1
|
Molina E, Tejero M, Duzenli OF, Kuoch H, Caine C, Krotova K, Paulaitis M, Aslanidi G. Insights in AAV-mediated antigen-specific immunity and a strategy for AAV vaccine dose reduction through AAV-extracellular vesicle association. Mol Ther Methods Clin Dev 2024; 32:101358. [PMID: 39559560 PMCID: PMC11570487 DOI: 10.1016/j.omtm.2024.101358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024]
Abstract
We previously showed therapeutic advantages of using a capsid-modified and encoded antigen-optimized AAV-based cancer vaccine to initiate strong antigen-specific immune responses and increase survival in a syngeneic mouse model of melanoma. In this study, we further explore AAV vaccine dose reduction and possible mechanisms of the immune response. Immunization with extracellular vesicle (EV)-associated AAV6-S663V encoded ovalbumin (OVA) or tyrosinase-related protein 1 (TRP-1) induced significantly higher levels of antigen-specific CD8+ T cells compared with standard AAV in mice. Importantly, a higher number of specific CD8+ T cells was achieved with EV-AAV several logs lower than optAAV-based doses. EV-optAAV-OVA was used in a dose 100 times lower, and EV-optTRP-1 10 times lower than optOVA and optTRP-1 correspondingly. Our data suggest that significant dose reduction for optimized AAV-based vaccines is possible without sacrificing efficiency. In addition, we studied the role of conventional type 1 dendritic cells (cDC1) in optimized AAV-based immunization using a C57BL/6-Irf8em1Kmm (Irf8 + 32-/-) mouse model lacking cDC1. Interestingly, we found that cDC1 are not essential for conveying effector T cell responses to AAV-encoded tumor antigens.
Collapse
Affiliation(s)
- Ester Molina
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Marcos Tejero
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | | | - Hisae Kuoch
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Colin Caine
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Karina Krotova
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Michael Paulaitis
- Department of Ophthalmology, Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - George Aslanidi
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 5455, USA
| |
Collapse
|
2
|
Kota N, Gonzalez DD, Liu HC, Viswanath D, Vander Pol R, Wood A, Di Trani N, Chua CYX, Grattoni A. Prophylactic and therapeutic cancer vaccine with continuous localized immunomodulation. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 62:102776. [PMID: 39102973 DOI: 10.1016/j.nano.2024.102776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/13/2024] [Accepted: 07/28/2024] [Indexed: 08/07/2024]
Abstract
Selective in vivo immune cell manipulation offers a promising strategy for cancer vaccines. In this context, spatiotemporal control over recruitment of specific cells, and their direct exposure to appropriate immunoadjuvants and antigens are key to effective cancer vaccines. We present an implantable 3D-printed cancer vaccine platform called the 'NanoLymph' that enables spatiotemporally-controlled recruitment and manipulation of immune cells in a subcutaneous site. Leveraging two reservoirs each for continuous immunoadjuvant release or antigen presentation, the NanoLymph attracts dendritic cells (DCs) on site and exposes them to tumor-associated antigens. Upon local antigen-specific activation, DCs are mobilized to initiate a systemic immune response. NanoLymph releasing granulocyte-macrophage colony-stimulating factor and CpG-oligodeoxynucleotides with irradiated whole cell tumor lysate inhibited tumor growth of B16F10 murine melanoma in a prophylactic and therapeutic vaccine setting. Overall, this study presents the NanoLymph as a versatile cancer vaccine development platform with replenishable and controlled local release of antigens and immunoadjuvants.
Collapse
Affiliation(s)
- Nikitha Kota
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Department of Biosciences, Rice University, Houston, TX, USA
| | | | - Hsuan-Chen Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Dixita Viswanath
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Robin Vander Pol
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Anthony Wood
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Nicola Di Trani
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | | | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Department of Surgery, Houston Methodist Hospital, Houston, TX, USA; Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
3
|
Daugherty-Lopès A, Pérez-Guijarro E, Gopalan V, Rappaport J, Chen Q, Huang A, Lam KC, Chin S, Ebersole J, Wu E, Needle GA, Church I, Kyriakopoulos G, Xie S, Zhao Y, Gruen C, Sassano A, Araya RE, Thorkelsson A, Smith C, Lee MP, Hannenhalli S, Day CP, Merlino G, Goldszmid RS. IMMUNE AND MOLECULAR CORRELATES OF RESPONSE TO IMMUNOTHERAPY REVEALED BY BRAIN-METASTATIC MELANOMA MODELS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609785. [PMID: 39372744 PMCID: PMC11451731 DOI: 10.1101/2024.08.26.609785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Despite the promising results of immune checkpoint blockade (ICB) therapy, outcomes for patients with brain metastasis (BrM) remain poor. Identifying resistance mechanisms has been hindered by limited access to patient samples and relevant preclinical models. Here, we developed two mouse melanoma BrM models that recapitulate the disparate responses to ICB seen in patients. We demonstrate that these models capture the cellular and molecular complexity of human disease and reveal key factors shaping the tumor microenvironment and influencing ICB response. BR1-responsive tumor cells express inflammatory programs that polarize microglia into reactive states, eliciting robust T cell recruitment. In contrast, BR3-resistant melanoma cells are enriched in neurological programs and exploit tolerance mechanisms to maintain microglia homeostasis and limit T cell infiltration. In humans, BR1 and BR3 expression signatures correlate positively or negatively with T cell infiltration and BrM patient outcomes, respectively. Our study provides clinically relevant models and uncovers mechanistic insights into BrM ICB responses, offering potential biomarkers and therapeutic targets to improve therapy efficacy.
Collapse
Affiliation(s)
- Amélie Daugherty-Lopès
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Eva Pérez-Guijarro
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Vishaka Gopalan
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jessica Rappaport
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Quanyi Chen
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Kelly Government Solutions, Bethesda, MD, USA
| | - April Huang
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Kelly Government Solutions, Bethesda, MD, USA
| | - Khiem C. Lam
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Sung Chin
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21701, USA
| | - Jessica Ebersole
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21701, USA
| | - Emily Wu
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Gabriel A. Needle
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Isabella Church
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - George Kyriakopoulos
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Shaojun Xie
- CCR-SF Bioinformatics Team, Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Yongmei Zhao
- CCR-SF Bioinformatics Team, Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Charli Gruen
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Antonella Sassano
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Romina E. Araya
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Andres Thorkelsson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Cari Smith
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21701, USA
| | - Maxwell P. Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Romina S. Goldszmid
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
4
|
Liao Z, Yao Y, Dong B, Le Y, Luo L, Miao F, Jiang S, Lei T. Involvement of interferon γ-producing mast cells in immune responses against melanocytes in vitiligo requires MrgX2 activation. Chin Med J (Engl) 2024:00029330-990000000-01263. [PMID: 39344472 DOI: 10.1097/cm9.0000000000003173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Increasing evidence indicates that oxidative stress and interferon γ (IFNγ)-driven cellular immune responses are responsible for the pathogenesis of vitiligo. However, the connection between oxidative stress and the local production of IFNγ in early vitiligo remains unexplored. The aim of this study was to identify the mechanism underlying the production of IFNγ by mast cells and its impact on vitiligo pathogenesis. METHODS Skin specimens from the central, marginal, and perilesional skin areas of active vitiligo lesions were collected to characterize changes of mast cells, CD8+ T cells, and IFNγ-producing cells. Cell supernatants from hydrogen peroxide (H2O2)-treated keratinocytes (KCs) were harvested to measure levels of soluble stem cell factor (sSCF) and matrix metalloproteinase (MMP)-9. A murine vitiligo model was established using Mas-related G protein-coupled receptor-B2 (MrgB2, mouse ortholog of human MrgX2) conditional knockout (MrgB2-/-) mice to investigate IFNγ production and inflammatory cell infiltrations in tail skin following the challenge with tyrosinase-related protein (Tyrp)-2 180 peptide. Potential interactions between the Tyrp-2 180 peptide and MrgX2 were predicted using molecular docking. The siRNAs targeting MrgX2 and the calcineurin inhibitor FK506 were also used to examine the signaling pathways involved in mast cell activation. RESULTS IFNγ-producing mast cells were closely aligned with the recruitment of CD8+ T cells in the early phase of vitiligo skin. sSCF released by KCs through stress-enhanced MMP9-dependent proteolytic cleavage recruited mast cells into sites of inflamed skin (Perilesion vs. lesion, 13.00 ± 4.00/HPF vs. 26.60 ± 5.72/HPF, P <0.05). Moreover, IFNγ-producing mast cells were also observed in mouse tail skin following challenge with Tyrp-2 180 (0 h vs. 48 h post-recall, 0.00 ± 0.00/HPF vs. 3.80 ± 1.92/HPF, P <0.05). The IFNγ+ mast cell and CD8+ T cell counts were lower in the skin of MrgB2-/-mice than in those of wild-type mice (WT vs. KO 48 h post-recall, 4.20 ± 0.84/HPF vs. 0.80 ± 0.84/HPF, P <0.05). CONCLUSION Mast cells activated by MrgX2 serve as a local IFNγ producer that bridges between innate and adaptive immune responses against MCs in early vitiligo. Targeting MrgX2-mediated mast cell activation may represent a new strategy for treating vitiligo.
Collapse
Affiliation(s)
- Zhikai Liao
- Department of Dermatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Trehan R, Zhu XB, Huang P, Wang X, Soliman M, Strepay D, Nur A, Kedei N, Arhin M, Ghabra S, Rodríguez-Matos F, Benmebarek MR, Ma C, Korangy F, Greten TF. A Paradoxical Tumor Antigen Specific Response in the Liver. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.614002. [PMID: 39372792 PMCID: PMC11451677 DOI: 10.1101/2024.09.19.614002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Functional tumor-specific CD8+ T cells are essential for an effective anti-tumor immune response and the efficacy of immune checkpoint inhibitor therapy. In comparison to other organ sites, we found higher numbers of tumor-specific CD8+ T cells in primary, metastatic liver tumors in murine tumor models. Despite their abundance, CD8+ T cells in the liver displayed an exhausted phenotype. Depletion of CD8+ T cells showed that liver tumor-reactive CD8+ T failed to control liver tumors but was effective against subcutaneous tumors. Similarly, analysis of single-cell RNA sequencing data from patients showed a higher frequency of exhausted tumor-reactive CD8+ T cells in liver metastasis compared to paired primary colon cancer. High-dimensional, multi-omic analysis combining proteomic CODEX and scRNA-seq data revealed enriched interaction of SPP1+ macrophages and CD8+ tumor-reactive T cells in profibrotic, alpha-SMA rich regions in the liver. Liver tumors grew less in Spp1-/- mice and the tumor-specific CD8+ T cells were less exhausted. Differential pseudotime trajectory inference analysis revealed extrahepatic signaling promoting an intermediate cell (IC) population in the liver, characterized by co-expression of VISG4, CSF1R, CD163, TGF-βR, IL-6R, SPP1. scRNA-seq of a third data set of premetastatic adenocarcinoma showed that enrichment of this population may predict liver metastasis. Our data suggests a mechanism by which extrahepatic tumors facilitate the formation of liver metastasis by promoting an IC population inhibiting tumor-reactive CD8+ T cell function.
Collapse
Affiliation(s)
- Rajiv Trehan
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xiao Bin Zhu
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Patrick Huang
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xin Wang
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marlaine Soliman
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dillon Strepay
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Amran Nur
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Noemi Kedei
- Collaborative Protein Technology Resource, OSTR, Office of the Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Martin Arhin
- Neurosurgery Unit for Pituitary and Inheritable Diseases, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Shadin Ghabra
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Francisco Rodríguez-Matos
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mohamed-Reda Benmebarek
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chi Ma
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Firouzeh Korangy
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Senior author
| | - Tim F. Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- NCI CCR Liver Cancer Program, National Institutes of Health, Bethesda, MD, USA
- Senior author
| |
Collapse
|
6
|
Pearson JRD, Puig-Saenz C, Thomas JE, Hardowar LD, Ahmad M, Wainwright LC, McVicar AM, Brentville VA, Tinsley CJ, Pockley AG, Durrant LG, McArdle SEB. TRP-2 / gp100 DNA vaccine and PD-1 checkpoint blockade combination for the treatment of intracranial tumors. Cancer Immunol Immunother 2024; 73:178. [PMID: 38954031 PMCID: PMC11219641 DOI: 10.1007/s00262-024-03770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024]
Abstract
Intracranial tumors present a significant therapeutic challenge due to their physiological location. Immunotherapy presents an attractive method for targeting these intracranial tumors due to relatively low toxicity and tumor specificity. Here we show that SCIB1, a TRP-2 and gp100 directed ImmunoBody® DNA vaccine, generates a strong TRP-2 specific immune response, as demonstrated by the high number of TRP2-specific IFNγ spots produced and the detection of a significant number of pentamer positive T cells in the spleen of vaccinated mice. Furthermore, vaccine-induced T cells were able to recognize and kill B16HHDII/DR1 cells after a short in vitro culture. Having found that glioblastoma multiforme (GBM) expresses significant levels of PD-L1 and IDO1, with PD-L1 correlating with poorer survival in patients with the mesenchymal subtype of GBM, we decided to combine SCIB1 ImmunoBody® with PD-1 immune checkpoint blockade to treat mice harboring intracranial tumors expressing TRP-2 and gp100. Time-to-death was significantly prolonged, and this correlated with increased CD4+ and CD8+ T cell infiltration in the tissue microenvironment (TME). However, in addition to PD-L1 and IDO, the GBM TME was found to contain a significant number of immunoregulatory T (Treg) cell-associated transcripts, and the presence of such cells is likely to significantly affect clinical outcome unless also tackled.
Collapse
Affiliation(s)
- Joshua R D Pearson
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Carles Puig-Saenz
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Jubini E Thomas
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Lydia D Hardowar
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Murrium Ahmad
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Louise C Wainwright
- Bioscience Support Facility, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Adam M McVicar
- Bioscience Support Facility, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Victoria A Brentville
- Scancell Ltd, Unit 202, Bellhouse Building, Oxford Science Park, Sanders Road, Oxford, OX4 4GA, UK
| | - Chris J Tinsley
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - A Graham Pockley
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Lindy G Durrant
- Scancell Ltd, Unit 202, Bellhouse Building, Oxford Science Park, Sanders Road, Oxford, OX4 4GA, UK
| | | |
Collapse
|
7
|
Song K, Pun SH. Design and Evaluation of Synthetic Delivery Formulations for Peptide-Based Cancer Vaccines. BME FRONTIERS 2024; 5:0038. [PMID: 38515636 PMCID: PMC10956738 DOI: 10.34133/bmef.0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/09/2024] [Indexed: 03/23/2024] Open
Abstract
With the recent advances in neoantigen identification, peptide-based cancer vaccines offer substantial potential in the field of immunotherapy. However, rapid clearance, low immunogenicity, and insufficient antigen-presenting cell (APC) uptake limit the efficacy of peptide-based cancer vaccines. This review explores the barriers hindering vaccine efficiency, highlights recent advancements in synthetic delivery systems, and features strategies for the key delivery steps of lymph node (LN) drainage, APC delivery, cross-presentation strategies, and adjuvant incorporation. This paper also discusses the design of preclinical studies evaluating vaccine efficiency, including vaccine administration routes and murine tumor models.
