A Cancer Nanovaccine for Co-Delivery of Peptide Neoantigens and Optimized Combinations of STING and TLR4 AgonistsClick to copy article linkArticle link copied!
- Jessalyn J. BaljonJessalyn J. BaljonDepartment of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United StatesMore by Jessalyn J. Baljon
- Alexander J. KwiatkowskiAlexander J. KwiatkowskiDepartment of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United StatesMore by Alexander J. Kwiatkowski
- Hayden M. PagendarmHayden M. PagendarmDepartment of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United StatesMore by Hayden M. Pagendarm
- Payton T. StonePayton T. StoneDepartment of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United StatesMore by Payton T. Stone
- Amrendra KumarAmrendra KumarDepartment of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United StatesMore by Amrendra Kumar
- Vijaya BhartiVijaya BhartiDepartment of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United StatesMore by Vijaya Bharti
- Jacob A. SchulmanJacob A. SchulmanDepartment of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United StatesMore by Jacob A. Schulman
- Kyle W. BeckerKyle W. BeckerDepartment of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United StatesMore by Kyle W. Becker
- Eric W. RothEric W. RothNorthwestern University Atomic and Nanoscale Characterization Experimental (NUANCE) Center, Northwestern University, Evanston, Illinois 60208, United StatesMore by Eric W. Roth
- Plamen P. ChristovPlamen P. ChristovVanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United StatesMore by Plamen P. Christov
- Sebastian JoyceSebastian JoyceDepartment of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United StatesDepartment of Veteran Affairs Tennessee Valley Healthcare System, Nashville, Tennessee 37212, United StatesVanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United StatesVanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United StatesMore by Sebastian Joyce
- John T. Wilson*John T. Wilson*Email: [email protected]Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United StatesDepartment of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United StatesVanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United StatesDepartment of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United StatesVanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United StatesVanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United StatesVanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United StatesMore by John T. Wilson
Abstract
Immune checkpoint blockade (ICB) has revolutionized cancer treatment and led to complete and durable responses, but only for a minority of patients. Resistance to ICB can largely be attributed to insufficient number and/or function of antitumor CD8+ T cells in the tumor microenvironment. Neoantigen targeted cancer vaccines can activate and expand the antitumor T cell repertoire, but historically, clinical responses have been poor because immunity against peptide antigens is typically weak, resulting in insufficient activation of CD8+ cytotoxic T cells. Herein, we describe a nanoparticle vaccine platform that can overcome these barriers in several ways. First, the vaccine can be reproducibly formulated using a scalable confined impingement jet mixing method to coload a variety of physicochemically diverse peptide antigens and multiple vaccine adjuvants into pH-responsive, vesicular nanoparticles that are monodisperse and less than 100 nm in diameter. Using this approach, we encapsulated synergistically acting adjuvants, cGAMP and monophosphoryl lipid A (MPLA), into the nanocarrier to induce a robust and tailored innate immune response that increased peptide antigen immunogenicity. We found that incorporating both adjuvants into the nanovaccine synergistically enhanced expression of dendritic cell costimulatory markers, pro-inflammatory cytokine secretion, and peptide antigen cross-presentation. Additionally, the nanoparticle delivery increased lymph node accumulation and uptake of peptide antigen by dendritic cells in the draining lymph node. Consequently, nanoparticle codelivery of peptide antigen, cGAMP, and MPLA enhanced the antigen-specific CD8+ T cell response and delayed tumor growth in several mouse models. Finally, the nanoparticle platform improved the efficacy of ICB immunotherapy in a murine colon carcinoma model. This work establishes a versatile nanoparticle vaccine platform for codelivery of peptide neoantigens and synergistic adjuvants to enhance responses to cancer vaccines.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Results and Discussion
Formulation of Nanoparticle Vaccine via Confined Impingement Jet Mixing
Co-Delivery of cGAMP and MPLA Enhances Co-Stimulatory Molecule Expression and Pro-Inflammatory Cytokine Secretion
Nanoparticle Delivery Enhances Lymphatic Accumulation and Uptake by Antigen Presenting Cells
Nanoparticle Co-Delivery of Antigen, cGAMP, and MPLA Enhances Antigen-Specific CD8+ T Cell Response
Nanoparticle Vaccine Inhibits Tumor Growth and Improves Survival in Mice
Nanoparticle Vaccine Enhances Functionality of Antigen-Specific CD8+ T Cell Response
Nanoparticle Vaccine Enhances Neoantigen Vaccine Efficacy and Response to ICB
Nanoparticle Vaccine Activates a Functional Antigen-Specific CD8+ T Cell Response Against Neoantigens
Conclusion
Materials and Methods
Polymer Synthesis and Characterization
Nanoparticle Formulation
Gal9 Recruitment Assay
In Vitro Evaluation of Adjuvant Activity
In Vitro Evaluation of BMDC Activation
In Vitro Evaluation of Antigen Cross-Presentation
In Vitro Evaluation of Peptide Uptake by BMDCs
Animal Care
Analysis of Nanoparticle Accumulation and Uptake in Lymph Node
Analysis of OVA-Specific CD8+ T Cell Response in Spleen
Vaccination and Splenocyte Isolation for Downstream T Cell Analysis Studies
Establishment of Batf3–/– Model and Evaluation of CD8+ T Cell Response
Analysis of Memory Phenotype
Intracellular Cytokine Staining of Splenocytes
Enzyme-Linked Immunosorbent Spot Assay (ELISpot)
Tumor Studies with EG7.OVA
Tumor Studies with MC38
Statistical Analysis
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsnano.3c04471.
Supplementary data describing polymer characterization, nanovaccine drug loading and sizing properties, BMDC costimulatory molecule expression and pro-inflammatory cytokine secretion, Loewe synergy scores, cellular uptake, tumor growth curves, and flow cytometry gating strategies (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We gratefully acknowledge C. Duvall for use of IVIS imaging equipment. Additionally, we thank the core facilities of the Vanderbilt Institute of Nanoscale Sciences and Engineering (VINSE) for use of dynamic light scattering, the VUMC Flow Cytometry Shared Resource supported by the Vanderbilt Ingram Cancer Center (P30 CA68485) and the Vanderbilt Digestive Disease Research Center (DK058404), and the Vanderbilt Mass Spectrometry Research Center (MSRC) for use of mass spectrometry. We also acknowledge M.J. Munson for providing the Gal9-mCherry H358 cells. Additionally, the cryoTEM work made use of the BioCryo facility of Northwestern University’s NUANCE center, which has received support from the SHyNE Resource (NSF ECCS-2025633), and IIN, and Northwestern’s MRSEC program (NSF DMR-1720139). 2′3′-cGAMP was provided by the Vanderbilt Institute of Chemical Biology Chemical Synthesis Core. Experiments were performed in the Vanderbilt High-Throughput Screening (HTS) Core Facility with assistance provided by Joshua A. Bauer and David Baughman. The HTS Core receives support from the Vanderbilt Institute of Chemical Biology and the Vanderbilt Ingram Cancer Center (P30 CA68485). This work was supported by grants from the National Science Foundation CBET-1554623 (J.T.W), the National Institutes of Health (NIH) R01CA266767 (J.T.W), R01DE027749 (S.J.), F31CA257275-01 (J.J.B.), the NIH Integrated in Engineering and Diabetes Training Grant (T32DK101003, H.M.P), the NIH Microenvironmental Influences in Cancer Training Grant (T32CA009592, A.J.K),the NIH Chemical-Biology Interface Training Grant (T32GM065086, P.T.S), a Vanderbilt Ingram Cancer Center Scholarship (J.T.W.), a Veteran’s Affairs RCS Award IK6 BX004595 (S.J.) and Merit Awards I01 BX001444 and BX001610 (S.J.), and a Stand Up To Cancer Innovative Research Grant, grant number SU2C-AACR-IRG- 20-17 (J.T.W.)Stand Up to Cancer (SU2C) is a program of the Entertainment Industry Foundation. Research grants are administered by the American Association for Cancer Research, the scientific partner of SU2C. H.M.P and P.T.S. acknowledge funding support through the National Science Foundation Graduate Research Fellowship Program under grant number 1937963. Any opinions, findings, and conclusions or recommendations expressed in this material are those of the author(s) and do not necessarily reflect the views of the National Science Foundation.
References
This article references 78 other publications.
- 1Korman, A. J.; Garrett-Thomson, S. C.; Lonberg, N. The Foundations of Immune Checkpoint Blockade and the Ipilimumab Approval Decennial. Nat. Rev. Drug Discov 2022, 21, 509– 528, DOI: 10.1038/s41573-021-00345-8Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXivValtr%252FJ&md5=4d70b7f025350759038f63b701a6c494The foundations of immune checkpoint blockade and the ipilimumab approval decennialKorman, Alan J.; Garrett-Thomson, Sarah C.; Lonberg, NilsNature Reviews Drug Discovery (2022), 21 (7), 509-528CODEN: NRDDAG; ISSN:1474-1776. (Nature Portfolio)Cancer immunity, and the potential for cancer immunotherapy, have been topics of scientific discussion and experimentation for over a hundred years. Several successful cancer immunotherapies - such as IL-2 and interferon-a (IFNa) - have appeared over the past 30 years. However, it is only in the past decade that immunotherapy has made a broad impact on patient survival in multiple high-incidence cancer indications. The emergence of immunotherapy as a new pillar of cancer treatment (adding to surgery, radiation, chemotherapy and targeted therapies) is due to the success of immune checkpoint blockade (ICB) drugs, the first of which - ipilimumab - was approved in 2011. ICB drugs block receptors and ligands involved in pathways that attenuate T cell activation - such as cytotoxic T lymphocyte antigen 4 (CTLA4), programed cell death 1 (PD1) and its ligand, PDL1 - and prevent, or reverse, acquired peripheral tolerance to tumor antigens. In this Review we mark the tenth anniversary of the approval of ipilimumab and discuss the foundational scientific history of ICB, together with the history of the discovery, development and elucidation of the mechanism of action of the first generation of drugs targeting the CTLA4 and PD1 pathways.
- 2Sharma, P.; Allison, J. P. The Future of Immune Checkpoint Therapy. Science 2015, 348, 56– 61, DOI: 10.1126/science.aaa8172Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXls1Wmurg%253D&md5=d63ae85b9c651ec64a3b5b002c609e35The future of immune checkpoint therapySharma, Padmanee; Allison, James P.Science (Washington, DC, United States) (2015), 348 (6230), 56-61CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)A review. Immune checkpoint therapy, which targets regulatory pathways in T cells to enhance antitumor immune responses, has led to important clin. advances and provided a new weapon against cancer. This therapy has elicited durable clin. responses and, in a fraction of patients, long-term remissions where patients exhibit no clin. signs of cancer for many years. The way forward for this class of novel agents lies in our ability to understand human immune responses in the tumor microenvironment. This will provide valuable information regarding the dynamic nature of the immune response and regulation of addnl. pathways that will need to be targeted through combination therapies to provide survival benefit for greater nos. of patients.
- 3Shum, B.; Larkin, J.; Turajlic, S. Predictive Biomarkers for Response to Immune Checkpoint Inhibition. Semin Cancer Biol. 2022, 79, 4– 17, DOI: 10.1016/j.semcancer.2021.03.036Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtlWqt7jK&md5=17802852fefce3fedc8ce6b596fd7510Predictive biomarkers for response to immune checkpoint inhibitionShum, Benjamin; Larkin, James; Turajlic, SamraSeminars in Cancer Biology (2022), 79 (), 4-17CODEN: SECBE7; ISSN:1044-579X. (Elsevier Ltd.)A review. Immune checkpoint inhibitors have transformed the prognosis and treatment paradigm of many cancer types, through the potential for durable responses. However, the majority of patients still do not benefit. Response to checkpoint inhibition is detd. by dynamic host, tumor and tumor microenvironment factors that display spatial and temporal variability, but our understanding of these interactions is incomplete. Through investigating biomarkers of resistance and response, opportunities arise to discover new therapeutic targets and shape personalised treatment strategies. Here we review approved and emerging biomarkers of response to immune checkpoint inhibitors, in particular the recent rapid progress in host and tumor genomics. It is unlikely that a single biomarker will precisely predict response, but multivariate multiomic markers may provide a balanced assessment of these factors and more accurately identify patients who will benefit. Further efforts are required to translate these groundbreaking discoveries into novel therapeutics and biomarker driven clin. trials, to provide durable treatment response to a greater population of patients.
- 4Jenkins, R. W.; Barbie, D. A.; Flaherty, K. T. Mechanisms of Resistance to Immune Checkpoint Inhibitors. Br. J. Cancer 2018, 118, 9– 16, DOI: 10.1038/bjc.2017.434Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlGmsQ%253D%253D&md5=98d6734fe4172247aa9402fd388159dfMechanisms of resistance to immune checkpoint inhibitorsJenkins, Russell W.; Barbie, David A.; Flaherty, Keith T.British Journal of Cancer (2018), 118 (1), 9-16CODEN: BJCAAI; ISSN:0007-0920. (Nature Research)Immune checkpoint inhibitors (ICI) targeting CTLA-4 and the PD-1/PD-L1 axis have shown unprecedented clin. activity in several types of cancer and are rapidly transforming the practice of medical oncol. Whereas cytotoxic chemotherapy and small mol. inhibitors ('targeted therapies') largely act on cancer cells directly, immune checkpoint inhibitors reinvigorate anti-tumor immune responses by disrupting co-inhibitory T-cell signalling. While resistance routinely develops in patients treated with conventional cancer therapies and targeted therapies, durable responses suggestive of long-lasting immunol. memory are commonly seen in large subsets of patients treated with ICI. However, initial response appears to be a binary event, with most non-responders to single-agent ICI therapy progressing at a rate consistent with the natural history of disease. In addn., late relapses are now emerging with longer follow-up of clin. trial populations, suggesting the emergence of acquired resistance. As robust biomarkers to predict clin. response and/or resistance remain elusive, the mechanisms underlying innate (primary) and acquired (secondary) resistance are largely inferred from pre-clin. studies and correlative clin. data. Improved understanding of mol. and immunol. mechanisms of ICI response (and resistance) may not only identify novel predictive and/or prognostic biomarkers, but also ultimately guide optimal combination/sequencing of ICI therapy in the clinic. Here we review the emerging clin. and pre-clin. data identifying novel mechanisms of innate and acquired resistance to immune checkpoint inhibition.
- 5Ye, Z.; Qian, Q.; Jin, H.; Qian, Q. Cancer Vaccine: Learning Lessons from Immune Checkpoint Inhibitors. J. Cancer 2018, 9, 263– 268, DOI: 10.7150/jca.20059Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MvhslGhtQ%253D%253D&md5=c878c5e7ffce77d94296fce9ae06908aCancer vaccine: learning lessons from immune checkpoint inhibitorsYe ZhenLong; Qian Qiming; Jin HuaJun; Qian QiJunJournal of Cancer (2018), 9 (2), 263-268 ISSN:1837-9664.Cancer vaccines have been exclusively studied all through the past decades, and have made exceptional achievements in cancer treatment. Few cancer vaccines have been approved by the US Food and Drug Administration (FDA), for instance, Provenge, which was approved for the treatment of prostate carcinoma in 2012. Moreover, more recently, T-VEC got approval for the treatment of melanoma. While, the overall therapeutic effects of cancer vaccines have been taken into consideration as below expectations, low antigenicity of targeting antigen and tumor heterogeneity are the two key limiting barriers encountered by the cancer vaccines. Nonetheless, recent developments in cancer immune-therapies together with associated technologies, for instance the unparalleled achievements bagged by immune checkpoint inhibitor based therapies and neo-antigen identification tools, envisage potential improvements in cancer vaccines in respect to the treatments of malignancies. This review brings forth measures for the purpose of refining therapeutic cancer vaccines by learning lessons from the success of PD-1 inhibitor based immune-therapies.
- 6Lin, M. J.; Svensson-Arvelund, J.; Lubitz, G. S.; Marabelle, A.; Melero, I.; Brown, B. D.; Brody, J. D. Cancer Vaccines: The Next Immunotherapy Frontier. Nat. Cancer 2022, 3, 911– 926, DOI: 10.1038/s43018-022-00418-6Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitlCit7jE&md5=47262a65ec90f090054e453a5bae8b98Cancer vaccines: the next immunotherapy frontierLin, Matthew J.; Svensson-Arvelund, Judit; Lubitz, Gabrielle S.; Marabelle, Aurelien; Melero, Ignacio; Brown, Brian D.; Brody, Joshua D.Nature Cancer (2022), 3 (8), 911-926CODEN: NCAADQ; ISSN:2662-1347. (Springer International Publishing AG)A review. After several decades, therapeutic cancer vaccines now show signs of efficacy and potential to help patients resistant to other std.-of-care immunotherapies, but they have yet to realize their full potential and expand the oncol. armamentarium. Here, we classify cancer vaccines by what is known of the included antigens, which tumors express those antigens and where the antigens colocalize with antigen-presenting cells, thus delineating predefined vaccines (shared or personalized) and anonymous vaccines (ex vivo or in situ). To expedite clin. development, we highlight the need for accurate immune monitoring of early trials to acknowledge failures and advance the most promising vaccines.
