51
|
Zottnick S, Voß AL, Riemer AB. Inducing Immunity Where It Matters: Orthotopic HPV Tumor Models and Therapeutic Vaccinations. Front Immunol 2020; 11:1750. [PMID: 32922389 PMCID: PMC7457000 DOI: 10.3389/fimmu.2020.01750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/30/2020] [Indexed: 12/24/2022] Open
Abstract
Anogenital and oropharyngeal cancers caused by human papillomavirus (HPV) infections account for 4.5% of all cancer cases worldwide. So far, only the initial infection with selected high-risk types can be prevented by prophylactic vaccination. Already existing persistent HPV infections, however, can currently only be treated by surgical removal of resulting lesions. Therapeutic HPV vaccination, promoting cell-based anti-HPV immunity, would be ideal to eliminate and protect against HPV-induced lesions and tumors. A multitude of vaccination approaches has been tested to date, many of which led to high amounts of HPV-specific T cells in vivo. However, growing evidence suggests that not the induction of systemic but of local immunity is paramount for tackling mucosal infections and tumors. Therefore, recent therapeutic vaccination studies have focused on how to induce tissue-resident T cells in the anogenital and oropharyngeal mucosa. These approaches include direct mucosal vaccinations and influencing the migration of systemic T cells toward the mucosa. The efficacy of these new vaccination approaches is best tested in vivo by utilizing orthotopic tumor models, i.e. HPV-positive tumors being located in the animal's mucosa. In line with this, we here review existing HPV tumor models and describe two novel tumorigenic cell lines for the MHC-humanized mouse model A2.DR1. These were used for the establishment of an HPV16 E6/E7-positive vaginal tumor model, suitable for testing therapeutic vaccines containing HLA-A2-restricted HPV16-derived epitopes. The newly developed MHC-humanized orthotopic HPV16-positive tumor model is likely to improve the translatability of in vivo findings to the clinical setting.
Collapse
Affiliation(s)
- Samantha Zottnick
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Alessa L Voß
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Angelika B Riemer
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| |
Collapse
|
52
|
Zhao X, Yang F, Mariz F, Osen W, Bolchi A, Ottonello S, Müller M. Combined prophylactic and therapeutic immune responses against human papillomaviruses induced by a thioredoxin-based L2-E7 nanoparticle vaccine. PLoS Pathog 2020; 16:e1008827. [PMID: 32886721 PMCID: PMC7498061 DOI: 10.1371/journal.ppat.1008827] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/17/2020] [Accepted: 07/21/2020] [Indexed: 01/08/2023] Open
Abstract
Global burden of cervical cancer, the most common cause of mortality caused by human papillomavirus (HPV), is expected to increase during the next decade, mainly because current alternatives for HPV vaccination and cervical cancer screening programs are costly to be established in low-and-middle income countries. Recently, we described the development of the broadly protective, thermostable vaccine antigen Trx-8mer-OVX313 based on the insertion of eight different minor capsid protein L2 neutralization epitopes into a thioredoxin scaffold from the hyperthermophilic archaeon Pyrococcus furiosus and conversion of the resulting antigen into a nanoparticle format (median radius ~9 nm) upon fusion with the heptamerizing OVX313 module. Here we evaluated whether the engineered thioredoxin scaffold, in addition to humoral immune responses, can induce CD8+ T-cell responses upon incorporation of MHC-I-restricted epitopes. By systematically examining the contribution of individual antigen modules, we demonstrated that B-cell and T-cell epitopes can be combined into a single antigen construct without compromising either immunogenicity. While CD8+ T-cell epitopes had no influence on B-cell responses, the L2 polytope (8mer) and OVX313-mediated heptamerization of the final antigen significantly increased CD8+ T-cell responses. In a proof-of-concept experiment, we found that vaccinated mice remained tumor-free even after two consecutive tumor challenges, while unvaccinated mice developed tumors. A cost-effective, broadly protective vaccine with both prophylactic and therapeutic properties represents a promising option to overcome the challenges associated with prevention and treatment of HPV-caused diseases. Currently, there are three licensed prophylactic vaccines available against HPV, but none of them shows a therapeutic effect on pre-existing infections. Thus, a prophylactic vaccine also endowed with a therapeutic activity presents application potentials to individuals regardless of their HPV-infection status. Such a dual-purpose vaccine would be particularly valuable for post-exposure prophylaxis and shields population from recurrent HPV infections. Here, we constructed a combined vaccine relying on L2- and E7-specific epitopes grafted onto the surface of a hyper-stable thioredoxin scaffold. The resulting antigen was converted into a nanoparticle format with the use of a heptamerization domain. Our data document that the modular design of the antigen allows combination of B-cell and T-cell epitopes in one antigen without compromising either’s immunogenicity. The antigen retains its ability to provide broad protection against different HPV types but also presents strong therapeutic effects in a mouse tumor model. Therefore, the vaccine is potentially capable of resolving productive infection as well as HPV-related malignancies, and thus benefitting both uninfected and already infected individuals. Moreover, our vaccine utilizes E. coli as protein producer and distribution does not require cold-chain, which reduces costs making it applicable to less-affluent countries.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/pharmacology
- Antigens, Viral/chemistry
- Antigens, Viral/pharmacology
- Archaeal Proteins/chemistry
- Archaeal Proteins/pharmacology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cancer Vaccines/chemistry
- Cancer Vaccines/pharmacology
- Epitopes, B-Lymphocyte/chemistry
- Epitopes, B-Lymphocyte/pharmacology
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/pharmacology
- Female
- Humans
- Immunity, Cellular/drug effects
- Mice
- Mice, Inbred BALB C
- Nanoparticles/chemistry
- Nanoparticles/therapeutic use
- Papillomaviridae/chemistry
- Papillomaviridae/immunology
- Papillomavirus Vaccines/chemistry
- Papillomavirus Vaccines/pharmacology
- Pyrococcus furiosus/chemistry
- Thioredoxins/chemistry
- Thioredoxins/pharmacology
- Uterine Cervical Neoplasms/immunology
- Uterine Cervical Neoplasms/virology
Collapse
Affiliation(s)
- Xueer Zhao
- German Cancer Research Center, Heidelberg, Germany
| | - Fan Yang
- German Cancer Research Center, Heidelberg, Germany
| | - Filipe Mariz
- German Cancer Research Center, Heidelberg, Germany
| | - Wolfram Osen
- German Cancer Research Center, Heidelberg, Germany
| | - Angelo Bolchi
- Department of Chemical Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Simone Ottonello
- Department of Chemical Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Martin Müller
- German Cancer Research Center, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
53
|
Vila-Leahey A, MacKay A, Portales-Cervantes L, Weir GM, Merkx-Jacques A, Stanford MM. Generation of highly activated, antigen-specific tumor-infiltrating CD8 + T cells induced by a novel T cell-targeted immunotherapy. Oncoimmunology 2020; 9:1782574. [PMID: 32923145 PMCID: PMC7458631 DOI: 10.1080/2162402x.2020.1782574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The induction of tumor-targeted, cytotoxic T lymphocytes has been recognized as a key component to successful immunotherapy. DPX-based treatment was previously shown to effectively recruit activated CD8+ T cells to the tumor. Herein, we analyze the unique phenotype of the CD8+ T cells recruited into the tumor in response to DPX-based therapy, and how combination with checkpoint inhibitors impacts T cell response. C3-tumor-bearing mice were treated with cyclophosphamide (CPA) for seven continuous days every other week, followed by DPX treatment along with anti-CTLA-4 and/or anti-PD-1. Efficacy, immunogenicity, and CD8+ T cells tumor infiltration were assessed. The expression of various markers, including checkpoint markers, peptide specificity, and proliferation and activation markers, was determined by flow cytometry. tSNE analysis of the flow data revealed a resident phenotype of CD8+ T cells (PD-1+TIM-3+CTLA-4+) within untreated tumors, whereas DPX/CPA treatment induced recruitment of a novel population of CD8+ T cells (PD-1+TIM-3+CTLA-4−) within tumors. Combination of anti-CTLA-4 (ipilimumab) with DPX/CPA versus DPX/CPA alone significantly increased survival and inhibition of tumor growth, without changing overall systemic immunogenicity. Addition of checkpoint inhibitors did not significantly change the phenotype of the newly recruited cells induced by DPX/CPA. Yet, anti-CTLA-4 treatment in combination with DPX/CPA enhanced a non-antigen specific response within the tumor. Finally, the tumor-recruited CD8+ T cells induced by DPX/CPA were highly activated, antigen-specific, and proliferative, while resident phenotype CD8+ T cells, seemingly initially exhausted, were reactivated with combination treatment. This study supports the potential of combining DPX/CPA with ipilimumab to further enhance survival clinically.
Collapse
Affiliation(s)
| | - Alecia MacKay
- Research and Development, IMV Inc, Dartmouth, NS, Canada
| | | | | | | | - Marianne M Stanford
- Research and Development, IMV Inc, Dartmouth, NS, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
54
|
HPV16-E7 Protein T Cell Epitope Prediction and Global Therapeutic Peptide Vaccine Design Based on Human Leukocyte Antigen Frequency: An In-Silico Study. Int J Pept Res Ther 2020; 27:365-378. [PMID: 32837456 PMCID: PMC7320846 DOI: 10.1007/s10989-020-10089-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2020] [Indexed: 02/08/2023]
Abstract
Cervical cancer is the second most common leading cause of women's death due to cancer worldwide, about 528,000 patients’ cases and 266,000 deaths per year, related to human papillomavirus (HPV). Peptide-based vaccines being safe, stable, and easy to produce have demonstrated great potential to develop therapeutic HPV vaccine. In this study, the major histocompatibility complex (MHC) class I, class II T cell epitopes of HPV16-E7 were predicted. Therefore, we designed a plan to find the most effective peptides to prompt appropriate immune responses. For this purpose, retrieving protein sequences, conserved region identification, phylogenic tree construction, T cell epitope prediction, epitope-predicted population coverage calculation, and molecular docking were performed consecutively and most effective immune response prompting peptides were selected. Based on different tools index, six CD8+ T cells and six CD4+ epitopes were chosen. This combination of 12 epitopes created a putative global vaccine with a 95.06% population coverage. These identified peptides can be employed further for peptide analysis and can be used as a peptide or poly-epitope candidates for therapeutic vaccine studies to treat HPV-associated cancers.
Collapse
|
55
|
Mehta NK, Pradhan RV, Soleimany AP, Moynihan KD, Rothschilds AM, Momin N, Rakhra K, Mata-Fink J, Bhatia SN, Wittrup KD, Irvine DJ. Pharmacokinetic tuning of protein-antigen fusions enhances the immunogenicity of T-cell vaccines. Nat Biomed Eng 2020; 4:636-648. [PMID: 32483299 DOI: 10.1038/s41551-020-0563-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 04/27/2020] [Indexed: 12/21/2022]
Abstract
The formulations of peptide-based antitumour vaccines being tested in clinical studies are generally associated with weak potency. Here, we show that pharmacokinetically tuning the responses of peptide vaccines by fusing the peptide epitopes to carrier proteins optimizes vaccine immunogenicity in mice. In particular, we show in immunized mice that the carrier protein transthyretin simultaneously optimizes three factors: efficient antigen uptake in draining lymphatics from the site of injection, protection of antigen payloads from proteolytic degradation and reduction of antigen presentation in uninflamed distal lymphoid organs. Optimizing these factors increases vaccine immunogenicity by up to 90-fold and maximizes the responses to viral antigens, tumour-associated antigens, oncofetal antigens and shared neoantigens. Protein-peptide epitope fusions represent a facile and generalizable strategy for enhancing the T-cell responses elicited by subunit vaccines.
Collapse
Affiliation(s)
- Naveen K Mehta
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roma V Pradhan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ava P Soleimany
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Harvard Graduate Program in Biophysics, Harvard University, Boston, MA, USA
| | - Kelly D Moynihan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Adrienne M Rothschilds
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Noor Momin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kavya Rakhra
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jordi Mata-Fink
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sangeeta N Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Harvard-MIT Health Sciences and Technology Program, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.,Howard Hughes Medical Institute, Cambridge, MA, USA
| | - K Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. .,The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA. .,Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Howard Hughes Medical Institute, Cambridge, MA, USA.
| |
Collapse
|
56
|
Panahi HA, Bolhassani A, Javadi G, Noormohammadi Z, Agi E. Development of multiepitope therapeutic vaccines against the most prevalent high-risk human papillomaviruses. Immunotherapy 2020; 12:459-479. [DOI: 10.2217/imt-2019-0196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: Our goal was the development of DNA- or peptide-based multiepitope vaccines targeting HPV E7, E6 and E5 oncoproteins in tumor mouse model. Materials & methods: After designing the multiepitope E7, E6 and E5 constructs from four types of high risk HPVs (16, 18, 31 & 45) using bioinformatics tools, mice vaccination was performed by different homologous and heterologous modalities in a prophylactic setting. Then, anti-tumor effects of the best prophylactic strategies were studied in a therapeutic setting. Results: In both prophylactic and therapeutic experiments, groups receiving homologous E7+E6+E5 polypeptide, and heterologous E7+E6+E5 DNA prime/polypeptide boost were successful in complete rejection of tumors. Conclusion: The designed multiepitope constructs can be considered as promising candidates to develop effective therapeutic HPV vaccines.