Collapse
Affiliation(s)
- Kefan Song
- Department of Bioengineering, University of Washington, USA
| | - Suzie H Pun
- Department of Bioengineering, University of Washington, USA
- Molecular Engineering & Sciences Institute, University of Washington, USA
| |
Collapse
|
8
|
Balasco N, Tagliamonte M, Buonaguro L, Vitagliano L, Paladino A. Structural and Dynamic-Based Characterization of the Recognition Patterns of E7 and TRP-2 Epitopes by MHC Class I Receptors through Computational Approaches. Int J Mol Sci 2024; 25:1384. [PMID: 38338663 PMCID: PMC10855917 DOI: 10.3390/ijms25031384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
A detailed comprehension of MHC-epitope recognition is essential for the design and development of new antigens that could be effectively used in immunotherapy. Yet, the high variability of the peptide together with the large abundance of MHC variants binding makes the process highly specific and large-scale characterizations extremely challenging by standard experimental techniques. Taking advantage of the striking predictive accuracy of AlphaFold, we report a structural and dynamic-based strategy to gain insights into the molecular basis that drives the recognition and interaction of MHC class I in the immune response triggered by pathogens and/or tumor-derived peptides. Here, we investigated at the atomic level the recognition of E7 and TRP-2 epitopes to their known receptors, thus offering a structural explanation for the different binding preferences of the studied receptors for specific residues in certain positions of the antigen sequences. Moreover, our analysis provides clues on the determinants that dictate the affinity of the same epitope with different receptors. Collectively, the data here presented indicate the reliability of the approach that can be straightforwardly extended to a large number of related systems.
Collapse
Affiliation(s)
- Nicole Balasco
- Institute of Molecular Biology and Pathology IBPM-CNR c/o Department Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| | - Maria Tagliamonte
- Immunological Models Lab, Istituto Nazionale Tumori—Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)—“Fond. G. Pascale”, Via Mariano Semmola 53, 80131 Napoli, Italy; (M.T.); (L.B.)
| | - Luigi Buonaguro
- Immunological Models Lab, Istituto Nazionale Tumori—Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)—“Fond. G. Pascale”, Via Mariano Semmola 53, 80131 Napoli, Italy; (M.T.); (L.B.)
| | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging IBB-CNR, Via Pietro Castellino 111, 80131 Napoli, Italy;
| | - Antonella Paladino
- Institute of Biostructures and Bioimaging IBB-CNR, Via Pietro Castellino 111, 80131 Napoli, Italy;
| |
Collapse
|
9
|
Ponomarev AV, Shubina IZ, Sokolova ZA, Baryshnikova MA, Kosorukov VS. Transplantable Murine Tumors in the Studies of Peptide Antitumor Vaccines. Oncol Rev 2024; 17:12189. [PMID: 38260723 PMCID: PMC10800450 DOI: 10.3389/or.2023.12189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Numerous studies have shown that antitumor vaccines based on synthetic peptides are safe and can induce both CD8+ and CD4+ tumor-specific T cell responses. However, clinical results are still scarce, and such approach to antitumor treatment has not gained a wide implication, yet. Recently, particular advances have been achieved due to tumor sequencing and the search for immunogenic neoantigens caused by mutations. One of the most important issues for peptide vaccines, along with the choice of optimal adjuvants and vaccination regimens, is the search for effective target antigens. Extensive studies of peptide vaccines, including those on murine models, are required to reveal the effective vaccine constructs. The review presents transplantable murine tumors with the detected peptides that showed antitumor efficacy as a vaccine compound.
Collapse
|
10
|
Ma S, Huis in't Veld RV, Hao Y, Gu Z, Rich C, Gelmi MC, Mulder AA, van Veelen PA, Vu TKH, van Hall T, Ossendorp FA, Jager MJ. Tumor Pigmentation Does Not Affect Light-Activated Belzupacap Sarotalocan Treatment but Influences Macrophage Polarization in a Murine Melanoma Model. Invest Ophthalmol Vis Sci 2024; 65:42. [PMID: 38271187 PMCID: PMC10829805 DOI: 10.1167/iovs.65.1.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/21/2023] [Indexed: 01/27/2024] Open
Abstract
Purpose Pigmentation in uveal melanoma is associated with increased malignancy and is known as a barrier for photodynamic therapy. We investigated the role of pigmentation in tumor behavior and the response to light-activated Belzupacap sarotalocan (Bel-sar) treatment in a pigmented (wild type) and nonpigmented (tyrosinase knock-out [TYR knock-out]) cell line in vitro and in a murine model. Methods The B16F10 (TYR knock-out) was developed using CRISPR/Cas9. After the treatment with light-activated Bel-sar, cytotoxicity and exposure of damage-associated molecular patterns (DAMPs) were measured by flow cytometry. Treated tumor cells were co-cultured with bone marrow-derived macrophages (BMDMs) and dendritic cells (DCs) to assess phagocytosis and activation. Both cell lines were injected subcutaneously in syngeneic C57BL/6 mice. Results Knock-out of the tyrosinase gene in B16F10 led to loss of pigmentation and immature melanosomes. Pigmented tumors contained more M1 and fewer M2 macrophages compared with amelanotic tumors. Bel-sar treatment induced near complete cell death, accompanied with enhanced exposure of DAMPs in both cell lines, resulting in enhanced phagocytosis of BMDMs and maturation of DCs. Bel-sar treatment induced a shift to M1 macrophages and delayed tumor growth in both in vivo tumor models. Following treatment, especially the pigmented tumors and their draining lymph nodes contained IFN-gamma positive CD8+T cells. Conclusions Pigmentation influenced the type of infiltrating macrophages in the tumor, with more M1 macrophages in pigmented tumors. Belzupacap sarotalocan treatment induced immunogenic cell death and tumor growth delay in pigmented as well as in nonpigmented models and stimulated M1 macrophage influx in both models.
Collapse
Affiliation(s)
- Sen Ma
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Ruben V. Huis in't Veld
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Department of Radiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Yang Hao
- Department of Radiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, China
| | - Zili Gu
- Department of Radiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Cadmus Rich
- Aura Biosciences, Inc., Boston, Massachusetts, United States
| | - Maria Chiara Gelmi
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Aat A. Mulder
- Department of Electron Microscopy, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Peter A. van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - T. Khanh H. Vu
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Oncology Institute, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Ferry A. Ossendorp
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| |
Collapse
|
11
|
Chen J, Xu Y, Zhou M, Xu S, Varley AJ, Golubovic A, Lu RXZ, Wang KC, Yeganeh M, Vosoughi D, Li B. Combinatorial design of ionizable lipid nanoparticles for muscle-selective mRNA delivery with minimized off-target effects. Proc Natl Acad Sci U S A 2023; 120:e2309472120. [PMID: 38060560 PMCID: PMC10723144 DOI: 10.1073/pnas.2309472120] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/30/2023] [Indexed: 12/17/2023] Open
Abstract
Ionizable lipid nanoparticles (LNPs) pivotal to the success of COVID-19 mRNA (messenger RNA) vaccines hold substantial promise for expanding the landscape of mRNA-based therapies. Nevertheless, the risk of mRNA delivery to off-target tissues highlights the necessity for LNPs with enhanced tissue selectivity. The intricate nature of biological systems and inadequate knowledge of lipid structure-activity relationships emphasize the significance of high-throughput methods to produce chemically diverse lipid libraries for mRNA delivery screening. Here, we introduce a streamlined approach for the rapid design and synthesis of combinatorial libraries of biodegradable ionizable lipids. This led to the identification of iso-A11B5C1, an ionizable lipid uniquely apt for muscle-specific mRNA delivery. It manifested high transfection efficiencies in muscle tissues, while significantly diminishing off-targeting in organs like the liver and spleen. Moreover, iso-A11B5C1 also exhibited reduced mRNA transfection potency in lymph nodes and antigen-presenting cells, prompting investigation into the influence of direct immune cell transfection via LNPs on mRNA vaccine effectiveness. In comparison with SM-102, while iso-A11B5C1's limited immune transfection attenuated its ability to elicit humoral immunity, it remained highly effective in triggering cellular immune responses after intramuscular administration, which is further corroborated by its strong therapeutic performance as cancer vaccine in a melanoma model. Collectively, our study not only enriches the high-throughput toolkit for generating tissue-specific ionizable lipids but also encourages a reassessment of prevailing paradigms in mRNA vaccine design. This study encourages rethinking of mRNA vaccine design principles, suggesting that achieving high immune cell transfection might not be the sole criterion for developing effective mRNA vaccines.
Collapse
Affiliation(s)
- Jingan Chen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ONM5S 3G9, Canada
| | - Yue Xu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Muye Zhou
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Shufen Xu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Andrew James Varley
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Alex Golubovic
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Rick Xing Ze Lu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Kevin Chang Wang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
| | - Mina Yeganeh
- Institute of Medical Science, University of Toronto, Toronto, ONM5G 1L7, Canada
| | - Daniel Vosoughi
- Institute of Medical Science, University of Toronto, Toronto, ONM5G 1L7, Canada
- Latner Thoracic Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ONM5G 2C4, Canada
| | - Bowen Li
- Institute of Biomedical Engineering, University of Toronto, Toronto, ONM5S 3G9, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ONM5S 3M2, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ONM5G 2C1, Canada
| |
Collapse
|
12
|
Bao Y, Qiao Y, Choi JE, Zhang Y, Mannan R, Cheng C, He T, Zheng Y, Yu J, Gondal M, Cruz G, Grove S, Cao X, Su F, Wang R, Chang Y, Kryczek I, Cieslik M, Green MD, Zou W, Chinnaiyan AM. Targeting the lipid kinase PIKfyve upregulates surface expression of MHC class I to augment cancer immunotherapy. Proc Natl Acad Sci U S A 2023; 120:e2314416120. [PMID: 38011559 PMCID: PMC10710078 DOI: 10.1073/pnas.2314416120] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/30/2023] [Indexed: 11/29/2023] Open
Abstract
Despite the remarkable clinical success of immunotherapies in a subset of cancer patients, many fail to respond to treatment and exhibit resistance. Here, we found that genetic or pharmacologic inhibition of the lipid kinase PIKfyve, a regulator of autophagic flux and lysosomal biogenesis, upregulated surface expression of major histocompatibility complex class I (MHC-I) in cancer cells via impairing autophagic flux, resulting in enhanced cancer cell killing mediated by CD8+ T cells. Genetic depletion or pharmacologic inhibition of PIKfyve elevated tumor-specific MHC-I surface expression, increased intratumoral functional CD8+ T cells, and slowed tumor progression in multiple syngeneic mouse models. Importantly, enhanced antitumor responses by Pikfyve-depletion were CD8+ T cell- and MHC-I-dependent, as CD8+ T cell depletion or B2m knockout rescued tumor growth. Furthermore, PIKfyve inhibition improved response to immune checkpoint blockade (ICB), adoptive cell therapy, and a therapeutic vaccine. High expression of PIKFYVE was also predictive of poor response to ICB and prognostic of poor survival in ICB-treated cohorts. Collectively, our findings show that targeting PIKfyve enhances immunotherapies by elevating surface expression of MHC-I in cancer cells, and PIKfyve inhibitors have potential as agents to increase immunotherapy response in cancer patients.
Collapse
Affiliation(s)
- Yi Bao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI48109
| | - Jae Eun Choi
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Rahul Mannan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Caleb Cheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Tongchen He
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Yang Zheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Jiali Yu
- Department of Surgery, University of Michigan, Ann Arbor, MI48109
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI48109
| | - Mahnoor Gondal
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI48109
| | - Gabriel Cruz
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
| | - Sara Grove
- Department of Surgery, University of Michigan, Ann Arbor, MI48109
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI48109
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Fengyun Su
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Rui Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Yu Chang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
| | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, MI48109
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI48109
| | - Marcin Cieslik
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI48109
| | - Michael D. Green
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI48109
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI48109
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI48109
| | - Weiping Zou
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI48109
- Department of Surgery, University of Michigan, Ann Arbor, MI48109
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI48109
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI48109
- Department of Pathology, University of Michigan, Ann Arbor, MI48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI48109
- HHMI, University of Michigan, Ann Arbor, MI48109
- Department of Urology, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
13
|
Mizuno S, Chuma Y, Shibuya Y, Horibata S, Baba T, Yokokawa E, Matsuo K. Culture filtrate proteins from BCG act as adjuvants for cytotoxic T lymphocyte induction. Front Immunol 2023; 14:1271228. [PMID: 37928526 PMCID: PMC10622798 DOI: 10.3389/fimmu.2023.1271228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Mycobacterium bovis bacilli Calmette-Guerin (BCG) is a licensed vaccine against tuberculosis. It requires attenuated live bacteria to be effective, possibly because actively secreted proteins play a critical role in inducing anti-tuberculosis immunity. BCG also functions as an effective adjuvant. Moreover, the effects of BCG components as adjuvants are not important as those of attenuated live BCG, which is used in cancer immunotherapy. However, the BCG secreted proteins have not been paid attention in anticancer immunity. To understand mycobacterial secreted proteins' function, we investigate immune responses to BCG culture filtrate proteins (CFP). Here, CFP strongly induce both antigen-specific CD4+ T cells and specific CD8+ T cells, which may be functional cytotoxic T lymphocytes (CTLs). In this study, we clearly demonstrate that CFP acts as an adjuvant for CTL induction against specific co-administered proteins and propose CFP as a new protein adjuvant. The CTL response shows potent anticancer effects in mice. These findings could provide insight into the contribution of mycobacterial secreted proteins in both anticancer and antimycobacterial immunity.
Collapse
Affiliation(s)
- Satoru Mizuno
- Research and Development Department, Japan BCG Laboratory, Kiyose, Tokyo, Japan
| | - Yasushi Chuma
- Research and Development Department, Japan BCG Laboratory, Kiyose, Tokyo, Japan
| | - Yukihiro Shibuya
- Research and Development Department, Japan BCG Laboratory, Kiyose, Tokyo, Japan
| | - Shigeo Horibata
- Research and Development Department, Japan BCG Laboratory, Kiyose, Tokyo, Japan
| | - Tomoe Baba
- Research and Development Department, Japan BCG Laboratory, Kiyose, Tokyo, Japan
| | - Emi Yokokawa
- Research and Development Department, Japan BCG Laboratory, Kiyose, Tokyo, Japan
| | - Kazuhiro Matsuo
- Research and Development Department, Japan BCG Laboratory, Kiyose, Tokyo, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
14
|
Inamdar S, Suresh AP, Mangal JL, Ng ND, Sundem A, Behbahani HS, Rubino TE, Shi X, Loa ST, Yaron JR, Hitosugi T, Green M, Gu H, Curtis M, Acharya AP. Succinate based polymers drive immunometabolism in dendritic cells to generate cancer immunotherapy. J Control Release 2023; 358:541-554. [PMID: 37182805 PMCID: PMC10324539 DOI: 10.1016/j.jconrel.2023.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023]
Abstract
Boosting the metabolism of immune cells while restricting cancer cell metabolism is challenging. Herein, we report that using biomaterials for the controlled delivery of succinate metabolite to phagocytic immune cells activates them and modulates their metabolism in the presence of metabolic inhibitors. In young immunocompetent mice, polymeric microparticles, with succinate incorporated in the backbone, induced strong pro-inflammatory anti-melanoma responses. Administration of poly(ethylene succinate) (PES MP)-based vaccines and glutaminase inhibitor to young immunocompetent mice with aggressive and large, established B16F10 melanoma tumors increased their survival three-fold, a result of increased cytotoxic T cells expressing RORγT (Tc17). Mechanistically, PES MPs directly modulate glutamine and glutamate metabolism, upregulate succinate receptor SUCNR1, activate antigen presenting cells through and HIF-1alpha, TNFa and TSLP-signaling pathways, and are dependent on alpha-ketoglutarate dehydrogenase for their activity, which demonstrates correlation of succinate delivery and these pathways. Overall, our findings suggest that immunometabolism-modifying PES MP strategies provide an approach for developing robust cancer immunotherapies.