- 7Curran, M. A.; Glisson, B. S. New Hope for Therapeutic Cancer Vaccines in the Era of Immune Checkpoint Modulation. Annu. Rev. Med. 2019, 70, 409– 424, DOI: 10.1146/annurev-med-050217-121900Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitVGksLzI&md5=58f81e23d229c445006c8672cf6b338bNew Hope for Therapeutic Cancer Vaccines in the Era of Immune Checkpoint ModulationCurran, Michael A.; Glisson, Bonnie S.Annual Review of Medicine (2019), 70 (), 409-424CODEN: ARMCAH; ISSN:0066-4219. (Annual Reviews)A review. The driver and passenger mutations accumulated in the process of malignant transformation offer an adequate spectrum of immune visible alterations to the cellular proteome and resulting peptidome to render these cancers targetable-and, in theory, rejectable-by the host T cell immune response. In addn., cancers often overexpress tissue-specific and developmental antigens to which immune tolerance is incomplete. Sometimes, virally transferred oncogenes drive malignant transformation and remain expressed throughout the cancer. Despite this state of antigenic sufficiency, cancer grows progressively and overcomes all efforts of the host immune system to contain it. While therapeutic cancer vaccination can mobilize high frequencies of tumor-specific T cells, these responses remain subject to intratumoral attenuation. Antibody modulation of T cell function through checkpoint blockade or costimulatory activation can restore survival, proliferation, and effector function to these tumor-infiltrating T cells and convert otherwise subtherapeutic vaccines into potentially curative cancer immunotherapeutics.
- 8Shae, D.; Baljon, J. J.; Wehbe, M.; Becker, K. W.; Sheehy, T. L.; Wilson, J. T. At the Bench: Engineering the Next Generation of Cancer Vaccines. J. Leukoc Biol. 2020, 108, 1435– 1453, DOI: 10.1002/JLB.5BT0119-016RGoogle Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1ShsrbN&md5=cd67858ef425a0f35c2b527fcf950ea3At the bench: Engineering the next generation of cancer vaccinesShae, Daniel; Baljon, Jessalyn J.; Wehbe, Mohamed; Becker, Kyle W.; Sheehy, Taylor L.; Wilson, John TannerJournal of Leukocyte Biology (2020), 108 (4), 1435-1453CODEN: JLBIE7; ISSN:1938-3673. (John Wiley & Sons, Inc.)A review. Cancer vaccines hold promise as an immunotherapeutic modality based on their potential to generate tumor antigen-specific T cell responses and long-lived antitumor responses capable of combating metastatic disease and recurrence. However, cancer vaccines have historically failed to deliver significant therapeutic benefit in the clinic, which we maintain is due in part to drug delivery challenges that have limited vaccine immunogenicity and efficacy. In this review, we examine some of the known and putative failure mechanisms of common first-generation clin. cancer vaccines, and describe how the rational design of materials engineered for vaccine delivery and immunomodulation can address these shortcomings. First, we outline vaccine design principles for augmenting cellular immunity to tumor antigens and describe how well-engineered materials can improve vaccine efficacy, highlighting recent innovations in vaccine delivery technol. that are primed for integration into neoantigen vaccine development pipelines. We also discuss the importance of sequencing, timing, and kinetics in mounting effective immune responses to cancer vaccines, and highlight examples of materials that potentiate antitumor immunity through spatiotemporal control of immunomodulation. Furthermore, we describe several engineering strategies for improving outcomes of in situ cancer vaccines, which leverage local, intratumoral delivery to stimulate systemic immunity. Finally, we highlight recent innovations leveraging nanotechnol. for increasing the immunogenicity of the tumor microenvironment (TME), which is crit. to enhancing tumor infiltration and function of T cells elicited in response to cancer vaccines. These immunoengineering strategies and tools complement ongoing advances in cancer vaccines as they reemerge as an important component of the immunotherapeutic armamentarium.
- 9Kleponis, J.; Skelton, R.; Zheng, L. Fueling the Engine and Releasing the Break: Combinational Therapy of Cancer Vaccines and Immune Checkpoint Inhibitors. Cancer Biol. Med. 2015, 12, 201– 208, DOI: 10.7497/j.issn.2095-3941.2015.0046Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXlslGjsbY%253D&md5=98c84619ccf7224c4d30882d091a89a5Fueling the engine and releasing the break: combinational therapy of cancer vaccines and immune checkpoint inhibitorsKleponis, Jennifer; Skelton, Richard; Zheng, LeiCancer Biology & Medicine (2015), 12 (3Spec.Iss.), 201-208CODEN: CBMADQ ISSN:. (Tianjin Medical University Cancer Institute and Hospital)Immune checkpoint inhibitors are increasingly drawing much attention in the therapeutic development for cancer treatment. However, many cancer patients do not respond to treatments with immune checkpoint inhibitors, partly because of the lack of tumor-infiltrating effector T cells. Cancer vaccines may prime patients for treatments with immune checkpoint inhibitors by inducing effector T-cell infiltration into the tumors and immune checkpoint signals. The combination of cancer vaccine and an immune checkpoint inhibitor may function synergistically to induce more effective antitumor immune responses, and clin. trials to test the combination are currently ongoing.
- 10Shemesh, C. S.; Hsu, J. C.; Hosseini, I.; Shen, B. Q.; Rotte, A.; Twomey, P.; Girish, S.; Wu, B. Personalized Cancer Vaccines: Clinical Landscape, Challenges, and Opportunities. Mol. Ther 2021, 29, 555– 570, DOI: 10.1016/j.ymthe.2020.09.038Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjsVCmsLY%253D&md5=020291133534cc7a2697ae7101562a6cPersonalized Cancer Vaccines: Clinical Landscape, Challenges, and OpportunitiesShemesh, Colby S.; Hsu, Joy C.; Hosseini, Iraj; Shen, Ben-Quan; Rotte, Anand; Twomey, Patrick; Girish, Sandhya; Wu, BenjaminMolecular Therapy (2021), 29 (2), 555-570CODEN: MTOHCK; ISSN:1525-0024. (Cell Press)A review. Tremendous innovation is underway among a rapidly expanding repertoire of promising personalized immune-based treatments. Therapeutic cancer vaccines (TCVs) are attractive systemic immunotherapies that activate and expand antigen-specific CD8+ and CD4+ T cells to enhance anti-tumor immunity. Our review highlights key issues impacting TCVs in clin. practice and reports on progress in development. We review the mechanism of action, immune-monitoring, dosing strategies, combinations, obstacles, and regulation of cancer vaccines. Most trials of personalized TCVs are ongoing and represent diverse platforms with predominantly early investigations of mRNA, DNA, or peptide-based targeting strategies against neoantigens in solid tumors, with many in combination immunotherapies. Multiple delivery systems, routes of administration, and dosing strategies are used. I.v. or i.m. administration is common, including delivery by lipid nanoparticles. Absorption and biodistribution impact antigen uptake, expression, and presentation, affecting the strength, speed, and duration of immune response. The emerging trials illustrate the complexity of developing this class of innovative immunotherapies. Methodical testing of the multiple potential factors influencing immune responses, as well as refined quant. methodologies to facilitate optimal dosing strategies, could help resolve uncertainty of therapeutic approaches. To increase the likelihood of success in bringing these medicines to patients, several unique development challenges must be overcome.
- 11Zhao, J.; Chen, Y.; Ding, Z. Y.; Liu, J. Y. Safety and Efficacy of Therapeutic Cancer Vaccines Alone or in Combination with Immune Checkpoint Inhibitors in Cancer Treatment. Front Pharmacol 2019, 10, 1184, DOI: 10.3389/fphar.2019.01184Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXpvVeqtLk%253D&md5=b9137c6f5bd286e8db5eb610319bab4bSafety and efficacy of therapeutic cancer vaccines alone or in combination with immune checkpoint inhibitors in cancer treatmentZhao, Jing; Chen, Ye; Ding, Zhen-Yu.; Liu, Ji-YanFrontiers in Pharmacology (2019), 10 (), 1184CODEN: FPRHAU; ISSN:1663-9812. (Frontiers Media S.A.)A review. Therapeutic cancer vaccines have proven to seldom induce dramatic clin. response when used alone, and therefore, they are being studied in combination with addnl. treatment modalities to achieve optimal treatment activities. Growing preclin. data show that combining vaccines and immune checkpoint inhibitors (ICIs) can prime intensified immunogenicity and modulate immunosuppressive tumor microenvironment. Herein, we focus on the safety and efficacy of approved and promising cancer vaccines alone or combined with ICIs in the treatment of several malignancies. Generally, the majority of clin. trials support the concept of synergy that combination therapy of vaccines and ICIs holds maximized potential to improve clin. outcomes. Importantly, the combination has acceptable safety and minimal addnl. toxicity compared with single-agent vaccines or ICIs. Addnl., the potential strategies of combining personalized tumor vaccines with ICIs will become priority option and future direction of vaccine development and application and the urgent need to develop effective biomarkers to screen appropriate patient populations and predict response to combination therapy.
- 12Blass, E.; Ott, P. A. Advances in the Development of Personalized Neoantigen-Based Therapeutic Cancer Vaccines. Nat. Rev. Clin Oncol 2021, 18, 215– 229, DOI: 10.1038/s41571-020-00460-2Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3srivFOqtQ%253D%253D&md5=82c721ffe87a85367bc1081598d18a5dAdvances in the development of personalized neoantigen-based therapeutic cancer vaccinesBlass Eryn; Ott Patrick A; Ott Patrick A; Ott Patrick A; Ott Patrick ANature reviews. Clinical oncology (2021), 18 (4), 215-229 ISSN:.Within the past decade, the field of immunotherapy has revolutionized the treatment of many cancers with the development and regulatory approval of various immune-checkpoint inhibitors and chimeric antigen receptor T cell therapies in diverse indications. Another promising approach to cancer immunotherapy involves the use of personalized vaccines designed to trigger de novo T cell responses against neoantigens, which are highly specific to tumours of individual patients, in order to amplify and broaden the endogenous repertoire of tumour-specific T cells. Results from initial clinical studies of personalized neoantigen-based vaccines, enabled by the availability of rapid and cost-effective sequencing and bioinformatics technologies, have demonstrated robust tumour-specific immunogenicity and preliminary evidence of antitumour activity in patients with melanoma and other cancers. Herein, we provide an overview of the complex process that is necessary to generate a personalized neoantigen vaccine, review the types of vaccine-induced T cells that are found within tumours and outline strategies to enhance the T cell responses. In addition, we discuss the current status of clinical studies testing personalized neoantigen vaccines in patients with cancer and considerations for future clinical investigation of this novel, individualized approach to immunotherapy.
- 13Schumacher, T. N.; Scheper, W.; Kvistborg, P. Cancer Neoantigens. Annu. Rev. Immunol. 2019, 37, 173– 200, DOI: 10.1146/annurev-immunol-042617-053402Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFejsLfP&md5=21b8e70354a6e489f6f56f526f2f08baCancer NeoantigensSchumacher, Ton N.; Scheper, Wouter; Kvistborg, PiaAnnual Review of Immunology (2019), 37 (), 173-200CODEN: ARIMDU; ISSN:0732-0582. (Annual Reviews)A review. Malignant transformation of cells depends on accumulation of DNA damage. Over the past years we have learned that the T cell-based immune system frequently responds to the neoantigens that arise as a consequence of this DNA damage. Furthermore, recognition of neoantigens appears an important driver of the clin. activity of both T cell checkpoint blockade and adoptive T cell therapy as cancer immunotherapies. Here we review the evidence for the relevance of cancer neoantigens in tumor control and the biol. properties of these antigens. We discuss recent technol. advances utilized to identify neoantigens, and the T cells that recognize them, in individual patients. Finally, we discuss strategies that can be employed to exploit cancer neoantigens in clin. interventions.
- 14Janes, M. E.; Gottlieb, A. P.; Park, K. S.; Zhao, Z.; Mitragotri, S. Cancer Vaccines in the Clinic. Bioeng Transl Med. 2024, 9, e10588 DOI: 10.1002/btm2.10588Google ScholarThere is no corresponding record for this reference.
- 15Baljon, J. J.; Wilson, J. T. Bioinspired Vaccines to Enhance Mhc Class-I Antigen Cross-Presentation. Curr. Opin Immunol 2022, 77, 102215, DOI: 10.1016/j.coi.2022.102215Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xhtl2jtr7M&md5=66ac8dde3eed78c8f43ac96caae336feBioinspired vaccines to enhance MHC class-I antigen cross-presentationBaljon, Jessalyn J.; Wilson, John T.Current Opinion in Immunology (2022), 77 (), 102215CODEN: COPIEL; ISSN:0952-7915. (Elsevier Ltd.)A review. Here, we briefly summarize known cross-presentation pathways and highlight how synthetic vaccines can be engineered to enhance MHC-I presentation of exogenous peptide and protein antigens by professional antigen-presenting cells (APCs). In particular, we summarize how mol. engineering and nanotechnol. are being harnessed to enhance antigen delivery to lymph nodes and to cross-presenting dendritic cells, to bypass endosomal trafficking of exogenous antigen to promote delivery of antigen to the cytosol of APCs, and to coordinate the delivery of antigen with immune-stimulating adjuvants that can act synergistically to augment antigen cross-presentation. Cross-presentation of exogenous antigen on MHC class-I is a crucial process for generating a CD8+ T cell response, and is therefore an important design consideration in the development of T-cell-engaging vaccines against viruses, intracellular bacteria, and cancers.
- 16Liu, W.; Tang, H.; Li, L.; Wang, X.; Yu, Z.; Li, J. Peptide-Based Therapeutic Cancer Vaccine: Current Trends in Clinical Application. Cell Prolif 2021, 54, e13025 DOI: 10.1111/cpr.13025Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVWrtLfK&md5=00a6a94c14b8864a77e1fcce50f5f785Peptide-based therapeutic cancer vaccine: Current trends in clinical applicationLiu, Wensi; Tang, Haichao; Li, Luanfeng; Wang, Xiangyi; Yu, Zhaojin; Li, JianpingCell Proliferation (2021), 54 (5), e13025CODEN: CPROEM; ISSN:0960-7722. (John Wiley & Sons Ltd.)A review. The peptide-based therapeutic cancer vaccines have attracted enormous attention in recent years as one of the effective treatments of tumor immunotherapy. Most of peptide-based vaccines are based on epitope peptides stimulating CD8+ T cells or CD4+ T helper cells to target tumor-assocd. antigens (TAAs) or tumor-specific antigens (TSAs). Some adjuvants and nanomaterials have been exploited to optimize the efficiency of immune response of the epitope peptide to improve its clin. application. At present, numerous peptide-based therapeutic cancer vaccines have been developed and achieved significant clin. benefits. Similarly, the combination of peptide-based vaccines and other therapies has demonstrated a superior efficacy in improving anti-cancer activity. We delve deeper into the choices of targets, design and screening of epitope peptides, clin. efficacy and adverse events of peptide-based vaccines, and strategies combination of peptide-based therapeutic cancer vaccines and other therapies. The review will provide a detailed overview and basis for future clin. application of peptide-based therapeutic cancer vaccines.
- 17Stephens, A. J.; Burgess-Brown, N. A.; Jiang, S. Beyond Just Peptide Antigens: The Complex World of Peptide-Based Cancer Vaccines. Front Immunol 2021, 12, 696791, DOI: 10.3389/fimmu.2021.696791Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFOitrvK&md5=d642fe90d7d555cd18312a800d8be80fBeyond just peptide antigens: the complex world of peptide-based cancer vaccinesStephens, Alexander J.; Burgess-Brown, Nicola A.; Jiang, ShisongFrontiers in Immunology (2021), 12 (), 696791CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)A review. Peptide-based cancer vaccines rely upon the strong activation of the adaptive immune response to elicit its effector function. They have shown to be highly specific and safe, but have yet to prove themselves as an efficacious treatment for cancer in the clinic. This is for a variety of reasons, including tumor heterogeneity, self-tolerance, and immune suppression. Importance has been placed on the overall design of peptide-based cancer vaccines, which have evolved from simple peptide derivs. of a cancer antigen, to complex drugs; incorporating overlapping regions, conjugates, and delivery systems to target and stimulate different components of antigen presenting cells, and to bolster antigen cross-presentation. Peptide-based cancer vaccines are increasingly becoming more personalised to an individuals tumor antigen repertoire and are often combined with existing cancer treatments. This strategy ultimately aids in combating the shortcomings of a more generalised vaccine strategy and provides a comprehensive treatment, taking into consideration cancer cell variability and its ability to avoid immune interrogation.