Collapse
Affiliation(s)
- Heidar Ali Panahi
- Department of Hepatitis & AIDS, Pasteur Institute of Iran, Tehran, Iran
- Department of Biology, School of Basic Sciences, Science & Research Branch, Islamic Azad University, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis & AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Gholamreza Javadi
- Department of Biology, School of Basic Sciences, Science & Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Noormohammadi
- Department of Biology, School of Basic Sciences, Science & Research Branch, Islamic Azad University, Tehran, Iran
| | - Elnaz Agi
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| |
Collapse
|
57
|
O'Brien-Moran Z, Bowen CV, Rioux JA, Brewer KD. Cell density quantification with TurboSPI: R 2* mapping with compensation for off-resonance fat modulation. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2019; 33:469-481. [PMID: 31872356 DOI: 10.1007/s10334-019-00817-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/10/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Tracking the migration of superparamagnetic iron oxide (SPIO)-labeled immune cells in vivo is valuable for understanding the immunogenic response to cancer and therapies. Quantitative cell tracking using TurboSPI-based R2* mapping is a promising development to improve accuracy in longitudinal studies on immune recruitment. However, off-resonance fat signal isochromats lead to modulations in the signal time-course that can be erroneously fit as R2* signal decay, overestimating the density of labeled cells, while excluding voxels with fat-typical modulations results in underestimation of cell density in voxels with mixed content. Approaches capable of accurate R2* estimation in the presence of fat are needed. METHODS We propose a dual-decay (separate R2f* and R2w* for fat and water) Dixon-based signal model that accounts for the presence of fat in a voxel to provide better estimates of SPIO-induced dephasing. This model was tested in silico, in phantoms with varying quantities of fat and SPIO-labeled cells, and in 5 mice injected with SPIO-labeled CD8+ T cells. RESULTS In silico single voxel simulations illustrate how the proposed dual-decay model provides stable R2w* estimates that are invariant to fat content. The proposed model outperforms previous methods when applied to in vitro samples of SPIO-labeled cells and oil prepared with oil content ≥ 15%. Preliminary in vivo results show that, compared to previous methods, the dual-decay model improves the balance of R2* mapping in fat-dense areas, which will yield more reliable analysis in future cell tracking studies. DISCUSSION The proposed model is a promising tool for quantitative TurboSPI R2* cell tracking, with further refinements offering the possibility of better specificity and sensitivity.
Collapse
Affiliation(s)
- Zoe O'Brien-Moran
- Biomedical Translational Imaging Centre, IWK Health Centre and Nova Scotia Health Authority, Halifax, NS, Canada.,Dalhousie University, Halifax, NS, Canada
| | - Chris Van Bowen
- Biomedical Translational Imaging Centre, IWK Health Centre and Nova Scotia Health Authority, Halifax, NS, Canada.,Dalhousie University, Halifax, NS, Canada
| | - James Allen Rioux
- Biomedical Translational Imaging Centre, IWK Health Centre and Nova Scotia Health Authority, Halifax, NS, Canada.,Dalhousie University, Halifax, NS, Canada
| | - Kimberly Dawn Brewer
- Biomedical Translational Imaging Centre, IWK Health Centre and Nova Scotia Health Authority, Halifax, NS, Canada. .,Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
58
|
Barsoumian HB, Batra L, Shrestha P, Bowen WS, Zhao H, Egilmez NK, Gomez-Gutierrez JG, Yolcu ES, Shirwan H. A Novel Form of 4-1BBL Prevents Cancer Development via Nonspecific Activation of CD4 + T and Natural Killer Cells. Cancer Res 2019; 79:783-794. [PMID: 30770367 DOI: 10.1158/0008-5472.can-18-2401] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/13/2018] [Accepted: 12/28/2018] [Indexed: 01/11/2023]
Abstract
Costimulation through 4-1BB (CD137) receptor generates robust CD8+ T-effector and memory responses. The only known ligand, 4-1BBL, is a trimeric transmembrane protein that has no costimulatory activity as a soluble molecule. Thus, agonistic antibodies to the receptor have been used for cancer immunotherapy in preclinical models and are currently being evaluated in the clinic. Here, we report that treatment with an oligomeric form of the ligand, SA-4-1BBL, as a single agent is able to protect mice against subsequent tumor challenge irrespective of the tumor type. Protection was long-lasting (>8 weeks) and a bona fide property of SA-4-1BBL, as treatment with an agonistic antibody to the 4-1BB receptor was ineffective in generating immune protection against tumor challenge. Mechanistically, SA-4-1BBL significantly expanded IFNγ-expressing, preexisting memory-like CD44+CD4+ T cells and NK cells in naïve mice as compared with the agonistic antibody. In vivo blockade of IFNγ or depletion of CD4+ T or NK cells, but not CD8+ T or B cells, abrogated the immunopreventive effects of SA-4-1BBL against cancer. SA-4-1BBL as a single agent also exhibited robust efficacy in controlling postsurgical recurrences. This work highlights unexpected features of SA-4-1BBL as a novel immunomodulator with implications for cancer immunoprevention and therapy. SIGNIFICANCE: This study demonstrates the unique and unexpected immunomodulatory features of SA-4-1BBL that bridge innate and adaptive immune responses with both preventive and therapeutic efficacy against cancer.
Collapse
Affiliation(s)
- Hampartsoum B Barsoumian
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Lalit Batra
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Pradeep Shrestha
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | | | - Hong Zhao
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Nejat K Egilmez
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | | | - Esma S Yolcu
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky. .,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky.,FasCure Therapeutics, LLC, Louisville, Kentucky
| | - Haval Shirwan
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky. .,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
59
|
Hancock G, Blight J, Lopez-Camacho C, Kopycinski J, Pocock M, Byrne W, Price MJ, Kemlo P, Evans RI, Bloss A, Saunders K, Kirton R, Andersson M, Hellner K, Reyes-Sandoval A, Dorrell L. A multi-genotype therapeutic human papillomavirus vaccine elicits potent T cell responses to conserved regions of early proteins. Sci Rep 2019; 9:18713. [PMID: 31822717 PMCID: PMC6904585 DOI: 10.1038/s41598-019-55014-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022] Open
Abstract
Despite an efficacious prophylactic human papillomavirus (HPV) vaccine there is still a considerable global burden of HPV-related disease. Therapeutic vaccines that could prevent cancers in at-risk women are urgently needed. Most candidate therapeutic vaccines have focused on two high-risk (hr) HPV genotypes, 16 and 18, and two viral targets, E6 and E7, which may limit global coverage and efficacy. We designed the synthetic gene '5GHPV3' by selecting conserved regions from each of the six early proteins and generating consensus sequences to represent five hrHPV genotypes. 5GHPV3 was delivered by plasmid DNA, chimpanzee adenovirus (ChAdOx1) and modified vaccinia Ankara (MVA) vectors in prime-boost regimens to mice. ChAdOx1-5GHPV3 / MVA-5GHPV3 induced higher magnitude and more durable HPV-specific T cell responses than other regimens. Vaccine-induced T cells were polyfunctional and persisted at high frequencies for at least six weeks. Importantly, HPV-specific effector CD8 + T cells were detected in the cervix following systemic administration of ChAdOx1-5GHPV3 / MVA-5GHPV3 and increased in frequency over time, indicating continued trafficking of T cells to the cervix. Finally, T cells specific for 5GHPV3 encoded antigens were detected by IFN-γ Elispot in women with current or past hrHPV infections, confirming the presence of epitopes relevant to natural immune control.
Collapse
Affiliation(s)
- Gemma Hancock
- Nuffield Department of Medicine, University of Oxford and Oxford NIHR Biomedical Research Centre, NDM Research Building, Old Road Campus, Oxford, UK.
| | - Joshua Blight
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, Oxford, UK
| | - Cesar Lopez-Camacho
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, Oxford, UK
| | - Jakub Kopycinski
- Nuffield Department of Medicine, University of Oxford and Oxford NIHR Biomedical Research Centre, NDM Research Building, Old Road Campus, Oxford, UK
| | - Mamatha Pocock
- Nuffield Department of Medicine, University of Oxford and Oxford NIHR Biomedical Research Centre, NDM Research Building, Old Road Campus, Oxford, UK
| | - Wendy Byrne
- Direct Delivery Team, NIHR Clinical Research Network Thames Valley and South Midlands, Nuffield Orthopaedic Centre, Block 8, Oxford, OX3 7LD, UK
| | - Michael J Price
- Nuffield Department of Medicine, University of Oxford and Oxford NIHR Biomedical Research Centre, NDM Research Building, Old Road Campus, Oxford, UK
| | - Phillip Kemlo
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, Oxford, UK
| | - Ranoromanana Ionitiana Evans
- Direct Delivery Team, NIHR Clinical Research Network Thames Valley and South Midlands, Nuffield Orthopaedic Centre, Block 8, Oxford, OX3 7LD, UK
| | - Angela Bloss
- Direct Delivery Team, NIHR Clinical Research Network Thames Valley and South Midlands, Nuffield Orthopaedic Centre, Block 8, Oxford, OX3 7LD, UK
| | - Kathryn Saunders
- Direct Delivery Team, NIHR Clinical Research Network Thames Valley and South Midlands, Nuffield Orthopaedic Centre, Block 8, Oxford, OX3 7LD, UK
| | - Richard Kirton
- Microbiology Department, Oxford University Hospital NHS Foundation Trust, Oxford, UK
| | - Monique Andersson
- Microbiology Department, Oxford University Hospital NHS Foundation Trust, Oxford, UK
| | - Karin Hellner
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, UK
| | - Arturo Reyes-Sandoval
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, Oxford, UK
| | - Lucy Dorrell
- Nuffield Department of Medicine, University of Oxford and Oxford NIHR Biomedical Research Centre, NDM Research Building, Old Road Campus, Oxford, UK
| |
Collapse
|
60
|
Westrich JA, Vermeer DW, Silva A, Bonney S, Berger JN, Cicchini L, Greer RO, Song JI, Raben D, Slansky JE, Lee JH, Spanos WC, Pyeon D. CXCL14 suppresses human papillomavirus-associated head and neck cancer through antigen-specific CD8 + T-cell responses by upregulating MHC-I expression. Oncogene 2019; 38:7166-7180. [PMID: 31417179 PMCID: PMC6856418 DOI: 10.1038/s41388-019-0911-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/23/2019] [Accepted: 05/26/2019] [Indexed: 12/30/2022]
Abstract
Evasion of the host immune responses is critical for both persistent human papillomavirus (HPV) infection and associated cancer progression. We have previously shown that expression of the homeostatic chemokine CXCL14 is significantly downregulated by the HPV oncoprotein E7 during cancer progression. Restoration of CXCL14 expression in HPV-positive head and neck cancer (HNC) cells dramatically suppresses tumor growth and increases survival through an immune-dependent mechanism in mice. While CXCL14 recruits natural killer (NK) and T cells to the tumor microenvironment, the mechanism by which CXCL14 mediates tumor suppression through NK and/or T cells remained undefined. Here, we report that CD8+ T cells are required for CXCL14-mediated tumor suppression. Using a CD8+ T cell receptor transgenic model, we show that the CXCL14-mediated antitumor CD8+ T cell responses require antigen specificity. Interestingly, CXCL14 expression restores major histocompatibility complex class I (MHC-I) expression on HPV-positive HNC cells downregulated by HPV, and knockdown of MHC-I expression in HNC cells results in loss of tumor suppression even with CXCL14 expression. These results suggest that CXCL14 enacts antitumor immunity through restoration of MHC-I expression on tumor cells and promoting antigen-specific CD8+ T cell responses to suppress HPV-positive HNC.