Collapse
Affiliation(s)
- Sahil Inamdar
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA; Department of Immunology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Abhirami P Suresh
- Biological Design, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA
| | - Joslyn L Mangal
- Biological Design, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA
| | - Nathan D Ng
- Molecular Biosciences and Biotechnology, The College of Liberal Arts and Sciences, Arizona State University, Tempe, AZ 85281, USA
| | - Alison Sundem
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA
| | - Hoda Shokrollahzadeh Behbahani
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA
| | - Thomas E Rubino
- Department of Cancer Biology, Mayo Clinic, Scottsdale, AZ 85259, USA; College of Medicine and Science, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Xiaojian Shi
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Sharon T Loa
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jordan R Yaron
- Biological Design, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA
| | - Taro Hitosugi
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew Green
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA; Biomedical Engineering, School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85281, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Marion Curtis
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Cancer Biology, Mayo Clinic, Scottsdale, AZ 85259, USA; College of Medicine and Science, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Abhinav P Acharya
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA; Biological Design, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA; Biomedical Engineering, School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85281, USA; Materials Science and Engineering, School for the Engineering of Matter, Transport, and energy, Arizona State University, Tempe, AZ 85281, USA; Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ 85281, USA; Biodesign Center for Biomaterials Innovation and Translation.
| |
Collapse
|
15
|
Reil KA, Tsuji S, Molina E, Nelson KL, McGuire KL, Giacalone MJ. Intralesional administration of VAX014 facilitates in situ immunization and potentiates immune checkpoint blockade in immunologically cold tumors. J Immunother Cancer 2023; 11:e006749. [PMID: 37290924 PMCID: PMC10254596 DOI: 10.1136/jitc-2023-006749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND Immunologically cold tumors with an 'immune desert' phenotype lack tumor-infiltrating lymphocytes (TILs) and are typically impervious to systemic immune checkpoint blockade (ICB). Intratumoral treatment of tumors with immunomodulatory agents can promote local tumor inflammation leading to improved T cell responses in injected tumors. Addition of systemic ICB increases response frequency and immune-mediated clearance of injected and distal non-injected lesions, and this promising approach is being widely investigated clinically. In this work, we evaluate and characterize the local and systemic antitumor immunotherapeutic activity of VAX014, a novel non-viral targeted oncolytic agent based on recombinant bacterial minicells, following intratumoral administration and in combination with systemic ICB. METHODS The immunotherapeutic activity of VAX014 following weekly intratumoral administration was investigated in multiple preclinical tumor models with B16F10 murine melanoma serving as the primary model for evaluation of immune desert tumors. Mice bearing a single intradermal tumor were used to evaluate tumor response and overall survival (OS), assess changes in immune cell populations, and explore global changes to immunotranscriptomes of injected tumors. Mice bearing bilateral intradermal tumors were then used to evaluate non-injected tumors for changes in TIL populations and phenotypes, compare immunotranscriptomes across treatment groups, and assess distal non-injected tumor response in the context of monotherapy or in combination with ICB. RESULTS VAX014 demonstrated strong immune-mediated tumor clearance of injected tumors coinciding with significantly elevated CD8+ TILs and upregulation of multiple immune pathways essential for antitumor immune responses. Modest activity against distal non-injected immune desert tumors was observed despite elevated levels of systemic antitumor lymphocytes. Combination with systemic CTLA-4 blockade improved survival and elevated TILs but did not improve clearance rates of non-injected tumors. Immunotranscriptomes of non-injected tumors from this treatment combination group exhibited upregulation of multiple immune pathways but also identified upregulation of PD-1. Further addition of systemic PD-1 blockade led to rapid clearance of non-injected tumors, enhanced OS, and provided durable protective immunological memory. CONCLUSIONS Intratumoral administration of VAX014 stimulates local immune activation and robust systemic antitumor lymphocytic responses. Combination with systemic ICB deepens systemic antitumor responses to mediate clearance of injected and distal non-injected tumors.
Collapse
Affiliation(s)
- Katherine A Reil
- Vaxiion Therapeutics, San Diego, California, USA
- Biology, San Diego State University College of Sciences, San Diego, California, USA
| | - Shingo Tsuji
- Vaxiion Therapeutics, San Diego, California, USA
| | - Elsa Molina
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| | - Kinsey L Nelson
- Vaxiion Therapeutics, San Diego, California, USA
- Biology, San Diego State University College of Sciences, San Diego, California, USA
| | - Kathleen L McGuire
- Biology, San Diego State University College of Sciences, San Diego, California, USA
| | | |
Collapse
|
16
|
Jiang H, Shin DH, Yi Y, Fan X, Gumin J, He J, Gillard AG, Lang FF, Gomez-Manzano C, Fueyo J. Adjuvant Therapy with Oncolytic Adenovirus Delta-24-RGDOX After Intratumoral Adoptive T-cell Therapy Promotes Antigen Spread to Sustain Systemic Antitumor Immunity. CANCER RESEARCH COMMUNICATIONS 2023; 3:1118-1131. [PMID: 37379361 PMCID: PMC10295804 DOI: 10.1158/2767-9764.crc-23-0054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 06/30/2023]
Abstract
Cancer cell heterogeneity and immunosuppressive tumor microenvironment (TME) pose a challenge in treating solid tumors with adoptive cell therapies targeting limited tumor-associated antigens (TAA), such as chimeric antigen receptor T-cell therapy. We hypothesize that oncolytic adenovirus Delta-24-RGDOX activates the TME and promote antigen spread to potentiate the abscopal effect of adoptive TAA-targeting T cells in localized intratumoral treatment. Herein, we used C57BL/6 mouse models with disseminated tumors derived from B16 melanoma cell lines to assess therapeutic effects and antitumor immunity. gp100-specific pmel-1 or ovalbumin (OVA)-specific OT-I T cells were injected into the first subcutaneous tumor, followed by three injections of Delta-24-RGDOX. We found TAA-targeting T cells injected into one subcutaneous tumor showed tumor tropism. Delta-24-RGDOX sustained the systemic tumor regression mediated by the T cells, leading to improved survival rate. Further analysis revealed that, in mice with disseminated B16-OVA tumors, Delta-24-RGDOX increased CD8+ leukocyte density within treated and untreated tumors. Importantly, Delta-24-RGDOX significantly reduced the immunosuppression of endogenous OVA-specific CTLs while increasing that of CD8+ leukocytes and, to a lesser extent, adoptive pmel-1 T cells. Consequently, Delta-24-RGDOX drastically increased the density of the OVA-specific CTLs in both tumors, and the combination synergistically enhanced the effect. Consistently, the splenocytes from the combination group showed a significantly stronger response against other TAAs (OVA and TRP2) than gp100, resulted in higher activity against tumor cells. Therefore, our data demonstrate that, as an adjuvant therapy followed TAA-targeting T cells in localized treatment, Delta-24-RGDOX activates TME and promotes antigen spread, leading to efficacious systemic antitumor immunity to overcome tumor relapse. Significance Adjuvant therapy with oncolytic viruses promotes antigen spread to potentiate localized intratumoral adoptive T-cell therapy with limited TAA targets, leading to sustainable systemic antitumor immunity to overcome tumor relapse.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dong Ho Shin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yanhua Yi
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xuejun Fan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joy Gumin
- Department of Neuro-Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiasen He
- Pediatric division, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Andrew G. Gillard
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frederick F. Lang
- Department of Neuro-Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
17
|
García-Ferreras R, Osuna-Pérez J, Ramírez-Santiago G, Méndez-Pérez A, Acosta-Moreno AM, Del Campo L, Gómez-Sánchez MJ, Iborra M, Herrero-Fernández B, González-Granado JM, Sánchez-Madrid F, Carrasco YR, Boya P, Martínez-Martín N, Veiga E. Bacteria-instructed B cells cross-prime naïve CD8 + T cells triggering effective cytotoxic responses. EMBO Rep 2023:e56131. [PMID: 37184882 DOI: 10.15252/embr.202256131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 04/14/2023] [Accepted: 04/25/2023] [Indexed: 05/16/2023] Open
Abstract
In addition to triggering humoral responses, conventional B cells have been described in vitro to cross-present exogenous antigens activating naïve CD8+ T cells. Nevertheless, the way B cells capture these exogenous antigens and the physiological roles of B cell-mediated cross-presentation remain poorly explored. Here, we show that B cells capture bacteria by trans-phagocytosis from previously infected dendritic cells (DC) when they are in close contact. Bacterial encounter "instructs" the B cells to acquire antigen cross-presentation abilities, in a process that involves autophagy. Bacteria-instructed B cells, henceforth referred to as BacB cells, rapidly degrade phagocytosed bacteria, process bacterial antigens and cross-prime naïve CD8+ T cells which differentiate into specific cytotoxic cells that efficiently control bacterial infections. Moreover, a proof-of-concept experiment shows that BacB cells that have captured bacteria expressing tumor antigens could be useful as novel cellular immunotherapies against cancer.
Collapse
Affiliation(s)
- Raquel García-Ferreras
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Jesús Osuna-Pérez
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Guillermo Ramírez-Santiago
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Almudena Méndez-Pérez
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Andrés M Acosta-Moreno
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Lara Del Campo
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
- Departamento de Biología Celular, Facultad de Odontología, Universidad Complutense de Madrid, Madrid, Spain
| | - María J Gómez-Sánchez
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
- Department of Immunology, School of Medicine, Complutense University of Madrid, 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Marta Iborra
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Beatriz Herrero-Fernández
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - José M González-Granado
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Francisco Sánchez-Madrid
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, Madrid, Spain
| | - Yolanda R Carrasco
- Department of Immunology & Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Patricia Boya
- Department of Neuroscience, University of Fribourg, Fribourg, Switzerland
| | | | - Esteban Veiga
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| |
Collapse
|
18
|
Estrada J, Zhan J, Mitchell P, Werner J, Beltran PJ, DeVoss J, Qing J, Cooke KS. OncoVEX mGM-CSFexpands tumor antigen-specific CD8+ T-cell response in preclinical models. J Immunother Cancer 2023; 11:jitc-2022-006374. [PMID: 37164449 PMCID: PMC10173969 DOI: 10.1136/jitc-2022-006374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) have demonstrated clinical efficacy in advanced melanoma, but only a subset of patients with inflamed tumors are responsive. Talimogene laherparepvec (T-VEC), a modified herpes simplex virus type 1 (HSV-1) expressing granulocyte-macrophage colony-stimulating factor (GM-CSF), is a first-in-class oncolytic immunotherapy approved for the treatment of melanoma and has been shown to inflame the tumor microenvironment. To evaluate the potential and mechanisms of T-VEC to elicit systemic antitumor immunity and overcome resistance to checkpoint inhibitors in murine tumor models, OncoVEXmGM-CSF was developed similarly to T-VEC, except the human GM-CSF transgene was replaced with murine GM-CSF. Previous work had demonstrated that OncoVEXmGM-CSF generated systemic antitumor immunity dependent on CD8+ T cells in an immune checkpoint-sensitive tumor cell model. METHODS A novel B16F10 syngeneic tumor model with both HSV-1-permissive subcutaneous tumors and HSV-1-refractory experimental lung metastasis was used to study the local and systemic effects of OncoVEXmGM-CSF treatment alone or in combination with checkpoint inhibitors. RESULTS Intratumoral injection of OncoVEXmGM-CSF in combination with an anti-CTLA-4 or anti-PD-1 blocking antibody led to increased tumor growth inhibition, a reduction in the number of lung metastases, and prolonged animal survival. OncoVEXmGM-CSF induced both neoantigen-specific and tumor antigen-specific T-cell responses. Furthermore, cured mice from the combination treatment of OncoVEXmGM-CSF and anti-CTLA-4 antibody rejected tumor rechallenges. CONCLUSIONS These data support the concept that T-VEC and checkpoint inhibition may be an effective combination to treat patients with advanced melanoma.
Collapse
|
19
|
Tumor antigen-loaded AAV vaccine drives protective immunity in a melanoma animal model. Mol Ther Methods Clin Dev 2023; 28:301-311. [PMID: 36851984 PMCID: PMC9957711 DOI: 10.1016/j.omtm.2023.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/29/2023] [Indexed: 02/04/2023]
Abstract
We previously described therapeutic opportunities provided by capsid- and expression cassette-optimized adeno-associated virus serotype 6 (AAV6) vectors to suppress tumor growth in both solid and metastatic mouse models by using artificial ovalbumin (OVA) immunogen. In the current study, we further elucidated the mechanism of function of a novel AAV-based vaccine loaded with the melanoma tumor-associated antigens premelanosome protein gp100, tyrosinase (Tyr), tyrosinase-related protein 1 (TRP1), and dopachrome tautomerase (TRP2). We showed that the AAV6-based vaccine creates cellular and humoral antigen-specific responses, while antigen expression at the site of vaccine injection was temporal, and the clearance of antigen coincided with T cell infiltration. Our data revealed the superior protective immune response of optimized AAV6-TRP1 compared with other self-antigens in a disease-free mouse model. We further assessed the ability of AAV6-TRP1 to protect animals from metastatic spread in the lungs and to extend animal survival by inhibiting solid tumor growth. Flow cytometry-based analysis indicated significant infiltration of CD8+ T cells and natural killer (NK) cells in the tumor site, as well as changes in the polarization of intratumoral macrophages. Altogether, our data strongly support the use of optimized AAV vectors for cancer vaccine development.
Collapse
|
20
|
Irie H, Morita K, Matsuda M, Koizumi M, Mochizuki S. Tyrosinase-Related Protein2 Peptide with Replacement of N-Terminus Residue by Cysteine Binds to H-2K b and Induces Antigen-Specific Cytotoxic T Lymphocytes after Conjugation with CpG-DNA. Bioconjug Chem 2023; 34:433-442. [PMID: 36708315 DOI: 10.1021/acs.bioconjchem.2c00592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Recent studies have shown the potent efficacy of peptide-based vaccines for cancer immunotherapy. Immunological performance is optimized through the co-delivery of adjuvant and antigenic peptide molecules to antigen-presenting cells simultaneously. In our previous study, we showed that a conjugate consisting of 40-mer CpG-DNA and an antigenic ovalbumin peptide through disulfide bonding could efficiently induce ovalbumin-specific cytotoxic T lymphocyte (CTL) responses in vivo. In this study, based on the conjugation design, we prepared a conjugate consisting of 30-mer CpG-DNA (CpG30) and a cancer antigenic peptide of Tyrosinase-related protein 2 (TRP2180-188) using a cysteine residue attached at the N-terminus of TRP2180-188. However, the immunization of mice with this conjugate did not induce efficient TRP2180-188-specific immune responses. It was thought that the resultant peptide (10-mer) cleaved from the conjugate might be too long to fit into the H-2Kb molecule because the optimal length for binding to it is 8-9 amino acids. We newly designed a conjugate consisting of CpG30 and the C-TRP2181-188 peptide (9-mer), in which the N-terminal serine residue of TRP2180-188 is replaced by a cysteine. By adjusting the peptide length, we succeeded in inducing strong TRP2180-188 peptide-specific CTL activity upon immunization with the CpG30-C-TRP2181-188 conjugate. Furthermore, various CpG30-C-TRP2181-188 conjugates having other CpG-DNA sequences or cysteine analogues also induced the same level of CTL activity. Therefore, CpG-C-peptide conjugates prepared by replacement of the amino acid residue at the N-terminus with a cysteine residue could be a new and effective platform for peptide vaccines for targeting specific antigens of cancers and infectious diseases.