- 18Yewdell, J. W. Designing CD8+ T Cell Vaccines: It’s Not Rocket Science (Yet). Curr. Opin Immunol 2010, 22, 402– 410, DOI: 10.1016/j.coi.2010.04.002Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXntV2jsbk%253D&md5=f6f6853cd382fab6b98cb14088f25b2fDesigning CD8+ T cell vaccines: it's not rocket science (yet)Yewdell, Jonathan W.Current Opinion in Immunology (2010), 22 (3), 402-410CODEN: COPIEL; ISSN:0952-7915. (Elsevier B.V.)A review. CD8+ T cells play important roles in clearing viral infections and eradicating tumors. Designing vaccines that elicit effective CD8+ T cell responses requires a thorough knowledge of the pathways of antigen presentation in vivo. Here, I review recent progress in understanding the activation of naive CD8+ T cells in vivo, with particular emphasis on cross-priming, the presentation of protein antigens acquired by dendritic cells from their environment. With the rapid advances in this area of research, the dawn of rational vaccine design is at hand.
- 19Khong, H.; Overwijk, W. W. Adjuvants for Peptide-Based Cancer Vaccines. J. Immunother Cancer 2016, 4, 56, DOI: 10.1186/s40425-016-0160-yGoogle Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2svjtl2ntw%253D%253D&md5=caffd368c197f407b12a8b1264be8038Adjuvants for peptide-based cancer vaccinesKhong Hiep; Overwijk Willem WJournal for immunotherapy of cancer (2016), 4 (), 56 ISSN:2051-1426.Cancer therapies based on T cells have shown impressive clinical benefit. In particular, immune checkpoint blockade therapies with anti-CTLA-4 and anti-PD-1/PD-L1 are causing dramatic tumor shrinkage and prolonged patient survival in a variety of cancers. However, many patients do not benefit, possibly due to insufficient spontaneous T cell reactivity against their tumors and/or lacking immune cell infiltration to tumor site. Such tumor-specific T cell responses could be induced through anti-cancer vaccination; but despite great success in animal models, only a few of many cancer vaccine trials have demonstrated robust clinical benefit. One reason for this difference may be the use of potent, effective vaccine adjuvants in animal models, vs. the use of safe, but very weak, vaccine adjuvants in clinical trials. As vaccine adjuvants dictate the type and magnitude of the T cell response after vaccination, it is critical to understand how they work to design safe, but also effective, cancer vaccines for clinical use. Here we discuss current insights into the mechanism of action and practical application of vaccine adjuvants, with a focus on peptide-based cancer vaccines.
- 20Gouttefangeas, C.; Rammensee, H. G. Personalized Cancer Vaccines: Adjuvants Are Important, Too. Cancer Immunol Immunother 2018, 67, 1911– 1918, DOI: 10.1007/s00262-018-2158-4Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXnsVGqsrc%253D&md5=f0bb1792ca78fcf2e11b427377a3cd44Personalized cancer vaccines: adjuvants are important, tooGouttefangeas, Cecile; Rammensee, Hans-GeorgCancer Immunology Immunotherapy (2018), 67 (12), 1911-1918CODEN: CIIMDN; ISSN:0340-7004. (Springer)Therapeutic cancer vaccines have shown limited clin. efficacy so far. Nevertheless, in the meantime, our understanding of immune cell function and the interactions of immune cells with growing tumors has advanced considerably. We are now in a position to invest this knowledge into the design of more powerful vaccines and therapy combinations aimed at increasing immunogenicity and decreasing tumor-induced immunosuppression. This review focuses essentially on peptide-based human vaccines. We will discuss two aspects that are crit. for increasing their intrinsic immunogenicity: the selection of the antigen(s) to be targeted, and the as yet unmet need for strong adjuvants.
- 21Temizoz, B.; Kuroda, E.; Ishii, K. J. Vaccine Adjuvants as Potential Cancer Immunotherapeutics. Int. Immunol. 2016, 28, 329– 338, DOI: 10.1093/intimm/dxw015Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFOru7fE&md5=92aa03134406c55673017cb24da45eb4Vaccine adjuvants as potential cancer immunotherapeuticsTemizoz, Burcu; Kuroda, Etsushi; Ishii, Ken J.International Immunology (2016), 28 (7), 329-338CODEN: INIMEN; ISSN:0953-8178. (Oxford University Press)Accumulated evidence obtained from various clin. trials and animal studies suggested that cancer vaccines need better adjuvants than those that are currently licensed, which include the most commonly used alum and incomplete Freund's adjuvant, because of either a lack of potent anti-tumor immunity or the induction of undesired immunity. Several clin. trials using immunostimulatory adjuvants, particularly agonistic as well as non-agonistic ligands for TLRs, C-type lectin receptors, retinoic acid-inducible gene I-like receptors and stimulator of interferon genes, have revealed their therapeutic potential not only as vaccine adjuvants but also as anti-tumor agents. Recently, combinations of such immunostimulatory or immunomodulatory adjuvants have shown superior efficacy over their singular use, suggesting that seeking optimal combinations of the currently available or well-characterized adjuvants may provide a better chance for the development of novel adjuvants for cancer immunotherapy.
- 22Kaur, A.; Baldwin, J.; Brar, D.; Salunke, D. B.; Petrovsky, N. Toll-Like Receptor (TLR) Agonists as a Driving Force Behind Next-Generation Vaccine Adjuvants and Cancer Therapeutics. Curr. Opin Chem. Biol. 2022, 70, 102172, DOI: 10.1016/j.cbpa.2022.102172Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xhs1yitbnL&md5=3150d579bdb7406e044934c74a7af2cdToll-like receptor (TLR) agonists as a driving force behind next-generation vaccine adjuvants and cancer therapeuticsKaur, Arshpreet; Baldwin, Jeremy; Brar, Deshkanwar; Salunke, Deepak B.; Petrovsky, NikolaiCurrent Opinion in Chemical Biology (2022), 70 (), 102172CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review until recently, the development of new human adjuvants was held back by a poor understanding of their mechanisms of action. The field was revolutionized by the discovery of the toll-like receptors (TLRs), innate immune receptors that directly or indirectly are responsible for detecting pathogen-assocd. mol. patterns (PAMPs) and respond to them by activating innate and adaptive immune pathways. Hundreds of ligands targeting various TLRs have since been identified and characterized as vaccine adjuvants. This work has important implications not only for the development of vaccines against infectious diseases but also for immuno-therapies against cancer, allergy, Alzheimer's disease, drug addiction and other diseases. Each TLR has its own specific tissue localization and downstream gene pathways, providing researchers the opportunity to precisely tailor adjuvants with specific immune effects. TLR agonists can be combined with other TLR or alternative adjuvants to create combination adjuvants with synergistic or modulatory effects. This review provides an introduction to the various classes of TLR adjuvants and their resp. pathways. It provides an overview of recent advancements in the TLR field in the past 2-3 years and discusses criteria for selecting specific TLR adjuvants based on considerations, such as disease mechanisms and correlates of protection, TLR immune biasing capabilities, route of administration, antigen compatibility, new vaccine technol. platforms, and age- and species-specific effects.
- 23Hu, H. G.; Li, Y. M. Emerging Adjuvants for Cancer Immunotherapy. Front Chem. 2020, 8, 601, DOI: 10.3389/fchem.2020.00601Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitFWit7vF&md5=03aa3cb231a4e3245efde5c3e6959b61Emerging adjuvants for cancer immunotherapyHu, Hong-Guo; Li, Yan-MeiFrontiers in Chemistry (Lausanne, Switzerland) (2020), 8 (), 601CODEN: FCLSAA; ISSN:2296-2646. (Frontiers Media S.A.)A review. Cancer is a life-threatening disease, and immunotherapies have been developed as a novel, potent treatment for cancer. Adjuvants, used alone or in combination with other agents, play crucial roles in immune activation. This is necessary for cancer immunotherapy, particularly in the construction of therapeutic cancer vaccines. Adjuvants activate antigen-presenting cells and promote the presentation of antigen epitopes on major histocompatibility complex mols., further enhancing adaptive immune responses, including cytotoxic T lymphocytes, to elicit cancer-cell death. However, the applications of adjuvants are limited by their poor efficacy or insufficient safety. In recent studies, researchers attempted to develop safe, efficacious adjuvants for cancer immunotherapy, and many compds. (including inorg. compds., org. mols., polymers, and colloids) have been identified and optimized as agonists of various pathways. In this , we focus on the discovery and structural design of emerging adjuvants and discuss how these findings benefit healthcare.
- 24Paston, S. J.; Brentville, V. A.; Symonds, P.; Durrant, L. G. Cancer Vaccines, Adjuvants, and Delivery Systems. Front Immunol 2021, 12, 627932, DOI: 10.3389/fimmu.2021.627932Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXpvVyhtrg%253D&md5=3d9e90696dda37b5ab117b67a282262dCancer vaccines, adjuvants, and delivery systemsPaston, Samantha j.; Brentville, Victoria a.; Symonds, Peter; Durrant, Lindy g.Frontiers in Immunology (2021), 12 (), 627932CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)A review. Vaccination was first pioneered in the 18th century by Edward Jenner and eventually led to the development of the smallpox vaccine and subsequently the eradication of smallpox. The impact of vaccination to prevent infectious diseases has been outstanding with many infections being prevented and a significant decrease in mortality worldwide. Cancer vaccines aim to clear active disease instead of aiming to prevent disease, the only exception being the recently approved vaccine that prevents cancers caused by the Human Papillomavirus. The development of therapeutic cancer vaccines has been disappointing with many early cancer vaccines that showed promise in preclin. models often failing to translate into efficacy in the clinic. In this review we provide an overview of the current vaccine platforms, adjuvants and delivery systems that are currently being investigated or have been approved. With the advent of immune checkpoint inhibitors, we also review the potential of these to be used with cancer vaccines to improve efficacy and help to overcome the immune suppressive tumor microenvironment.
- 25Keskin, D. B.; Anandappa, A. J.; Sun, J.; Tirosh, I.; Mathewson, N. D.; Li, S.; Oliveira, G.; Giobbie-Hurder, A.; Felt, K.; Gjini, E. Neoantigen Vaccine Generates Intratumoral T Cell Responses in Phase Ib Glioblastoma Trial. Nature 2019, 565, 234– 239, DOI: 10.1038/s41586-018-0792-9Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFyksLvE&md5=665425d605411ebfd4764314e7d1f220Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trialKeskin, Derin B.; Anandappa, Annabelle J.; Sun, Jing; Tirosh, Itay; Mathewson, Nathan D.; Li, Shuqiang; Oliveira, Giacomo; Giobbie-Hurder, Anita; Felt, Kristen; Gjini, Evisa; Shukla, Sachet A.; Hu, Zhuting; Li, Letitia; Le, Phuong M.; Allesoee, Rosa L.; Richman, Alyssa R.; Kowalczyk, Monika S.; Abdelrahman, Sara; Geduldig, Jack E.; Charbonneau, Sarah; Pelton, Kristine; Iorgulescu, J. Bryan; Elagina, Liudmila; Zhang, Wandi; Olive, Oriol; McCluskey, Christine; Olsen, Lars R.; Stevens, Jonathan; Lane, William J.; Salazar, Andres M.; Daley, Heather; Wen, Patrick Y.; Chiocca, E. Antonio; Harden, Maegan; Lennon, Niall J.; Gabriel, Stacey; Getz, Gad; Lander, Eric S.; Regev, Aviv; Ritz, Jerome; Neuberg, Donna; Rodig, Scott J.; Ligon, Keith L.; Suva, Mario L.; Wucherpfennig, Kai W.; Hacohen, Nir; Fritsch, Edward F.; Livak, Kenneth J.; Ott, Patrick A.; Wu, Catherine J.; Reardon, David A.Nature (London, United Kingdom) (2019), 565 (7738), 234-239CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Neoantigens, which are derived from tumor-specific protein-coding mutations, are exempt from central tolerance, can generate robust immune responses1,2 and can function as bona fide antigens that facilitate tumor rejection3. Here we demonstrate that a strategy that uses multi-epitope, personalized neoantigen vaccination, which has previously been tested in patients with high-risk melanoma4-6, is feasible for tumors such as glioblastoma, which typically have a relatively low mutation load1,7 and an immunol. 'cold' tumor microenvironment8. We used personalized neoantigen-targeting vaccines to immunize patients newly diagnosed with glioblastoma following surgical resection and conventional radiotherapy in a phase I/Ib study. Patients who did not receive dexamethasone-a highly potent corticosteroid that is frequently prescribed to treat cerebral edema in patients with glioblastoma-generated circulating polyfunctional neoantigen-specific CD4+ and CD8+ T cell responses that were enriched in a memory phenotype and showed an increase in the no. of tumor-infiltrating T cells. Using single-cell T cell receptor anal., we provide evidence that neoantigen-specific T cells from the peripheral blood can migrate into an intracranial glioblastoma tumor. Neoantigen-targeting vaccines thus have the potential to favorably alter the immune milieu of glioblastoma.
- 26Ott, P. A.; Hu, Z.; Keskin, D. B.; Shukla, S. A.; Sun, J.; Bozym, D. J.; Zhang, W.; Luoma, A.; Giobbie-Hurder, A.; Peter, L.; Chen, C.; Olive, O.; Carter, T. A.; Li, S.; Lieb, D. J.; Eisenhaure, T.; Gjini, E.; Stevens, J.; Lane, W. J.; Javeri, I. An Immunogenic Personal Neoantigen Vaccine for Patients with Melanoma. Nature 2017, 547, 217– 221, DOI: 10.1038/nature22991Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFaqt7rO&md5=e235523b95d4f6ae4aeceb1047460ab3An immunogenic personal neoantigen vaccine for patients with melanomaOtt, Patrick A.; Hu, Zhuting; Keskin, Derin B.; Shukla, Sachet A.; Sun, Jing; Bozym, David J.; Zhang, Wandi; Luoma, Adrienne; Giobbie-Hurder, Anita; Peter, Lauren; Chen, Christina; Olive, Oriol; Carter, Todd A.; Li, Shuqiang; Lieb, David J.; Eisenhaure, Thomas; Gjini, Evisa; Stevens, Jonathan; Lane, William J.; Javeri, Indu; Nellaiappan, Kaliappanadar; Salazar, Andres M.; Daley, Heather; Seaman, Michael; Buchbinder, Elizabeth I.; Yoon, Charles H.; Harden, Maegan; Lennon, Niall; Gabriel, Stacey; Rodig, Scott J.; Barouch, Dan H.; Aster, Jon C.; Getz, Gad; Wucherpfennig, Kai; Neuberg, Donna; Ritz, Jerome; Lander, Eric S.; Fritsch, Edward F.; Hacohen, Nir; Wu, Catherine J.Nature (London, United Kingdom) (2017), 547 (7662), 217-221CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Effective anti-tumor immunity in humans has been assocd. with the presence of T cells directed at cancer neoantigens, a class of HLA-bound peptides that arise from tumor-specific mutations. They are highly immunogenic because they are not present in normal tissues and hence bypass central thymic tolerance. Although neoantigens were long-envisioned as optimal targets for an anti-tumor immune response, their systematic discovery and evaluation only became feasible with the recent availability of massively parallel sequencing for detection of all coding mutations within tumors, and of machine learning approaches to reliably predict those mutated peptides with high-affinity binding of autologous human leukocyte antigen (HLA) mols. We hypothesized that vaccination with neoantigens can both expand pre-existing neoantigen-specific T-cell populations and induce a broader repertoire of new T-cell specificities in cancer patients, tipping the intra-tumoral balance in favor of enhanced tumor control. Here we demonstrate the feasibility, safety, and immunogenicity of a vaccine that targets up to 20 predicted personal tumor neoantigens. Vaccine-induced polyfunctional CD4+ and CD8+ T cells targeted 58 (60%) and 15 (16%) of the 97 unique neoantigens used across patients, resp. These T cells discriminated mutated from wild-type antigens, and in some cases directly recognized autologous tumor. Of six vaccinated patients, four had no recurrence at 25 mo after vaccination, while two with recurrent disease were subsequently treated with anti-PD-1 (anti-programmed cell death-1) therapy and experienced complete tumor regression, with expansion of the repertoire of neoantigen-specific T cells. These data provide a strong rationale for further development of this approach, alone and in combination with checkpoint blockade or other immunotherapies.
- 27Swartz, M. A.; Hirosue, S.; Hubbell, J. A. Engineering Approaches to Immunotherapy. Science Translational Medicine 2012, 4, 148rv149, DOI: 10.1126/scitranslmed.3003763Google ScholarThere is no corresponding record for this reference.