Collapse
Affiliation(s)
- Joseph A Westrich
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.,Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Daniel W Vermeer
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD, 57104, USA
| | - Alexa Silva
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Stephanie Bonney
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Jennifer N Berger
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Louis Cicchini
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Robert O Greer
- Departments of Pathology and Dermatology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.,Division of Oral and Maxillofacial Pathology, University of Colorado School of Dental Medicine, Aurora, CO, 80045, USA
| | - John I Song
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - David Raben
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Jill E Slansky
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - John H Lee
- Chan Soon-Shiong Institute for Medicine, El Segundo, CA, 90245, USA
| | - William C Spanos
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD, 57104, USA
| | - Dohun Pyeon
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA. .,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
61
|
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
|
62
|
Marijt KA, Sluijter M, Blijleven L, Tolmeijer SH, Scheeren FA, van der Burg SH, van Hall T. Metabolic stress in cancer cells induces immune escape through a PI3K-dependent blockade of IFNγ receptor signaling. J Immunother Cancer 2019; 7:152. [PMID: 31196219 PMCID: PMC6567539 DOI: 10.1186/s40425-019-0627-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/20/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND T-cell mediated immunotherapy brought clinical success for many cancer patients. Nonetheless, downregulation of MHC class I antigen presentation, frequently occurring in solid cancers, limits the efficacy of these therapies. Unraveling the mechanisms underlying this type of immune escape is therefore of great importance. We here investigated the immunological effects of metabolic stress in cancer cells as a result of nutrient deprivation. METHODS TC1 and B16F10 tumor cell lines were cultured under oxygen- and glucose-deprivation conditions that mimicked the tumor microenvironment of solid tumors. Presentation of peptide antigens by MHC class I molecules was measured by flow cytometry and via activation of tumor-specific CD8 T cell clones. The proficiency of the IFNy-STAT1 pathway was investigated by Western blots on phosphorylated proteins, transfection of constitutive active STAT1 constructs and qPCR of downstream targets. Kinase inhibitors for PI3K were used to examine its role in IFNy receptor signal transduction. RESULTS Combination of oxygen- and glucose-deprivation resulted in decreased presentation of MHC class I antigens on cancer cells, even in the presence of the stimulatory cytokine IFNy. This unresponsiveness to IFNy was the result of failure to phosphorylate the signal transducer STAT1. Forced expression of constitutive active STAT1 fully rescued the MHC class I presentation. Furthermore, oxygen- and glucose-deprivation increased PI3K activity in tumor cells. Pharmacological inhibition of this pathway not only restored signal transduction through IFNy-STAT1 but also improved MHC class I presentation. Importantly, PI3K inhibitors also rendered tumor cells sensitive for recognition by CD8 T cells in culture conditions of metabolic stress. CONCLUSIONS These data revealed a strong impact of metabolic stress on the presentation of tumor antigens by MHC class I and suggest that this type of tumor escape takes place at hypoxic areas even during times of active T cell immunity and IFNy release.
Collapse
Affiliation(s)
- Koen A Marijt
- Department of Medical Oncology, Oncode Institute, C7-P, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands.
| | - Marjolein Sluijter
- Department of Medical Oncology, Oncode Institute, C7-P, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands
| | - Laura Blijleven
- Department of Medical Oncology, Oncode Institute, C7-P, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands
| | - Sofie H Tolmeijer
- Department of Medical Oncology, Oncode Institute, C7-P, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands
| | - Ferenc A Scheeren
- Department of Medical Oncology, Oncode Institute, C7-P, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, C7-P, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, C7-P, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, the Netherlands
| |
Collapse
|
63
|
Da Silva DM, Skeate JG, Chavez-Juan E, Lühen KP, Wu JM, Wu CM, Kast WM, Hwang K. Therapeutic efficacy of a human papillomavirus type 16 E7 bacterial exotoxin fusion protein adjuvanted with CpG or GPI-0100 in a preclinical mouse model for HPV-associated disease. Vaccine 2019; 37:2915-2924. [PMID: 31010714 DOI: 10.1016/j.vaccine.2019.04.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 12/18/2022]
Abstract
Persistent human papillomavirus (HPV) infection is causally linked to the development of several human cancers, including cervical, vulvar, vaginal, anal, penile, and oropharyngeal cancers. To address the need for a therapeutic vaccine against HPV-associated diseases, here we test and compare the immunogenicity and therapeutic efficacy of a bacterial exotoxin fusion protein covalently linked to the HPV16 E7 oncoprotein adjuvanted with CpG or GPI-0100 in the C3.43 preclinical HPV16-transformed tumor model. We show that TVGV-1 protein vaccine adjuvanted with either CpG or GPI-0100 adjuvant induces a high frequency of E7-specific CD8+ T cells, and both adjuvants are able to assist the immune response in inducing polyfunctional cytokine-secreting lytic T cells that show therapeutic efficacy against well-established C3.43 tumors. CpG-adjuvanted TVGV-1 resulted in higher frequencies of IFNγ secreting and degranulating E7-specific T cells compared to GPI-0100-adjuvanted TVGV-1, resulting in marginally increased in vivo efficacy. Despite minor differences in immune response outcomes, we consider both CpG ODN and GPI-0100 to be promising vaccine adjuvants to increase the immunogenicity and therapeutic efficacy of the TVGV-1 protein for HPV16-driven cancers.
Collapse
Affiliation(s)
- Diane M Da Silva
- Department of Obstetrics & Gynecology, University of Southern California, Los Angeles, CA 90033, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
| | - Joseph G Skeate
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Elena Chavez-Juan
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Kim P Lühen
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Jiun-Ming Wu
- TheVax Genetics Vaccine Co., Ltd, Zhubei City, Hsinchu County 302, Taiwan, ROC
| | - Chia-Mao Wu
- TheVax Genetics Vaccine Co., Ltd, Zhubei City, Hsinchu County 302, Taiwan, ROC
| | - W Martin Kast
- Department of Obstetrics & Gynecology, University of Southern California, Los Angeles, CA 90033, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - KinKai Hwang
- TheVax Genetics Vaccine Co., Ltd, Irvine, CA 92618, USA
| |
Collapse
|
64
|
Sow HS, Ren J, Camps M, Ossendorp F, Ten Dijke P. Combined Inhibition of TGF-β Signaling and the PD-L1 Immune Checkpoint Is Differentially Effective in Tumor Models. Cells 2019; 8:cells8040320. [PMID: 30959852 PMCID: PMC6523576 DOI: 10.3390/cells8040320] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 12/14/2022] Open
Abstract
Antibodies blocking the programmed death-ligand 1 (PD-L1) have shown impressive and durable responses in clinical studies. However, this type of immunotherapy is only effective in a subset of patients and not sufficient for rejection of all tumor types. In this study, we explored in two mouse tumor models whether the antitumor effect could be enhanced by the combined blockade of PD-L1 and transforming growth factor-β (TGF-β), a potent immunosuppressive cytokine. The effect of anti-PD-L1 mouse monoclonal (mAb) and a TGF-β type I receptor small molecule kinase inhibitor (LY364947) was evaluated in the highly immunogenic mouse MC38 colon adenocarcinoma and the poorly immunogenic mouse KPC1 pancreatic tumor model. In the MC38 tumor model, LY364947 monotherapy did not show any antitumor effect, whereas treatment with anti-PD-L1 mAb significantly delayed tumor outgrowth. However, combination therapy showed the strongest therapeutic efficacy, resulting in improved long-term survival compared with anti-PD-L1 mAb monotherapy. This improved survival was associated with an increased influx of CD8+ T cells in the tumor microenvironment. In the KPC1 tumor model, LY364947 did not enhance the antitumor effect of anti-PD-L1 mAb. Despite this, delayed KPC1 tumor outgrowth was observed in the LY364947-treated group and this treatment led to a significant reduction of CD4+ T cells in the tumor microenvironment. Together, our data indicate that an additive anti-tumor response of dual targeting PD-L1 and TGF-β is dependent on the tumor model used, highlighting the importance of selecting appropriate cancer types, using in-depth analysis of the tumor microenvironment, which can benefit from combinatorial immunotherapy regimens.
Collapse
Affiliation(s)
- Heng Sheng Sow
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
| | - Jiang Ren
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
| | - Marcel Camps
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
| |
Collapse
|
65
|
Yang Y, Che Y, Zhao Y, Wang X. Prevention and treatment of cervical cancer by a single administration of human papillomavirus peptide vaccine with CpG oligodeoxynucleotides as an adjuvant in vivo. Int Immunopharmacol 2019; 69:279-288. [PMID: 30743204 DOI: 10.1016/j.intimp.2019.01.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/10/2019] [Accepted: 01/15/2019] [Indexed: 12/11/2022]
Abstract
No licensed therapeutic human papillomavirus (HPV) vaccine is currently available, so it remains a high priority to develop a therapeutic HPV vaccine or prophylactic/therapeutic HPV vaccine for cervical cancer. In this current study, we designed an HPV vaccine including CpG oligodeoxynucleotides 1826 as an adjuvant and HPV16 E7 43-77 peptide as antigen, which contains a CD8 T cell epitope (E7 49-57), and two CD4 T cell epitopes (E7 43-77 and E7 50-62). The prophylactic and therapeutic effect on cervical cancer induced by a single administration of vaccine, were comprehensively evaluated by examining the tumor size and the percentage of tumor-free/bearing mice. The cellular immunity and modulation of immunosuppressive cells induced by the vaccine were evaluated by examining intracellular cytokine staining (ICS) of splenocytes and FCM, respectively. Antigen-specific cytotoxic T-lymphocyte (CTL) responses were investigated using in vivo cytolytic assay. The results showed that the single administration of vaccine elicited significant prophylactic as well as therapeutic effect on cervical cancer. The increased cellular immunity mediated by CD4 + IFN-γ + T cells and CD8 + IFN-γ + T cells, and the decreased numbers of immunosuppressive cells including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) were induced by the vaccine. Antigen-specific CTL response was also induced by vaccination. These findings suggested that significant anti-tumor effect of the vaccine may result from the induction of increased cellular immunity and decreased immunosuppressive cells.
Collapse
Affiliation(s)
- Yang Yang
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yuxin Che
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yan Zhao
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang, China
| | - Xuelian Wang
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
66
|
Wang W, Liu Z, Zhou X, Guo Z, Zhang J, Zhu P, Yao S, Zhu M. Ferritin nanoparticle-based SpyTag/SpyCatcher-enabled click vaccine for tumor immunotherapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 16:69-78. [DOI: 10.1016/j.nano.2018.11.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/23/2018] [Accepted: 11/19/2018] [Indexed: 01/21/2023]
|
67
|
Beyranvand Nejad E, Ratts RB, Panagioti E, Meyer C, Oduro JD, Cicin-Sain L, Früh K, van der Burg SH, Arens R. Demarcated thresholds of tumor-specific CD8 T cells elicited by MCMV-based vaccine vectors provide robust correlates of protection. J Immunother Cancer 2019; 7:25. [PMID: 30704520 PMCID: PMC6357411 DOI: 10.1186/s40425-019-0500-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/08/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The capacity of cytomegalovirus (CMV) to elicit long-lasting strong T cell responses, and the ability to engineer the genome of this DNA virus positions CMV-based vaccine vectors highly suitable as a cancer vaccine platform. Defined immune thresholds for tumor protection and the factors affecting such thresholds have not well been investigated in cancer immunotherapy. We here determined using CMV as a vaccine platform whether critical thresholds of vaccine-specific T cell responses can be established that relate to tumor protection, and which factors control such thresholds. METHODS We generated CMV-based vaccine vectors expressing the E7 epitope and tested these in preclinical models of HPV16-induced cancer. Vaccination was applied via different doses and routes (intraperitoneal (IP), subcutaneous (SC) and intranasal (IN)). The magnitude, kinetics and phenotype of the circulating tumor-specific CD8+ T cell response were determined. Mice were subsequently challenged with tumor cells, and the tumor protection was monitored. RESULTS Immunization with CMV-based vaccines via the IP or SC route eliciting vaccine-induced CD8+ T cell responses of > 0.3% of the total circulating CD8 T cell population fully protects mice against lethal tumor challenge. However, low dose inoculations via the IP or SC route or IN vaccination elicited vaccine-induced CD8+ T cell responses that did not reach protective thresholds for tumor protection. In addition, whereas weak pre-existing immunity did not alter the protective thresholds of the vaccine-specific T cell response following subsequent immunization with CMV-based vaccine vectors, strong pre-existing immunity inhibited the development of vaccine-induced T cells and their control on tumor progression. CONCLUSIONS This study highlight the effectiveness of CMV-based vaccine vectors, and shows that demarcated thresholds of vaccine-specific T cells could be defined that correlate to tumor protection. Together, these results may hold importance for cancer vaccine development to achieve high efficacy in vaccine recipients.
Collapse
Affiliation(s)
- Elham Beyranvand Nejad
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands.,Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Eleni Panagioti
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands.,Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jennifer D Oduro
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Luka Cicin-Sain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute for Virology, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site, Hannover/Braunschweig, Germany
| | | | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands.
| |
Collapse
|
68
|
Curran MA, Glisson BS. New Hope for Therapeutic Cancer Vaccines in the Era of Immune Checkpoint Modulation. Annu Rev Med 2019; 70:409-424. [PMID: 30379596 DOI: 10.1146/annurev-med-050217-121900] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The driver and passenger mutations accumulated in the process of malignant transformation offer an adequate spectrum of immune visible alterations to the cellular proteome and resulting peptidome to render these cancers targetable-and, in theory, rejectable-by the host T cell immune response. In addition, cancers often overexpress tissue-specific and developmental antigens to which immune tolerance is incomplete. Sometimes, virally transferred oncogenes drive malignant transformation and remain expressed throughout the cancer. Despite this state of antigenic sufficiency, cancer grows progressively and overcomes all efforts of the host immune system to contain it. While therapeutic cancer vaccination can mobilize high frequencies of tumor-specific T cells, these responses remain subject to intratumoral attenuation. Antibody modulation of T cell function through checkpoint blockade or costimulatory activation can restore survival, proliferation, and effector function to these tumor-infiltrating T cells and convert otherwise subtherapeutic vaccines into potentially curative cancer immunotherapeutics.