Collapse
Affiliation(s)
- Hitomi Irie
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka 808-0135, Japan
| | - Koji Morita
- Modality Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Miyu Matsuda
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka 808-0135, Japan
| | - Makoto Koizumi
- Modality Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Shinichi Mochizuki
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka 808-0135, Japan
| |
Collapse
|
21
|
Cho H, Binder J, Weeratna R, Dermyer M, Dai S, Boccia A, Li W, Li S, Jooss K, Merson J, Hollingsworth RE. Preclinical development of a vaccine-based immunotherapy regimen (VBIR) that induces potent and durable T cell responses to tumor-associated self-antigens. Cancer Immunol Immunother 2023; 72:287-300. [PMID: 35829790 PMCID: PMC10992523 DOI: 10.1007/s00262-022-03245-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 06/16/2022] [Indexed: 01/26/2023]
Abstract
The development of therapeutic cancer vaccines remains an active area, although previous approaches have yielded disappointing results. We have built on lessons from previous cancer vaccine approaches and immune checkpoint inhibitor research to develop VBIR, a vaccine-based immunotherapy regimen. Assessment of various technologies led to selection of a heterologous vaccine using chimpanzee adenovirus (AdC68) for priming followed by boosts with electroporation of DNA plasmid to deliver T cell antigens to the immune system. We found that priming with AdC68 rapidly activates and expands antigen-specific T cells and does not encounter pre-existing immunity as occurs with the use of a human adenovirus vaccine. The AdC68 vector does, however, induce new anti-virus immune responses, limiting its use for boosting. To circumvent this, boosting with DNA encoding the same antigens can be done repetitively to augment and maintain vaccine responses. Using mouse and monkey models, we found that the activation of both CD4 and CD8 T cells was amplified by combination with anti-CTLA-4 and anti-PD-1 antibodies. These antibodies were administered subcutaneously to target their distribution to vaccination sites and to reduce systemic exposure which may improve their safety. VBIR can break tolerance and activate T cells recognizing tumor-associated self-antigens. This activation lasts more than a year after completing treatment in monkeys, and inhibits tumor growth to a greater degree than is observed using the individual components in mouse cancer models. These results have encouraged the testing of this combination regimen in cancer patients with the aim of increasing responses beyond current therapies.
Collapse
Affiliation(s)
- Helen Cho
- Pfizer Cancer Vaccines and Immunotherapeutics, 10777 Science Center Drive, San Diego, CA, USA
- Samyang Holdings Corporation, 295 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Joe Binder
- Pfizer Cancer Vaccines and Immunotherapeutics, 10777 Science Center Drive, San Diego, CA, USA
| | - Risini Weeratna
- Pfizer Vaccines and Immunotherapeutics, 340 Terry Fox Drive, Suite 200, Ottawa, ON, Canada
- National Research Council of Canada, Ottawa, ON, Canada
| | - Michael Dermyer
- Pfizer Cancer Vaccines and Immunotherapeutics, 10777 Science Center Drive, San Diego, CA, USA
| | - Stanley Dai
- Pfizer Cancer Vaccines and Immunotherapeutics, 10777 Science Center Drive, San Diego, CA, USA
- Nektar Therapeutics, 455 Mission Bay Boulevard, South San Francisco, CA, 94158, USA
| | - Antionio Boccia
- Pfizer Cancer Vaccines and Immunotherapeutics, 10777 Science Center Drive, San Diego, CA, USA
| | - Wei Li
- Pfizer Cancer Vaccines and Immunotherapeutics, 10777 Science Center Drive, San Diego, CA, USA
| | - Shangjin Li
- Pfizer Cancer Vaccines and Immunotherapeutics, 10777 Science Center Drive, San Diego, CA, USA
| | - Karin Jooss
- Pfizer Cancer Vaccines and Immunotherapeutics, 10777 Science Center Drive, San Diego, CA, USA
- Gritstone Oncology Inc., 5858 Horton Street, Suite 210, Emeryville, CA, USA
| | - James Merson
- Pfizer Cancer Vaccines and Immunotherapeutics, 10777 Science Center Drive, San Diego, CA, USA
- Johnson & Johnson, 5000 Shoreline Ct., South San Francisco, CA, USA
| | - Robert E Hollingsworth
- Pfizer Cancer Vaccines and Immunotherapeutics, 10777 Science Center Drive, San Diego, CA, USA.
| |
Collapse
|
22
|
Facile repurposing of peptide-MHC-restricted antibodies for cancer immunotherapy. Nat Biotechnol 2023:10.1038/s41587-022-01567-w. [PMID: 36593402 DOI: 10.1038/s41587-022-01567-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 10/13/2022] [Indexed: 01/03/2023]
Abstract
Monoclonal antibodies (Abs) that recognize major histocompatability complex (MHC)-presented tumor antigens in a manner similar to T cell receptors (TCRs) have great potential as cancer immunotherapeutics. However, isolation of 'TCR-mimic' (TCRm) Abs is laborious because Abs have not evolved the structurally nuanced peptide-MHC restriction of αβ-TCRs. Here, we present a strategy for rapid isolation of highly peptide-specific and 'MHC-restricted' Abs by re-engineering preselected Abs that engage peptide-MHC in a manner structurally similar to that of conventional αβ-TCRs. We created structure-based libraries focused on the peptide-interacting residues of TCRm Ab complementarity-determining region (CDR) loops, and rapidly generated MHC-restricted Abs to both mouse and human tumor antigens that specifically killed target cells when formatted as IgG, bispecific T cell engager (BiTE) and chimeric antigen receptor-T (CAR-T). Crystallographic analysis of one selected pMHC-restricted Ab revealed highly peptide-specific recognition, validating the engineering strategy. This approach can yield tumor antigen-specific antibodies in several weeks, potentially enabling rapid clinical translation.
Collapse
|
23
|
Corogeanu D, Zaki K, Beavil AJ, Arnold JN, Diebold SS. Antibody conjugates for targeted delivery of Toll-like receptor 9 agonist to the tumor tissue. PLoS One 2023; 18:e0282831. [PMID: 36913398 PMCID: PMC10010539 DOI: 10.1371/journal.pone.0282831] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 02/23/2023] [Indexed: 03/14/2023] Open
Abstract
Imiquimod, a Toll-like receptor 7 (TLR7) agonist is routinely used for topical administration in basal cell carcinoma and stage zero melanoma. Similarly, the TLR agonist Bacillus Calmette-Guérin is used for the local treatment of bladder cancer and clinical trials showed treatment efficacy of intratumoral injections with TLR9 agonists. However, when administered systemically, endosomal TLR agonists cause adverse responses due to broad immune activation. Hence, strategies for targeted delivery of TLR agonists to the tumor tissue are needed to enable the widespread use of endosomal TLR agonists in the context of tumor immunotherapy. One strategy for targeted delivery of TLR agonist is their conjugation to tumor antigen-specific therapeutic antibodies. Such antibody-TLR agonist conjugates act synergistically by inducing local TLR-mediated innate immune activation which complements the anti-tumor immune mechanisms induced by the therapeutic antibody. In this study, we explored different conjugation strategies for TLR9 agonists to immunoglobulin G (IgG). We evaluated biochemical conjugation of immunostimulatory CpG oligodesoxyribonucleotides (ODN) to the HER2-specific therapeutic antibody Trastuzumab with different cross-linkers comparing stochastic with site-specific conjugation. The physiochemical make-up and biological activities of the generated Trastuzumab-ODN conjugates were characterized in vitro and demonstrated that site-specific conjugation of CpG ODN is crucial for maintaining the antigen-binding capabilities of Trastuzumab. Furthermore, site-specific conjugate was effective in promoting anti-tumor immune responses in vivo in a pseudo-metastasis mouse model with engineered human HER2-transgenic tumor cells. In this in vivo model, co-delivery of Trastuzumab and CpG ODN in form of site-specific conjugates was superior to co-injection of unconjugated Trastuzumab, CpG ODN or stochastic conjugate in promoting T cell activation and expansion. Thereby, this study highlights that site-specific conjugation of CpG ODN to therapeutic antibodies targeting tumor markers is a feasible and more reliable approach for generation of conjugates which retain and combine the functional properties of the adjuvant and the antibody.
Collapse
Affiliation(s)
- Diana Corogeanu
- National Institute for Biological Standards and Control (NIBSC), Biotherapeutics Division, Medicines and Healthcare products Regulatory Agency, Potters Bar, United Kingdom
| | - Kam Zaki
- National Institute for Biological Standards and Control (NIBSC), Advanced Therapies Division, Medicines and Healthcare products Regulatory Agency, Potters Bar, United Kingdom
| | - Andrew J Beavil
- King's College London, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, Guy's Hospital, London, United Kingdom
| | - James N Arnold
- King's College London, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, Guy's Hospital, London, United Kingdom
| | - Sandra S Diebold
- National Institute for Biological Standards and Control (NIBSC), Biotherapeutics Division, Medicines and Healthcare products Regulatory Agency, Potters Bar, United Kingdom
| |
Collapse
|
24
|
Kremenovic M, Chan AA, Feng B, Bäriswyl L, Robatel S, Gruber T, Tang L, Lee DJ, Schenk M. BCG hydrogel promotes CTSS-mediated antigen processing and presentation, thereby suppressing metastasis and prolonging survival in melanoma. J Immunother Cancer 2022; 10:jitc-2021-004133. [PMID: 35732347 PMCID: PMC9226922 DOI: 10.1136/jitc-2021-004133] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2022] [Indexed: 12/31/2022] Open
Abstract
Background The use of intralesional Mycobacterium bovis BCG (intralesional live BCG) for the treatment of metastatic melanoma resulted in regression of directly injected, and occasionally of distal lesions. However, intralesional-BCG is less effective in patients with visceral metastases and did not significantly improve overall survival. Methods We generated a novel BCG lysate and developed it into a thermosensitive PLGA-PEG-PLGA hydrogel (BCG hydrogel), which was injected adjacent to the tumor to assess its antitumor effect in syngeneic tumor models (B16F10, MC38). The effect of BCG hydrogel treatment on contralateral tumors, lung metastases, and survival was assessed to evaluate systemic long-term efficacy. Gene expression profiles of tumor-infiltrating immune cells and of tumor-draining lymph nodes from BCG hydrogel-treated mice were analyzed by single-cell RNA sequencing (scRNA-seq) and CD8+ T cell receptor (TCR) repertoire diversity was assessed by TCR-sequencing. To confirm the mechanistic findings, RNA-seq data of biopsies obtained from in-transit cutaneous metastases of patients with melanoma who had received intralesional-BCG therapy were analyzed. Results Here, we show that BCG lysate exhibits enhanced antitumor efficacy compared to live mycobacteria and promotes a proinflammatory tumor microenvironment and M1 macrophage (MΦ) polarization in vivo. The underlying mechanisms of BCG lysate-mediated tumor immunity are dependent on MΦ and dendritic cells (DCs). BCG hydrogel treatment induced systemic immunity in melanoma-bearing mice with suppression of lung metastases and improved survival. Furthermore, BCG hydrogel promoted cathepsin S (CTSS) activity in MΦ and DCs, resulting in enhanced antigen processing and presentation of tumor-associated antigens. Finally, BCG hydrogel treatment was associated with increased frequencies of melanoma-reactive CD8+ T cells. In human patients with melanoma, intralesional-BCG treatment was associated with enhanced M1 MΦ, mature DC, antigen processing and presentation, as well as with increased CTSS expression which positively correlated with patient survival. Conclusions These findings provide mechanistic insights as well as rationale for the clinical translation of BCG hydrogel as cancer immunotherapy to overcome the current limitations of immunotherapies for the treatment of patients with melanoma.
Collapse
Affiliation(s)
- Mirela Kremenovic
- Experimental Pathology, University of Bern Institute of Pathology, Bern, Switzerland.,Graduate School Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Alfred A Chan
- Division of Dermatology, Department of Medicine, The Lundquist Institute, Torrance, California, USA
| | - Bing Feng
- Institute of Bioengineering and Institute of Materials Science and Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, VD, Switzerland
| | - Lukas Bäriswyl
- Experimental Pathology, University of Bern Institute of Pathology, Bern, Switzerland
| | - Steve Robatel
- Experimental Pathology, University of Bern Institute of Pathology, Bern, Switzerland.,Graduate School Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Thomas Gruber
- Experimental Pathology, University of Bern Institute of Pathology, Bern, Switzerland
| | - Li Tang
- Institute of Bioengineering and Institute of Materials Science and Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, VD, Switzerland
| | - Delphine J Lee
- Division of Dermatology, Department of Medicine, The Lundquist Institute, Torrance, California, USA
| | - Mirjam Schenk
- Experimental Pathology, University of Bern Institute of Pathology, Bern, Switzerland
| |
Collapse
|
25
|
Kozaka S, Wakabayashi R, Kamiya N, Goto M. Lyotropic liquid crystal-based transcutaneous peptide delivery system: Evaluation of skin permeability and potential for transcutaneous vaccination. Acta Biomater 2022; 138:273-284. [PMID: 34774785 DOI: 10.1016/j.actbio.2021.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/13/2021] [Accepted: 11/05/2021] [Indexed: 12/27/2022]
Abstract
Transcutaneous drug delivery is a promising method in terms of drug repositioning and reformulation because of its non-invasive and easy-to-use features. To overcome the skin barrier, which is the biggest challenge in transcutaneous drug delivery, a number of techniques, such as microemulsion, solid-in-oil dispersions and liposomes, have been studied extensively. However, the low viscosity of these formulations limits drug retention on the skin and reduces patient acceptability. Although viscosity can be increased by adding a thickening reagent, such an addition often alters formulation nanostructures and drug solubility, and importantly, decreases skin permeability. In this study, a gel-like lyotropic liquid crystal (LLC) was used as a tool to enhance skin permeability. In particular, we prepared 1-monolinolein (ML)-based LLCs with different water contents. All LLCs significantly enhanced skin permeation of a peptide drug, an epitope peptide of melanoma, despite their high viscoelasticity. Fourier transform infra-red spectroscopic analysis of the skin surface treated with the LLCs revealed that the gyroid geometry more strongly interacted with the lamellar structure inside the stratum corneum (SC) than the diamond geometry. Finally, as the result of the in vivo tumor challenge experiment using B16F10 melanoma-bearing mice, the LLC with the gyroid geometry showed stronger vaccine effect against tumor than a subcutaneous injection. Collectively, ML-based LLCs, especially with the gyroid geometry, are a promising strategy to deliver biomacromolecules into skin. STATEMENT OF SIGNIFICANCE: Transcutaneous drug delivery is a promising method for drug repositioning and reformulation because of its non-invasive and easy-to-use features. To overcome the skin barrier, which is the biggest challenge in transcutaneous drug delivery, we used a gel-like lyotropic liquid crystal (LLC) as a novel tool to enhance skin permeability. In this paper, we demonstrated that an LLC with a specific liquid crystalline structure has the highest skin permeation enhancement effect for a peptide antigen as a model drug. Moreover, the peptide antigen-loaded LLC showed a vaccine effect that was comparable to a subcutaneous injection in vivo. This study provides a basis for designing a transcutaneous delivery system of peptide drugs with LLC.