- 28Knight, F. C.; Gilchuk, P.; Kumar, A.; Becker, K. W.; Sevimli, S.; Jacobson, M. E.; Suryadevara, N.; Wang-Bishop, L.; Boyd, K. L.; Crowe, J. E., Jr.; Joyce, S.; Wilson, J. T. Mucosal Immunization with a pH-Responsive Nanoparticle Vaccine Induces Protective CD8(+) Lung-Resident Memory T Cells. ACS Nano 2019, 13, 10939– 10960, DOI: 10.1021/acsnano.9b00326Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvVCnt7%252FO&md5=44662b0a177490dcb700c98b7fd56e5eMucosal Immunization with a pH-Responsive Nanoparticle Vaccine Induces Protective CD8+ Lung-Resident Memory T CellsKnight, Frances C.; Gilchuk, Pavlo; Kumar, Amrendra; Becker, Kyle W.; Sevimli, Sema; Jacobson, Max E.; Suryadevara, Naveenchandra; Wang-Bishop, Lihong; Boyd, Kelli L.; Crowe, James E.; Joyce, Sebastian; Wilson, John T.ACS Nano (2019), 13 (10), 10939-10960CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Tissue-resident memory T cells (TRM) patrol nonlymphoid organs and provide superior protection against pathogens that commonly infect mucosal and barrier tissues, such as the lungs, intestine, liver, and skin. Thus, there is a need for vaccine technologies that can induce a robust, protective TRM response in these tissues. Nanoparticle (NP) vaccines offer important advantages over conventional vaccines; however, there has been minimal investigation into the design of NP-based vaccines for eliciting TRM responses. Here, we describe a pH-responsive polymeric nanoparticle vaccine for generating antigen-specific CD8+ TRM cells in the lungs. With a single intranasal dose, the NP vaccine elicited airway- and lung-resident CD8+ TRM cells and protected against respiratory virus challenge in both sublethal (vaccinia) and lethal (influenza) infection models for up to 9 wk after immunization. In elucidating the contribution of material properties to the resulting TRM response, we found that the pH-responsive activity of the carrier was important, as a structurally analogous non-pH-responsive control carrier elicited significantly fewer lung-resident CD8+ T cells. We also demonstrated that dual-delivery of protein antigen and nucleic acid adjuvant on the same NP substantially enhanced the magnitude, functionality, and longevity of the antigen-specific CD8+ TRM response in the lungs. Compared to administration of sol. antigen and adjuvant, the NP also mediated retention of vaccine cargo in pulmonary antigen-presenting cells (APCs), enhanced APC activation, and increased prodn. of TRM-related cytokines. Overall, these data suggest a promising vaccine platform technol. for rapid generation of protective CD8+ TRM cells in the lungs.
- 29Kuai, R.; Ochyl, L. J.; Bahjat, K. S.; Schwendeman, A.; Moon, J. J. Designer Vaccine Nanodiscs for Personalized Cancer Immunotherapy. Nat. Mater. 2017, 16, 489– 496, DOI: 10.1038/nmat4822Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFGitLvO&md5=247bc6ef1a21f3a1bce8addcbb7a6a0dDesigner vaccine nanodiscs for personalized cancer immunotherapyKuai, Rui; Ochyl, Lukasz J.; Bahjat, Keith S.; Schwendeman, Anna; Moon, James J.Nature Materials (2017), 16 (4), 489-496CODEN: NMAACR; ISSN:1476-1122. (Nature Publishing Group)Despite the tremendous potential of peptide-based cancer vaccines, their efficacy has been limited in humans. Recent innovations in tumor exome sequencing have signalled the new era of personalized immunotherapy with patient-specific neoantigens, but a general methodol. for stimulating strong CD8α+ cytotoxic T-lymphocyte (CTL) responses remains lacking. Here we demonstrate that high-d. lipoprotein-mimicking nanodiscs coupled with antigen (Ag) peptides and adjuvants can markedly improve Ag/adjuvant co-delivery to lymphoid organs and sustain Ag presentation on dendritic cells. Strikingly, nanodiscs elicited up to 47-fold greater frequencies of neoantigen-specific CTLs than sol. vaccines and even 31-fold greater than perhaps the strongest adjuvant in clin. trials (i.e., CpG in Montanide). Moreover, multi-epitope vaccination generated broad-spectrum T-cell responses that potently inhibited tumor growth. Nanodiscs eliminated established MC-38 and B16F10 tumors when combined with anti-PD-1 and anti-CTLA-4 therapy. These findings represent a new powerful approach for cancer immunotherapy and suggest a general strategy for personalized nanomedicine.
- 30Li, A. W.; Sobral, M. C.; Badrinath, S.; Choi, Y.; Graveline, A.; Stafford, A. G.; Weaver, J. C.; Dellacherie, M. O.; Shih, T. Y.; Ali, O. A.; Kim, J.; Wucherpfennig, K. W.; Mooney, D. J. A Facile Approach to Enhance Antigen Response for Personalized Cancer Vaccination. Nat. Mater. 2018, 17, 528– 534, DOI: 10.1038/s41563-018-0028-2Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXltFOrsrs%253D&md5=3723b6388b35a6a546eb78ace8db9e9fA facile approach to enhance antigen response for personalized cancer vaccinationLi, Aileen Weiwei; Sobral, Miguel C.; Badrinath, Soumya; Choi, Youngjin; Graveline, Amanda; Stafford, Alexander G.; Weaver, James C.; Dellacherie, Maxence O.; Shih, Ting-Yu; Ali, Omar A.; Kim, Jaeyun; Wucherpfennig, Kai W.; Mooney, David J.Nature Materials (2018), 17 (6), 528-534CODEN: NMAACR; ISSN:1476-1122. (Nature Research)Existing strategies to enhance peptide immunogenicity for cancer vaccination generally require direct peptide alteration, which, beyond practical issues, may impact peptide presentation and result in vaccine variability. Here, we report a simple adsorption approach using polyethyleneimine (PEI) in a mesoporous silica microrod (MSR) vaccine to enhance antigen immunogenicity. The MSR-PEI vaccine significantly enhanced host dendritic cell activation and T-cell response over the existing MSR vaccine and bolus vaccine formulations. Impressively, a single injection of the MSR-PEI vaccine using an E7 peptide completely eradicated large, established TC-1 tumors in about 80% of mice and generated immunol. memory. When immunized with a pool of B16F10 or CT26 neoantigens, the MSR-PEI vaccine eradicated established lung metastases, controlled tumor growth and synergized with anti-CTLA4 therapy. Our findings from three independent tumor models suggest that the MSR-PEI vaccine approach may serve as a facile and powerful multi-antigen platform to enable robust personalized cancer vaccination.
- 31Liu, H.; Moynihan, K. D.; Zheng, Y.; Szeto, G. L.; Li, A. V.; Huang, B.; Van Egeren, D. S.; Park, C.; Irvine, D. J. Structure-Based Programming of Lymph-Node Targeting in Molecular Vaccines. Nature 2014, 507, 519– 522, DOI: 10.1038/nature12978Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXkvV2ru70%253D&md5=2890c3d27d8b989930052ba6bbed3294Structure-based programming of lymph-node targeting in molecular vaccinesLiu, Haipeng; Moynihan, Kelly D.; Zheng, Yiran; Szeto, Gregory L.; Li, Adrienne V.; Huang, Bonnie; Van Egeren, Debra S.; Park, Clara; Irvine, Darrell J.Nature (London, United Kingdom) (2014), 507 (7493), 519-522CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)In cancer patients, visual identification of sentinel lymph nodes (LNs) is achieved by the injection of dyes that bind avidly to endogenous albumin, targeting these compds. to LNs, where they are efficiently filtered by resident phagocytes. Here we translate this 'albumin hitchhiking' approach to mol. vaccines, through the synthesis of amphiphiles (amph-vaccines) comprising an antigen or adjuvant cargo linked to a lipophilic albumin-binding tail by a soly.-promoting polar polymer chain. Administration of structurally optimized CpG-DNA/peptide amph-vaccines in mice resulted in marked increases in LN accumulation and decreased systemic dissemination relative to their parent compds., leading to 30-fold increases in T-cell priming and enhanced anti-tumor efficacy while greatly reducing systemic toxicity. Amph-vaccines provide a simple, broadly applicable strategy to simultaneously increase the potency and safety of subunit vaccines.
- 32Lynn, G. M.; Sedlik, C.; Baharom, F.; Zhu, Y.; Ramirez-Valdez, R. A.; Coble, V. L.; Tobin, K.; Nichols, S. R.; Itzkowitz, Y.; Zaidi, N.; Gammon, J. M.; Blobel, N. J.; Denizeau, J.; de la Rochere, P.; Francica, B. J.; Decker, B.; Maciejewski, M.; Cheung, J.; Yamane, H.; Smelkinson, M. G. Peptide-TLR-7/8a Conjugate Vaccines Chemically Programmed for Nanoparticle Self-Assembly Enhance CD8 T-Cell Immunity to Tumor Antigens. Nat. Biotechnol. 2020, 38, 320– 332, DOI: 10.1038/s41587-019-0390-xGoogle Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXotFGltw%253D%253D&md5=2b4321aa1d0eec8e7a4b2d5aca33a648Peptide-TLR-7/8a conjugate vaccines chemically programmed for nanoparticle self-assembly enhance CD8 T-cell immunity to tumor antigensLynn, Geoffrey M.; Sedlik, Christine; Baharom, Faezzah; Zhu, Yaling; Ramirez-Valdez, Ramiro A.; Coble, Vincent L.; Tobin, Kennedy; Nichols, Sarah R.; Itzkowitz, Yaakov; Zaidi, Neeha; Gammon, Joshua M.; Blobel, Nicolas J.; Denizeau, Jordan; de la Rochere, Philippe; Francica, Brian J.; Decker, Brennan; Maciejewski, Mateusz; Cheung, Justin; Yamane, Hidehiro; Smelkinson, Margery G.; Francica, Joseph R.; Laga, Richard; Bernstock, Joshua D.; Seymour, Leonard W.; Drake, Charles G.; Jewell, Christopher M.; Lantz, Olivier; Piaggio, Eliane; Ishizuka, Andrew S.; Seder, Robert A.Nature Biotechnology (2020), 38 (3), 320-332CODEN: NABIF9; ISSN:1087-0156. (Nature Research)Personalized cancer vaccines targeting patient-specific neoantigens are a promising cancer treatment modality; however, neoantigen physicochem. variability can present challenges to manufg. personalized cancer vaccines in an optimal format for inducing anticancer T cells. Here, we developed a vaccine platform (SNP-7/8a) based on charge-modified peptide-TLR-7/8a conjugates that are chem. programmed to self-assemble into nanoparticles of uniform size (∼20 nm) irresp. of the peptide antigen compn. This approach provided precise loading of diverse peptide neoantigens linked to TLR-7/8a (adjuvant) in nanoparticles, which increased uptake by and activation of antigen-presenting cells that promote T-cell immunity. Vaccination of mice with SNP-7/8a using predicted neoantigens (n = 179) from three tumor models induced CD8 T cells against ∼50% of neoantigens with high predicted MHC-I binding affinity and led to enhanced tumor clearance. SNP-7/8a delivering in silico-designed mock neoantigens also induced CD8 T cells in nonhuman primates. Altogether, SNP-7/8a is a generalizable approach for codelivering peptide antigens and adjuvants in nanoparticles for inducing anticancer T-cell immunity.
- 33Shae, D.; Baljon, J. J.; Wehbe, M.; Christov, P. P.; Becker, K. W.; Kumar, A.; Suryadevara, N.; Carson, C. S.; Palmer, C. R.; Knight, F. C.; Joyce, S.; Wilson, J. T. Co-Delivery of Peptide Neoantigens and Stimulator of Interferon Genes Agonists Enhances Response to Cancer Vaccines. ACS Nano 2020, 14, 9904– 9916, DOI: 10.1021/acsnano.0c02765Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVCmsLjL&md5=e52e9db129f9916ccbb51b96ead541fcCo-delivery of Peptide Neoantigens and Stimulator of Interferon Genes Agonists Enhances Response to Cancer VaccinesShae, Daniel; Baljon, Jessalyn J.; Wehbe, Mohamed; Christov, Plamen P.; Becker, Kyle W.; Kumar, Amrendra; Suryadevara, Naveenchandra; Carson, Carcia S.; Palmer, Christian R.; Knight, Frances C.; Joyce, Sebastian; Wilson, John T.ACS Nano (2020), 14 (8), 9904-9916CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Cancer vaccines targeting patient-specific neoantigens have emerged as a promising strategy for improving responses to immune checkpoint blockade. However, neoantigenic peptides are poorly immunogenic and inept at stimulating CD8+ T cell responses, motivating a need for new vaccine technologies that enhance their immunogenicity. The stimulator of interferon genes (STING) pathway is an endogenous mechanism by which the innate immune system generates an immunol. context for priming and mobilizing neoantigen-specific T cells. Owing to this crit. role in tumor immune surveillance, a synthetic cancer nanovaccine platform (nanoSTING-vax) was developed that mimics immunogenic cancer cells in its capacity to efficiently promote co-delivery of peptide antigens and the STING agonist, cGAMP. The co-loading of cGAMP and peptides into pH-responsive, endosomolytic polymersomes promoted the coordinated delivery of both cGAMP and peptide antigens to the cytosol, thereby eliciting inflammatory cytokine prodn., co-stimulatory marker expression, and antigen cross-presentation. Consequently, nanoSTING-vax significantly enhanced CD8+ T cell responses to a range of peptide antigens. Therapeutic immunization with nanoSTING-vax, in combination with immune checkpoint blockade, inhibited tumor growth in multiple murine tumor models, even leading to complete tumor rejection and generation of durable antitumor immune memory. Collectively, this work establishes nanoSTING-vax as a versatile platform for enhancing immune responses to neoantigen-targeted cancer vaccines.
- 34Tornesello, A. L.; Tagliamonte, M.; Tornesello, M. L.; Buonaguro, F. M.; Buonaguro, L. Nanoparticles to Improve the Efficacy of Peptide-Based Cancer Vaccines. Cancers (Basel) 2020, 12, 1049, DOI: 10.3390/cancers12041049Google ScholarThere is no corresponding record for this reference.
- 35Viswanath, D. I.; Liu, H. C.; Huston, D. P.; Chua, C. Y. X.; Grattoni, A. Emerging Biomaterial-Based Strategies for Personalized Therapeutic in Situ Cancer Vaccines. Biomaterials 2022, 280, 121297, DOI: 10.1016/j.biomaterials.2021.121297Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXislWkurfI&md5=6c16a058af7c31ba986ce1a85033ee82Emerging biomaterial-based strategies for personalized therapeutic in situ cancer vaccinesViswanath, Dixita Ishani; Liu, Hsuan-Chen; Huston, David P.; Chua, Corrine Ying Xuan; Grattoni, AlessandroBiomaterials (2022), 280 (), 121297CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)A review. Landmark successes in oncoimmunol. have led to development of therapeutics boosting the host immune system to eradicate local and distant tumors with impactful tumor redn. in a subset of patients. However, current immunotherapy modalities often demonstrate limited success when involving immunol. cold tumors and solid tumors. Here, we describe the role of various biomaterials to formulate cancer vaccines as a form of cancer immunotherapy, seeking to utilize the host immune system to activate and expand tumor-specific T cells. Biomaterial-based cancer vaccines enhance the cancer-immunity cycle by harnessing cellular recruitment and activation against tumor-specific antigens. In this review, we discuss biomaterial-based vaccine strategies to induce lymphocytic responses necessary to mediate anti-tumor immunity. We focus on strategies that selectively attract dendritic cells via immunostimulatory gradients, activate them against presented tumor-specific antigens, and induce effective cross-presentation to T cells in secondary lymphoid organs, thereby generating immunity. We posit that personalized cancer vaccines are promising targets to generate long-term systemic immunity against patient- and tumor-specific antigens to ensure long-term cancer remission.
- 36Wu, S.; Xia, Y.; Hu, Y.; Ma, G. Bio-Mimic Particles for the Enhanced Vaccinations: Lessons Learnt from the Natural Traits and Pathogenic Invasion. Adv. Drug Deliv Rev. 2021, 176, 113871, DOI: 10.1016/j.addr.2021.113871Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhs1KgsL7O&md5=106fa4408f1ebb0c26ae41f4a4c8df36Bio-mimic particles for the enhanced vaccinations: Lessons learnt from the natural traits and pathogenic invasionWu, Sihua; Xia, Yufei; Hu, Yuning; Ma, GuanghuiAdvanced Drug Delivery Reviews (2021), 176 (), 113871CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)A review. In the combat against pathogens, the immune systems were evolved with the immune recognitions against the various danger signals, which responded vigorously upon the pathogen invasions and elicited potent antibodies or T cell engagement against the re-infections. Envisage with the prevailing pandemics and increasing demands for cancer vaccines, bio-mimic particles were developed to imitate the natural traits of the pathogens, which conferred the optimal strategies to stimulate the immune engagement and let to the increased vaccine efficacy. Here, the recent development in bio-mimic particles, as well as the natural cues from the pathogens were discussed. As such, the designing principles that adapted from the physiochem. properties of the pathogens were unfolded as the surface characteristics (hydrophobic, nano-pattern, antigen display, charge), properties (size, shape, softness) and the delivered components (peptide, protein, nuclear acids, toll-like receptor (TLR) agonist, antibody). Addnl., the strategies for the efficient delivery, regarding the biodistribution, internalization and presentation of the antigens were also illustrated. Through reviewing the state-of-art in biomimetic particles, the lesson learnt from the natural traits and pathogenic invasion may shed light on the rational design for the enhanced vaccinations.