Collapse
Affiliation(s)
- Michael A Curran
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA; .,University of Texas Health Science Center at Houston Graduate School of Biomedical Science, Houston, Texas 77054, USA
| | - Bonnie S Glisson
- Department of Thoracic Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
69
|
Xiang SD, Wilson KL, Goubier A, Heyerick A, Plebanski M. Design of Peptide-Based Nanovaccines Targeting Leading Antigens From Gynecological Cancers to Induce HLA-A2.1 Restricted CD8 + T Cell Responses. Front Immunol 2018; 9:2968. [PMID: 30631324 PMCID: PMC6315164 DOI: 10.3389/fimmu.2018.02968] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/03/2018] [Indexed: 01/02/2023] Open
Abstract
Gynecological cancers are a leading cause of mortality in women. CD8+ T cell immunity largely correlates with enhanced survival, whereas inflammation is associated with poor prognosis. Previous studies have shown polystyrene nanoparticles (PSNPs) are biocompatible, do not induce inflammation and when used as vaccine carriers for model peptides induce CD8+ T cell responses. Herein we test the immunogenicity of 24 different peptides, from three leading vaccine target proteins in gynecological cancers: the E7 protein of human papilloma virus (HPV); Wilms Tumor antigen 1 (WT1) and survivin (SV), in PSNP conjugate vaccines. Of relevance to vaccine development was the finding that a minimal CD8+ T cell peptide epitope from HPV was not able to induce HLA-A2.1 specific CD8+ T cell responses in transgenic humanized mice using conventional adjuvants such as CpG, but was nevertheless able to generate strong immunity when delivered as part of a specific longer peptide conjugated to PSNPs vaccines. Conversely, in most cases, when the minimal CD8+ T cell epitopes were able to induce immune responses (with WT1 or SV super agonists) in CpG, they also induced responses when conjugated to PSNPs. In this case, extending the sequence around the CD8+ T cell epitope, using the natural protein context, or engineering linker sequences proposed to enhance antigen processing, had minimal effects in enhancing or changing the cross-reactivity pattern induced by the super agonists. Nanoparticle approaches, such as PSNPs, therefore may offer an alternative vaccination strategy when conventional adjuvants are unable to elicit the desired CD8+ T cell specificity. The findings herein also offer sequence specific insights into peptide vaccine design for nanoparticle-based vaccine carriers.
Collapse
Affiliation(s)
- Sue D Xiang
- Department of Immunology, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia.,PX Biosolutions Pty Ltd., South Melbourne, VIC, Australia.,Ovarian Cancer Biomarker Laboratory, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Kirsty L Wilson
- Department of Immunology, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Anne Goubier
- PX Biosolutions Pty Ltd., South Melbourne, VIC, Australia
| | - Arne Heyerick
- PX Biosolutions Pty Ltd., South Melbourne, VIC, Australia
| | - Magdalena Plebanski
- Department of Immunology, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia.,PX Biosolutions Pty Ltd., South Melbourne, VIC, Australia.,School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
70
|
Zom GG, Willems MMJHP, Khan S, van der Sluis TC, Kleinovink JW, Camps MGM, van der Marel GA, Filippov DV, Melief CJM, Ossendorp F. Novel TLR2-binding adjuvant induces enhanced T cell responses and tumor eradication. J Immunother Cancer 2018; 6:146. [PMID: 30541631 PMCID: PMC6292168 DOI: 10.1186/s40425-018-0455-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 11/16/2018] [Indexed: 02/07/2023] Open
Abstract
Background Ligands for the Toll-like receptor (TLR) family can induce activation of cells of the innate immune system and are widely studied for their potential to enhance adaptive immunity. Conjugation of TLR2-ligand Pam3CSK4 to synthetic long peptides (SLPs) was shown to strongly enhance the induction of antitumor immunity. To further improve cancer vaccination, we have previously shown that the novel TLR2-L Amplivant (AV), a modified Pam3CSK4, potentiates the maturation effects on murine DCs. In the current study, we further assessed the immunological properties of AV. Methods Naïve mice were vaccinated with a conjugate of either Pam3CSK4 or AV and an SLP to assess specific T cell priming efficiency in vivo. The potency of AV and Pam3CSK4, either as free compounds or conjugated to different SLPs, to mature murine DCs was compared by stimulating murine dendritic cells overnight followed by ELISA and flow cytometry analysis. Murine tumor experiments were carried out by vaccinating mice carrying established HPV16 E6 and E7-expressing tumors and subsequently analyzing myeloid and lymphoid cells infiltrating the tumor microenvironment. Furthermore, tumor outgrowth after vaccination was monitored to enable comparison of the efficiency to induce antitumor immunity by Pam3CSK-SLP and AV-SLP conjugates. To enhance therapeutic efficacy, AV-SLP conjugate vaccination was combined with ablative therapies to assess whether synergism between such therapies would occur. Results SLPs conjugated to AV induce stronger DC maturation, in vivo T cell priming and antitumor immunity compared to conjugates with Pam3CSK4. Interestingly, AV-SLP conjugates modulate the macrophage populations in the tumor microenvironment, correlating with a therapeutic effect in an aggressive murine tumor model. The potency of AV-SLP conjugates in cancer vaccination operates optimally in combination with chemotherapy or photodynamic therapy. Conclusion These data allow further optimization of vaccination-based immunotherapy of cancer by use of the improved TLR2-ligand Amplivant. Electronic supplementary material The online version of this article (10.1186/s40425-018-0455-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gijs G Zom
- Department of Immunohematology and Blood Transfusion, section Tumorimmunology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Selina Khan
- Department of Immunohematology and Blood Transfusion, section Tumorimmunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tetje C van der Sluis
- Department of Immunohematology and Blood Transfusion, section Tumorimmunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Willem Kleinovink
- Department of Immunohematology and Blood Transfusion, section Tumorimmunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marcel G M Camps
- Department of Immunohematology and Blood Transfusion, section Tumorimmunology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Dmitri V Filippov
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Cornelis J M Melief
- Department of Immunohematology and Blood Transfusion, section Tumorimmunology, Leiden University Medical Center, Leiden, The Netherlands.,ISA Pharmaceuticals BV, Leiden, The Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, section Tumorimmunology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
71
|
Panahi HA, Bolhassani A, Javadi G, Noormohammadi Z. A comprehensive in silico analysis for identification of therapeutic epitopes in HPV16, 18, 31 and 45 oncoproteins. PLoS One 2018; 13:e0205933. [PMID: 30356257 PMCID: PMC6200245 DOI: 10.1371/journal.pone.0205933] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/11/2018] [Indexed: 11/25/2022] Open
Abstract
Human papillomaviruses (HPVs) are a group of circular double-stranded DNA viruses, showing severe tropism to mucosal tissues. A subset of HPVs, especially HPV16 and 18, are the primary etiological cause for several epithelial cell malignancies, causing about 5.2% of all cancers worldwide. Due to the high prevalence and mortality, HPV-associated cancers have remained as a significant health problem in human society, making an urgent need to develop an effective therapeutic vaccine against them. Achieving this goal is primarily dependent on the identification of efficient tumor-associated epitopes, inducing a robust cell-mediated immune response. Previous information has shown that E5, E6, and E7 early proteins are responsible for the induction and maintenance of HPV-associated cancers. Therefore, the prediction of major histocompatibility complex (MHC) class I T cell epitopes of HPV16, 18, 31 and 45 oncoproteins was targeted in this study. For this purpose, a two-step plan was designed to identify the most probable CD8+ T cell epitopes. In the first step, MHC-I and II binding, MHC-I processing, MHC-I population coverage and MHC-I immunogenicity prediction analyses, and in the second step, MHC-I and II protein-peptide docking, epitope conservation, and cross-reactivity with host antigens' analyses were carried out successively by different tools. Finally, we introduced five probable CD8+ T cell epitopes for each oncoprotein of the HPV genotypes (60 epitopes in total), which obtained better scores by an integrated approach. These predicted epitopes are valuable candidates for in vitro or in vivo therapeutic vaccine studies against the HPV-associated cancers. Additionally, this two-step plan that each step includes several analyses to find appropriate epitopes provides a rational basis for DNA- or peptide-based vaccine development.
Collapse
Affiliation(s)
- Heidar Ali Panahi
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Gholamreza Javadi
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Noormohammadi
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
72
|
Critical role for the Ly49 family of class I MHC receptors in adaptive natural killer cell responses. Proc Natl Acad Sci U S A 2018; 115:11579-11584. [PMID: 30249666 DOI: 10.1073/pnas.1722374115] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Adaptive natural killer (NK) cell memory represents a new frontier in immunology. Work over the last decade has discovered and confirmed the existence of NK cells with antigen-specific memories, which had previously been considered a unique property of T and B cells. These findings have shown that antigen-specific NK cells gain their specificity without the use of RAG proteins, representing a novel mechanism for generating antigen specificity, but the details of this mechanism have remained a mystery. We have discovered that members of the Ly49 family of surface receptors are critically involved in both the sensitization and the challenge phases of an NK cell memory response, as is antigen presentation from their binding partner, the class I MHC. Moreover, we demonstrate that the Ly49-interacting component of a presented antigen dictates the specificity of the NK cell memory response, implicating Ly49 receptors themselves in antigen-specific recognition. Finally, we demonstrate that adaptive NK cell memories can protect against an otherwise lethal melanoma without T cell or B cell support. These findings offer insight into the mechanism behind NK cell antigen specificity and demonstrate the clinical potential of this adaptive immune cell.
Collapse
|
73
|
Jang HY, Han BS, Kwon B, Sin JI. Optimized Gemcitabine Therapy in Combination with E7 Peptide Immunization Elicits Tumor Cure by Preventing Ag-Specific CTL Inhibition in Animals with Large Established Tumors. DNA Cell Biol 2018; 37:850-860. [PMID: 30227079 DOI: 10.1089/dna.2018.4319] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The role of chemotherapeutic agents in tumor immunotherapy is still controversial. In this study, we test using a TC-1 tumor model whether gemcitabine plus E7 peptide vaccine regimens (E7 peptides+CpG-ODN+anti-4-1BB Abs) may result in tumor cure in mice with large established tumors, with a focus on their effects on Ag-specific cytotoxic T lymphocyte (CTL) and myeloid-derived suppressor cell levels. Gemcitabine inhibited tumor growth by its direct cytotoxicity to tumor cells in vivo. E7 peptide vaccine regimens enhanced Ag-specific CTL lytic and antitumor therapeutic activity. Initial combination therapy using gemcitabine and E7 peptide vaccine regimens resulted in tumor regression with tumor relapse in animals with large established tumors, which appeared to result from the suppression of Ag-specific CTL activity by gemcitabine treatment. However, optimization of gemcitabine therapy by reducing its dose and frequency led to complete tumor regression without any recurring tumors in all tested mice even after discontinuation of therapy, possibly due to Ag-specific CTL responses. Thus, this study shows that the optimal dose and therapy frequency of gemcitabine are critical for achieving tumor cure in tumor-bearing animals undergoing E7 peptide vaccine regimen therapy, mainly by preventing CTL suppression. These findings may have implications for designing peptide-based therapeutic vaccines in cancer patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Ho-Young Jang
- 1 Department of Microbiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, South Korea
| | - Baek-Sang Han
- 2 BK21 Plus Graduate Program, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, South Korea
| | - Byungsuk Kwon
- 3 School of Biological Sciences, University of Ulsan, Ulsan, South Korea
| | - Jeong-Im Sin
- 1 Department of Microbiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, South Korea.,2 BK21 Plus Graduate Program, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, South Korea
| |
Collapse
|
74
|
Galliverti G, Tichet M, Domingos-Pereira S, Hauert S, Nardelli-Haefliger D, Swartz MA, Hanahan D, Wullschleger S. Nanoparticle Conjugation of Human Papillomavirus 16 E7-long Peptides Enhances Therapeutic Vaccine Efficacy against Solid Tumors in Mice. Cancer Immunol Res 2018; 6:1301-1313. [PMID: 30131378 DOI: 10.1158/2326-6066.cir-18-0166] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/16/2018] [Accepted: 08/16/2018] [Indexed: 11/16/2022]
Abstract
Treatment of patients bearing human papillomavirus (HPV)-related cancers with synthetic long-peptide (SLP) therapeutic vaccines has shown promising results in clinical trials against premalignant lesions, whereas responses against later stage carcinomas have remained elusive. We show that conjugation of a well-documented HPV-E7 SLP to ultra-small polymeric nanoparticles (NP) enhances the antitumor efficacy of therapeutic vaccination in different mouse models of HPV+ cancers. Immunization of TC-1 tumor-bearing mice with a single dose of NP-conjugated E7LP (NP-E7LP) generated a larger pool of E7-specific CD8+ T cells with increased effector functions than unconjugated free E7LP. At the tumor site, NP-E7LP prompted a robust infiltration of CD8+ T cells that was not accompanied by concomitant accumulation of regulatory T cells (Tregs), resulting in a higher CD8+ T-cell to Treg ratio. Consequently, the amplified immune response elicited by the NP-E7LP formulation led to increased regression of large, well-established tumors, resulting in a significant percentage of complete responses that were not achievable by immunizing with the non-NP-conjugated long-peptide. The partial responses were characterized by distinct phases of regression, stable disease, and relapse to progressive growth, establishing a platform to investigate adaptive resistance mechanisms. The efficacy of NP-E7LP could be further improved by therapeutic activation of the costimulatory receptor 4-1BB. This NP-E7LP formulation illustrates a "solid-phase" antigen delivery strategy that is more effective than a conventional free-peptide ("liquid") vaccine, further highlighting the potential of using such formulations for therapeutic vaccination against solid tumors. Cancer Immunol Res; 6(11); 1301-13. ©2018 AACR.