Collapse
|
26
|
Wu J, Yang H, Xu JC, Hu Z, Gu WF, Chen ZY, Xia JX, Lowrie DB, Lu SH, Fan XY. Mycobacterium tuberculosis Rv3628 isan effective adjuvant via activationof dendritic cells for cancer immunotherapy. MOLECULAR THERAPY-ONCOLYTICS 2021; 23:288-302. [PMID: 34786473 PMCID: PMC8571481 DOI: 10.1016/j.omto.2021.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/24/2021] [Accepted: 10/07/2021] [Indexed: 12/30/2022]
Abstract
Tumor antigens (Ags) are weakly immunogenic and elicit inadequate immune responses, thus induction of antigen-specific immune activation via the maturation of dendritic cells (DCs) is a strategy used for cancer immunotherapy. In this study, we examined the effect of Rv3628 from Mycobacterium tuberculosis (Mtb) on activation of DCs and anti-tumor immunity in vivo. Intravenous injection of mice with Rv3628 promoted DC activation of spleen and lymph nodes. More importantly, Rv3628 also induced activation of DCs and enhanced Ag presentation in tumor-bearing mice. In mice bearing ovalbumin (OVA)-expressing tumors, combination treatment with Rv3628 and OVA peptide promoted OVA-specific T cell activation and accumulation of interferon (IFN)-γ and tumor necrosis factor (TNF)-α-producing OT-I and OT-II cells in tumor-draining lymph nodes. Moreover, three different tumor Ags in three different tumor models showed enhanced anti-tumor activity with Rv3628 as adjuvant, including inhibition of growth of OVA-expressing B16 melanoma, CT26 carcinoma, and B16 melanoma tumors, and a synergistic effect with anti-programmed cell death protein 1 (PD-1) antibody treatment. Additionally, potential application against human tumors was indicated by similar activation of human peripheral blood DCs by Rv3628. Taken together, these data demonstrate that Rv3628 could be an effective adjuvant in tumor immunotherapy via enhanced capacity of DC activation and Ag presentation.
Collapse
Affiliation(s)
- Juan Wu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China.,TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai 201508, China
| | - Heng Yang
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China
| | - Jin-Chuan Xu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China
| | - Zhidong Hu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China.,TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai 201508, China
| | - Wen-Fei Gu
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhen-Yan Chen
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China
| | - Jing-Xian Xia
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China
| | - Douglas B Lowrie
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China
| | - Shui-Hua Lu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China.,TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai 201508, China
| | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China.,TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai 201508, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
27
|
Kyjacova L, Saup R, Rothley M, Schmaus A, Wagner T, Boßerhoff A, Garvalov BK, Thiele W, Sleeman JP. Quantitative Detection of Disseminated Melanoma Cells by Trp-1 Transcript Analysis Reveals Stochastic Distribution of Pulmonary Metastases. J Clin Med 2021; 10:jcm10225459. [PMID: 34830742 PMCID: PMC8618565 DOI: 10.3390/jcm10225459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/29/2022] Open
Abstract
A better understanding of the process of melanoma metastasis is required to underpin the development of novel therapies that will improve patient outcomes. The use of appropriate animal models is indispensable for investigating the mechanisms of melanoma metastasis. However, reliable and practicable quantification of metastases in experimental mice remains a challenge, particularly if the metastatic burden is low. Here, we describe a qRT-PCR-based protocol that employs the melanocytic marker Trp-1 for the sensitive quantification of melanoma metastases in the murine lung. Using this protocol, we were able to detect the presence of as few as 100 disseminated melanoma cells in lung tissue. This allowed us to quantify metastatic burden in a spontaneous syngeneic B16-F10 metastasis model, even in the absence of visible metastases, as well as in the autochthonous Tg(Grm1)/Cyld−/− melanoma model. Importantly, we also observed an uneven distribution of disseminated melanoma cells amongst the five lobes of the murine lung, which varied considerably from animal to animal. Together, our findings demonstrate that the qRT-PCR-based detection of Trp-1 allows the quantification of low pulmonary metastatic burden in both transplantable and autochthonous murine melanoma models, and show that the analysis of lung metastasis in such models needs to take into account the stochastic distribution of metastatic lesions amongst the lung lobes.
Collapse
Affiliation(s)
- Lenka Kyjacova
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
| | - Rafael Saup
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
| | - Melanie Rothley
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT)-Campus North, D-76344 Karlsruhe, Germany
| | - Anja Schmaus
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT)-Campus North, D-76344 Karlsruhe, Germany
| | - Tabea Wagner
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
| | - Anja Boßerhoff
- Institute of Biochemistry, Faculty of Medicine, Friedrich-Alexander University Erlangen-Nürnberg (FAU), D-91054 Erlangen, Germany;
| | - Boyan K. Garvalov
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
| | - Wilko Thiele
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT)-Campus North, D-76344 Karlsruhe, Germany
| | - Jonathan P. Sleeman
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT)-Campus North, D-76344 Karlsruhe, Germany
- Correspondence: ; Tel.: +49-621-383-71595
| |
Collapse
|
28
|
Howell LM, Forbes NS. Bacteria-based immune therapies for cancer treatment. Semin Cancer Biol 2021; 86:1163-1178. [PMID: 34547442 DOI: 10.1016/j.semcancer.2021.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/03/2021] [Accepted: 09/12/2021] [Indexed: 12/23/2022]
Abstract
Engineered bacterial therapies that target the tumor immune landscape offer a new class of cancer immunotherapy. Salmonella enterica and Listeria monocytogenes are two species of bacteria that have been engineered to specifically target tumors and serve as delivery vessels for immunotherapies. Therapeutic bacteria have been engineered to deliver cytokines, gene silencing shRNA, and tumor associated antigens that increase immune activation. Bacterial therapies stimulate both the innate and adaptive immune system, change the immune dynamics of the tumor microenvironment, and offer unique strategies for targeting tumors. Bacteria have innate adjuvant properties, which enable both the delivered molecules and the bacteria themselves to stimulate immune responses. Bacterial immunotherapies that deliver cytokines and tumor-associated antigens have demonstrated clinical efficacy. Harnessing the diverse set of mechanisms that Salmonella and Listeria use to alter the tumor-immune landscape has the potential to generate many new and effective immunotherapies.
Collapse
Affiliation(s)
- Lars M Howell
- Department of Chemical Engineering, University of Massachusetts, Amherst, United States
| | - Neil S Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, United States.
| |
Collapse
|
29
|
Johnson LR, Lee DY, Eacret JS, Ye D, June CH, Minn AJ. The immunostimulatory RNA RN7SL1 enables CAR-T cells to enhance autonomous and endogenous immune function. Cell 2021; 184:4981-4995.e14. [PMID: 34464586 PMCID: PMC11338632 DOI: 10.1016/j.cell.2021.08.004] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 05/27/2021] [Accepted: 08/04/2021] [Indexed: 12/31/2022]
Abstract
Poor tumor infiltration, development of exhaustion, and antigen insufficiency are common mechanisms that limit chimeric antigen receptor (CAR)-T cell efficacy. Delivery of pattern recognition receptor agonists is one strategy to improve immune function; however, targeting these agonists to immune cells is challenging, and off-target signaling in cancer cells can be detrimental. Here, we engineer CAR-T cells to deliver RN7SL1, an endogenous RNA that activates RIG-I/MDA5 signaling. RN7SL1 promotes expansion and effector-memory differentiation of CAR-T cells. Moreover, RN7SL1 is deployed in extracellular vesicles and selectively transferred to immune cells. Unlike other RNA agonists, transferred RN7SL1 restricts myeloid-derived suppressor cell (MDSC) development, decreases TGFB in myeloid cells, and fosters dendritic cell (DC) subsets with costimulatory features. Consequently, endogenous effector-memory and tumor-specific T cells also expand, allowing rejection of solid tumors with CAR antigen loss. Supported by improved endogenous immunity, CAR-T cells can now co-deploy peptide antigens with RN7SL1 to enhance efficacy, even when heterogenous CAR antigen tumors lack adequate neoantigens.
Collapse
Affiliation(s)
- Lexus R Johnson
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Mark Foundation Center for Immunotherapy, Immune Signaling, and Radiation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Y Lee
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacqueline S Eacret
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Darwin Ye
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Mark Foundation Center for Immunotherapy, Immune Signaling, and Radiation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Andy J Minn
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Mark Foundation Center for Immunotherapy, Immune Signaling, and Radiation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
30
|
LncRNA FENDRR Expression Correlates with Tumor Immunogenicity. Genes (Basel) 2021; 12:genes12060897. [PMID: 34200642 PMCID: PMC8226633 DOI: 10.3390/genes12060897] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 01/06/2023] Open
Abstract
FENDRR (Fetal-lethal non-coding developmental regulatory RNA, LncRNA FOXF1-AS1) is a recently identified tumor suppressor long non-coding (LncRNA) RNA, and its expression has been linked with epigenetic modulation of the target genes involved in tumor immunity. In this study, we aimed to understand the role of FENDRR in predicting immune-responsiveness and the inflammatory tumor environment. Briefly, FENDRR expression and its relationship to immune activation signals were assessed in murine cell lines. Data suggested that tumor cells (e.g., C26 colon, 4T1 breast) that typically upregulate immune activation genes and the MHC class I molecule exhibited high FENDRR expression levels. Conversely, tumor cells with a generalized downregulation of immune-related gene expression (e.g., B16F10 melanoma) demonstrated low to undetectable FENDRR levels. Mechanistically, the modulation of FENDRR expression enhanced the inflammatory and WNT signaling pathways in tumors. Our early data suggest that FENDRR can play an important role in the development of immune-relevant phenotypes in tumors, and thereby improve cancer immunotherapy.
Collapse
|
31
|
Yin Q, Yu W, Grzeskowiak CL, Li J, Huang H, Guo J, Chen L, Wang F, Zhao F, von Boehmer L, Metzner TJ, Leppert JT, Chien YH, Kuo CJ, Davis MM. Nanoparticle-enabled innate immune stimulation activates endogenous tumor-infiltrating T cells with broad antigen specificities. Proc Natl Acad Sci U S A 2021; 118:e2016168118. [PMID: 34021082 PMCID: PMC8166076 DOI: 10.1073/pnas.2016168118] [Citation(s) in RCA: 12] [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] [Indexed: 12/17/2022] Open
Abstract
Tumors are often infiltrated by T lymphocytes recognizing either self- or mutated antigens but are generally inactive, although they often show signs of prior clonal expansion. Hypothesizing that this may be due to peripheral tolerance, we formulated nanoparticles containing innate immune stimulants that we found were sufficient to activate self-specific CD8+ T cells and injected them into two different mouse tumor models, B16F10 and MC38. These nanoparticles robustly activated and/or expanded antigen-specific CD8+ tumor-infiltrating T cells, along with a decrease in regulatory CD4+ T cells and an increase in Interleukin-17 producers, resulting in significant tumor growth retardation or elimination and the establishment of immune memory in surviving mice. Furthermore, nanoparticles with modification of stimulating human T cells enabled the robust activation of endogenous T cells in patient-derived tumor organoids. These results indicate that breaking peripheral tolerance without regard to the antigen specificity creates a promising pathway for cancer immunotherapy.
Collapse
Affiliation(s)
- Qian Yin
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305
| | - Wong Yu
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA 94305
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA 94305
| | - Caitlin L Grzeskowiak
- Division of Hematology, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305
| | - Jing Li
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305
| | - Huang Huang
- HHMI, Stanford University School of Medicine, Stanford, CA 94305
| | - Jing Guo
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA 94305
| | - Liang Chen
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305
| | - Feng Wang
- HHMI, Stanford University School of Medicine, Stanford, CA 94305
| | - Fan Zhao
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA 94305
| | - Lotta von Boehmer
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305
| | - Thomas J Metzner
- Department of Urology, School of Medicine, Stanford University, Stanford, CA 94305
| | - John T Leppert
- Department of Urology, School of Medicine, Stanford University, Stanford, CA 94305
| | - Yueh-Hsiu Chien
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA 94305
| | - Calvin J Kuo
- Division of Hematology, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305
| | - Mark M Davis
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305;
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA 94305
- HHMI, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
32
|
Truckenbrod EN, Burrack KS, Knutson TP, Borges da Silva H, Block KE, O'Flanagan SD, Stagliano KR, Hurwitz AA, Fulton RB, Renkema KR, Jameson SC. CD8 + T cell self-tolerance permits responsiveness but limits tissue damage. eLife 2021; 10:65615. [PMID: 33929324 PMCID: PMC8147182 DOI: 10.7554/elife.65615] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/29/2021] [Indexed: 01/25/2023] Open
Abstract
Self-specific CD8+T cells can escape clonal deletion, but the properties and capabilities of such cells in a physiological setting are unclear. We characterized polyclonal CD8+ T cells specific for the melanocyte antigen tyrosinase-related protein 2 (Trp2) in mice expressing or lacking this enzyme (due to deficiency in Dct, which encodes Trp2). Phenotypic and gene expression profiles of pre-immune Trp2/Kb-specific cells were similar; the size of this population was only slightly reduced in wild-type (WT) compared to Dct-deficient (Dct-/-) mice. Despite comparable initial responses to Trp2 immunization, WT Trp2/Kb-specific cells showed blunted expansion and less readily differentiated into a CD25+proliferative population. Functional self-tolerance clearly emerged when assessing immunopathology: adoptively transferred WT Trp2/Kb-specific cells mediated vitiligo much less efficiently. Hence, CD8+ T cell self-specificity is poorly predicted by precursor frequency, phenotype, or even initial responsiveness, while deficient activation-induced CD25 expression and other gene expression characteristics may help to identify functionally tolerant cells.
Collapse
Affiliation(s)
| | - Kristina S Burrack
- Center for Immunology, University of Minnesota, Saint Paul, United States
| | - Todd P Knutson
- Minnesota Supercomputing Institute, University of Minnesota, Saint Paul, United States
| | | | - Katharine E Block
- Center for Immunology, University of Minnesota, Saint Paul, United States
| | | | - Katie R Stagliano
- Center for Immunology, University of Minnesota, Saint Paul, United States
| | - Arthur A Hurwitz
- Center for Immunology, University of Minnesota, Saint Paul, United States
| | - Ross B Fulton
- Center for Immunology, University of Minnesota, Saint Paul, United States
| | - Kristin R Renkema
- Center for Immunology, University of Minnesota, Saint Paul, United States
| | - Stephen C Jameson
- Center for Immunology, University of Minnesota, Saint Paul, United States
| |
Collapse
|
33
|
Strobel SB, Machiraju D, Hülsmeyer I, Becker JC, Paschen A, Jäger D, Wels WS, Bachmann M, Hassel JC. Expression of Potential Targets for Cell-Based Therapies on Melanoma Cells. Life (Basel) 2021; 11:life11040269. [PMID: 33805080 PMCID: PMC8064084 DOI: 10.3390/life11040269] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 01/17/2023] Open
Abstract
Tumor antigen-specific redirection of cytotoxic T cells (CTLs) or natural killer (NK) cells including chimeric antigen receptor (CAR-) and T cell receptor (TCR-) cell therapy is currently being evaluated in different tumor entities including melanoma. Expression of melanoma-specific antigen recognized by the respective CAR or TCR directly or presented by HLA molecules is an indispensable prerequisite for this innovative therapy. In this study, we investigated in 168 FFPE tumor specimens of patients with stage I-IV melanoma the protein expression of HER2, TRP2, ABCB5, gp100, p53, and GD2 by immunohistochemistry (IHC). These results were correlated with clinical parameters. Membrane expression of HER2 and GD2 was also investigated in ten melanoma cell lines by flow cytometry for which corresponding tumors were analyzed by IHC. Our results demonstrated that gp100 was the most frequently overexpressed protein (61%), followed by TRP2 (50%), GD2 (38%), p53 (37%), ABCB5 (17%), and HER2 (3%). TRP2 expression was higher in primary tumors compared to metastases (p = 0.005). Accordingly, TRP2 and ABCB5 expression was significantly associated with lower tumor thickness of the primary (p = 0.013 and p = 0.025). There was no association between protein expression levels and survival in advanced melanoma patients. Flow cytometric analysis revealed abundant surface expression of GD2 and HER2 in all melanoma cell lines. The discordant HER2 expression in situ and in vitro suggests a tissue culture associated induction. In summary, our data support the use of gp100 and GD2 as a potential target for developing engineered TCR- or CAR-cell therapies, respectively, against melanoma.