- 37Rosenthal, J. A.; Chen, L.; Baker, J. L.; Putnam, D.; DeLisa, M. P. Pathogen-Like Particles: Biomimetic Vaccine Carriers Engineered at the Nanoscale. Curr. Opin Biotechnol 2014, 28, 51– 58, DOI: 10.1016/j.copbio.2013.11.005Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtF2ktLfK&md5=e2b314a29050e3a7f370dcf0137aa250Pathogen-like particles: biomimetic vaccine carriers engineered at the nanoscaleRosenthal, Joseph A.; Chen, Linxiao; Baker, Jenny L.; Putnam, David; DeLisa, Matthew P.Current Opinion in Biotechnology (2014), 28 (), 51-58CODEN: CUOBE3; ISSN:0958-1669. (Elsevier B.V.)Vaccine adjuvants are an essential component of vaccine design, helping to generate immunity to pathogen antigens in the absence of infection. Recent advances in nanoscale engineering have created a new class of particulate bionanotechnol. that uses biomimicry to better integrate adjuvant and antigen. These pathogen-like particles, or PLPs, can come from a variety of sources, ranging from fully synthetic platforms to biol. derived, self-assembling systems. By employing molecularly engineered targeting and stimulation of key immune cells, recent studies utilizing PLPs as vaccine delivery platforms have shown great promise against high-impact, unsolved vaccine targets ranging from bacterial and viral pathogens to cancer and addiction.
- 38Gutjahr, A.; Papagno, L.; Nicoli, F.; Kanuma, T.; Kuse, N.; Cabral-Piccin, M. P.; Rochereau, N.; Gostick, E.; Lioux, T.; Perouzel, E. The STING Ligand cGamp Potentiates the Efficacy of Vaccine-Induced CD8+ T Cells. JCI Insight 2019, 4 (7), e125107, DOI: 10.1172/jci.insight.125107Google ScholarThere is no corresponding record for this reference.
- 39He, Y.; Hong, C.; Fletcher, S. J.; Berger, A. G.; Sun, X.; Yang, M.; Huang, S.; Belcher, A. M.; Irvine, D. J.; Li, J.; Hammond, P. T. Peptide-Based Cancer Vaccine Delivery Via the STINGΔTM-cGamp Complex. Adv. Healthc Mater. 2022, 11, e2200905 DOI: 10.1002/adhm.202200905Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsFGqt7jK&md5=48caf8a595d73d5693dcfc610067baf7Peptide-Based Cancer Vaccine Delivery via the STINGΔTM-cGAMP ComplexHe, Yanpu; Hong, Celestine; Fletcher, Samantha J.; Berger, Adam G.; Sun, Xin; Yang, Mengdi; Huang, Shengnan; Belcher, Angela M.; Irvine, Darrell J.; Li, Jiahe; Hammond, Paula T.Advanced Healthcare Materials (2022), 11 (15), 2200905CODEN: AHMDBJ; ISSN:2192-2640. (Wiley-VCH Verlag GmbH & Co. KGaA)With the advent of bioinformatic tools in efficiently predicting neo-antigens, peptide vaccines have gained tremendous attention in cancer immunotherapy. However, the delivery of peptide vaccines remains a major challenge, primarily due to ineffective transport to lymph nodes and low immunogenicity. Here, a strategy for peptide vaccine delivery is reported by first fusing the peptide to the cytosolic domain of the stimulator of interferon genes protein (STINGΔTM), then complexing the peptide-STINGΔTM protein with STING agonist 2'3' cyclic guanosine monophosphate-adenosine monophosphate (cGAMP). The process results in the formation of self-assembled cGAMP-peptide-STINGΔTM tetramers, which enables efficient lymphatic trafficking of the peptide. Moreover, the cGAMP-STINGΔTM complex acts not only as a protein carrier for the peptide, but also as a potent adjuvant capable of triggering STING signaling independent of endogenous STING protein-an esp. important attribute considering that certain cancer cells epigenetically silence their endogenous STING expression. With model antigen SIINFEKL, it is demonstrated that the platform elicits effective STING signaling in vitro, draining lymph node targeting in vivo, effective T cell priming in vivo as well as antitumoral immune response in a mouse colon carcinoma model, providing a versatile soln. to the challenges faced in peptide vaccine delivery.
- 40Li, X. D.; Wu, J.; Gao, D.; Wang, H.; Sun, L.; Chen, Z. J. Pivotal Roles of cGAS-cGAMP Signaling in Antiviral Defense and Immune Adjuvant Effects. Science 2013, 341, 1390– 1394, DOI: 10.1126/science.1244040Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVCmu7vK&md5=46c48534439d7b6442911d63f7c3fa51Pivotal Roles of cGAS-cGAMP Signaling in Antiviral Defense and Immune Adjuvant EffectsLi, Xiao-Dong; Wu, Jiaxi; Gao, Daxing; Wang, Hua; Sun, Lijun; Chen, Zhijian J.Science (Washington, DC, United States) (2013), 341 (6152), 1390-1394CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Invasion of microbial DNA into the cytoplasm of animal cells triggers a cascade of host immune reactions that help clear the infection; however, self DNA in the cytoplasm can cause autoimmune diseases. Biochem. approaches led to the identification of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) as a cytosolic DNA sensor that triggers innate immune responses. Here, we show that cells from cGAS-deficient (cGas-/-) mice, including fibroblasts, macrophages, and dendritic cells, failed to produce type I interferons and other cytokines in response to DNA transfection or DNA virus infection. cGas-/- mice were more susceptible to lethal infection with herpes simplex virus 1 (HSV1) than wild-type mice. We also show that cGAMP is an adjuvant that boosts antigen-specific T cell activation and antibody prodn. in mice.
- 41Van Herck, S.; Feng, B.; Tang, L. Delivery of STING Agonists for Adjuvanting Subunit Vaccines. Adv. Drug Deliv Rev. 2021, 179, 114020, DOI: 10.1016/j.addr.2021.114020Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVyrsrnM&md5=991bac587ff443dcb8ebbbdef18d31aeDelivery of STING agonists for adjuvanting subunit vaccinesVan Herck, Simon; Feng, Bing; Tang, LiAdvanced Drug Delivery Reviews (2021), 179 (), 114020CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)A review. Adjuvant is an essential component in subunit vaccines. Many agonists of pathogen recognition receptors have been developed as potent adjuvants to optimize the immunogenicity and efficacy of vaccines. Recently discovered cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway has attracted much attention as it is a key mediator for modulating immune responses. Vaccines adjuvanted with STING agonists are found to mediate a robust immune defense against infections and cancer. In this review, we first discuss the mechanisms of STING agonists in the context of vaccination. Next, we present recent progress in novel STING agonist discovery and the delivery strategies. We next highlight recent work in optimizing the efficacy while minimizing toxicity of STING agonist-assisted subunit vaccines for protection against infectious diseases or treatment of cancer. Finally, we share our perspectives of current issues and future directions in further developing STING agonists for adjuvanting subunit vaccines.
- 42Embgenbroich, M.; Burgdorf, S. Current Concepts of Antigen Cross-Presentation. Front Immunol 2018, 9, 1643, DOI: 10.3389/fimmu.2018.01643Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlSisLvP&md5=c6d5b139383717e9d5ed31dafc8a55d3Current concepts of antigen cross-presentationEmbgenbroich, Maria; Burgdorf, SvenFrontiers in Immunology (2018), 9 (), 1643/1-1643/10CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)Dendritic cells have the ability to efficiently present internalized antigens on major histocompatibility complex (MHC) I mols. This process is termed cross-presentation and is important role in the generation of an immune response against viruses and tumors, after vaccinations or in the induction of immune tolerance. The mol. mechanisms enabling cross-presentation have been topic of intense debate since many years. However, a clear view on these mechanisms remains difficult, partially due to important remaining questions, controversial results and discussions. Here, we give an overview of the current concepts of antigen cross-presentation and focus on a description of the major cross-presentation pathways, the role of retarded antigen degrdn. for efficient cross-presentation, the dislocation of antigens from endosomal compartment into the cytosol, the reverse transport of proteasome-derived peptides for loading on MHC I and the translocation of the cross-presentation machinery from the ER to endosomes. We try to highlight recent advances, discuss some of the controversial data and point out some of the major open questions in the field.
- 43Tom, J. K.; Albin, T. J.; Manna, S.; Moser, B. A.; Steinhardt, R. C.; Esser-Kahn, A. P. Applications of Immunomodulatory Immune Synergies to Adjuvant Discovery and Vaccine Development. Trends Biotechnol 2019, 37, 373– 388, DOI: 10.1016/j.tibtech.2018.10.004Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitV2itbrN&md5=c5510e87652758ab52910904679d93f2Applications of Immunomodulatory Immune Synergies to Adjuvant Discovery and Vaccine DevelopmentTom, Janine K.; Albin, Tyler J.; Manna, Saikat; Moser, Brittany A.; Steinhardt, Rachel C.; Esser-Kahn, Aaron P.Trends in Biotechnology (2019), 37 (4), 373-388CODEN: TRBIDM; ISSN:0167-7799. (Elsevier Ltd.)A review. Pathogens comprise a diverse set of immunostimulatory mols. that activate the innate immune system during infection. The immune system recognizes distinct combinations of pathogenic mols. leading to multiple immune activation events that cooperate to produce enhanced immune responses, known as 'immune synergies'. Effective immune synergies are essential for the clearance of pathogens, thus inspiring novel adjuvant design to improve vaccines. We highlight current vaccine adjuvants and the importance of immune synergies to adjuvant and vaccine design. The focus is on new technologies used to study and apply immune synergies to adjuvant and vaccine development. Finally, we discuss how recent findings can be applied to the future design and characterization of synergistic adjuvants and vaccines.
- 44Collier, M. A.; Junkins, R. D.; Gallovic, M. D.; Johnson, B. M.; Johnson, M. M.; Macintyre, A. N.; Sempowski, G. D.; Bachelder, E. M.; Ting, J. P.; Ainslie, K. M. Acetalated Dextran Microparticles for Codelivery of STING and TLR7/8 Agonists. Mol. Pharmaceutics 2018, 15, 4933– 4946, DOI: 10.1021/acs.molpharmaceut.8b00579Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvVertLvM&md5=72cfae51f29f59ae7c4a6b11a36d7acbAcetalated Dextran Microparticles for Codelivery of STING and TLR7/8 AgonistsCollier, Michael A.; Junkins, Robert D.; Gallovic, Matthew D.; Johnson, Brandon M.; Johnson, Monica M.; Macintyre, Andrew N.; Sempowski, Gregory D.; Bachelder, Eric M.; Ting, Jenny P.-Y.; Ainslie, Kristy M.Molecular Pharmaceutics (2018), 15 (11), 4933-4946CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Vaccines are the most effective tool for preventing infectious diseases; however, subunit vaccines, considered the safest type, suffer from poor immunogenicity and require adjuvants to create a strong and sustained immune response. As adjuvants, pathogen-assocd. mol. patterns (PAMPs) offer potent immunostimulatory properties and defined mechanisms of action through their cognate pattern recognition receptors (PRRs). Their activity can be further enhanced through combining two or more PAMPs, particularly those that activate multiple immune signaling pathways. However, the cytosolic localization of many PRRs requires intracellular delivery of PAMPs for optimal biol. activity, which is particularly true of the stimulator of interferon genes (STING) PRR. Using acetalated dextran (Ace-DEX) microparticles (MPs) encapsulating STING agonist 3'3'-cyclic GMP-AMP (cGAMP) combined with sol. PAMPS, we screened the effect of codelivery of adjuvants using primary mouse bone marrow derived dendritic cells (BMDCs). We identified that codelivery of cGAMP MPs and sol. Toll-like receptor 7/8 (TLR7/8) agonist resiquimod (R848) elicited the broadest cytokine response. CGAMP and R848 were then coencapsulated within Ace-DEX MPs via electrospray. Using the model antigen ovalbumin, we obsd. that Ace-DEX MPs coencapsulating cGAMP and R848 (cGAMP/R848 Ace-DEX MPs) induced antigen-specific cellular immunity, and a balanced Th1/Th2 humoral response that was greater than cGAMP Ace-DEX MPs alone and PAMPs delivered in sep. MPs. These data indicate that polymeric Ace-DEX MPs loaded with STING and TLR7/8 agonists represent a potent cellular and humoral vaccine adjuvant.
- 45Barman, S.; Borriello, F.; Brook, B.; Pietrasanta, C.; De Leon, M.; Sweitzer, C.; Menon, M.; van Haren, S. D.; Soni, D.; Saito, Y.; Nanishi, E.; Yi, S.; Bobbala, S.; Levy, O.; Scott, E. A.; Dowling, D. J. Shaping Neonatal Immunization by Tuning the Delivery of Synergistic Adjuvants Via Nanocarriers. ACS Chem. Biol. 2022, 17, 2559– 2571, DOI: 10.1021/acschembio.2c00497Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xit1alsb3K&md5=779648f1160a984f3c8b7b98026bf77cShaping Neonatal Immunization by Tuning the Delivery of Synergistic Adjuvants via NanocarriersBarman, Soumik; Borriello, Francesco; Brook, Byron; Pietrasanta, Carlo; De Leon, Maria; Sweitzer, Cali; Menon, Manisha; van Haren, Simon D.; Soni, Dheeraj; Saito, Yoshine; Nanishi, Etsuro; Yi, Sijia; Bobbala, Sharan; Levy, Ofer; Scott, Evan A.; Dowling, David J.ACS Chemical Biology (2022), 17 (9), 2559-2571CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Adjuvanted nanocarrier-based vaccines hold substantial potential for applications in novel early-life immunization strategies. Here, via mouse and human age-specific in vitro modeling, we identified the combination of a small-mol. STING agonist (2'3'-cyclic GMP-AMP, cGAMP) and a TLR7/8 agonist (CL075) to drive the synergistic activation of neonatal dendritic cells and precision CD4 T-helper (Th) cell expansion via the IL-12/IFNγ axis. We further demonstrate that the vaccination of neonatal mice with quadrivalent influenza recombinant hemagglutinin (rHA) and an admixt. of two polymersome (PS) nanocarriers sep. encapsulating cGAMP (cGAMP-PS) and CL075 (CL075-PS) drove robust Th1 bias, high frequency of T follicular helper (TFH) cells, and germinal center (GC) B cells along with the IgG2c-skewed humoral response in vivo. Dual-loaded cGAMP/CL075-PSs did not outperform admixed cGAMP-PS and CL075-PS in vivo. These data validate an optimally designed adjuvantation system via age-selected small-mol. synergy and a multicomponent nanocarrier formulation as an effective approach to induce type 1 immune responses in early life.
- 46Kuai, R.; Sun, X.; Yuan, W.; Ochyl, L. J.; Xu, Y.; Hassani Najafabadi, A.; Scheetz, L.; Yu, M. Z.; Balwani, I.; Schwendeman, A.; Moon, J. J. Dual TLR Agonist Nanodiscs as a Strong Adjuvant System for Vaccines and Immunotherapy. J. Controlled Release 2018, 282, 131– 139, DOI: 10.1016/j.jconrel.2018.04.041Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXosFWrtr0%253D&md5=00b84399e940517a6e19c016bd9fc318Dual TLR agonist nanodiscs as a strong adjuvant system for vaccines and immunotherapyKuai, Rui; Sun, Xiaoqi; Yuan, Wenmin; Ochyl, Lukasz J.; Xu, Yao; Hassani Najafabadi, Alireza; Scheetz, Lindsay; Yu, Min-Zhi; Balwani, Ishina; Schwendeman, Anna; Moon, James J.Journal of Controlled Release (2018), 282 (), 131-139CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Recent studies have shown that certain combinations of Toll-like receptor (TLR) agonists can induce synergistic immune activation. However, it remains challenging to achieve such robust responses in vivo in a manner that is effective, facile, and amenable for clin. translation. Here, we show that MPLA, a TLR4 agonist, and CpG, a TLR9 agonist, can be efficiently co-loaded into synthetic high-d. lipoprotein nanodiscs, forming a potent adjuvant system (ND-MPLA/CpG) that can be readily combined with a variety of subunit antigens, including proteins and peptides. ND-MPLA/CpG significantly enhanced activation of dendritic cells, compared with free dual adjuvants or nanodiscs delivering a single TLR agonist. Importantly, mice immunized with phys. mixts. of protein antigens ND-MPLA/CpG generated strong humoral responses, including induction of IgG responses against protein convertase subtilisin/kexin 9 (PCSK9), leading to 17-30% redn. of the total plasma cholesterol levels. Moreover, ND-MPLA/CpG exerted strong anti-tumor efficacy in multiple murine tumor models. Compared with free adjuvants, ND-MPLA/CpG admixed with ovalbumin markedly improved antigen-specific CD8+ T cell responses by 8-fold and promoted regression of B16F10-OVA melanoma (P<0.0001). Furthermore, ND-MPLA/CpG admixed with E7 peptide antigen elicited ∼20% E7-specific CD8+ T cell responses and achieved complete regression of established TC-1 tumors in all treated animals. Taken together, our work highlights the simplicity, versatility, and potency of dual TLR agonist nanodiscs for applications in vaccines and cancer immunotherapy.