Collapse
Affiliation(s)
- Gabriele Galliverti
- Institute of Bioengineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Mélanie Tichet
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, EPFL, Lausanne, Switzerland
| | | | - Sylvie Hauert
- Institute of Bioengineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland.,Institute for Molecular Engineering, University of Chicago, Chicago, Illinois
| | | | - Melody A Swartz
- Institute of Bioengineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland. .,Institute for Molecular Engineering, University of Chicago, Chicago, Illinois.,The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Douglas Hanahan
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, EPFL, Lausanne, Switzerland.
| | - Stephan Wullschleger
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, EPFL, Lausanne, Switzerland.
| |
Collapse
|
75
|
Woodham AW, Cheloha RW, Ling J, Rashidian M, Kolifrath SC, Mesyngier M, Duarte JN, Bader JM, Skeate JG, Da Silva DM, Kast WM, Ploegh HL. Nanobody-Antigen Conjugates Elicit HPV-Specific Antitumor Immune Responses. Cancer Immunol Res 2018; 6:870-880. [PMID: 29792298 DOI: 10.1158/2326-6066.cir-17-0661] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/19/2018] [Accepted: 05/16/2018] [Indexed: 12/29/2022]
Abstract
High-risk human papillomavirus-associated cancers express viral oncoproteins (e.g., E6 and E7) that induce and maintain the malignant phenotype. The viral origin of these proteins makes them attractive targets for development of a therapeutic vaccine. Camelid-derived single-domain antibody fragments (nanobodies or VHHs) that recognize cell surface proteins on antigen-presenting cells (APC) can serve as targeted delivery vehicles for antigens attached to them. Such VHHs were shown to induce CD4+ and CD8+ T-cell responses against model antigens conjugated to them via sortase, but antitumor responses had not yet been investigated. Here, we tested the ability of an anti-CD11b VHH (VHHCD11b) to target APCs and serve as the basis for a therapeutic vaccine to induce CD8+ T-cell responses against HPV+ tumors. Mice immunized with VHHCD11b conjugated to an H-2Db-restricted immunodominant E7 epitope (E749-57) had more E7-specific CD8+ T cells compared with those immunized with E749-57 peptide alone. These CD8+ T cells acted prophylactically and conferred protection against a subsequent challenge with HPV E7-expressing tumor cells. In a therapeutic setting, VHHCD11b-E749-57 vaccination resulted in greater numbers of CD8+ tumor-infiltrating lymphocytes compared with mice receiving E749-57 peptide alone in HPV+ tumor-bearing mice, as measured by in vivo noninvasive VHH-based immune-positron emission tomography (immunoPET), which correlated with tumor regression and survival outcome. Together, these results demonstrate that VHHs can serve as a therapeutic cancer vaccine platform for HPV-induced cancers. Cancer Immunol Res; 6(7); 870-80. ©2018 AACR.
Collapse
Affiliation(s)
- Andrew W Woodham
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts. .,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Ross W Cheloha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Jingjing Ling
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Mohammad Rashidian
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Stephen C Kolifrath
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Maia Mesyngier
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Joao N Duarte
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts
| | - Justin M Bader
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Joseph G Skeate
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, California
| | - Diane M Da Silva
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, California.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - W Martin Kast
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, California.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California.,Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, California
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts.
| |
Collapse
|
76
|
Franconi R, Massa S, Illiano E, Muller A, Cirilli A, Accardd L, Bonito PDI, Giorgi C, Venuti A. Exploiting the Plant Secretory Pathway to Improve the Anticancer Activity of a Plant-Derived HPV16 E7 Vaccine. Int J Immunopathol Pharmacol 2018. [DOI: 10.1177/205873920601900119] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The human papillomavirus 16 (HPV16) E7 oncoprotein can be considered a ‘tumor-specific antigen’ and, therefore, it represents a promising target for a therapeutic vaccine against HPV-associated tumors. Efficient production of E7 protein with a plant-based transient expression system has been already described and it was demonstrated that E7-containing crude plant extracts confer partial protection against tumor challenge in a mouse model system. Before adopting the plant-based system as a cost-effective method for the production of an E7-based anti-cancer vaccine, some aspects, such as the oncoprotein yield, need further investigation. In the present study, we report the transient expression, mediated by a potato virus X (PVX)-derived vector, of the E7 protein targeted to the secretory system of Nicotiana benthamiana plants by using a plant-derived signal sequence. Targeting the antigen to the secretory pathway enhanced the E7 protein expression levels about five-fold. Mice immunized by s.c. administration with crude foliar extracts containing E7 showed strong stimulation of cell-mediated immune response after five boosters, as detected by ELISPOT. After challenging with the E7-expressing C3 tumor cells, tumor growth was completely inhibited in 80% of the vaccinated animals and a drastic reduction of tumor burden was observed in the remaining tumor-affected mice. These data demonstrate that, by enhancing E7 yield, it is possible to improve the anti-cancer activity of the plant-based experimental vaccine and open the way for a large-scale production of the E7 protein which could be purified or used as ‘in planta’ formulation, also suitable for oral therapeutic vaccination.
Collapse
Affiliation(s)
- R. Franconi
- ENEA, Italian National Agency for New Technologies, Energy and the Environment, BIOTEC, Laboratory of Plant Genetics and Genomics, C.R. Casaccia, P.O. Box 2400 I-00100 Roma, Italy
| | - S. Massa
- ENEA, Italian National Agency for New Technologies, Energy and the Environment, BIOTEC, Laboratory of Plant Genetics and Genomics, C.R. Casaccia, P.O. Box 2400 I-00100 Roma, Italy
| | - E. Illiano
- ENEA, Italian National Agency for New Technologies, Energy and the Environment, BIOTEC, Laboratory of Plant Genetics and Genomics, C.R. Casaccia, P.O. Box 2400 I-00100 Roma, Italy
| | - A. Muller
- Laboratory of Virology, Regina Elena Cancer Institute, Via delle Messi d'Oro 156, Roma, Italy
| | - A. Cirilli
- Laboratory of Virology, Regina Elena Cancer Institute, Via delle Messi d'Oro 156, Roma, Italy
| | - L. Accardd
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy
| | - P. DI Bonito
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy
| | - C. Giorgi
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy
| | - A. Venuti
- Laboratory of Virology, Regina Elena Cancer Institute, Via delle Messi d'Oro 156, Roma, Italy
| |
Collapse
|
77
|
Allen A, Wang C, Caproni LJ, Sugiyarto G, Harden E, Douglas LR, Duriez PJ, Karbowniczek K, Extance J, Rothwell PJ, Orefo I, Tite JP, Stevenson FK, Ottensmeier CH, Savelyeva N. Linear doggybone DNA vaccine induces similar immunological responses to conventional plasmid DNA independently of immune recognition by TLR9 in a pre-clinical model. Cancer Immunol Immunother 2018; 67:627-638. [PMID: 29330557 PMCID: PMC5860099 DOI: 10.1007/s00262-017-2111-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 12/20/2017] [Indexed: 12/29/2022]
Abstract
Vaccination with DNA that encodes cancer antigens is a simple and convenient way to raise immunity against cancer and has already shown promise in the clinical setting. Conventional plasmid DNA is commonly used which together with the encoded antigen also includes bacterial immunostimulatory CpG motifs to target the DNA sensor Toll-like receptor 9. Recently DNA vaccines using doggybone DNA (dbDNA™), have been developed without the use of bacteria. The cell-free process relies on the use of Phi29 DNA polymerase to amplify the template followed by protelomerase TelN to complete individual closed linear DNA. The resulting DNA contains the required antigenic sequence, a promoter and a poly A tail but lacks bacterial sequences such as an antibiotic resistance gene, prompting the question of immunogenicity. Here we compared the ability of doggybone DNA vaccine with plasmid DNA vaccine to induce adaptive immunity using clinically relevant oncotargets E6 and E7 from HPV. We demonstrate that despite the inability to trigger TLR9, doggybone DNA was able to induce similar levels of cellular and humoral immunity as plasmid DNA, with suppression of established TC-1 tumours.
Collapse
Affiliation(s)
- Alex Allen
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Chuan Wang
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Lisa J Caproni
- Touchlight Genetics Ltd, Morelands and Riverdale Buildings, Lower Sunbury Road, Hampton, London, TW12 2ER, UK
| | - Gessa Sugiyarto
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Elena Harden
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Leon R Douglas
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Patrick J Duriez
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Kinga Karbowniczek
- Touchlight Genetics Ltd, Morelands and Riverdale Buildings, Lower Sunbury Road, Hampton, London, TW12 2ER, UK
| | - Jon Extance
- Touchlight Genetics Ltd, Morelands and Riverdale Buildings, Lower Sunbury Road, Hampton, London, TW12 2ER, UK
| | - Paul J Rothwell
- Touchlight Genetics Ltd, Morelands and Riverdale Buildings, Lower Sunbury Road, Hampton, London, TW12 2ER, UK
| | - Ifeayinwa Orefo
- Touchlight Genetics Ltd, Morelands and Riverdale Buildings, Lower Sunbury Road, Hampton, London, TW12 2ER, UK
| | - John P Tite
- Touchlight Genetics Ltd, Morelands and Riverdale Buildings, Lower Sunbury Road, Hampton, London, TW12 2ER, UK
| | - Freda K Stevenson
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Christian H Ottensmeier
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Natalia Savelyeva
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK.
| |
Collapse
|
78
|
Brewer KD, Weir GM, Dude I, Davis C, Parsons C, Penwell A, Rajagopalan R, Sammatur L, Bowen CV, Stanford MM. Unique depot formed by an oil based vaccine facilitates active antigen uptake and provides effective tumour control. J Biomed Sci 2018; 25:7. [PMID: 29374458 PMCID: PMC5787234 DOI: 10.1186/s12929-018-0413-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 01/18/2018] [Indexed: 01/04/2023] Open
Abstract
Background Oil emulsions are commonly used as vaccine delivery platforms to facilitate slow release of antigen by forming a depot at the injection site. Antigen is trapped in the aqueous phase and as the emulsion degrades in vivo the antigen is passively released. DepoVax™ is a unique oil based delivery system that directly suspends the vaccine components in the oil diluent that forces immune cells to actively take up components from the formulation in the absence of passive release. The aim of this study was to use magnetic resonance imaging (MRI) with additional biological markers to evaluate and understand differences in clearance between several different delivery systems used in peptide-based cancer vaccines. Methods C57BL/6 mice were implanted with a cervical cancer model and vaccinated 5 days post-implant with either DepoVax (DPX), a water-in-oil emulsion (w/o), a squalene oil-in-water emulsion (squal o/w) or a saponin/liposome emulsion (sap/lip) containing iron oxide-labeled targeted antigen. MRI was then used to monitor antigen clearance, the site of injection, tumour and inguinal lymph node volumes and other gross anatomical changes. HLA-A2 transgenic mice were also vaccinated to evaluate immune responses of human directed peptides. Results We demonstrated differences in antigen clearance between DPX and w/o both in regard to how quickly the antigen was cleared and the pattern in which it was cleared. We also found differences in lymph node responses between DPX and both squal o/w and sap/lip. Conclusions These studies underline the unique mechanism of action of this clinical stage vaccine delivery system. Electronic supplementary material The online version of this article (10.1186/s12929-018-0413-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kimberly D Brewer
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada.,Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada.,Department of Physics and Atmospheric Science, Dalhousie University, Halifax, NS, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada.,School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| | | | - Iulia Dude
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | - Christa Davis
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | - Cathryn Parsons
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | | | | | | | - Chris V Bowen
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada.,Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada.,Department of Physics and Atmospheric Science, Dalhousie University, Halifax, NS, Canada.,School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| | - Marianne M Stanford
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada.,Immunovaccine Inc., Halifax, NS, Canada
| |
Collapse
|
79
|
Nano-Pulse Stimulation induces immunogenic cell death in human papillomavirus-transformed tumors and initiates an adaptive immune response. PLoS One 2018; 13:e0191311. [PMID: 29324830 PMCID: PMC5764415 DOI: 10.1371/journal.pone.0191311] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/02/2018] [Indexed: 02/01/2023] Open
Abstract
Nano-Pulse Stimulation (NPS) is a non-thermal pulsed electric field modality that has been shown to have cancer therapeutic effects. Here we applied NPS treatment to the human papillomavirus type 16 (HPV 16)-transformed C3.43 mouse tumor cell model and showed that it is effective at eliminating primary tumors through the induction of immunogenic cell death while subsequently increasing the number of tumor-infiltrating lymphocytes within the tumor microenvironment. In vitro NPS treatment of C3.43 cells resulted in a doubling of activated caspase 3/7 along with the translocation of phosphatidylserine (PS) to the outer leaflet of the plasma membrane, indicating programmed cell death activity. Tumor-bearing mice receiving standard NPS treatment showed an initial decrease in tumor volume followed by clearing of tumors in most mice, and a significant increase in overall survival. Intra-tumor analysis of mice that were unable to clear tumors showed an inverse correlation between the number of tumor infiltrating lymphocytes and the size of the tumor. Approximately half of the mice that cleared established tumors were protected against tumor re-challenge on the opposite flank. Selective depletion of CD8+ T cells eliminated this protection, suggesting that NPS treatment induces an adaptive immune response generating CD8+ T cells that recognize tumor antigen(s) associated with the C3.43 tumor model. This method may be utilized in the future to not only ablate primary tumors, but also to induce an anti-tumor response driven by effector CD8+ T cells capable of protecting individuals from disease recurrence.