Collapse
Affiliation(s)
- Sophia B. Strobel
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; (S.B.S.); (D.M.)
| | - Devayani Machiraju
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; (S.B.S.); (D.M.)
| | - Ingrid Hülsmeyer
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (I.H.); (J.C.B.); (A.P.); (W.S.W.)
| | - Jürgen C. Becker
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (I.H.); (J.C.B.); (A.P.); (W.S.W.)
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), 45141 Essen, Germany
- Department of Dermatology, University Hospital Essen, 45147 Essen, Germany
| | - Annette Paschen
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (I.H.); (J.C.B.); (A.P.); (W.S.W.)
- Department of Dermatology, University Hospital Essen, 45147 Essen, Germany
| | - Dirk Jäger
- National Center for Tumor Diseases (NCT) Heidelberg, Department of Medical Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany;
- National Center for Tumor Diseases, German Cancer Research Center, Clinical Cooperation Unit Applied Tumor Immunity, 69120 Heidelberg, Germany
| | - Winfried S. Wels
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (I.H.); (J.C.B.); (A.P.); (W.S.W.)
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60590 Frankfurt am Main, Germany
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany;
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), University Hospital ‘Carl Gustav Carus’, TU Dresden, 01307 Dresden, Germany
- Tumor Immunology, University Cancer Center (UCC) ‘Carl Gustav Carus’, TU Dresden, 01307 Dresden, Germany
| | - Jessica C. Hassel
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; (S.B.S.); (D.M.)
- Correspondence:
| |
Collapse
|
34
|
Morante V, Borghi M, Farina I, Michelini Z, Grasso F, Gallinaro A, Cecchetti S, Di Virgilio A, Canitano A, Pirillo MF, Bona R, Cara A, Negri D. Integrase-Defective Lentiviral Vector Is an Efficient Vaccine Platform for Cancer Immunotherapy. Viruses 2021; 13:v13020355. [PMID: 33672349 PMCID: PMC7927015 DOI: 10.3390/v13020355] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Integrase-defective lentiviral vectors (IDLVs) have been used as a safe and efficient delivery system in several immunization protocols in murine and non-human primate preclinical models as well as in recent clinical trials. In this work, we validated in preclinical murine models our vaccine platform based on IDLVs as delivery system for cancer immunotherapy. To evaluate the anti-tumor activity of our vaccine strategy we generated IDLV delivering ovalbumin (OVA) as a non-self-model antigen and TRP2 as a self-tumor associated antigen (TAA) of melanoma. Results demonstrated the ability of IDLVs to eradicate and/or controlling tumor growth after a single immunization in preventive and therapeutic approaches, using lymphoma and melanoma expressing OVA. Importantly, LV-TRP2 but not IDLV-TRP2 was able to break tolerance efficiently and prevent tumor growth of B16F10 melanoma cells. In order to improve the IDLV efficacy, the human homologue of murine TRP2 was used, showing the ability to break tolerance and control the tumor growth. These results validate the use of IDLV for cancer therapy.
Collapse
Affiliation(s)
- Valeria Morante
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.M.); (M.B.); (I.F.); (F.G.)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.M.); (M.B.); (I.F.); (F.G.)
| | - Iole Farina
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.M.); (M.B.); (I.F.); (F.G.)
| | - Zuleika Michelini
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (Z.M.); (A.G.); (A.C.); (M.F.P.); (R.B.)
| | - Felicia Grasso
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.M.); (M.B.); (I.F.); (F.G.)
| | - Alessandra Gallinaro
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (Z.M.); (A.G.); (A.C.); (M.F.P.); (R.B.)
| | - Serena Cecchetti
- Confocal Microscopy Unit NMR, Confocal Microscopy Area Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Antonio Di Virgilio
- Center for Animal Research and Welfare, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Andrea Canitano
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (Z.M.); (A.G.); (A.C.); (M.F.P.); (R.B.)
| | - Maria Franca Pirillo
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (Z.M.); (A.G.); (A.C.); (M.F.P.); (R.B.)
| | - Roberta Bona
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (Z.M.); (A.G.); (A.C.); (M.F.P.); (R.B.)
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (Z.M.); (A.G.); (A.C.); (M.F.P.); (R.B.)
- Correspondence: (A.C.); (D.N.)
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.M.); (M.B.); (I.F.); (F.G.)
- Correspondence: (A.C.); (D.N.)
| |
Collapse
|
35
|
Levy ES, Chang R, Zamecnik CR, Dhariwala MO, Fong L, Desai TA. Multi-Immune Agonist Nanoparticle Therapy Stimulates Type I Interferons to Activate Antigen-Presenting Cells and Induce Antigen-Specific Antitumor Immunity. Mol Pharm 2021; 18:1014-1025. [PMID: 33541072 DOI: 10.1021/acs.molpharmaceut.0c00984] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer immunity is mediated by a delicate orchestration between the innate and adaptive immune system both systemically and within the tumor microenvironment. Although several adaptive immunity molecular targets have been proven clinically efficacious, stand-alone innate immunity targeting agents have not been successful in the clinic. Here, we report a nanoparticle optimized for systemic administration that combines immune agonists for TLR9, STING, and RIG-I with a melanoma-specific peptide to induce antitumor immunity. These immune agonistic nanoparticles (iaNPs) significantly enhance the activation of antigen-presenting cells to orchestrate the development and response of melanoma-sensitized T-cells. iaNP treatment not only suppressed tumor growth in an orthotopic solid tumor model, but also significantly reduced tumor burden in a metastatic animal model. This combination biomaterial-based approach to coordinate innate and adaptive anticancer immunity provides further insights into the benefits of stimulating multiple activation pathways to promote tumor regression, while also offering an important platform to effectively and safely deliver combination immunotherapies for cancer.
Collapse
Affiliation(s)
- Elizabeth S Levy
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California 94158, United States
| | - Ryan Chang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California 94158, United States.,Department of Medicine, University of California San Francisco, San Francisco, California 94143, United States
| | - Colin R Zamecnik
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California 94158, United States
| | - Miqdad O Dhariwala
- Department of Dermatology, University of California San Francisco, San Francisco, California 94143, United Stats
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California 94143, United States.,Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, California 94143, United States
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
36
|
Lam H, McNeil LK, Starobinets H, DeVault VL, Cohen RB, Twardowski P, Johnson ML, Gillison ML, Stein MN, Vaishampayan UN, DeCillis AP, Foti JJ, Vemulapalli V, Tjon E, Ferber K, DeOliveira DB, Broom W, Agnihotri P, Jaffee EM, Wong KK, Drake CG, Carroll PM, Davis TA, Flechtner JB. An Empirical Antigen Selection Method Identifies Neoantigens That Either Elicit Broad Antitumor T-cell Responses or Drive Tumor Growth. Cancer Discov 2021; 11:696-713. [PMID: 33504579 DOI: 10.1158/2159-8290.cd-20-0377] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 09/15/2020] [Accepted: 11/13/2020] [Indexed: 11/16/2022]
Abstract
Neoantigens are critical targets of antitumor T-cell responses. The ATLAS bioassay was developed to identify neoantigens empirically by expressing each unique patient-specific tumor mutation individually in Escherichia coli, pulsing autologous dendritic cells in an ordered array, and testing the patient's T cells for recognition in an overnight assay. Profiling of T cells from patients with lung cancer revealed both stimulatory and inhibitory responses to individual neoantigens. In the murine B16F10 melanoma model, therapeutic immunization with ATLAS-identified stimulatory neoantigens protected animals, whereas immunization with peptides associated with inhibitory ATLAS responses resulted in accelerated tumor growth and abolished efficacy of an otherwise protective vaccine. A planned interim analysis of a clinical study testing a poly-ICLC adjuvanted personalized vaccine containing ATLAS-identified stimulatory neoantigens showed that it is well tolerated. In an adjuvant setting, immunized patients generated both CD4+ and CD8+ T-cell responses, with immune responses to 99% of the vaccinated peptide antigens. SIGNIFICANCE: Predicting neoantigens in silico has progressed, but empirical testing shows that T-cell responses are more nuanced than straightforward MHC antigen recognition. The ATLAS bioassay screens tumor mutations to uncover preexisting, patient-relevant neoantigen T-cell responses and reveals a new class of putatively deleterious responses that could affect cancer immunotherapy design.This article is highlighted in the In This Issue feature, p. 521.
Collapse
Affiliation(s)
- Hubert Lam
- Genocea Biosciences Inc., Cambridge, Massachusetts
| | | | | | | | - Roger B Cohen
- University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Maura L Gillison
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark N Stein
- Columbia University Medical Center, New York, New York
| | | | | | - James J Foti
- Genocea Biosciences Inc., Cambridge, Massachusetts
| | | | - Emily Tjon
- Genocea Biosciences Inc., Cambridge, Massachusetts
| | - Kyle Ferber
- Genocea Biosciences Inc., Cambridge, Massachusetts
| | | | - Wendy Broom
- Genocea Biosciences Inc., Cambridge, Massachusetts
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Singh MP, Sethuraman SN, Miller C, Malayer J, Ranjan A. Boiling histotripsy and in-situ CD40 stimulation improve the checkpoint blockade therapy of poorly immunogenic tumors. Theranostics 2021; 11:540-554. [PMID: 33391491 PMCID: PMC7738858 DOI: 10.7150/thno.49517] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Advanced stage cancers with a suppressive tumor microenvironment (TME) are often refractory to immune checkpoint inhibitor (ICI) therapy. Recent studies have shown that focused ultrasound (FUS) TME-modulation can synergize ICI therapy, but enhancing survival outcomes in poorly immunogenic tumors remains challenging. Here, we investigated the role of focused ultrasound based boiling histotripsy (HT) and in-situ anti-CD40 agonist antibody (αCD40) combinatorial therapy in enhancing therapeutic efficacy against ICI refractory murine melanoma. Methods: Unilateral and bilateral large (~330-400 mm3) poorly immunogenic B16F10 melanoma tumors were established in the flank regions of mice. Tumors were exposed to single local HT followed by an in-situ administration of αCD40 (HT+ αCD40: HT40). Inflammatory signatures post treatment were assessed using pan-cancer immune profiling and flow cytometry. The ability of HT40 ± ICI to enhance local and systemic effects was determined by immunological characterization of the harvested tissues, and by tumor growth delay of local and distant untreated tumors 4-6 weeks post treatment. Results: Immune profiling revealed that HT40 upregulated a variety of inflammatory markers in the tumors. Immunologically, HT40 treated tumors showed an increased population of granzyme B+ expressing functional CD8+ T cells (~4-fold) as well as an increased M1 to M2 macrophage ratio (~2-3-fold) and CD8+ T: regulatory T cell ratio (~5-fold) compared to the untreated control. Systemically, the proliferation rates of the melanoma-specific memory T cell population were significantly enhanced by HT40 treatment. Finally, the combination of HT40 and ICI therapy (anti-CTLA-4 and anti-PD-L1) caused superior inhibition of distant untreated tumors, and prolonged survival rates compared to the control. Conclusions: Data suggest that HT40 reprograms immunologically cold tumors and sensitizes them to ICI therapy. This approach may be clinically useful for treating advanced stage melanoma cancers.
Collapse
Affiliation(s)
- Mohit Pratap Singh
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078
| | - Sri Nandhini Sethuraman
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078
| | - Craig Miller
- Department of Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078
| | - Jerry Malayer
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078
| | - Ashish Ranjan
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078
| |
Collapse
|
38
|
Bassi G, Favalli N, Vuk M, Catalano M, Martinelli A, Trenner A, Porro A, Yang S, Tham CL, Moroglu M, Yue WW, Conway SJ, Vogt PK, Sartori AA, Scheuermann J, Neri D. A Single-Stranded DNA-Encoded Chemical Library Based on a Stereoisomeric Scaffold Enables Ligand Discovery by Modular Assembly of Building Blocks. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001970. [PMID: 33240760 PMCID: PMC7675038 DOI: 10.1002/advs.202001970] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/17/2020] [Indexed: 06/11/2023]
Abstract
A versatile and Lipinski-compliant DNA-encoded library (DEL), comprising 366 600 glutamic acid derivatives coupled to oligonucleotides serving as amplifiable identification barcodes is designed, constructed, and characterized. The GB-DEL library, constructed in single-stranded DNA format, allows de novo identification of specific binders against several pharmaceutically relevant proteins. Moreover, hybridization of the single-stranded DEL with a set of known protein ligands of low to medium affinity coupled to a complementary DNA strand results in self-assembled selectable chemical structures, leading to the identification of affinity-matured compounds.
Collapse
Affiliation(s)
- Gabriele Bassi
- Department of Chemistry and Applied BiosciencesETH ZürichZürich8092Switzerland
| | - Nicholas Favalli
- Department of Chemistry and Applied BiosciencesETH ZürichZürich8092Switzerland
| | - Miriam Vuk
- Department of Chemistry and Applied BiosciencesETH ZürichZürich8092Switzerland
| | - Marco Catalano
- Department of Chemistry and Applied BiosciencesETH ZürichZürich8092Switzerland
| | - Adriano Martinelli
- Department of Chemistry and Applied BiosciencesETH ZürichZürich8092Switzerland
| | - Anika Trenner
- Institute of Molecular Cancer ResearchUniversity of ZürichZürich8006Switzerland
| | - Antonio Porro
- Institute of Molecular Cancer ResearchUniversity of ZürichZürich8006Switzerland
| | - Su Yang
- Scripps Research InstituteDepartment of Molecular MedicineLa JollaCA92037USA
| | - Chuin Lean Tham
- Structural Genomic Consortium (SGC)Nuffield Department of MedicineUniversity of OxfordOxfordOX1 2JDUK
| | - Mustafa Moroglu
- Department of ChemistryChemistry Research LaboratoryUniversity of OxfordMansfield RoadOxfordOX1 3TAUK
| | - Wyatt W. Yue
- Structural Genomic Consortium (SGC)Nuffield Department of MedicineUniversity of OxfordOxfordOX1 2JDUK
| | - Stuart J. Conway
- Department of ChemistryChemistry Research LaboratoryUniversity of OxfordMansfield RoadOxfordOX1 3TAUK
| | - Peter K. Vogt
- Scripps Research InstituteDepartment of Molecular MedicineLa JollaCA92037USA
| | | | - Jörg Scheuermann
- Department of Chemistry and Applied BiosciencesETH ZürichZürich8092Switzerland
| | - Dario Neri
- Department of Chemistry and Applied BiosciencesETH ZürichZürich8092Switzerland
| |
Collapse
|
39
|
Gruber T, Kremenovic M, Sadozai H, Rombini N, Baeriswyl L, Maibach F, Modlin RL, Gilliet M, von Werdt D, Hunger RE, Seyed Jafari SM, Parisi G, Abril-Rodriguez G, Ribas A, Schenk M. IL-32γ potentiates tumor immunity in melanoma. JCI Insight 2020; 5:138772. [PMID: 32841222 PMCID: PMC7526542 DOI: 10.1172/jci.insight.138772] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/13/2020] [Indexed: 12/19/2022] Open
Abstract
Myeloid cells orchestrate the antitumor immune response and influence the efficacy of immune checkpoint blockade (ICB) therapies. We and others have previously shown that IL-32 mediates DC differentiation and macrophage activation. Here, we demonstrate that IL-32 expression in human melanoma positively correlates with overall survival, response to ICB, and an immune-inflamed tumor microenvironment (TME) enriched in mature DC, M1 macrophages, and CD8+ T cells. Treatment of B16F10 murine melanomas with IL-32 increased the frequencies of activated, tumor-specific CD8+ T cells, leading to the induction of systemic tumor immunity. Our mechanistic in vivo studies revealed a potentially novel role of IL-32 in activating intratumoral DC and macrophages to act in concert to prime CD8+ T cells and recruit them into the TME through CCL5. Thereby, IL-32 treatment reduced tumor growth and rendered ICB-resistant B16F10 tumors responsive to anti-PD-1 therapy without toxicity. Furthermore, increased baseline IL-32 gene expression was associated with response to nivolumab and pembrolizumab in 2 independent cohorts of patients with melanoma, implying that IL-32 is a predictive biomarker for anti-PD-1 therapy. Collectively, this study suggests IL-32 as a potent adjuvant in immunotherapy to enhance the efficacy of ICB in patients with non-T cell-inflamed TME.