- 47Zhang, B. D.; Wu, J. J.; Li, W. H.; Hu, H. G.; Zhao, L.; He, P. Y.; Zhao, Y. F.; Li, Y. M. STING and TLR7/8 Agonists-Based Nanovaccines for Synergistic Antitumor Immune Activation. Nano Res. 2022, 15, 6328– 6339, DOI: 10.1007/s12274-022-4282-xGoogle Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtVeitLbO&md5=0103b8d2efebe7669675a9d6ee1e51b5STING and TLR7/8 agonists-based nanovaccines for synergistic antitumor immune activationZhang, Bo-Dou; Wu, Jun-Jun; Li, Wen-Hao; Hu, Hong-Guo; Zhao, Lang; He, Pei-Yang; Zhao, Yu-Fen; Li, Yan-MeiNano Research (2022), 15 (7), 6328-6339CODEN: NRAEB5; ISSN:1998-0000. (Springer GmbH)Immunostimulatory therapies based on pattern recognition receptors (PRRs) have emerged as an effective approach in the fight against cancer, with the ability to recruit tumor-specific lymphocytes in a low-immunogenicity tumor environment. The agonist cyclic dinucleotides (CDNs) of the stimulator of interferon gene (STING) are a group of very promising anticancer mols. that increase tumor immunogenicity by activating innate immunity. However, the tumor immune efficacy of CDNs is limited by several factors, including relatively narrow cytokine prodn., inefficient delivery to STING, and rapid clearance. In addn., a single adjuvant mol. is unable to elicit a broad cytokine response and thus cannot further amplify the anticancer effect. To address this problem, two or more agonist mols. are often used together to synergistically enhance immune efficacy. In this work, we found that a combination of the STING agonist CDGSF and the Toll-like receptor 7/8 (TLR7/8) agonist 522 produced a broader cytokine response. Subsequently, we developed multicomponent nanovaccines (MCNVs) consisting of a PC7A polymer as a nanocarrier encapsulating the antigen OVA and adjuvant mols. These MCNVs activate bone marrow-derived dendritic cells (BMDCs) to produce multiple proinflammatory factors that promote antigen cross-presentation to stimulate specific antitumor T-cell responses. In in vivo expts., we obsd. that MCNVs triggered a strong T-cell response in tumor-infiltrating lymphocytes, resulting in significant tumor regression and, notably, a 100% survival rate in mice through 25 days without other partnering therapies. These data suggest that our nanovaccines have great potential to advance cancer immunotherapy with increased durability and potency.
- 48Pradhan, P.; Toy, R.; Jhita, N.; Atalis, A.; Pandey, B.; Beach, A.; Blanchard, E. L.; Moore, S. G.; Gaul, D. A.; Santangelo, P. J. TRAF6-IRF5 Kinetics, TRIF, and Biophysical Factors Drive Synergistic Innate Responses to Particle-Mediated MPLA-CpG Co-Presentation. Sci. Adv. 2021, 7, abd4235, DOI: 10.1126/sciadv.abd4235Google ScholarThere is no corresponding record for this reference.
- 49Temizoz, B.; Kuroda, E.; Ohata, K.; Jounai, N.; Ozasa, K.; Kobiyama, K.; Aoshi, T.; Ishii, K. J. TLR9 and STING Agonists Synergistically Induce Innate and Adaptive Type-II IFN. Eur. J. Immunol. 2015, 45, 1159– 1169, DOI: 10.1002/eji.201445132Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXks12gsr4%253D&md5=01e7129b3bc00d09165a64e1fa103859TLR9 and STING agonists synergistically induce innate and adaptive type-II IFNTemizoz, Burcu; Kuroda, Etsushi; Ohata, Keiichi; Jounai, Nao; Ozasa, Koji; Kobiyama, Kouji; Aoshi, Taiki; Ishii, Ken J.European Journal of Immunology (2015), 45 (4), 1159-1169CODEN: EJIMAF; ISSN:0014-2980. (Wiley-VCH Verlag GmbH & Co. KGaA)Agonists for TLR9 and Stimulator of IFN Gene (STING) act as vaccine adjuvants that induce type-1 immune responses. However, currently available CpG oligodeoxynucleotide (ODN) (K-type) induces IFNs only weakly and STING ligands rather induce type-2 immune responses, limiting their potential therapeutic applications. Here, we show a potent synergism between TLR9 and STING agonists. Together, they make an effective type-1 adjuvant and an anticancer agent. The synergistic effect between CpG ODN (K3) and STING-ligand cyclic GMP-AMP (cGAMP), culminating in NK cell IFN-γ (type-II IFN) prodn., is due to the concurrent effects of IL-12 and type-I IFNs, which are differentially regulated by IRF3/7, STING, and MyD88. The combination of CpG ODN with cGAMP is a potent type-1 adjuvant, capable of inducing strong Th1-type responses, as demonstrated by enhanced antigen-specific IgG2c and IFN-γ prodn., as well as cytotoxic CD8+ T-cell responses. In our murine tumor models, intratumoral injection of CpG ODN and cGAMP together reduced tumor size significantly compared with the singular treatments, acting as an antigen-free anticancer agent. Thus, the combination of CpG ODN and a STING ligand may offer therapeutic application as a potent type-II IFN inducer.
- 50Kim, J. Y.; Rosenberger, M. G.; Chen, S.; Ip, C. K.; Bahmani, A.; Chen, Q.; Shen, J.; Tang, Y.; Wang, A.; Kenna, E.; Son, M.; Tay, S.; Ferguson, A. L.; Esser-Kahn, A. P. Discovery of New States of Immunomodulation for Vaccine Adjuvants Via High Throughput Screening: Expanding Innate Responses to PRRs. ACS Cent Sci. 2023, 9, 427– 439, DOI: 10.1021/acscentsci.2c01351Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXjs1ygtb4%253D&md5=b2ef1701e544feba43084f5fc8b5d3adDiscovery of New States of Immunomodulation for Vaccine Adjuvants via High Throughput Screening: Expanding Innate Responses to PRRsKim, Jeremiah Y.; Rosenberger, Matthew G.; Chen, Siquan; IP, Carman KM; Bahmani, Azadeh; Chen, Qing; Shen, Jinjing; Tang, Yifeng; Wang, Andrew; Kenna, Emma; Son, Minjun; Tay, Savas; Ferguson, Andrew L.; Esser-Kahn, Aaron P.ACS Central Science (2023), 9 (3), 427-439CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)Stimulation of the innate immune system is crucial in both effective vaccinations and immunotherapies. This is often achieved through adjuvants, mols. that usually activate pattern recognition receptors (PRRs) and stimulate two innate immune signaling pathways: the nuclear factor kappa-light-chain-enhancer of activated B-cells pathway (NF-κB) and the interferon regulatory factors pathway (IRF). Here, we demonstrate the ability to alter and improve adjuvant activity via the addn. of small mol. "immunomodulators". By modulating signaling activity instead of receptor binding, these mols. allow the customization of select innate responses. We demonstrate both inhibition and enhancement of the products of the NF-κB and IRF pathways by several orders of magnitude. Some modulators apply generally across many receptors, while others focus specifically on individual receptors. Modulators boost correlates of a protective immune responses in a com. flu vaccine model and reduced correlates of reactogenicity in a typhoid vaccine model. These modulators have a range of applications: from adjuvanticity in prophylactics to enhancement of immunotherapy.
- 51Nihesh, N.; Manna, S.; Studnitzer, B.; Shen, J.; Esser-Kahn, A. P. A Synthetic Pathogen Mimetic Molecule Induces a Highly Amplified Synergistic Immune Response Via Activation of Multiple Signaling Pathways. Chem. Sci. 2021, 12, 6646– 6651, DOI: 10.1039/D1SC00964HGoogle Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXnvVGitr8%253D&md5=26cbbad0fab4671e7767affb9acdd81aA synthetic pathogen mimetic molecule induces a highly amplified synergistic immune response via activation of multiple signaling pathwaysNihesh, Naorem; Manna, Saikat; Studnitzer, Bradley; Shen, Jingjing; Esser-Kahn, Aaron P.Chemical Science (2021), 12 (19), 6646-6651CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)The current understanding of how the immune system processes complex information during natural infections is yet to be exploited for the mol. design of potent immune activators. Here, we address this challenge by design of a pathogen-mimetic mol. that simultaneously co-activates cell-surface active, endosomal and cytosolic immune receptors.
- 52Taylor, D.; Meyer, C. T.; Graves, D.; Sen, R.; Fu, J.; Tran, E.; Mirza, B.; Rodriguez, G.; Lang, C.; Feng, H.; Quaranta, V.; Wilson, J. T.; Kim, Y. J.; Korrer, M. J. MuSyC Dosing of Adjuvanted Cancer Vaccines Optimizes Antitumor Responses. Front Immunol 2022, 13, 936129, DOI: 10.3389/fimmu.2022.936129Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitlChu73M&md5=4f4dfbaf0362bfb299001faff278e0f6MuSyC dosing of adjuvanted cancer vaccines optimizes antitumor responsesTaylor, David; Meyer, Christian T.; Graves, Diana; Sen, Rupashree; Fu, Juan; Tran, Emily; Mirza, Bilal; Rodriguez, Gabriel; Lang, Cara; Feng, Hanwen; Quaranta, Vito; Wilson, John T.; Kim, Young J.; Korrer, Michael J.Frontiers in Immunology (2022), 13 (), 936129CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)With the clin. approval of T-cell-dependent immune checkpoint inhibitors for many cancers, therapeutic cancer vaccines have re-emerged as a promising immunotherapy. Cancer vaccines require the addn. of immunostimulatory adjuvants to increase vaccine immunogenicity, and increasingly multiple adjuvants are used in combination to bolster further and shape cellular immunity to tumor antigens. However, rigorous quantification of adjuvants' synergistic interactions is challenging due to partial redundancy in costimulatory mols. and cytokine prodn., leading to the common assumption that combining both adjuvants at the max. tolerated dose results in optimal efficacy. Herein, we examine this max. dose assumption and find combinations of these doses are suboptimal. Instead, we optimized dendritic cell activation by extending the Multidimensional Synergy of Combinations (MuSyC) framework that measures the synergy of efficacy and potency between two vaccine adjuvants. Initially, we performed a preliminary in vitro screening of clin. translatable adjuvant receptor targets (TLR, STING, NLL, and RIG-I). We detd. that STING agonist (CDN) plus TLR4 agonist (MPL-A) or TLR7/8 agonist (R848) as the best pairwise combinations for dendritic cell activation. In addn., we found that the combination of R848 and CDN is synergistically efficacious and potent in activating both murine and human antigen-presenting cells (APCs) in vitro. These two selected adjuvants were then used to est. a MuSyC-dose optimized for in vivo T-cell priming using ovalbumin-based peptide vaccines. Finally, using B16 melanoma and MOC1 head and neck cancer models, MuSyC-dose-based adjuvating of cancer vaccines improved the antitumor response, increased tumor infiltrating lymphocytes, and induced novel myeloid tumor infiltration changes. Further, the MuSyC-dose-based adjuvants approach did not cause addnl. wt. changes or increased plasma cytokine levels compared to CDN alone. Collectively, our findings offer a proof of principle that our MuSyC-extended approach can be used to optimize cancer vaccine formulations for immunotherapy.
- 53Hanson, M. C.; Crespo, M. P.; Abraham, W.; Moynihan, K. D.; Szeto, G. L.; Chen, S. H.; Melo, M. B.; Mueller, S.; Irvine, D. J. Nanoparticulate STING Agonists Are Potent Lymph Node-Targeted Vaccine Adjuvants. J. Clin Invest 2015, 125, 2532– 2546, DOI: 10.1172/JCI79915Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2MfgsVChtw%253D%253D&md5=b840cee3cc090db0b723072b6936b05cNanoparticulate STING agonists are potent lymph node-targeted vaccine adjuvantsHanson Melissa C; Crespo Monica P; Abraham Wuhbet; Moynihan Kelly D; Szeto Gregory L; Chen Stephanie H; Melo Mariane B; Mueller Stefanie; Irvine Darrell JThe Journal of clinical investigation (2015), 125 (6), 2532-46 ISSN:.Cyclic dinucleotides (CDNs) are agonists of stimulator of IFN genes (STING) and have potential as vaccine adjuvants. However, cyclic di-GMP (cdGMP) injected s.c. shows minimal uptake into lymphatics/draining lymph nodes (dLNs) and instead is rapidly distributed to the bloodstream, leading to systemic inflammation. Here, we encapsulated cdGMP within PEGylated lipid nanoparticles (NP-cdGMP) to redirect this adjuvant to dLNs. Compared with unformulated CDNs, encapsulation blocked systemic dissemination and markedly enhanced dLN accumulation in murine models. Delivery of NP-cdGMP increased CD8+ T cell responses primed by peptide vaccines and enhanced therapeutic antitumor immunity. A combination of a poorly immunogenic liposomal HIV gp41 peptide antigen and NP-cdGMP robustly induced type I IFN in dLNs, induced a greater expansion of vaccine-specific CD4+ T cells, and greatly increased germinal center B cell differentiation in dLNs compared with a combination of liposomal HIV gp41 and soluble CDN. Further, NP-cdGMP promoted durable antibody titers that were substantially higher than those promoted by the well-studied TLR agonist monophosphoryl lipid A and comparable to a much larger dose of unformulated cdGMP, without the systemic toxicity of the latter. These results demonstrate that nanoparticulate delivery safely targets CDNs to the dLNs and enhances the efficacy of this adjuvant. Moreover, this approach can be broadly applied to other small-molecule immunomodulators of interest for vaccines and immunotherapy.
- 54Atukorale, P. U.; Raghunathan, S. P.; Raguveer, V.; Moon, T. J.; Zheng, C.; Bielecki, P. A.; Wiese, M. L.; Goldberg, A. L.; Covarrubias, G.; Hoimes, C. J.; Karathanasis, E. Nanoparticle Encapsulation of Synergistic Immune Agonists Enables Systemic Codelivery to Tumor Sites and IFNβ-Driven Antitumor Immunity. Cancer Res. 2019, 79, 5394– 5406, DOI: 10.1158/0008-5472.CAN-19-0381Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtFSqtb0%253D&md5=469de56cb58341e82e13aa77c48c6515Nanoparticle encapsulation of synergistic immune agonists enables systemic codelivery to tumor sites and IFNβ-driven antitumor immunityAtukorale, Prabhani U.; Raghunathan, Shruti P.; Raguveer, Vanitha; Moon, Tayior J.; Zheng, Carolyn; Bielecki, Peter A.; Wiese, Michelle L.; Goldberg, Amy L.; Covarrubias, Gil; Hoimes, Christopher J.; Karathanasis, EfstathiosCancer Research (2019), 79 (20), 5394-5406CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Effective cancer immunotherapy depends on the robust activation of tumor-specific antigen-presenting cells (APC). Immune agonists encapsulated within nanoparticles (NP) can be delivered to tumor sites to generate powerful antitumor immune responses with minimal off-target dissemination. Systemic delivery enables widespread access to the microvasculature and draining to the APC-rich perivasculature. We developed an immuno-nanoparticle (immuno-NP) coloaded with cyclic diguanylate monophosphate, an agonist of the stimulator of interferon genes pathway, and monophosphoryl lipid A, and a Toll-like receptor 4 agonist, which synergize to produce high levels of type I ΙΡΝβ. Usinga murine model of metastatic triple-neg. breast cancer. systemic delivery of these immuno-NPs resulted in significant therapeutic outcomes due to extensive upregulation of APCs and natural killer cells in the blood and tumor compared with control treatments. These results indicate that NPs can facilitate systemic delivery of multiple immune potentiating cargoes for effective APC-driven local and systemic antitumor immunity.
- 55Pandey, S.; Gruenbaum, A.; Kanashova, T.; Mertins, P.; Cluzel, P.; Chevrier, N. Pairwise Stimulations of Pathogen-Sensing Pathways Predict Immune Responses to Multi-Adjuvant Combinations. Cell Syst 2020, 11, 495– 508, DOI: 10.1016/j.cels.2020.10.001Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisVaqu7jK&md5=04738bf46febbbc04848669c0147be24Pairwise Stimulations of Pathogen-Sensing Pathways Predict Immune Responses to Multi-adjuvant CombinationsPandey, Surya; Gruenbaum, Adam; Kanashova, Tamara; Mertins, Philipp; Cluzel, Philippe; Chevrier, NicolasCell Systems (2020), 11 (5), 495-508.e10CODEN: CSEYA4; ISSN:2405-4712. (Cell Press)The immune system makes decisions in response to combinations of multiple microbial inputs. We do not understand the combinatorial logic governing how higher-order combinations of microbial signals shape immune responses. Here, using coculture expts. and statistical analyses, we discover a general property for the combinatorial sensing of microbial signals, whereby the effects of triplet combinations of microbial signals on immune responses can be predicted by combining the effects of single and pairs. Mechanistically, we find that singles and pairs dictate the information signaled by triplets in mouse and human DCs at the levels of transcription, chromatin, and protein secretion. We exploit this simplifying property to develop cell-based immunotherapies prepd. with adjuvant combinations that trigger protective responses in mouse models of cancer. We conclude that the processing of multiple input signals by innate immune cells is governed by pairwise effects, which will inform the rationale combination of adjuvants to manipulate immunity.