Collapse
|
80
|
Brewer KD, DeBay DR, Dude I, Davis C, Lake K, Parsons C, Rajagopalan R, Weir G, Stanford MM, Mansour M, Bowen CV. Using lymph node swelling as a potential biomarker for successful vaccination. Oncotarget 2018; 7:35655-35669. [PMID: 27232944 PMCID: PMC5094952 DOI: 10.18632/oncotarget.9580] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/12/2016] [Indexed: 12/14/2022] Open
Abstract
There is currently a lack of biomarkers to help properly assess novel immunotherapies at both the preclinical and clinical stages of development. Recent work done by our group indicated significant volume changes in the vaccine draining right lymph node (RLN) volumes of mice that had been vaccinated with DepoVaxTM, a lipid-based vaccine platform that was developed to enhance the potency of peptide-based vaccines. These changes in lymph node (LN) volume were unique to vaccinated mice.To better assess the potential of volumetric LN markers for multiple vaccination platforms, we evaluated 100 tumor bearing mice and assessed their response to vaccination with either a DepoVax based vaccine (DPX) or a water-in-oil emulsion (w/o), and compared them to untreated controls. MRI was used to longitudinally monitor LN and tumor volumes weekly over 4 weeks. We then evaluated changes in LN volumes occurring in response to therapy as a potential predictive biomarker for treatment success.We found that for both vaccine types, DPX and w/o, the %RLN volumetric increase over baseline and the ratio of RLN/LLN were strong predictors of successful tumor suppression (LLN is left inguinal LN). The area under the curve (AUC) was greatest, between 0.75-0.85, two (%RLN) or three (RLN/LLN) weeks post-vaccination. For optimized critical thresholds we found these biomarkers consistently had sensitivity >90% and specificity >70% indicating strong prognostic potential. Vaccination with DepoVax had a more pronounced effect on draining lymph nodes than w/o emulsion vaccines, which correlated with a higher anti-tumor activity in DPX-treated mice.
Collapse
Affiliation(s)
- Kimberly D Brewer
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada.,Department of Radiology, Dalhousie University, Halifax, NS, Canada.,Department of Physics, Dalhousie University, Halifax, NS, Canada
| | - Drew R DeBay
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | - Iulia Dude
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | - Christa Davis
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | - Kerry Lake
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | - Cathryn Parsons
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | | | | | - Marianne M Stanford
- Immunovaccine Inc., Halifax, NS, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | | | - Chris V Bowen
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada.,Department of Radiology, Dalhousie University, Halifax, NS, Canada.,Department of Physics, Dalhousie University, Halifax, NS, Canada.,School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
81
|
|
82
|
A novel intracellular antibody against the E6 oncoprotein impairs growth of human papillomavirus 16-positive tumor cells in mouse models. Oncotarget 2017; 7:15539-53. [PMID: 26788990 PMCID: PMC4941259 DOI: 10.18632/oncotarget.6925] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023] Open
Abstract
Single-chain variable fragments (scFvs) expressed as “intracellular antibodies” (intrabodies) can target intracellular antigens to hamper their function efficaciously and specifically. Here we use an intrabody targeting the E6 oncoprotein of Human papillomavirus 16 (HPV16) to address the issue of a non-invasive therapy for HPV cancer patients. A scFv against the HPV16 E6 was selected by Intracellular Antibody Capture Technology and expressed as I7nuc in the nucleus of HPV16-positive SiHa, HPV-negative C33A and 293T cells. Colocalization of I7nuc and recombinant E6 was observed in different cell compartments, obtaining evidence of E6 delocalization ascribable to I7nuc. In SiHa cells, I7nuc expressed by pLNCX retroviral vector was able to partially inhibit degradation of the main E6 target p53, and induced p53 accumulation in nucleus. When analyzing in vitro activity on cell proliferation and survival, I7nuc was able to decrease growth inducing late apoptosis and necrosis of SiHa cells. Finally, I7nuc antitumor activity was demonstrated in two pre-clinical models of HPV tumors. C57BL/6 mice were injected subcutaneously with HPV16-positive TC-1 or C3 tumor cells, infected with pLNCX retroviral vector expressing or non-expressing I7nuc. All the mice injected with I7nuc-expressing cells showed a clear delay in tumor onset; 60% and 40% of mice receiving TC-1 and C3 cells, respectively, remained tumor-free for 17 weeks of follow-up, whereas 100% of the controls were tumor-bearing 20 days post-inoculum. Our data support the therapeutic potential of E6-targeted I7nuc against HPV tumors.
Collapse
|
83
|
Tremblay ML, Davis C, Bowen CV, Stanley O, Parsons C, Weir G, Karkada M, Stanford MM, Brewer KD. Using MRI cell tracking to monitor immune cell recruitment in response to a peptide-based cancer vaccine. Magn Reson Med 2017; 80:304-316. [PMID: 29193231 DOI: 10.1002/mrm.27018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/26/2017] [Accepted: 10/28/2017] [Indexed: 12/18/2022]
Abstract
PURPOSE MRI cell tracking can be used to monitor immune cells involved in the immunotherapy response, providing insight into the mechanism of action, temporal progression of tumor growth, and individual potency of therapies. To evaluate whether MRI could be used to track immune cell populations in response to immunotherapy, CD8+ cytotoxic T cells, CD4+ CD25+ FoxP3+ regulatory T cells, and myeloid-derived suppressor cells were labeled with superparamagnetic iron oxide particles. METHODS Superparamagnetic iron oxide-labeled cells were injected into mice (one cell type/mouse) implanted with a human papillomavirus-based cervical cancer model. Half of these mice were also vaccinated with DepoVaxTM (ImmunoVaccine, Inc., Halifax, Nova Scotia, Canada), a lipid-based vaccine platform that was developed to enhance the potency of peptide-based vaccines. RESULTS MRI visualization of CD8+ cytotoxic T cells, regulatory T cells, and myeloid-derived suppressor cells was apparent 24 h post-injection, with hypointensities due to iron-labeled cells clearing approximately 72 h post-injection. Vaccination resulted in increased recruitment of CD8+ cytotoxic T cells, and decreased recruitment of myeloid-derived suppressor cells and regulatory T cells to the tumor. We also found that myeloid-derived suppressor cell and regulatory T cell recruitment were positively correlated with final tumor volume. CONCLUSION This type of analysis can be used to noninvasively study changes in immune cell recruitment in individual mice over time, potentially allowing improved application and combination of immunotherapies. Magn Reson Med 80:304-316, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
| | - Christa Davis
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada
| | - Chris V Bowen
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada.,Department of Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Olivia Stanley
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada
| | - Cathryn Parsons
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada
| | | | - Mohan Karkada
- Wyss Institute at Harvard Medical School, Boston, Massachusetts, USA
| | - Marianne M Stanford
- Immunovaccine Inc., Halifax, Nova Scotia, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kimberly D Brewer
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada.,Department of Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia, Canada.,School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
84
|
In vivo electroporation enhances vaccine-mediated therapeutic control of human papilloma virus-associated tumors by the activation of multifunctional and effector memory CD8 + T cells. Vaccine 2017; 35:7240-7249. [PMID: 29174677 DOI: 10.1016/j.vaccine.2017.11.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/01/2017] [Accepted: 11/07/2017] [Indexed: 12/11/2022]
Abstract
In vivo electroporation (EP) has reignited the clinical interest on DNA vaccines as immunotherapeutic approaches to control different types of cancer. EP has been associated with increased immune response potency, but its capacity in influencing immunomodulation remains unclear. Here we evaluated the impact of in vivo EP on the induction of cellular immune responses and therapeutic effects of a DNA vaccine targeting human papillomavirus-induced tumors. Our results demonstrate that association of EP with the conventional intramuscular administration route promoted a more efficient activation of multifunctional and effector memory CD8+ T cells with enhanced cytotoxic activity. Furthermore, EP increased tumor infiltration of CD8+ T cells and avoided tumor recurrences. Finally, our results demonstrated that EP promotes local migration of antigen presenting cells that enhances with vaccine co-delivery. Altogether the present evidences shed further light on the in vivo electroporation action and its impact on the immunogenicity of DNA vaccines.
Collapse
|
85
|
Li J, Chen S, Ge J, Lu F, Ren S, Zhao Z, Pu X, Chen X, Sun J, Gu Y. A novel therapeutic vaccine composed of a rearranged human papillomavirus type 16 E6/E7 fusion protein and Fms-like tyrosine kinase-3 ligand induces CD8 + T cell responses and antitumor effect. Vaccine 2017; 35:6459-6467. [PMID: 29029939 DOI: 10.1016/j.vaccine.2017.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/26/2017] [Accepted: 09/03/2017] [Indexed: 01/15/2023]
Abstract
The development of cervical cancer is mainly caused by infection with high risk genotypes of human papillomavirus, particularly type 16 (HPV16), which accounts for more than 50% of cervical cancer. The two early viral oncogenes, E6 and E7, are continuously expressed in cervical cancer cells and are necessary to maintain the malignant cellular phenotype, thus providing ideal targets for immunotherapy of cervical cancer. In this study, a novel vaccine strategy was developed based on a rationally shuffled HPV16 E6/E7 fusion protein, the addition of Fms-like tyrosine kinase-3 ligand (Flt3L) or the N domain of calreticulin (NCRT), and the usage of a CpG adjuvant. Four recombinant proteins were constructed: m16E6E7 (mutant E6/E7 fusion protein), rm16E6E7 (rearranged mutant HPV16 E6/E7 fusion protein), Flt3L-RM16 (Flt3L fused to rm16E6E7), and NCRT-RM16 (NCRT fused to rm16E6E7). Our results suggest that Flt3L-RM16 was the most potent of these proteins in terms of inducing E6- and E7-specific CD8+ T cell responses. Additionally, Flt3L-RM16 significantly induced regression of established E6/E7-expressing TC-1 tumors. Higher doses of Flt3L-RM16 trended toward higher levels of antitumor activity, but these differences did not reach statistical significance. In summary, this study found that Flt3L-RM16 fusion protein is a promising therapeutic vaccine for immunotherapy of HPV16-associated cervical cancer.