Collapse
Affiliation(s)
- Thomas Gruber
- Institute of Pathology, Experimental Pathology, and.,Graduate School Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Mirela Kremenovic
- Institute of Pathology, Experimental Pathology, and.,Graduate School Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Hassan Sadozai
- Institute of Pathology, Experimental Pathology, and.,Graduate School Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | | | | | - Robert L Modlin
- Division of Dermatology, Department of Medicine and Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Michel Gilliet
- Department of Dermatology, Lausanne University Hospital, Lausanne, Switzerland
| | - Diego von Werdt
- Institute of Pathology, Experimental Pathology, and.,Graduate School Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Robert E Hunger
- Department of Dermatology, Inselspital, University Hospital of Bern, University of Bern, Bern, Switzerland
| | - S Morteza Seyed Jafari
- Department of Dermatology, Inselspital, University Hospital of Bern, University of Bern, Bern, Switzerland
| | - Giulia Parisi
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, and UCLA Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| | - Gabriel Abril-Rodriguez
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, and UCLA Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| | - Antoni Ribas
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, and UCLA Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| | | |
Collapse
|
40
|
Yang K, Oak AS, Slominski RM, Brożyna AA, Slominski AT. Current Molecular Markers of Melanoma and Treatment Targets. Int J Mol Sci 2020; 21:ijms21103535. [PMID: 32429485 PMCID: PMC7278971 DOI: 10.3390/ijms21103535] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/08/2020] [Accepted: 05/13/2020] [Indexed: 12/17/2022] Open
Abstract
Melanoma is a deadly skin cancer that becomes especially difficult to treat after it metastasizes. Timely identification of melanoma is critical for effective therapy, but histopathologic diagnosis can frequently pose a significant challenge to this goal. Therefore, auxiliary diagnostic tools are imperative to facilitating prompt recognition of malignant lesions. Melanoma develops as result of a number of genetic mutations, with UV radiation often acting as a mutagenic risk factor. Novel methods of genetic testing have improved detection of these molecular alterations, which subsequently revealed important information for diagnosis and prognosis. Rapid detection of genetic alterations is also significant for choosing appropriate treatment and developing targeted therapies for melanoma. This review will delve into the understanding of various mutations and the implications they may pose for clinical decision making.
Collapse
Affiliation(s)
- Kevin Yang
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (K.Y.); (A.S.O.)
| | - Allen S.W. Oak
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (K.Y.); (A.S.O.)
| | - Radomir M. Slominski
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Anna A. Brożyna
- Department of Human Biology, Institute of Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Toruń, Poland;
| | - Andrzej T. Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (K.Y.); (A.S.O.)
- Comprehensive Cancer Center, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Veteran Administration Medical Center, Birmingham, AL 35294, USA
- Correspondence:
| |
Collapse
|
41
|
Pérez-Guijarro E, Yang HH, Araya RE, El Meskini R, Michael HT, Vodnala SK, Marie KL, Smith C, Chin S, Lam KC, Thorkelsson A, Iacovelli AJ, Kulaga A, Fon A, Michalowski AM, Hugo W, Lo RS, Restifo NP, Sharan SK, Van Dyke T, Goldszmid RS, Weaver Ohler Z, Lee MP, Day CP, Merlino G. Multimodel preclinical platform predicts clinical response of melanoma to immunotherapy. Nat Med 2020; 26:781-791. [PMID: 32284588 DOI: 10.1038/s41591-020-0818-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/03/2020] [Indexed: 02/07/2023]
Abstract
Although immunotherapy has revolutionized cancer treatment, only a subset of patients demonstrate durable clinical benefit. Definitive predictive biomarkers and targets to overcome resistance remain unidentified, underscoring the urgency to develop reliable immunocompetent models for mechanistic assessment. Here we characterize a panel of syngeneic mouse models, representing a variety of molecular and phenotypic subtypes of human melanomas and exhibiting their diverse range of responses to immune checkpoint blockade (ICB). Comparative analysis of genomic, transcriptomic and tumor-infiltrating immune cell profiles demonstrated alignment with clinical observations and validated the correlation of T cell dysfunction and exclusion programs with resistance. Notably, genome-wide expression analysis uncovered a melanocytic plasticity signature predictive of patient outcome in response to ICB, suggesting that the multipotency and differentiation status of melanoma can determine ICB benefit. Our comparative preclinical platform recapitulates melanoma clinical behavior and can be employed to identify mechanisms and treatment strategies to improve patient care.
Collapse
Affiliation(s)
- Eva Pérez-Guijarro
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Howard H Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Romina E Araya
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rajaa El Meskini
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Helen T Michael
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Suman Kumar Vodnala
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Lyell Immunopharma, South San Francisco, CA, USA
| | - Kerrie L Marie
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cari Smith
- Laboratory Animal Science Program, Leidos Biomedical Research Inc, Frederick, MD, USA
| | - Sung Chin
- Laboratory Animal Science Program, Leidos Biomedical Research Inc, Frederick, MD, USA
| | - Khiem C Lam
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andres Thorkelsson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anthony J Iacovelli
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Alan Kulaga
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Anyen Fon
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Aleksandra M Michalowski
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Willy Hugo
- Division of Dermatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Roger S Lo
- Division of Dermatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Nicholas P Restifo
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Lyell Immunopharma, South San Francisco, CA, USA
| | - Shyam K Sharan
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.,Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Terry Van Dyke
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.,Path Forward Solutions, Frederick, MD, USA
| | - Romina S Goldszmid
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zoe Weaver Ohler
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Maxwell P Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
42
|
Tähtinen S, Feola S, Capasso C, Laustio N, Groeneveldt C, Ylösmäki EO, Ylösmäki L, Martins B, Fusciello M, Medeot M, Tagliamonte M, Chiaro J, Hamdan F, Peltonen K, Ranki T, Buonaguro L, Cerullo V. Exploiting Preexisting Immunity to Enhance Oncolytic Cancer Immunotherapy. Cancer Res 2020; 80:2575-2585. [PMID: 32107211 DOI: 10.1158/0008-5472.can-19-2062] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 12/06/2019] [Accepted: 02/24/2020] [Indexed: 11/16/2022]
Abstract
Because of the high coverage of international vaccination programs, most people worldwide have been vaccinated against common pathogens, leading to acquired pathogen-specific immunity with a robust memory T-cell repertoire. Although CD8+ antitumor cytotoxic T lymphocytes (CTL) are the preferred effectors of cancer immunotherapy, CD4+ T-cell help is also required for an optimal antitumor immune response to occur. Hence, we investigated whether the pathogen-related CD4+ T-cell memory populations could be reengaged to support the CTLs, converting a weak primary antitumor immune response into a stronger secondary one. To this end, we used our PeptiCRAd technology that consists of an oncolytic adenovirus coated with MHC-I-restricted tumor-specific peptides and developed it further by introducing pathogen-specific MHC-II-restricted peptides. Mice preimmunized with tetanus vaccine were challenged with B16.OVA tumors and treated with the newly developed hybrid TT-OVA-PeptiCRAd containing both tetanus toxoid- and tumor-specific peptides. Treatment with the hybrid PeptiCRAd significantly enhanced antitumor efficacy and induced TT-specific, CD40 ligand-expressing CD4+ T helper cells and maturation of antigen-presenting cells. Importantly, this approach could be extended to naturally occurring tumor peptides (both tumor-associated antigens and neoantigens), as well as to other pathogens beyond tetanus, highlighting the usefulness of this technique to take full advantage of CD4+ memory T-cell repertoires when designing immunotherapeutic treatment regimens. Finally, the antitumor effect was even more prominent when combined with the immune checkpoint inhibitor anti-PD-1, strengthening the rationale behind combination therapy with oncolytic viruses. SIGNIFICANCE: These findings establish a novel technology that enhances oncolytic cancer immunotherapy by capitalizing on pre-acquired immunity to pathogens to convert a weak antitumor immune response into a much stronger one.
Collapse
Affiliation(s)
- Siri Tähtinen
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy iCAN Digital Precision Cancer Medicine Flagship, Helsinki University, Viikinkaari 5E, Finland
| | - Sara Feola
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy iCAN Digital Precision Cancer Medicine Flagship, Helsinki University, Viikinkaari 5E, Finland
| | - Cristian Capasso
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy iCAN Digital Precision Cancer Medicine Flagship, Helsinki University, Viikinkaari 5E, Finland
| | - Netta Laustio
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy iCAN Digital Precision Cancer Medicine Flagship, Helsinki University, Viikinkaari 5E, Finland
| | | | - Erkko O Ylösmäki
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy iCAN Digital Precision Cancer Medicine Flagship, Helsinki University, Viikinkaari 5E, Finland
| | - Leena Ylösmäki
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy iCAN Digital Precision Cancer Medicine Flagship, Helsinki University, Viikinkaari 5E, Finland
| | - Beatriz Martins
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy iCAN Digital Precision Cancer Medicine Flagship, Helsinki University, Viikinkaari 5E, Finland
| | - Manlio Fusciello
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy iCAN Digital Precision Cancer Medicine Flagship, Helsinki University, Viikinkaari 5E, Finland
| | - Marta Medeot
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Maria Tagliamonte
- Cancer Immunoregulatory, Istituto Nazionale Tumori (IRCCS) G. Pascale, Naples, Italy
| | - Jacopo Chiaro
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy iCAN Digital Precision Cancer Medicine Flagship, Helsinki University, Viikinkaari 5E, Finland
| | - Firas Hamdan
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy iCAN Digital Precision Cancer Medicine Flagship, Helsinki University, Viikinkaari 5E, Finland
| | - Karita Peltonen
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy iCAN Digital Precision Cancer Medicine Flagship, Helsinki University, Viikinkaari 5E, Finland
| | | | - Luigi Buonaguro
- Cancer Immunoregulatory, Istituto Nazionale Tumori (IRCCS) G. Pascale, Naples, Italy
| | - Vincenzo Cerullo
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy iCAN Digital Precision Cancer Medicine Flagship, Helsinki University, Viikinkaari 5E, Finland. .,Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.,Department of Molecular Medicine and Medical Biotechnology, Naples University "Federico II", Naples, Italy
| |
Collapse
|
43
|
Measles Vaccines Designed for Enhanced CD8 + T Cell Activation. Viruses 2020; 12:v12020242. [PMID: 32098134 PMCID: PMC7077255 DOI: 10.3390/v12020242] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/16/2020] [Accepted: 02/19/2020] [Indexed: 12/26/2022] Open
Abstract
Priming and activation of CD8+ T cell responses is crucial to achieve anti-viral and anti-tumor immunity. Live attenuated measles vaccine strains have been used successfully for immunization for decades and are currently investigated in trials of oncolytic virotherapy. The available reverse genetics systems allow for insertion of additional genes, including heterologous antigens. Here, we designed recombinant measles vaccine vectors for priming and activation of antigen-specific CD8+ T cells. For proof-of-concept, we used cytotoxic T lymphocyte (CTL) lines specific for the melanoma-associated differentiation antigen tyrosinase-related protein-2 (TRP-2), or the model antigen chicken ovalbumin (OVA), respectively. We generated recombinant measles vaccine vectors with TRP-2 and OVA epitope cassette variants for expression of the full-length antigen or the respective immunodominant CD8+ epitope, with additional variants mediating secretion or proteasomal degradation of the epitope. We show that these recombinant measles virus vectors mediate varying levels of MHC class I (MHC-I)-restricted epitope presentation, leading to activation of cognate CTLs, as indicated by secretion of interferon-gamma (IFNγ) in vitro. Importantly, the recombinant OVA vaccines also mediate priming of naïve OT-I CD8+ T cells by dendritic cells. While all vaccine variants can prime and activate cognate T cells, IFNγ release was enhanced using a secreted epitope variant and a variant with epitope strings targeted to the proteasome. The principles presented in this study will facilitate the design of recombinant vaccines to elicit CD8+ responses against pathogens and tumor antigens.
Collapse
|
44
|
Wang Z, Chen T, Lin W, Zheng W, Chen J, Huang F, Xie X. Functional tumor specific CD8 + T cells in spleen express a high level of PD-1. Int Immunopharmacol 2020; 80:106242. [PMID: 32014811 DOI: 10.1016/j.intimp.2020.106242] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/13/2020] [Accepted: 01/21/2020] [Indexed: 01/11/2023]
Abstract
The inhibitory effects of programmed cell death 1 (PD-1) receptor on tumor specific T cells were mainly investigated at tumor site. While PD-1 expression can be rapidly unregulated upon T cell activation at lymphoid tissues, little is known about where PD-1 signal exerts its inhibitory function in tumor-bearing host. To address this issue, we assessed the effects of PD-1 on vaccine induced activation of splenic CD8 + T cells in mice. The vaccine consisted of mice CD8 + T cell epitope peptide and poly IC. After vaccination, spleen or tumor tissues were dissociated, IFN-γ synthesis and PD-1 expression by CD8 + T cells were detected by flow cytometry. We found that CD8 + T cells could be successfully activated in spleen after immunization, characterized by the capability of producing IFN-γ when encountering relevant peptide. These activated splenic CD8 + T cells also expressed a high level of PD-1. Although PD-L1 expression in spleen parenchyma was also increased after vaccination, PD-1 blockade did not affect the activation of splenic CD8 + T cells, but enhanced the anti-tumor effects of peptide vaccine. This synergetic effect of peptide vaccine plus PD-1 blockade was coupled with increased aggregation of IFN-γ + CD8 + tumor infiltrated lymphocytes (TILs), rather than CD4 + TILs. The results indicated that for tumor-bearing host, PD-1 pathway exerted its inhibitory function at tumor site and PD-1 expression on the splenic CD8 + T cells correlated positively with IFN-γ synthesis.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/administration & dosage
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- B7-H1 Antigen/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/administration & dosage
- Cell Line, Tumor/transplantation
- Drug Resistance, Neoplasm/immunology
- Epitopes, T-Lymphocyte/immunology
- Female
- Gene Expression Regulation, Neoplastic/immunology
- Immunotherapy/methods
- Interferon-gamma/metabolism
- Intramolecular Oxidoreductases/administration & dosage
- Intramolecular Oxidoreductases/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Poly I-C/immunology
- Programmed Cell Death 1 Receptor/metabolism
- Specific Pathogen-Free Organisms
- Spleen/cytology
- Spleen/immunology
- Spleen/metabolism
- Spleen/pathology
- Up-Regulation/immunology
- Vaccines, Subunit/administration & dosage
Collapse
Affiliation(s)
- Zili Wang
- Department of Oncology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Ting Chen
- Department of Oncology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Wanzun Lin
- Department of Oncology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Weili Zheng
- Department of Oncology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Junying Chen
- Central Lab, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Fei Huang
- Central Lab, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Xianhe Xie
- Department of Oncology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China.
| |
Collapse
|
45
|
Kozaka S, Tahara Y, Wakabayashi R, Nakata T, Ueda T, Kamiya N, Goto M. Transcutaneous Cancer Vaccine Using a Reverse Micellar Antigen Carrier. Mol Pharm 2019; 17:645-655. [PMID: 31833775 DOI: 10.1021/acs.molpharmaceut.9b01104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Skin dendritic cells (DCs) such as Langerhans cells and dermal dendritic cells have a pivotal role in inducing antigen-specific immunity; therefore, transcutaneous cancer vaccines are a promising strategy to prophylactically prevent the onset of a variety of diseases, including cancers. The largest obstacle to delivering antigen to these skin DC subsets is the barrier function of the stratum corneum. Although reverse micellar carriers are commonly used to enhance skin permeability to hydrophilic drugs, the transcutaneous delivery of antigen, proteins, or peptides has not been achieved to date because of the large molecular weight of drugs. To achieve effective antigen delivery to skin DCs, we developed a novel strategy using a surfactant as a skin permeation enhancer in a reverse micellar carrier. In this study, glyceryl monooleate (MO) was chosen as a skin permeation enhancer, and the MO-based reverse micellar carrier enabled the successful delivery of antigen to Langerhans cells and dermal dendritic cells. Moreover, transcutaneous vaccination with the MO-based reverse micellar carrier significantly inhibited tumor growth, indicating that it is a promising vaccine platform against tumors.
