of E7-specific vaccine or E7-specific CD8(+) T-cell adoptive transfer against the immune-resistant tumors. Thus, our data indicate that the activation of PI3K/Akt pathway represents a new mechanism of immune escape and has important implications for the development of a novel strategy in cancer immunotherapy against immune-resistant tumor cells.
Collapse
|
44
|
Tseng CW, Monie A, Wu CY, Huang B, Wang MC, Hung CF, Wu TC. Treatment with proteasome inhibitor bortezomib enhances antigen-specific CD8+ T-cell-mediated antitumor immunity induced by DNA vaccination. J Mol Med (Berl) 2008; 86:899-908. [PMID: 18542898 PMCID: PMC2535907 DOI: 10.1007/s00109-008-0370-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Revised: 05/05/2008] [Accepted: 05/13/2008] [Indexed: 01/22/2023]
Abstract
There is an urgent need to develop new innovative therapies for the control of cancer. Antigen-specific immunotherapy and the employment of proteasome inhibitors have emerged as two potentially plausible approaches for the control of cancer. In the current study, we explored the combination of the DNA vaccine encoding calreticulin (CRT) linked to human papillomavirus type 16 E7 antigen (CRT/E7) with the proteasome inhibitor, bortezomib, for their ability to generate E7-specific immune responses and antitumor effects in vaccinated mice. We found that the combination of treatment with bortezomib and CRT/E7(detox) DNA generated more potent E7-specific CD8+ T cell immune responses and better therapeutic effects against TC-1 tumors in tumor-bearing mice compared to monotherapy. Furthermore, we found that treatment with bortezomib led to increased apoptosis of TC-1 tumor cells and could render the TC-1 tumor cells more susceptible to lysis by E7-specific CD8+ T cells. Our data have significant implications for future clinical translation.
Collapse
Affiliation(s)
- Chih-Wen Tseng
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Dell K, Klein C, Gissmann L. Comparison of DNA- and mRNA-Transfected Mouse Dendritic Cells as Potential Vaccines against the Human Papillomavirus Type 16 associated Oncoprotein E7. Antivir Ther 2008. [DOI: 10.1177/135965350801300409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Dendritic cells (DCs) mediate the generation of strong cytotoxic T-lymphocyte (CTL) responses by functioning in antigen presentation and exerting adjuvant properties. We compared several activation markers and parameters of biological activity of DNA- and mRNA-transfected DCs in vitro and in vivo. Methods CpG-matured, bone marrow derived C57BL/6 mouse DCs were electroporated either with enhanced green fluorescence protein (EGFP) or human papillomavirus type 16 (HPV16) E7 expression plasmids or in vitro transcribed mRNAs encoding for the codon-optimized E7 or a shuffled version thereof. Activation marker expression and antigen presentation was analysed by fluorescence-activated cell sorting. The migratory behaviour of transfected DCs were investigated by in vitro chemotaxis experiments and cytokine expression by ELISA. CTL-priming capacity of transfected DCs were determined by vaccination of mice. Results mRNA transfection produced a two- to fourfold increase of the activation markers CD40, CD80, CD86 and MHC I and MHC II molecules. Predominately antigen-expressing DCs migrated after mRNA transfection. Furthermore, mRNA-transfected DCs were capable of inducing a chemokine gradient. After maturation, electroporation and activation with soluble CD40 ligand and interferon-γ, DCs displayed a T-helper cell type 2 cytokine expression pattern. Nevertheless, E7-transfected DCs were able to prime E7-specific CTL responses in vivo. The highest E7-specific CTL frequencies were found in mice immunized with mRNA-transfected DCs. The in vitro expanded CTLs exerted functional E7-specific cytotoxic activity. Conclusions Genetically modified DCs are suitable vehicles for the induction of E7-specific CTL responses in mice and hence could help to eradicate HPV-associated lesions in humans.