- 56Andrade, W. A.; Agarwal, S.; Mo, S.; Shaffer, S. A.; Dillard, J. P.; Schmidt, T.; Hornung, V.; Fitzgerald, K. A.; Kurt-Jones, E. A.; Golenbock, D. T. Type I Interferon Induction by Neisseria Gonorrhoeae: Dual Requirement of Cyclic GMP-AMP Synthase and Toll-Like Receptor 4. Cell Rep 2016, 15, 2438– 2448, DOI: 10.1016/j.celrep.2016.05.030Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xpt1entbg%253D&md5=55421b21e0d3fce79cfbfe853151908dType I Interferon Induction by Neisseriagonorrhoeae: Dual Requirement of Cyclic GMP-AMP Synthase and Toll-like Receptor 4Andrade, Warrison A.; Agarwal, Sarika; Mo, Shunyan; Shaffer, Scott A.; Dillard, Joseph P.; Schmidt, Tobias; Hornung, Veit; Fitzgerald, Katherine A.; Kurt-Jones, Evelyn A.; Golenbock, Douglas T.Cell Reports (2016), 15 (11), 2438-2448CODEN: CREED8; ISSN:2211-1247. (Cell Press)The innate immune system is the first line of defense against Neisseria gonorrhoeae (GC). Exposure of cells to GC lipooligosaccharides induces a strong immune response, leading to type I interferon (IFN) prodn. via TLR4/MD-2. In addn. to living freely in the extracellular space, GC can invade the cytoplasm to evade detection and elimination. Double-stranded DNA introduced into the cytosol binds and activates the enzyme cyclic-GMP-AMP synthase (cGAS), which produces 2'3'-cGAMP and triggers STING/TBK-1/IRF3 activation, resulting in type I IFN expression. Here, we reveal a cytosolic response to GC DNA that also contributes to type I IFN induction. We demonstrate that complete IFN-β induction by live GC depends on both cGAS and TLR4. Type I IFN is detrimental to the host, and dysregulation of iron homeostasis genes may explain lower bacteria survival in cGAS-/- and TLR4-/- cells. Collectively, these observations reveal cooperation between TLRs and cGAS in immunity to GC infection.
- 57Kocabas, B. B.; Almacioglu, K.; Bulut, E. A.; Gucluler, G.; Tincer, G.; Bayik, D.; Gursel, M.; Gursel, I. Dual-Adjuvant Effect of pH-Sensitive Liposomes Loaded with STING and TLR9 Agonists Regress Tumor Development by Enhancing Th1 Immune Response. J. Controlled Release 2020, 328, 587– 595, DOI: 10.1016/j.jconrel.2020.09.040Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitVCks7bE&md5=2eb22cda1e9a367c6e5e6757380a13efDual-adjuvant effect of pH-sensitive liposomes loaded with STING and TLR9 agonists regress tumor development by enhancing Th1 immune responseKocabas, Banu Bayyurt; Almacioglu, Kubra; Bulut, Esin Alpdundar; Gucluler, Gozde; Tincer, Gizem; Bayik, Defne; Gursel, Mayda; Gursel, IhsanJournal of Controlled Release (2020), 328 (), 587-595CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Nucleic acid-based pattern recognition receptor agonists are effective adjuvants and immunotherapeutic agents. Rather than single applications, ligand combinations could synergistically potentiate immune responses by elevating cytokine and chemokine prodn. via triggering multiple signaling pathways. However, short half-lives of such labile ligands due to nuclease attack and limited cellular uptake due to their structure significantly hamper their in vivo performances. More importantly, simultaneous delivery and activity presentation of protein antigen and nucleic acid ligands critically limit the clin. development of these constructs. In this work, we approached this problem by co-encapsulating a model antigen ovalbumin along with TLR9 and STING ligands within liposomes, a well-established drug delivery system that enables payload stability and enhanced cellular activity upon internalization. Moreover, by loading dual ligands we postulated to achieve heightened Th-1 immune response that would yield pronounced protective vaccine efficacy. We show that, pH-sensitive liposomes co-encapsulating CpG ODN and cGAMP induced synergistic innate immune response by elevating type I and type II interferon levels. Most importantly, this vaccine formulation led to ∼70% regression of established melanoma tumor. pH-sensitive liposomal vaccine administration elevated IgG2c/IgG1 antibody ratio, indicative of augmented OVA-specific Th1-biased immunity. Importantly, while the frequency of tumor-specific IFN-γ producing CD8+ T-cells was significantly increased, the M2-type anti-inflammatory macrophage levels were decreased in the tumor bed. In conclusion, our strategy induces reversal of immunosuppressive tumor microenvironment, while enhancing effective anti-tumor immune-response. We propose that this could be coupled with std. therapies during combating tumor eradication.
- 58Toy, R.; Keenum, M. C.; Pradhan, P.; Phang, K.; Chen, P.; Chukwu, C.; Nguyen, L. A. H.; Liu, J.; Jain, S.; Kozlowski, G.; Hosten, J.; Suthar, M. S.; Roy, K. TLR7 and RIG-I Dual-Adjuvant Loaded Nanoparticles Drive Broadened and Synergistic Responses in Dendritic Cells in Vitro and Generate Unique Cellular Immune Responses in Influenza Vaccination. J. Controlled Release 2021, 330, 866– 877, DOI: 10.1016/j.jconrel.2020.10.060Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitlCku7rE&md5=4985b3db7e0bf52f50272a1d4e1238dcTLR7 and RIG-I dual-adjuvant loaded nanoparticles drive broadened and synergistic responses in dendritic cells in vitro and generate unique cellular immune responses in influenza vaccinationToy, Randall; Keenum, M. Cole; Pradhan, Pallab; Phang, Katelynn; Chen, Patrick; Chukwu, Chinwendu; Nguyen, Lily Anh H.; Liu, Jiaying; Jain, Sambhav; Kozlowski, Gabrielle; Hosten, Justin; Suthar, Mehul S.; Roy, KrishnenduJournal of Controlled Release (2021), 330 (), 866-877CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Although the existing flu vaccines elicit strong antigen-specific antibody responses, they fail to provide effective, long term protection - partly due to the absence of robust cellular memory immunity. We hypothesized that co-administration of combination adjuvants, mirroring the flu-virus related innate signaling pathways, could elicit strong cellular immunity. Here, we show that the small mol. adjuvant R848 and the RNA adjuvant PUUC, targeting endosomal TLR7s and cytoplasmic RLRs resp., when delivered together in polymer nanoparticles (NP), elicits a broadened immune responses in mouse bone marrow-derived dendritic cells (mBMDCs) and a synergistic response in both mouse and human plasmacytoid dendritic cells (pDCs). In mBMDCs, NP-R848-PUUC induced both NF-κB and interferon signaling. Interferon responses to co-delivered R848 and PUUC were additive in human peripheral blood mononuclear cells (PBMCs) and synergistic in human FLT3-differentiated mBMDCs and CAL-1 pDCs. Vaccination with NPs loaded with H1N1 Flu antigen, R848, and PUUC increased percentage of CD8+ T-cells in the lungs, percentage of antigen-specific CD4-T-cells in the spleen, and enhanced overall cytokine-secreting T cell percentages upon antigen restimulation. Also, in the spleen, T lymphopenia, esp. after in vitro restimulation with dual adjuvants, was obsd., indicating highly antigen-reactive T cells. Our results demonstrate that simultaneous engagement of TLR7 and RIG-I pathways using particulate carriers is a potential approach to improve cellular immunity in flu vaccination.
- 59Hou, Y.; Wang, Y.; Tang, Y.; Zhou, Z.; Tan, L.; Gong, T.; Zhang, L.; Sun, X. Co-Delivery of Antigen and Dual Adjuvants by Aluminum Hydroxide Nanoparticles for Enhanced Immune Responses. J. Controlled Release 2020, 326, 120– 130, DOI: 10.1016/j.jconrel.2020.06.021Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXht1Kgu7bL&md5=ec7771ba4a626526fd70d75d7fa0af14Co-delivery of antigen and dual adjuvants by aluminum hydroxide nanoparticles for enhanced immune responsesHou, Yingying; Wang, Ying; Tang, Yao; Zhou, Zixuan; Tan, Lu; Gong, Tao; Zhang, Ling; Sun, XunJournal of Controlled Release (2020), 326 (), 120-130CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Adjuvants that contain pathogen-assocd. mol. patterns (PAMPs) can enhance vaccination efficacy by binding to pattern recognition receptors (PRRs), thereby stimulating immune responses. Particularly effective may be the combination of multiple PAMPs that activate different PRRs, which occurs with natural pathogens. Here we hypothesized the enhanced effects would occur in two adjuvants that stimulate different PRRs: CpG oligodeoxynucleotide (CpG-ODN), which is Toll-like receptor 9 agonist; and 5'-triphosphate, short, double-stranded RNA (3pRNA), which activates retinoic acid-inducible gene I (RIG-I). The model antigen ovalbumin (OVA) was loaded and adjuvants were surface-adsorbed to aluminum hydroxide nanoparticles (hereafter NP-3pRNA-CpG) by electrostatic interaction with an av. size of 120 nm and a neg. surface charge for targeting lymph nodes. These nanoparticles were efficiently internalized by antigen-presenting cells (APCs) in the lymph nodes, and the resulting APC activation and antigen cross-presentation generated strong humoral immunity and cytotoxic T lymphocyte responses and IFN-γ secretion. NP-3pRNA-CpG significantly suppressed B16-OVA tumor growth and prolonged survival of tumor-bearing mice in therapeutic and prophylactic models, illustrating the enhanced effects of CpG-ODN and 3pRNA. Our study highlights the potential of combining multiple adjuvants for effective vaccine design.
- 60Pagendarm, H. M.; Stone, P. T.; Kimmel, B. R.; Baljon, J. J.; Aziz, M. H.; Pastora, L. E.; Hubert, L.; Roth, E. W.; Almunif, S.; Scott, E. A.; Wilson, J. T. Engineering Endosomolytic Nanocarriers of Diverse Morphologies Using Confined Impingement Jet Mixing. Nanoscale 2023, 15, 16016– 16029, DOI: 10.1039/D3NR02874GGoogle Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXhvF2qsLnK&md5=04a78b16072f0ff7e9436c3c62991260Engineering endosomolytic nanocarriers of diverse morphologies using confined impingement jet mixingPagendarm, Hayden M.; Stone, Payton T.; Kimmel, Blaise R.; Baljon, Jessalyn J.; Aziz, Mina H.; Pastora, Lucinda E.; Hubert, Lauren; Roth, Eric W.; Almunif, Sultan; Scott, Evan A.; Wilson, John T.Nanoscale (2023), 15 (39), 16016-16029CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)The clin. translation of many biomol. therapeutics has been hindered by undesirable pharmacokinetic (PK) properties, inadequate membrane permeability, poor endosomal escape and cytosolic delivery, and/or susceptibility to degrdn. Overcoming these challenges merits the development of nanoscale drug carriers (nanocarriers) to improve the delivery of therapeutic cargo. Herein, we implement a flash nanopptn. (FNP) approach to produce nanocarriers of diverse vesicular morphologies by using various mol. wt. PEG-bl-DEAEMA-co-BMA (PEG-DB) polymers. We demonstrated that FNP can produce uniform (PDI < 0.1) particles after 5 impingements, and that by varying the copolymer hydrophilic mass fraction, FNP enables access to a diverse variety of nanoarchitectures including micelles, unilamellar vesicles (polymersomes), and multi-compartment vesicles (MCVs). We synthesized a library of 2 kDa PEG block copolymers, with DEAEMA-co-BMA second block mol. wts. of 3, 6, 12, 15, 20, and 30 kDa. All formulations were both pH responsive, endosomolytic, and capable of loading and cytosolically delivering small neg. charged mols. - albeit to different degrees. Using a B16. F10 melanoma model, we showcased the therapeutic potential of a lead FNP formulated PEG-DB nanocarrier, encapsulating the cyclic dinucleotide (CDN) cGAMP to activate the stimulator of interferon genes (STING) pathway in a therapeutically relevant context. Collectively, these data demonstrate that an FNP process can be used to formulate pH-responsive nanocarriers of diverse morphologies using a PEG-DB polymer system. As FNP is an industrially scalable process, these data address the crit. translational challenge of producing PEG-DB nanoparticles at scale. Furthermore, the diverse morphologies produced may specialize in the delivery of distinct biomol. cargos for other therapeutic applications, implicating the therapeutic potential of this platform in an array of disease applications.
- 61Manganiello, M. J.; Cheng, C.; Convertine, A. J.; Bryers, J. D.; Stayton, P. S. Diblock Copolymers with Tunable pH Transitions for Gene Delivery. Biomaterials 2012, 33, 2301– 2309, DOI: 10.1016/j.biomaterials.2011.11.019Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xnt1entw%253D%253D&md5=ec63b85f138fe9934ba5508bf04e594fDiblock copolymers with tunable pH transitions for gene deliveryManganiello, Matthew J.; Cheng, Connie; Convertine, Anthony J.; Bryers, James D.; Stayton, Patrick S.Biomaterials (2012), 33 (7), 2301-2309CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)A series of diblock copolymers contg. an endosomal-releasing segment composed of diethylaminoethyl methacrylate (DEAEMA) and Bu methacrylate (BMA) were synthesized via reversible addn.-fragmentation chain transfer (RAFT) polymn. The materials were designed to condense plasmid DNA (pDNA) through electrostatic interactions with a cationic poly(N,N-dimethylaminoethyl methacrylate) (DMAEMA) first block. The pDMAEMA was employed as a macro chain transfer agent (macroCTA) for the synthesis of a series in which the relative feed ratios of DEAEMA and BMA were systematically varied from 20% to 70% BMA. The resultant diblock copolymers exhibited low polydispersity (PDI ≤ 1.06) with similar mol. wts. (Mn = 19.3-23.1 kDa). Dynamic light scattering (DLS) measurements in combination with 1H NMR D2O studies demonstrated that the free copolymers assemble into core-shell micelles at physiol. pH. Redn. of the soln. pH to values representative of endosomal/lysosomal compartments induced an increase in the net cationic charge of the core through protonation of the DEAEMA residues. This protonation promotes micelle destabilization and exposure of the hydrophobic BMA residues that destabilize biol. membranes. The pH value at which this micelle-to-unimer transition occurred was dependent on the hydrophobic content of the copolymer, with higher BMA-contg. copolymer compns. exhibiting pH-induced transitions to the membrane-destabilizing state at successively lower pH values. The ability of the diblock copolymers to deliver pDNA was subsequently investigated using a GFP expression vector in two monocyte cell lines. High levels of DNA transfection were obsd. for the copolymer compns. exhibiting the sharpest pH transitions and membrane destabilizing activities, demonstrating the importance of tuning the endosomal-releasing segment compn.
- 62Allen, S.; Osorio, O.; Liu, Y. G.; Scott, E. Facile Assembly and Loading of Theranostic Polymersomes Via Multi-Impingement Flash Nanoprecipitation. J. Controlled Release 2017, 262, 91– 103, DOI: 10.1016/j.jconrel.2017.07.026Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1egsbjK&md5=a52b3f1d74b3a23fe99693893e9eab06Facile assembly and loading of theranostic polymersomes via multi-impingement flash nanoprecipitationAllen, Sean; Osorio, Omar; Liu, Yu-Gang; Scott, EvanJournal of Controlled Release (2017), 262 (), 91-103CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Flash nanopptn. has proven to be a powerful tool for the rapid and scalable assembly of solid-core nanoparticles from block copolymers. To date, FNP has not been applied for the fabrication of complex or vesicular nanoarchitectures capable of encapsulating hydrophilic mols. or bioactive protein therapeutics. Here, we present FNP as a single customizable method for the assembly of bicontinuous nanospheres, filomicelles and vesicular, multilamellar and tubular polymersomes from poly(ethylene glycol)-bl-poly(propylene sulfide) block copolymers. Multiple impingements of polymersomes assembled via FNP were shown to decrease vesicle diam. and polydispersity, allowing gram-scale fabrication of monodisperse polymersomes within minutes. Furthermore, we demonstrate that FNP supports simultaneous loading of both hydrophobic and hydrophilic mols. resp. into the polymersome membrane and aq. lumen, and encapsulated enzymes were found to be released and remain active following vesicle lysis. As an example application, theranostic polymersomes were generated via FNP that were dual loaded with the immunosuppressant rapamycin and a fluorescent dye to link targeted immune cells with the elicited immunomodulation of T cells. By expanding the capabilities of FNP, we present a rapid, scalable and reproducible method of nanofabrication for a wide range of nanoarchitectures that are typically challenging to assemble and load with therapeutics for controlled delivery and theranostic strategies.