Collapse
Affiliation(s)
- Jianqiang Li
- Jiangsu Theravac Bio-pharmaceutical CO., Ltd, Nanjing, China.
| | - Si Chen
- Jiangsu Theravac Bio-pharmaceutical CO., Ltd, Nanjing, China.
| | - Jun Ge
- Jiangsu Theravac Bio-pharmaceutical CO., Ltd, Nanjing, China.
| | - Feng Lu
- Jiangsu Theravac Bio-pharmaceutical CO., Ltd, Nanjing, China.
| | - Sulin Ren
- Jiangsu Theravac Bio-pharmaceutical CO., Ltd, Nanjing, China.
| | - Zhiqiang Zhao
- Suzhou Yuankang Bio-pharmaceutical Co., Ltd., Suzhou, China.
| | - Xiuying Pu
- Suzhou Yuankang Bio-pharmaceutical Co., Ltd., Suzhou, China.
| | - Xiaoxiao Chen
- Jiangsu Theravac Bio-pharmaceutical CO., Ltd, Nanjing, China.
| | - Jiaojiao Sun
- Jiangsu Theravac Bio-pharmaceutical CO., Ltd, Nanjing, China.
| | - Yueqing Gu
- China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
86
|
Porchia BFMM, Moreno ACR, Ramos RN, Diniz MO, de Andrade LHTM, Rosa DS, Barbuto JAM, Boscardin SB, Ferreira LCS. Herpes Simplex Virus Glycoprotein D Targets a Specific Dendritic Cell Subset and Improves the Performance of Vaccines to Human Papillomavirus-Associated Tumors. Mol Cancer Ther 2017; 16:1922-1933. [PMID: 28522585 DOI: 10.1158/1535-7163.mct-17-0071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 04/05/2017] [Accepted: 05/09/2017] [Indexed: 11/16/2022]
Abstract
Cervical cancer is a major public health problem and one of the leading causes of cancer deaths in women. Virtually all cases of cervical cancer, as well as a growing share of anal and head/neck tumors, are associated with human papillomavirus (HPV) infection. Despite the effectiveness, the available prophylactic vaccines do not benefit women with cervical lesions or cancer. Therefore, the search of new immunotherapeutic approaches to treat HPV-induced tumors is still a priority. The present study characterizes a therapeutic antitumor vaccine based on the genetic fusion of the Herpes simplex virus-1 (HSV-1) glycoprotein D (gD) with the E7 oncoprotein from HPV-16 (gDE7). Two subcutaneous doses of gDE7, admixed with poly (I:C), conferred complete and long-lasting therapeutic antitumor protection on mice previously challenged with tumor cells expressing the HPV-16 oncoproteins. The vaccine induced multifunctional E7-specific CD8+ T cells with cytotoxic activity and effector memory phenotype (CD44+ CD62Llow). In addition, gDE7 admixed with poly (I:C) vaccination controlled the expansion of tumor-induced regulatory T cells and myeloid-derived suppressor cells. More importantly, gDE7 activated mouse CD11c+ CD8α+ and human BDCA3+ dendritic cells (DC), specialized in antigen cross-presentation to CD8+ T cells, under in vitro conditions. These results indicated that the activation of a specific DC population, mediated by gD, improved the antigen-specific immune responses and the therapeutic performance induced by antitumor vaccines. These results open perspectives for the clinical testing of gDE7-based vaccines under the concept of active immunization as a tool for the therapeutic control of cancer. Mol Cancer Ther; 16(9); 1922-33. ©2017 AACR.
Collapse
Affiliation(s)
- Bruna F M M Porchia
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana Carolina R Moreno
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rodrigo N Ramos
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mariana O Diniz
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Laís Helena T M de Andrade
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniela S Rosa
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo-UNIFESP, São Paulo, Brazil
| | - José Alexandre M Barbuto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Silvia B Boscardin
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luís Carlos S Ferreira
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
87
|
Chahal JS, Fang T, Woodham AW, Khan OF, Ling J, Anderson DG, Ploegh HL. An RNA nanoparticle vaccine against Zika virus elicits antibody and CD8+ T cell responses in a mouse model. Sci Rep 2017; 7:252. [PMID: 28325910 PMCID: PMC5427874 DOI: 10.1038/s41598-017-00193-w] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/13/2017] [Indexed: 02/06/2023] Open
Abstract
The Zika virus (ZIKV) outbreak in the Americas and South Pacific poses a significant burden on human health because of ZIKV's neurotropic effects in the course of fetal development. Vaccine candidates against ZIKV are coming online, but immunological tools to study anti-ZIKV responses in preclinical models, particularly T cell responses, remain sparse. We deployed RNA nanoparticle technology to create a vaccine candidate that elicited ZIKV E protein-specific IgG responses in C57BL/6 mice as assayed by ELISA. Using this tool, we identified a unique H-2Db-restricted epitope to which there was a CD8+ T cell response in mice immunized with our modified dendrimer-based RNA nanoparticle vaccine. These results demonstrate that this approach can be used to evaluate new candidate antigens and identify immune correlates without the use of live virus.
Collapse
Affiliation(s)
- Jasdave S Chahal
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA, 02142, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Tao Fang
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA, 02142, USA
| | - Andrew W Woodham
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA, 02142, USA
| | - Omar F Khan
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Jingjing Ling
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA, 02142, USA
| | - Daniel G Anderson
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Hidde L Ploegh
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA, 02142, USA.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
88
|
Esquerré M, Bouillette-Marussig M, Goubier A, Momot M, Gonindard C, Keller H, Navarro A, Bissery MC. GTL001, a bivalent therapeutic vaccine against human papillomavirus 16 and 18, induces antigen-specific CD8+ T cell responses leading to tumor regression. PLoS One 2017; 12:e0174038. [PMID: 28301611 PMCID: PMC5354464 DOI: 10.1371/journal.pone.0174038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 03/02/2017] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Prophylactic vaccines are available for women and girls not yet infected with HPV, but women already infected with HPV need a treatment to prevent progression to high-grade cervical lesions and cancer. GTL001 is a bivalent therapeutic vaccine for eradicating HPV-infected cells that contains HPV16 E7 and HPV18 E7 both fused to detoxified adenylate cyclase from Bordetella pertussis, which binds specifically to CD11b+ antigen-presenting cells. This study examined the ability of therapeutic vaccination with GTL001 adjuvanted with topical imiquimod cream to induce functional HPV16 E7- and HPV18 E7-specific CD8+ T cell responses. METHODS Binding of GTL001 to human CD11b was assessed by a cell-based competition binding assay. Cellular immunogenicity of intradermal vaccination with GTL001 was assessed in C57BL/6 mice by enzyme-linked immunospot assay and in vivo killing assays. In vivo efficacy of GTL001 vaccination was investigated in the TC-1 murine HPV16 E7-expressing tumor model. RESULTS GTL001 bound specifically to the human CD11b/CD18 receptor. GTL001 adjuvanted with topical 5% imiquimod cream induced HPV16 E7 and HPV18 E7-specific CD8+ T cell responses. This CD8+ T-cell response mediated in vivo killing of HPV E7-expressing cells. In the HPV16 E7-expressing tumor model, GTL001 adjuvanted with imiquimod but not imiquimod alone or a combination of unconjugated HPV16 E7 and HPV18 E7 caused complete tumor regression. CONCLUSIONS GTL001 adjuvanted with topical 5% imiquimod is immunogenic and induces HPV16 E7 and HPV18 E7-specific CD8+ T cell responses that can kill HPV E7-expressing cells and eliminate HPV E7-expressing tumors.
Collapse
|
89
|
GTL001 and bivalent CyaA-based therapeutic vaccine strategies against human papillomavirus and other tumor-associated antigens induce effector and memory T-cell responses that inhibit tumor growth. Vaccine 2017; 35:1509-1516. [PMID: 28196735 DOI: 10.1016/j.vaccine.2017.01.074] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/06/2017] [Accepted: 01/30/2017] [Indexed: 02/02/2023]
Abstract
GTL001 is a bivalent therapeutic vaccine containing human papillomavirus (HPV) 16 and HPV18 E7 proteins inserted in the Bordetella pertussis adenylate cyclase (CyaA) vector intended to prevent cervical cancer in HPV-infected women with normal cervical cytology or mild abnormalities. To be effective, therapeutic cervical cancer vaccines should induce both a T cell-mediated effector response against HPV-infected cells and a robust CD8+ T-cell memory response to prevent potential later infection. We examined the ability of GTL001 and related bivalent CyaA-based vaccines to induce, in parallel, effector and memory CD8+ T-cell responses to both vaccine antigens. Intradermal vaccination of C57BL/6 mice with GTL001 adjuvanted with a TLR3 agonist (polyinosinic-polycytidylic acid) or a TLR7 agonist (topical 5% imiquimod cream) induced strong HPV16 E7-specific T-cell responses capable of eradicating HPV16 E7-expressing tumors. Tumor-free mice also had antigen-specific memory T-cell responses that protected them against a subsequent challenge with HPV18 E7-expressing tumor cells. In addition, vaccination with bivalent vaccines containing CyaA-HPV16 E7 and CyaA fused to a tumor-associated antigen (melanoma-specific antigen A3, MAGEA3) or to a non-viral, non-tumor antigen (ovalbumin) eradicated HPV16 E7-expressing tumors and protected against a later challenge with MAGEA3- and ovalbumin-expressing tumor cells, respectively. These results show that CyaA-based bivalent vaccines such as GTL001 can induce both therapeutic and prophylactic anti-tumor T-cell responses. The CyaA platform can be adapted to different antigens and adjuvants, and therefore may be useful for developing other therapeutic vaccines.
Collapse
|
90
|
Abstract
Aurora kinase A (AURKA) is a centrosomal protein that is overexpressed in a number of human malignancies and can contribute to tumor progression. As we used this protein as a target of DNA immunization, we increased its immunogenicity by the addition of the PADRE helper epitope and decreased its potential oncogenicity by mutagenesis of the kinase domain. For in vitro analysis of induced immune responses in mice, we identified the Aurka(220-228) nonapeptide representing an H-2Kb epitope. As DNA vaccination against the Aurka self-antigen by a gene gun did not show any antitumor effect, we combined DNA immunization with anti-CD25 treatment that depletes mainly regulatory T cells. Whereas 1 anti-CD25 dose injected before DNA vaccination did not enhance the activation of Aurka-specific splenocytes, 3 doses administered on days of immunizations augmented about 10-fold immunity against Aurka. However, an opposite effect was found for antitumor immunity-only 1 anti-CD25 dose combined with DNA vaccination reduced tumor growth. Moreover, the administration of 3 doses of anti-CD25 antibody alone accelerated tumor growth. Analysis of tumor-infiltrating cells showed that 3 anti-CD25 doses not only efficiently depleted regulatory T cells but also activated helper T cells and CD3(-)CD25(+) cells. Next, we found that blockade of the PD-1 receptor initiated 1 week after the first immunization was necessary for significant inhibition of tumor growth with therapeutic DNA vaccination against Aurka combined with depletion of CD25 cells. Our results suggest that combined cancer immunotherapy should be carefully evaluated to achieve the optimal antitumor effect.
Collapse
|
91
|
Kavunja HW, Lang S, Sungsuwan S, Yin Z, Huang X. Delivery of foreign cytotoxic T lymphocyte epitopes to tumor tissues for effective antitumor immunotherapy against pre-established solid tumors in mice. Cancer Immunol Immunother 2016; 66:451-460. [PMID: 28011995 DOI: 10.1007/s00262-016-1948-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 12/18/2016] [Indexed: 11/26/2022]
Abstract
Cytotoxic T lymphocyte (CTL) can have remarkable abilities to kill tumor cells. However, the establishment of successful CTL-based anticancer therapy has met with many challenges. Within tumor cells, there exist subpopulations with low or no expression of the targeted antigen (termed as antigen-loss variants). In addition, tumor cells can downregulate the levels of major histocompatibility complex class I (MHC-I) molecules on cell surface due to immune pressure. As a result, some tumor cells can escape the immune pressure bestowed by CTLs, resulting in treatment failure. To address these difficulties, a new approach is developed to deliver foreign high-affinity CTL epitopes to tumor tissues utilizing pH-responsive "smart" microparticles (MPs). These MPs could encapsulate CTL peptide epitope, release the peptide under acidic condition encountered in tumor tissues and enhance CTL activation. Mice bearing pre-established tumor as "antigen-loss variant" solid tumor models were administered intratumorally with MPs containing the CTL peptide, which showed 100% survival following the treatment. In contrast, all control mice died from tumor. Significant protection from tumor-induced death was also observed with systemic administration of CTL peptide-MPs. The therapeutic efficacy can be attributed to enhanced delivery of the epitope to tumor tissues, presentation of the epitope by tumor cells as well as tumor stromal cells and/or generation of epitope-specific CTLs by the peptide-containing MPs. These findings offer a promising new direction for treating established solid tumor using CTL therapy.