Collapse
Affiliation(s)
- Kerstin Dell
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Corinna Klein
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lutz Gissmann
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
46
|
Hung CF, Ma B, Monie A, Tsen SW, Wu TC. Therapeutic human papillomavirus vaccines: current clinical trials and future directions. Expert Opin Biol Ther 2008; 8:421-39. [PMID: 18352847 DOI: 10.1517/14712598.8.4.421] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Cervical cancer is the second largest cause of cancer deaths in women worldwide. It is now evident that persistent infection with high-risk human papillomavirus (HPV) is necessary for the development and maintenance of cervical cancer. Thus, effective vaccination against HPV represents an opportunity to restrain cervical cancer and other important cancers. The FDA recently approved the HPV vaccine Gardasil for the preventive control of HPV, using HPV virus-like particles (VLP) to generate neutralizing antibodies against major capsid protein, L1. However, prophylactic HPV vaccines do not have therapeutic effects against pre-existing HPV infections and HPV-associated lesions. Furthermore, due to the considerable burden of HPV infections worldwide, it would take decades for preventive vaccines to affect the prevalence of cervical cancer. Thus, in order to speed up the control of cervical cancer and treat current infections, the continued development of therapeutic vaccines against HPV is critical. Therapeutic HPV vaccines can potentially eliminate pre-existing lesions and malignant tumors by generating cellular immunity against HPV-infected cells that express early viral proteins such as E6 and E7. OBJECTIVE This review discusses the future directions of therapeutic HPV vaccine approaches for the treatment of established HPV-associated malignancies, with emphasis on current progress of HPV vaccine clinical trials. METHODS Relevant literature is discussed. RESULTS/CONCLUSION Though their development has been challenging, many therapeutic HPV vaccines have been shown to induce HPV-specific antitumor immune responses in preclinical animal models and several promising strategies have been applied in clinical trials. With continued progress in the field of vaccine development, HPV therapeutic vaccines may provide a potentially promising approach for the control of lethal HPV-associated malignancies.
Collapse
Affiliation(s)
- Chien-Fu Hung
- The Johns Hopkins University School of Medicine, Department of Pathology, CRBII 309, 1550 Orleans Street, Baltimore, Maryland 21231, USA
| | | | | | | | | |
Collapse
|
47
|
Herd KA, Wiethe C, Tindle RW. Co-immunisation with DNA encoding RANK/RANKL or 4-1BBL costimulatory molecules does not enhance effector or memory CTL responses afforded by immunisation with a tumour antigen-encoding DNA vaccine. Vaccine 2007; 25:5209-19. [PMID: 17544551 DOI: 10.1016/j.vaccine.2007.04.083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2006] [Revised: 04/15/2007] [Accepted: 04/25/2007] [Indexed: 10/23/2022]
Abstract
T cell mediated immune responses are induced following interaction of MHC-presented epitope on professional antigen presenting cells such as dendritic cells (DCs) with cognate T cell receptor. Up-regulation of receptor-ligand pairs of costimulatory molecules linking DC to T cell enhances the resulting T cell responses. This 'second signalling' occurs through the B7 molecules CD80/86 expressed by DCs, and importantly through members of the TNF ligand/TNF receptor superfamilies. We have previously shown that co-expression of RANK/RANKL or 41BB-L in addition to tumour antigen in dendritic cells augmented cognate effector and memory tumour antigen-directed cytotoxic T cell responses when the DCs were used to immunise mice. Here, we examined whether co-immunisation with naked plasmid DNAs encoding antigen and these costimulatory molecule(s), would enhance antigen specific T cell responses. We demonstrate that co-immunisation with DNAs encoding tumour antigen and costimulatory molecules failed to enhance antigen-directed CTL responses, or tumour protection, afforded by immunisation with DNA encoding tumour antigen alone.
Collapse
MESH Headings
- 4-1BB Ligand/genetics
- 4-1BB Ligand/metabolism
- Animals
- Antigens, Neoplasm/genetics
- Cell Line
- Cell Line, Tumor
- Cells, Cultured
- DNA/administration & dosage
- DNA/immunology
- Female
- Flow Cytometry
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Humans
- Immunization
- Male
- Mice
- Mice, Inbred C57BL
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/prevention & control
- Plasmids/administration & dosage
- Plasmids/genetics
- Plasmids/immunology
- Proteins/genetics
- Proteins/metabolism
- RANK Ligand/genetics
- RANK Ligand/metabolism
- Receptor Activator of Nuclear Factor-kappa B/genetics
- Receptor Activator of Nuclear Factor-kappa B/metabolism
- Survival Analysis
- T-Lymphocytes/cytology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Karen A Herd
- Sir Albert Sakzewski Virus Research Centre, Royal Children's Hospital, Brisbane, Australia
| | | | | |
Collapse
|
48
|
Kang TH, Lee JH, Noh KH, Han HD, Shin BC, Choi EY, Peng S, Hung CF, Wu TC, Kim TW. Enhancing dendritic cell vaccine potency by combining a BAK/BAX siRNA-mediated antiapoptotic strategy to prolong dendritic cell life with an intracellular strategy to target antigen to lysosomal compartments. Int J Cancer 2007; 120:1696-703. [PMID: 17230516 DOI: 10.1002/ijc.22377] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Dendritic cell (DC)-based vaccines have become important in immunotherapeutics as a measure for generating antitumor immune responses. We have previously demonstrated that linkage of the antigen gene to a lysosomal targeting signal, a sorting signal of the lysosome-associated membrane protein type 1 (LAMP-1), enhances the potency of DC-based vaccines. DCs have a limited life span, hindering their long-term ability to prime antigen-specific T cells. In this study, we attempted to further improve the potency of a DC vaccine that targets human papilloma virus 16 (HPV16) E7 to a lysosomal compartment (DC-Sig/E7/LAMP-1) by combining a strategy to prolong DC life. We show that small interfering RNA-targeting Bak and Bax proteins can be used to allow transfected DCs to resist being killed by T cells. This is done by downregulating these proapoptotic proteins, which have been known as so-called gate keepers in mitochondria-mediated apoptosis. DCs expressing intact E7 or Sig/E7/LAMP-1 became resistant to attack by CD8+ T cells after transfection with BAK/BAX siRNA, leading to enhanced E7-specific T cell activation in vitro and in vivo. More importantly, vaccination with E7-presenting DCs transfected with BAK/BAX siRNA generated a strong therapeutic effect against an E7-expressing tumor in vaccinated mice, compared with DCs transfected with control siRNA. Our data indicate that a combination of strategies to enhance intracellular Ag processing and to prolong DC life may offer a promising strategy for improving DC vaccine potency.