Collapse
Affiliation(s)
| | | | | | - Takahiro Nakata
- Kobayashi Pharmaceutical Co., Ltd. , 1-30-3 Toyokawa , Ibaraki , Osaka 567-0057 , Japan
| | - Taro Ueda
- Kobayashi Pharmaceutical Co., Ltd. , 1-30-3 Toyokawa , Ibaraki , Osaka 567-0057 , Japan
| | | | | |
Collapse
|
46
|
Miao L, Li L, Huang Y, Delcassian D, Chahal J, Han J, Shi Y, Sadtler K, Gao W, Lin J, Doloff JC, Langer R, Anderson DG. Delivery of mRNA vaccines with heterocyclic lipids increases anti-tumor efficacy by STING-mediated immune cell activation. Nat Biotechnol 2019; 37:1174-1185. [PMID: 31570898 DOI: 10.1038/s41587-019-0247-3] [Citation(s) in RCA: 433] [Impact Index Per Article: 72.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 07/13/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022]
Abstract
Therapeutic messenger RNA vaccines enable delivery of whole antigens, which can be advantageous over peptide vaccines. However, optimal efficacy requires both intracellular delivery, to allow antigen translation, and appropriate immune activation. Here, we developed a combinatorial library of ionizable lipid-like materials to identify mRNA delivery vehicles that facilitate mRNA delivery in vivo and provide potent and specific immune activation. Using a three-dimensional multi-component reaction system, we synthesized and evaluated the vaccine potential of over 1,000 lipid formulations. The top candidate formulations induced a robust immune response, and were able to inhibit tumor growth and prolong survival in melanoma and human papillomavirus E7 in vivo tumor models. The top-performing lipids share a common structure: an unsaturated lipid tail, a dihydroimidazole linker and cyclic amine head groups. These formulations induce antigen-presenting cell maturation via the intracellular stimulator of interferon genes (STING) pathway, rather than through Toll-like receptors, and result in limited systemic cytokine expression and enhanced anti-tumor efficacy.
Collapse
Affiliation(s)
- Lei Miao
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Linxian Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong, China
| | - Yuxuan Huang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Derfogail Delcassian
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA.,Division of Regenerative Medicine and Cellular Therapy, University of Nottingham, Nottingham, UK
| | - Jasdave Chahal
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jinsong Han
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong, China
| | - Yunhua Shi
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kaitlyn Sadtler
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA
| | - Wenting Gao
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jiaqi Lin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joshua C Doloff
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA.,Departments of Biomedical & Materials Science Engineering, Translational Tissue Engineering Center, Wilmer Eye Institute and the Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA. .,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
47
|
Lopes A, Feola S, Ligot S, Fusciello M, Vandermeulen G, Préat V, Cerullo V. Oncolytic adenovirus drives specific immune response generated by a poly-epitope pDNA vaccine encoding melanoma neoantigens into the tumor site. J Immunother Cancer 2019; 7:174. [PMID: 31291991 PMCID: PMC6621971 DOI: 10.1186/s40425-019-0644-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 06/19/2019] [Indexed: 02/08/2023] Open
Abstract
Background DNA vaccines against cancer held great promises due to the generation of a specific and long-lasting immune response. However, when used as a single therapy, they are not able to drive the generated immune response into the tumor, because of the immunosuppressive microenvironment, thus limiting their use in humans. To enhance DNA vaccine efficacy, we combined a new poly-epitope DNA vaccine encoding melanoma tumor associated antigens and B16F1-specific neoantigens with an oncolytic virus administered intratumorally. Methods Genomic analysis were performed to find specific mutations in B16F1 melanoma cells. The antigen gene sequences were designed according to these mutations prior to the insertion in the plasmid vector. Mice were injected with B16F1 tumor cells (n = 7–9) and therapeutically vaccinated 2, 9 and 16 days after the tumor injection. The virus was administered intratumorally at day 10, 12 and 14. Immune cell infiltration analysis and cytokine production were performed by flow cytometry, PCR and ELISPOT in the tumor site and in the spleen of animals, 17 days after the tumor injection. Results The combination of DNA vaccine and oncolytic virus significantly increased the immune activity into the tumor. In particular, the local intratumoral viral therapy increased the NK infiltration, thus increasing the production of different cytokines, chemokines and enzymes involved in the adaptive immune system recruitment and cytotoxic activity. On the other side, the DNA vaccine generated antigen-specific T cells in the spleen, which migrated into the tumor when recalled by the local viral therapy. The complementarity between these strategies explains the dramatic tumor regression observed only in the combination group compared to all the other control groups. Conclusions This study explores the immunological mechanism of the combination between an oncolytic adenovirus and a DNA vaccine against melanoma. It demonstrates that the use of a rational combination therapy involving DNA vaccination could overcome its poor immunogenicity. In this way, it will be possible to exploit the great potential of DNA vaccination, thus allowing a larger use in the clinic. Electronic supplementary material The online version of this article (10.1186/s40425-019-0644-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alessandra Lopes
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, B-1200, Brussels, Belgium
| | - Sara Feola
- University of Helsinki, Biocenter 2, Viikinkari 5E, Helsinki, Finland
| | - Sophie Ligot
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, B-1200, Brussels, Belgium
| | - Manlio Fusciello
- University of Helsinki, Biocenter 2, Viikinkari 5E, Helsinki, Finland
| | - Gaëlle Vandermeulen
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, B-1200, Brussels, Belgium
| | - Véronique Préat
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, B-1200, Brussels, Belgium.
| | - Vincenzo Cerullo
- University of Helsinki, Biocenter 2, Viikinkari 5E, Helsinki, Finland.
| |
Collapse
|
48
|
Scott AC, Dündar F, Zumbo P, Chandran SS, Klebanoff CA, Shakiba M, Trivedi P, Menocal L, Appleby H, Camara S, Zamarin D, Walther T, Snyder A, Femia MR, Comen EA, Wen HY, Hellmann MD, Anandasabapathy N, Liu Y, Altorki NK, Lauer P, Levy O, Glickman MS, Kaye J, Betel D, Philip M, Schietinger A. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 2019; 571:270-274. [PMID: 31207604 PMCID: PMC7698992 DOI: 10.1038/s41586-019-1324-y] [Citation(s) in RCA: 736] [Impact Index Per Article: 122.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 06/04/2019] [Indexed: 01/18/2023]
Abstract
Tumour-specific CD8 T cell dysfunction is a differentiation state that is distinct from the functional effector or memory T cell states1–6. Here we identify the nuclear factor TOX as a crucial regulator of the differentiation of tumour-specific T (TST) cells. We show that TOX is highly expressed in dysfunctional TST cells from tumours and in exhausted T cells during chronic viral infection. Expression of TOX is driven by chronic T cell receptor stimulation and NFAT activation. Ectopic expression of TOX in effector T cells in vitro induced a transcriptional program associated with T cell exhaustion. Conversely, deletion of Tox in TST cells in tumours abrogated the exhaustion program: Tox-deleted TST cells did not upregulate genes for inhibitory receptors (such as Pdcd1, Entpd1, Havcr2, Cd244 and Tigit), the chromatin of which remained largely inaccessible, and retained high expression of transcription factors such as TCF-1. Despite their normal, ‘non-exhausted’ immunophenotype, Tox-deleted TST cells remained dysfunctional, which suggests that the regulation of expression of inhibitory receptors is uncoupled from the loss of effector function. Notably, although Tox-deleted CD8 T cells differentiated normally to effector and memory states in response to acute infection, Tox-deleted TST cells failed to persist in tumours. We hypothesize that the TOX-induced exhaustion program serves to prevent the overstimulation of T cells and activation-induced cell death in settings of chronic antigen stimulation such as cancer.
Collapse
Affiliation(s)
- Andrew C Scott
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Friederike Dündar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.,Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Paul Zumbo
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.,Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Smita S Chandran
- Parker Institute for Cancer Immunotherapy, New York, NY, USA.,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher A Klebanoff
- Parker Institute for Cancer Immunotherapy, New York, NY, USA.,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Mojdeh Shakiba
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Prerak Trivedi
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laura Menocal
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Heather Appleby
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Steven Camara
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dmitriy Zamarin
- Parker Institute for Cancer Immunotherapy, New York, NY, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tyler Walther
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexandra Snyder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew R Femia
- Parker Institute for Cancer Immunotherapy, New York, NY, USA.,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth A Comen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Hannah Y Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew D Hellmann
- Parker Institute for Cancer Immunotherapy, New York, NY, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Niroshana Anandasabapathy
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.,Department of Dermatology, Weill Cornell Medical College, New York, NY, USA
| | - Yong Liu
- Department of Dermatology, Weill Cornell Medical College, New York, NY, USA
| | - Nasser K Altorki
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, USA
| | | | - Olivier Levy
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael S Glickman
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Jonathan Kaye
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA.,Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.,Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Mary Philip
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Andrea Schietinger
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA. .,Parker Institute for Cancer Immunotherapy, New York, NY, USA.
| |
Collapse
|
49
|
Pierrat MJ, Marsaud V, Mauviel A, Javelaud D. Transcriptional repression of the tyrosinase-related protein 2 gene by transforming growth factor-β and the Kruppel-like transcription factor GLI2. J Dermatol Sci 2019; 94:321-329. [PMID: 31208857 DOI: 10.1016/j.jdermsci.2019.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 02/21/2019] [Accepted: 04/01/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND Tyrosinase-Related Protein 2 (TRP2) is an enzyme involved in melanogenesis, that also exerts proliferative, anti-apoptotic and immunogenic functions in melanoma cells. TRP2 transcription is regulated by the melanocytic master transcription factor MITF. GLI2, a transcription factor that acts downstream of Hedgehog signaling, is also a direct transcriptional target of the TGF-β/SMAD pathway that contributes to melanoma progression and exerts transcriptional antagonistic activities against MITF. OBJECTIVES To characterize the molecular events responsible for TGF-β and GLI2 repression of TRP2 expression. METHODS In silico promoter analysis, transient cell transfection experiments with 5'-end TRP2 promoter deletion constructs, chromatin immuno-precipitation, and site-directed promoter mutagenesis were used to dissect the molecular mechanisms of TRP2 gene regulation by TGF-β and GLI2. RESULTS We demonstrate that TGF-β and GLI2-specific TRP2 repression involves direct mechanisms that occur in addition to MITF downregulation by TGF-β and GLI2. We identify two functional GLI2 binding sites within the TRP2 promoter that are critical for TGF-β and GLI2 responsiveness, one of them overlapping a CREB binding site. GLI2 and CREB competing for the same cis-element is associated with opposite transcriptional outcome. CONCLUSION Our results further refine the understanding of how TGF-β and GLI2 control the phenotypic plasticity of melanoma cells. In particular, we identify critical GLI2-binding cis-elements within the TRP2 promoter region that allow for its transcriptional repression independently from MITF concomitant downregulation.
Collapse
Affiliation(s)
- Marie-Jeanne Pierrat
- Institut Curie, PSL Research University, INSERM U1021, CNRS UMR3347, Team "TGF-ß and Oncogenesis", Equipe Labellisée LIGUE 2016, F-91400, Orsay, France; Université Paris-Sud, F-91400, Orsay, France
| | - Véronique Marsaud
- Institut Curie, PSL Research University, INSERM U1021, CNRS UMR3347, Team "TGF-ß and Oncogenesis", Equipe Labellisée LIGUE 2016, F-91400, Orsay, France; Université Paris-Sud, F-91400, Orsay, France
| | - Alain Mauviel
- Institut Curie, PSL Research University, INSERM U1021, CNRS UMR3347, Team "TGF-ß and Oncogenesis", Equipe Labellisée LIGUE 2016, F-91400, Orsay, France; Université Paris-Sud, F-91400, Orsay, France.
| | - Delphine Javelaud
- Institut Curie, PSL Research University, INSERM U1021, CNRS UMR3347, Team "TGF-ß and Oncogenesis", Equipe Labellisée LIGUE 2016, F-91400, Orsay, France; Université Paris-Sud, F-91400, Orsay, France.
| |
Collapse
|
50
|
van Dinther D, Lopez Venegas M, Veninga H, Olesek K, Hoogterp L, Revet M, Ambrosini M, Kalay H, Stöckl J, van Kooyk Y, den Haan JMM. Activation of CD8⁺ T Cell Responses after Melanoma Antigen Targeting to CD169⁺ Antigen Presenting Cells in Mice and Humans. Cancers (Basel) 2019; 11:cancers11020183. [PMID: 30764534 PMCID: PMC6406251 DOI: 10.3390/cancers11020183] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/28/2019] [Accepted: 01/31/2019] [Indexed: 12/30/2022] Open
Abstract
The lack of tumor-reactive T cells is one reason why immune checkpoint inhibitor therapies still fail in a significant proportion of melanoma patients. A vaccination that induces melanoma-specific T cells could potentially enhance the efficacy of immune checkpoint inhibitors. Here, we describe a vaccination strategy in which melanoma antigens are targeted to mouse and human CD169 and thereby induce strong melanoma antigen-specific T cell responses. CD169 is a sialic acid receptor expressed on a subset of mouse splenic macrophages that captures antigen from the blood and transfers it to dendritic cells (DCs). In human and mouse spleen, we detected CD169+ cells at an equivalent location using immunofluorescence microscopy. Immunization with melanoma antigens conjugated to antibodies (Abs) specific for mouse CD169 efficiently induced gp100 and Trp2-specific T cell responses in mice. In HLA-A2.1 transgenic mice targeting of the human MART-1 peptide to CD169 induced strong MART-1-specific HLA-A2.1-restricted T cell responses. Human gp100 peptide conjugated to Abs specific for human CD169 bound to CD169-expressing monocyte-derived DCs (MoDCs) and resulted in activation of gp100-specific T cells. Together, these data indicate that Ab-mediated antigen targeting to CD169 is a potential strategy for the induction of melanoma-specific T cell responses in mice and in humans.
Collapse
Affiliation(s)
- Dieke van Dinther
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands.
| | - Miguel Lopez Venegas
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands.
- DC4U B.V., De Corridor 21E, 3621 ZA Breukelen, The Netherlands.
| | - Henrike Veninga
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands.
| | - Katarzyna Olesek
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands.
| | - Leoni Hoogterp
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands.
| | - Mirjam Revet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands.
| | - Martino Ambrosini
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands.
- DC4U B.V., De Corridor 21E, 3621 ZA Breukelen, The Netherlands.
| | - Hakan Kalay
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands.
| | - Johannes Stöckl
- Institute of Immunology, Center of Pathophysiology, Immunology and Infectiology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Yvette van Kooyk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands.
- DC4U B.V., De Corridor 21E, 3621 ZA Breukelen, The Netherlands.
| | - Joke M M den Haan
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands.
| |
Collapse
|