Collapse
Affiliation(s)
- Herbert W Kavunja
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
| | - Shuyao Lang
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
| | - Suttipun Sungsuwan
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
| | - Zhaojun Yin
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA.
| |
Collapse
|
92
|
Paolini F, Curzio G, Cordeiro MN, Massa S, Mariani L, Pimpinelli F, de Freitas AC, Franconi R, Venuti A. HPV 16 E5 oncoprotein is expressed in early stage carcinogenesis and can be a target of immunotherapy. Hum Vaccin Immunother 2016; 13:291-297. [PMID: 27929754 DOI: 10.1080/21645515.2017.1264777] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
HPV16 persistent infection is a well-known condition that precedes human cancer development. High risk HPV E5 proteins cooperate with E6/E7 oncogenes to promote hyper-proliferation of infected cells leading to possible cancer progression. Thus, presence of E5 viral transcripts could be a key marker of active infection and, in turn, a target of immunotherapy. Purpose of the study is to detect E5 transcripts in clinical samples and to explore the activity of novel anti-HPV16 E5 DNA vaccines. HPV transcripts were detected by PCR with specific primers encompassing the splice-donor sites of E5 transcript. For E5-based immunotherapies, 2 E5-based versions of DNA vaccines carrying whole E5 gene or a synthetic multiepitope gene were improved by fusion to sequence of PVX coat protein. These vaccines were challenged with a new luminescent animal model based on C3-Luc cell line. E5 transcripts were detected in clinical samples of women with HPV positive low-grade SIL, demonstrating the validity of our test. In C3 pre-clinical mouse model, vaccine candidates were able to induce a strong cellular immunity as indicated by ELISPOT assays. In addition, E5-CP vaccines elicited strong anti-tumor effects as showed by decreased tumor growth monitored by animal imaging. The tumor growth inhibition was comparable to those obtained with anti-E7 DNA vaccines. In conclusion, detection of E5 transcripts in clinical samples indicates that E5 is a possible target of immunotherapy. Data from pre-clinical model demonstrate that E5 genetic immunization is feasible, efficacious and could be utilized in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Silvia Massa
- c ENEA, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, C.R. Casaccia , Rome , Italy
| | - Luciano Mariani
- a Regina Elena National Cancer Institute, HPV Unit , Rome , Italy
| | | | | | - Rosella Franconi
- c ENEA, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, C.R. Casaccia , Rome , Italy
| | - Aldo Venuti
- a Regina Elena National Cancer Institute, HPV Unit , Rome , Italy
| |
Collapse
|
93
|
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: 42] [Impact Index Per Article: 4.7] [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
|
94
|
Devaraj K, Gillison ML, Wu TC. Development of HPV Vaccines for HPV-associated Head and Neck Squamous Cell Carcinoma. ACTA ACUST UNITED AC 2016; 14:345-62. [PMID: 14530303 DOI: 10.1177/154411130301400505] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
High-risk genotypes of the human papillomavirus (HPV), particularly HPV type 16, are found in a distinct subset of head and neck squamous cell carcinomas (HNSCC). Thus, these HPV-associated HNSCC may be prevented or treated by vaccines designed to induce appropriate HPV virus-specific immune responses. Infection by HPV may be prevented by neutralizing antibodies specific for the viral capsid proteins. In clinical trials, vaccines comprised of HPV virus-like particles (VLPs) have shown great promise as prophylactic HPV vaccines. However, given that capsid proteins are not expressed at detectable levels by infected basal keratinocytes, vaccines with therapeutic potential must target other non-structural viral antigens. Two HPV oncogenic proteins, E6 and E7, are important in the induction and maintenance of cellular transformation and are co-expressed in the majority of HPV-containing carcinomas. Therefore, therapeutic vaccines targeting these proteins may have potential to control HPV-associated malignancies. Various candidate therapeutic HPV vaccines are currently being tested whereby E6 and/or E7 is administered in live vectors, in peptides or protein, in nucleic acid form, as components of chimeric VLPs, or in cell-based vaccines. Encouraging results from experimental vaccination systems in animal models have led to several prophylactic and therapeutic vaccine clinical trials. Should they fulfill their promise, these vaccines may prevent HPV infection or control its potentially life-threatening consequences in humans.
Collapse
Affiliation(s)
- Kalpana Devaraj
- Department of Pathology, The Johns Hopkins Medical Institutions, 720 Rutland Avenue, Ross Building 512, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
95
|
Arenas-Ramirez N, Zou C, Popp S, Zingg D, Brannetti B, Wirth E, Calzascia T, Kovarik J, Sommer L, Zenke G, Woytschak J, Regnier CH, Katopodis A, Boyman O. Improved cancer immunotherapy by a CD25-mimobody conferring selectivity to human interleukin-2. Sci Transl Med 2016; 8:367ra166. [DOI: 10.1126/scitranslmed.aag3187] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 06/08/2016] [Accepted: 09/08/2016] [Indexed: 12/31/2022]
|
96
|
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
|
97
|
Weir GM, Hrytsenko O, Quinton T, Berinstein NL, Stanford MM, Mansour M. Anti-PD-1 increases the clonality and activity of tumor infiltrating antigen specific T cells induced by a potent immune therapy consisting of vaccine and metronomic cyclophosphamide. J Immunother Cancer 2016; 4:68. [PMID: 27777777 PMCID: PMC5067905 DOI: 10.1186/s40425-016-0169-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/21/2016] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Future cancer immunotherapies will combine multiple treatments to generate functional immune responses to cancer antigens through synergistic, multi-modal mechanisms. In this study we explored the combination of three distinct immunotherapies: a class I restricted peptide-based cancer vaccine, metronomic cyclophosphamide (mCPA) and anti-PD-1 treatment in a murine tumor model expressing HPV16 E7 (C3). METHODS Mice were implanted with C3 tumors subcutaneously. Tumor bearing mice were treated with mCPA (20 mg/kg/day PO) for seven continuous days on alternating weeks, vaccinated with HPV16 E749-57 peptide antigen formulated in the DepoVax (DPX) adjuvanting platform every second week, and administered anti-PD-1 (200 μg/dose IP) after each vaccination. Efficacy was measured by following tumor growth and survival. Immunogenicity was measured by IFN-γ ELISpot of spleen, vaccine draining lymph nodes and tumor draining lymph nodes. Tumor infiltration was measured by flow cytometry for CD8α+ peptide-specific T cells and RT-qPCR for cytotoxic proteins. The clonality of tumor infiltrating T cells was measured by TCRβ sequencing using genomic DNA. RESULTS Untreated C3 tumors had low expression of PD-L1 in vivo and anti-PD-1 therapy alone provided no protection from tumor growth. Treatment with DPX/mCPA could delay tumor growth, and tri-therapy with DPX/mCPA/anti-PD-1 provided long-term control of tumors. We found that treatment with DPX/mCPA/anti-PD-1 enhanced systemic antigen-specific immune responses detected in the spleen as determined by IFN-γ ELISpot compared to those in the DPX/mCPA group, but immune responses in tumor-draining lymph nodes were not increased. Although no increases in antigen-specific CD8α+ TILs could be detected, there was a trend for increased expression of cytotoxic genes within the tumor microenvironment as well as an increase in clonality in mice treated with DPX/mCPA/anti-PD-1 compared to those with anti-PD-1 alone or DPX/mCPA. Using a library of antigen-specific CD8α+ T cell clones, we found that antigen-specific clones were more frequently expanded in the DPX/mCPA/anti-PD-1 treated group. CONCLUSIONS These results demonstrate how the efficacy of anti-PD-1 may be improved by combination with a potent and targeted T cell activating immune therapy.
Collapse
MESH Headings
- Administration, Metronomic
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- B7-H1 Antigen/genetics
- B7-H1 Antigen/metabolism
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Clonal Evolution/drug effects
- Clonal Evolution/immunology
- Cyclophosphamide/administration & dosage
- Cytotoxicity, Immunologic
- Disease Models, Animal
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/immunology
- Female
- Gene Expression
- Humans
- Immunomodulation/drug effects
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mice
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/pathology
- Neoplasms/therapy
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/metabolism
- T-Cell Antigen Receptor Specificity/drug effects
- T-Cell Antigen Receptor Specificity/immunology
- T-Lymphocyte Subsets/drug effects
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
| | - Olga Hrytsenko
- Immunovaccine Inc., 1344 Summer St., Halifax, NS B3H 0A8 Canada
| | - Tara Quinton
- Immunovaccine Inc., 1344 Summer St., Halifax, NS B3H 0A8 Canada
| | - Neil L. Berinstein
- Sunnybrook Research Institute, 2075 Bayview Ave., Toronto, ON M4N 3M5 Canada
- University of Toronto, 27 King’s College Cir, Toronto, ON M5S 1A1 Canada
| | - Marianne M. Stanford
- Immunovaccine Inc., 1344 Summer St., Halifax, NS B3H 0A8 Canada
- Department of Microbiology & Immunology, Dalhousie University, 5850 College St., Room 7C, Halifax, NS B3H 4R2 Canada
| | - Marc Mansour
- Immunovaccine Inc., 1344 Summer St., Halifax, NS B3H 0A8 Canada
| |
Collapse
|
98
|
Sabah SN, Gazi MA, Sthity RA, Husain AB, Quyyum SA, Rahman M, Islam MR. Designing of Epitope-Focused Vaccine by Targeting E6 and E7 Conserved Protein Sequences: An Immuno-Informatics Approach in Human Papillomavirus 58 Isolates. Interdiscip Sci 2016; 10:251-260. [PMID: 27640170 DOI: 10.1007/s12539-016-0184-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 08/02/2016] [Accepted: 09/02/2016] [Indexed: 10/21/2022]
Abstract
Human papillomavirus (HPV) is a DNA virus that belongs to the papillomavirus family and is capable of infecting humans. Currently, few vaccines are available to prevent infection by HPV. However, they are not so much effective and provide little benefit to women who have already been infected with HPV. The aim of this study was to design epitope-based vaccines of HPV58 by targeting E6 and E7 proteins of HPV58. Proteomic sequences were retrieved from different isolates at different time periods and later analyzed by performing alignment of these sequences. To ensure the capacity of humoral and cell-mediated immunity, both B cell and T cell immunity were checked for the peptides. For E6 protein, the peptide sequence from 48 to 54 amino acids and one 9-m epitope ETSVHEIEL were the most potential B cell and T cell epitopes, respectively. This peptide could interact with as many as eight MHC-1 alleles and showed high population coverage up to 90.31 %. On the other hand, the peptide region for the E7 protein ranged from 27 to 33 amino acids and two 9-m epitopes QAQPATANY, SSDEDEIGL were found as the most potential B cell and T cell epitopes, respectively. The peptide sequences could interact with as many as seven MHC-1 alleles and showed population coverage up to 90.31 %. Furthermore, conservancy analysis was also performed using in silico tools and showed a conservancy of 100 % for all the selected epitopes. In addition to this, the allergenicity of the epitopes was also evaluated. Although the study requires further in vitro and in vivo screening, this epitope-focused peptide vaccine designing opens up a new skyline that holds a prospective future in HPV research.
Collapse
Affiliation(s)
| | - Md Amran Gazi
- Nutrition and Clinical Services Division, Centre for Nutrition and Food Security, International Centre for Diarrhoeal Disease Research, GPO Box 128, Dhaka, 1000, Bangladesh.
| | - Rahvia Alam Sthity
- Immunobiology, Nutrition and Toxicology Laboratory, International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - Amena Binte Husain
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Salwa Abdul Quyyum
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Mustafizur Rahman
- Center for Bio-Medical Research, Manarat University, Dhaka, Bangladesh
| | - Md Rezaul Islam
- International Max Planck Research School, University of Göttingen, 37075, Göttingen, Germany.
| |
Collapse
|
99
|
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
|
100
|
Beyranvand Nejad E, van der Sluis TC, van Duikeren S, Yagita H, Janssen GM, van Veelen PA, Melief CJM, van der Burg SH, Arens R. Tumor Eradication by Cisplatin Is Sustained by CD80/86-Mediated Costimulation of CD8+ T Cells. Cancer Res 2016; 76:6017-6029. [PMID: 27569212 DOI: 10.1158/0008-5472.can-16-0881] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 08/01/2016] [Indexed: 11/16/2022]
Abstract
Certain cytotoxic chemotherapeutic drugs are immunogenic, stimulating tumor immunity through mechanisms that are not completely understood. Here we show how the DNA-damaging drug cisplatin modulates tumor immunity. At the maximum tolerated dose (MTD), cisplatin cured 50% of mice with established murine TC-1 or C3 tumors, which are preclinical models of human papillomavirus (HPV)-associated cancer. Notably, the curative benefit of cisplatin relied entirely upon induction of tumor-specific CD8+ T cells. Mechanistic investigations showed that cisplatin stimulated tumor infiltration of inflammatory antigen-presenting cells (APC) expressing relatively higher levels of the T-cell costimulatory ligands CD70, CD80, and CD86. Cell death triggered by cisplatin was associated with the release of at least 19 proteins in the tumor environment that could act as damage-associated molecular patterns and upregulate costimulatory molecules, either alone or in concert, but the responsible proteins remain unknown. Essentially, the curative effect of cisplatin was abrogated in mice lacking expression of CD80 and CD86 on APCs. Furthermore, cisplatin treatment was improved by CTLA-4 blockade, which increases the availability of CD80/86 to bind to CD28. In contrast, there was no effect of CD27 stimulation, which replaces CD70 interaction. At the cisplatin MTD, cure rates could also be increased by vaccination with synthetic long peptides, whereas cures could also be achieved at similar rates at 80% of the MTD with reduced side effects. Our findings reveal an essential basis for the immunogenic properties of cisplatin, which are mediated by the induction of costimulatory signals for CD8+ T-cell-dependent tumor destruction. Cancer Res; 76(20); 6017-29. ©2016 AACR.
Collapse
Affiliation(s)
- Elham Beyranvand Nejad
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Tetje C van der Sluis
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Suzanne van Duikeren
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - George M Janssen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter A van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Cornelis J M Melief
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands. ISA Pharmaceuticals, Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|