Collapse
Affiliation(s)
- Tae Heung Kang
- Laboratory of Infection and Immunology, Graduate School of Medicine, Korea University, Ansan, Gyeonggi, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Brulet JM, Maudoux F, Thomas S, Thielemans K, Burny A, Leo O, Bex F, Hallez S. DNA vaccine encoding endosome-targeted human papillomavirus type 16 E7 protein generates CD4+ T cell-dependent protection. Eur J Immunol 2007; 37:376-84. [PMID: 17274002 DOI: 10.1002/eji.200636233] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Human papillomavirus type 16 is commonly implicated in cervical cancers. The viral genome encodes potential targets like the oncoprotein E7, expressed in transformed cells but thought to represent a poorly immunogenic antigen. We describe in this work a DNA-based vaccination protocol aimed at inducing an efficient anti-E7 immune response in vivo. Plasmids allowing the expression of the E7 protein in distinct cellular compartments were generated and assayed in an in vivo model of tumor growth. Our data demonstrate that mice vaccinated with a plasmid encoding for an E7 protein fused to a domain of the MHC class II-associated invariant chain (IiE7) were protected against tumor challenge. Mice immunized against an ubiquitinated form of E7 (Ub(Ala)E7) failed to control tumor growth. Protection induced by IiE7 was correlated with the development of CD8+ CTL and required the presence of CD4+ cells. In vitro studies confirmed that the IiE7 fusion protein was expressed at high levels in the endosomal compartment of transfected cells, while the natural and the ubiquitin-modified form of E7 were mainly nuclear. The present study suggests that an efficient anti-tumor response can be induced in vivo by DNA constructs encoding for E7 protein forms localizing at the endosomal compartment.
Collapse
Affiliation(s)
- Jean-Marc Brulet
- Laboratory of Chemical Biology, Institut de Biologie et de Médecine Moléculaires, Université Libre de Bruxelles, Gosselies, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Hung CF, Tsai YC, He L, Wu TC. Control of mesothelin-expressing ovarian cancer using adoptive transfer of mesothelin peptide-specific CD8+ T cells. Gene Ther 2007; 14:921-9. [PMID: 17377599 PMCID: PMC3183576 DOI: 10.1038/sj.gt.3302913] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cancer immunotherapy targeting mesothelin represents a potentially plausible approach for the control of ovarian cancer as most ovarian cancers express high levels of mesothelin. In the current study, we created a mesothelin-positive luciferase-expressing ovarian cancer model, MOSEC/luc. This luciferase-expressing tumor model allowed us to quantitate tumor distribution and tumor load in tumor-challenged mice using a non-invasive bioluminescence imaging system. In addition, we identified an H-2D(b)-restricted mesothelin peptide-specific cytotoxic T-lymphocyte (CTL) epitope (amino acid (aa) 406-414) that was endogenously processed and presented by MOSEC/luc tumor cells. We showed that adoptive transfer of mesothelin peptide (aa406-414)-specific CD8(+) T cells led to the control of MOSEC/luc tumor cells. The MOSEC/luc tumor model and the newly identified H-2D(b)-restricted murine mesothelin-specific CTL epitope (aa406-414) will be very useful for the development of immunotherapy for ovarian cancer as well as for the development of quantitative CD8(+) T cell-mediated immunological assays.
Collapse
Affiliation(s)
- C-F Hung
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA.
| | | | | | | |
Collapse
|