- 63Han, J.; Zhu, Z.; Qian, H.; Wohl, A. R.; Beaman, C. J.; Hoye, T. R.; Macosko, C. W. A Simple Confined Impingement Jets Mixer for Flash Nanoprecipitation. J. Pharm. Sci. 2012, 101, 4018– 4023, DOI: 10.1002/jps.23259Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XpvVers70%253D&md5=c5e9605fc18c6dfbb6818cfe3a8888f6A simple confined impingement jets mixer for flash nanoprecipitationHan, Jing; Zhu, Zhengxi; Qian, Haitao; Wohl, Adam R.; Beaman, Charles J.; Hoye, Thomas R.; Macosko, Christopher W.Journal of Pharmaceutical Sciences (2012), 101 (10), 4018-4023CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)Johnson and Prud'homme (2003. AICHE J 49:2264-2282) introduced the confined impingement jets (CIJ) mixer to prep. nanoparticles loaded with hydrophobic compds. (e.g., drugs, inks, fragrances, or pheromones) via flash nanopptn. (FNP). We have modified the original CIJ design to allow hand operation, eliminating the need for a syringe pump, and we added a second antisolvent diln. stage. Impingement mixing requires equal flow momentum from two opposing jets, one contg. the drug in org. solvent and the other contg. an antisolvent, typically water. The subsequent diln. step in the new design allows rapid quenching with high antisolvent concn. that enhances nanoparticle stability. This new CIJ with diln. (CIJ-D) mixer is a simple, cheap, and efficient device to produce nanoparticles. We have made 55 nm diam. β-carotene nanoparticles using the CIJ-D mixer. They are stable and reproducible in terms of particle size and distribution. We have also compared the performance of our CIJ-D mixer with the vortex mixer, which can operate at unequal flow rates (Liu et al., 2008. Chem Eng Sci 63:2829-2842), to make β-carotene-contg. particles over a series of turbulent conditions. On the basis of dynamic light scattering measurements, the new CIJ-D mixer produces stable particles of a size similar to the vortex mixer. Our CIJ-D design requires less vol. and provides an easily operated and inexpensive tool to produce nanoparticles via FNP and to evaluate new nanoparticle formulation. © 2012 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 64Johnson, B. K.; Prud’homme, R. K. Mechanism for Rapid Self-Assembly of Block Copolymer Nanoparticles. Phys. Rev. Lett. 2003, 91, 118302, DOI: 10.1103/PhysRevLett.91.118302Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXnt1Oqurw%253D&md5=465c945d7870908649f6f231a6677a98Mechanism for Rapid Self-Assembly of Block Copolymer NanoparticlesJohnson, Brian K.; Prud'homme, Robert K.Physical Review Letters (2003), 91 (11), 118302/1-118302/4CODEN: PRLTAO; ISSN:0031-9007. (American Physical Society)Amphiphilic block copolymers in soln. spontaneously self-assemble when the solvent quality for one block is selectively decreased. We demonstrate that, for supersatn. ratio changes [d(S)/dt] over 105 per s from equil., nanoparticles are obtained with a formation mechanism and size dependent on the jumping rate and magnitude. The threshold rate for homogeneous pptn. is detd. by the induction time of a particle, equiv. to the diffusion limited fusion of copolymer chains to form a corona of overlapping sol. brushes. Via detn. of the induction time with a novel confined impinging jets mixer and use of a scaling relation, the interfacial free energy of a block copolymer nanoparticle was measured for the first time.
- 65Daniel, S.; Kis, Z.; Kontoravdi, C.; Shah, N. Quality by Design for Enabling RNA Platform Production Processes. Trends Biotechnol 2022, 40, 1213– 1228, DOI: 10.1016/j.tibtech.2022.03.012Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XpvVahurs%253D&md5=7b64c147111978920111452f652888dcQuality by Design for enabling RNA platform production processesDaniel, Simon; Kis, Zoltan; Kontoravdi, Cleo; Shah, NilayTrends in Biotechnology (2022), 40 (10), 1213-1228CODEN: TRBIDM; ISSN:0167-7799. (Elsevier Ltd.)A review. RNA-based products have emerged as one of the most promising and strategic technologies for global vaccination, infectious disease control, and future therapy development. The assessment of crit. quality attributes (CQAs), product-process interactions, relevant process anal. technologies, and process modeling capabilities can feed into a robust Quality by Design (QbD) framework for future development, design, and control of manufg. processes. QbD implementation will help the RNA technol. reach its full potential and will be central to the development, pre-qualification, and regulatory approval of rapid response, disease-agnostic RNA platform prodn. processes.
- 66Warne, N.; Ruesch, M.; Siwik, P.; Mensah, P.; Ludwig, J.; Hripcsak, M.; Godavarti, R.; Prigodich, A.; Dolsten, M. Delivering 3 Billion Doses of Comirnaty in 2021. Nat. Biotechnol. 2023, 41, 183– 188, DOI: 10.1038/s41587-022-01643-1Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXis1Sjs7o%253D&md5=c0125b7d1eb92de3d174e35c99a37aecDelivering 3 billion doses of Comirnaty in 2021Warne, Nicholas; Ruesch, Margaret; Siwik, Pamela; Mensah, Paul; Ludwig, John; Hripcsak, Michael; Godavarti, Ranga; Prigodich, Andrew; Dolsten, MikaelNature Biotechnology (2023), 41 (2), 183-188CODEN: NABIF9; ISSN:1087-0156. (Nature Portfolio)Pfizer created a 'light-speed' approach to meet the challenge of vaccinating the world against COVID-19. It involved developing new strategies for all aspects of vaccine development, from sourcing materials and scaling up manufg. to transportation and dosing.
- 67Chang, T. Z.; Stadmiller, S. S.; Staskevicius, E.; Champion, J. A. Effects of Ovalbumin Protein Nanoparticle Vaccine Size and Coating on Dendritic Cell Processing. Biomater Sci. 2017, 5, 223– 233, DOI: 10.1039/C6BM00500DGoogle Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvF2gurnK&md5=2847bc4357609887cd5580a2ae9eef75Effects of ovalbumin protein nanoparticle vaccine size and coating on dendritic cell processingChang, Timothy Z.; Stadmiller, Samantha S.; Staskevicius, Erika; Champion, Julie A.Biomaterials Science (2017), 5 (2), 223-233CODEN: BSICCH; ISSN:2047-4849. (Royal Society of Chemistry)Nanoparticle vaccine delivery platforms are a promising technol. for enhancing vaccine immunogenicity. Protein nanoparticles (PNPs), made entirely from antigen, have been shown to induce protective immune responses against influenza. However, the fundamental mechanisms by which PNPs enhance component protein immunogenicity are not understood. Here, we investigate the role of size and coating of model ovalbumin (OVA) PNPs on particle uptake and trafficking, as well as on inflammation and maturation factor expression in dendritic cells (DCs) in vitro. OVA PNPs enhance antigen uptake in a size-independent manner, and experience attenuated endosomal acidification as compared to sol. OVA. OVA PNPs also trigger Fc receptor upregulation. Expression of cytokines IL-1β and TNF-α were PNP size- and coating-dependent, with small (∼270 nm) nanoparticles triggering greater inflammatory cytokine prodn. than large (∼560 nm) particles. IL-1β expression by DCs in response to PNP stimulation implies activation of the inflammasome, a pathway known to be activated by certain types of nanoparticulate adjuvants. The attenuated acidification and pro-inflammatory profile generated by PNPs in DCs demonstrate that phys. biomaterial properties can modulate dendritic cell-mediated antigen processing and adjuvancy. In addn. to nanoparticles' enhancement of DC antigen uptake, our work suggests that vaccine nanoparticle size and coating are uptake-independent modulators of immunogenicity.
- 68Thomas, S. N.; Schudel, A. Overcoming Transport Barriers for Interstitial-, Lymphatic-, and Lymph Node-Targeted Drug Delivery. Curr. Opin Chem. Eng. 2015, 7, 65– 74, DOI: 10.1016/j.coche.2014.11.003Google Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2srotF2rsQ%253D%253D&md5=3253ad4ef935260a93f650e74b980196Overcoming transport barriers for interstitial-, lymphatic-, and lymph node-targeted drug deliveryThomas Susan N; Schudel AlexCurrent opinion in chemical engineering (2015), 7 (), 65-74 ISSN:2211-3398.Despite drug formulation improving circulation times and targeting, efficacy is stymied by inadequate penetration into and retention within target tissues. This review highlights the barriers restricting delivery to the connective tissue interstitium, lymphatics, and lymph nodes as well as advances in engineering drug carriers to overcome these delivery challenges. Three-dimensional tissue physiology is discussed in the context of providing material design principles for delivery to these tissues; in particular the influence of interstitial and lymphatic flows as well as differential permeabilities of the blood and lymphatic capillaries. Key examples of materials with different characteristics developed to overcome these transport barriers are discussed as well as potential areas for further development.
- 69Munson, M. J.; O’Driscoll, G.; Silva, A. M.; Lazaro-Ibanez, E.; Gallud, A.; Wilson, J. T.; Collen, A.; Esbjorner, E. K.; Sabirsh, A. A High-Throughput Galectin-9 Imaging Assay for Quantifying Nanoparticle Uptake, Endosomal Escape and Functional RNA Delivery. Commun. Biol. 2021, 4, 211, DOI: 10.1038/s42003-021-01728-8Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtV2iurvI&md5=2f6c6521ab150ffdcc94a614266cd245A high-throughput Galectin-9 imaging assay for quantifying nanoparticle uptake, endosomal escape and functional RNA deliveryMunson, Michael J.; O'Driscoll, Gwen; Silva, Andreia M.; Lazaro-Ibanez, Elisa; Gallud, Audrey; Wilson, John T.; Collen, Anna; Esbjoerner, Elin K.; Sabirsh, AlanCommunications Biology (2021), 4 (1), 211CODEN: CBOIDQ; ISSN:2399-3642. (Nature Research)RNA-based therapies have great potential to treat many undruggable human diseases. However, their efficacy, in particular for mRNA, remains hampered by poor cellular delivery and limited endosomal escape. Development and optimization of delivery vectors, such as lipid nanoparticles (LNPs), are impeded by limited screening methods to probe the intracellular processing of LNPs in sufficient detail. We have developed a high-throughput imaging-based endosomal escape assay utilizing a Galectin-9 reporter and fluorescently labeled mRNA to probe correlations between nanoparticle-mediated uptake, endosomal escape frequency, and mRNA translation. Furthermore, this assay has been integrated within a screening platform for optimization of lipid nanoparticle formulations. We show that Galectin-9 recruitment is a robust, quant. reporter of endosomal escape events induced by different mRNA delivery nanoparticles and small mols. We identify nanoparticles with superior escape properties and demonstrate cell line variances in endosomal escape response, highlighting the need for fine-tuning of delivery formulations for specific applications.
- 70Shae, D.; Becker, K. W.; Christov, P.; Yun, D. S.; Lytton-Jean, A. K. R.; Sevimli, S.; Ascano, M.; Kelley, M.; Johnson, D. B.; Balko, J. M.; Wilson, J. T. Endosomolytic Polymersomes Increase the Activity of Cyclic Dinucleotide STING Agonists to Enhance Cancer Immunotherapy. Nat. Nanotechnol 2019, 14, 269– 278, DOI: 10.1038/s41565-018-0342-5Google Scholar70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXmtVGitbo%253D&md5=4b95f1d3ac44492fd975fc9e37bcec58Endosomolytic polymersomes increase the activity of cyclic dinucleotide STING agonists to enhance cancer immunotherapyShae, Daniel; Becker, Kyle W.; Christov, Plamen; Yun, Dong Soo; Lytton-Jean, Abigail K. R.; Sevimli, Sema; Ascano, Manuel; Kelley, Mark; Johnson, Douglas B.; Balko, Justin M.; Wilson, John T.Nature Nanotechnology (2019), 14 (3), 269-278CODEN: NNAABX; ISSN:1748-3387. (Nature Research)Cyclic dinucleotide (CDN) agonists of stimulator of interferon genes (STING) are a promising class of immunotherapeutics that activate innate immunity to increase tumor immunogenicity. However, the efficacy of CDNs is limited by drug delivery barriers, including poor cellular targeting, rapid clearance and inefficient transport to the cytosol where STING is localized. Here, we describe STING-activating nanoparticles (STING-NPs)-rationally designed polymersomes for enhanced cytosolic delivery of the endogenous CDN ligand for STING, 2'3' cGMP-adenosine monophosphate (cGAMP). STING-NPs increase the biol. potency of cGAMP, enhance STING signaling in the tumor microenvironment and sentinel lymph node, and convert immunosuppressive tumors to immunogenic, tumoricidal microenvironments. This leads to enhanced therapeutic efficacy of cGAMP, inhibition of tumor growth, increased rates of long-term survival, improved response to immune checkpoint blockade and induction of immunol. memory that protects against tumor rechallenge. We validate STING-NPs in freshly isolated human melanoma tissue, highlighting their potential to improve clin. outcomes of immunotherapy.
- 71Zheng, S.; Wang, W.; Aldahdooh, J.; Malyutina, A.; Shadbahr, T.; Tanoli, Z.; Pessia, A.; Tang, J. Synergyfinder Plus: Toward Better Interpretation and Annotation of Drug Combination Screening Datasets. Genomics Proteomics Bioinformatics 2022, 20, 587– 596, DOI: 10.1016/j.gpb.2022.01.004Google Scholar71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2M7jslymtA%253D%253D&md5=586c7516cf1d71a67d4788d4e640754eSynergyFinder Plus: Toward Better Interpretation and Annotation of Drug Combination Screening DatasetsZheng Shuyu; Wang Wenyu; Aldahdooh Jehad; Malyutina Alina; Shadbahr Tolou; Tanoli Ziaurrehman; Pessia Alberto; Tang JingGenomics, proteomics & bioinformatics (2022), 20 (3), 587-596 ISSN:.Combinatorial therapies have been recently proposed to improve the efficacy of anticancer treatment. The SynergyFinder R package is a software used to analyze pre-clinical drug combination datasets. Here, we report the major updates to the SynergyFinder R package for improved interpretation and annotation of drug combination screening results. Unlike the existing implementations, the updated SynergyFinder R package includes five main innovations. 1) We extend the mathematical models to higher-order drug combination data analysis and implement dimension reduction techniques for visualizing the synergy landscape. 2) We provide a statistical analysis of drug combination synergy and sensitivity with confidence intervals and P values. 3) We incorporate a synergy barometer to harmonize multiple synergy scoring methods to provide a consensus metric for synergy. 4) We evaluate drug combination synergy and sensitivity to provide an unbiased interpretation of the clinical potential. 5) We enable fast annotation of drugs and cell lines, including their chemical and target information. These annotations will improve the interpretation of the mechanisms of action of drug combinations. To facilitate the use of the R package within the drug discovery community, we also provide a web server at www.synergyfinderplus.org as a user-friendly interface to enable a more flexible and versatile analysis of drug combination data.
- 72Mouries, J.; Moron, G.; Schlecht, G.; Escriou, N.; Dadaglio, G.; Leclerc, C. Plasmacytoid Dendritic Cells Efficiently Cross-Prime Naive T Cells in Vivo after TLR Activation. Blood 2008, 112, 3713– 3722, DOI: 10.1182/blood-2008-03-146290Google Scholar72https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlCntrvF&md5=6484bd424f61a2bcf69ee7076b403ea6Plasmacytoid dendritic cells efficiently cross-prime naive T cells in vivo after TLR activationMouries, Juliette; Moron, Gabriel; Schlecht, Geraldine; Escriou, Nicolas; Dadaglio, Gilles; Leclerc, ClaudeBlood (2008), 112 (9), 3713-3722CODEN: BLOOAW; ISSN:0006-4971. (American Society of Hematology)Cross-presentation is a crucial mechanism in tumoral and microbial immunity because it allows internalized cell assocd. or exogenous antigens (Ags) to be delivered into the major histocompatibility complex I pathway. This pathway is important for the development of CD8+ T-cell responses and for the induction of tolerance. In mice, cross-presentation is considered to be a unique property of CD8α+ conventional dendritic cells (DCs). Here we show that splenic plasmacytoid DCs (pDCs) efficiently capture exogenous Ags in vivo but are not able to cross-present these Ags at steady state. However, in vitro and in vivo stimulation by Toll-like receptor-7, or -9 or viruses licenses pDCs to cross-present sol. or particulate Ags by a transporter assocd. with antigen processing-dependent mechanism. Induction of cross-presentation confers to pDCs the ability to generate efficient effector CD8+ T-cell responses against exogenous Ags in vivo, showing that pDCs may play a crucial role in induction of adaptive immune responses against pathogens that do not infect tissues of hemopoietic origin. This study provides the first evidence for an in vivo role of splenic pDCs in Ag cross-presentation and T-cell cross-priming and suggests that pDCs may constitute an attractive target to boost the efficacy of vaccines based on cytotoxic T lymphocyte induction.
- 73Villadangos, J. A.; Young, L. Antigen-Presentation Properties of Plasmacytoid Dendritic Cells. Immunity 2008, 29, 352– 361, DOI: 10.1016/j.immuni.2008.09.002Google Scholar73https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtF2jtLvL&md5=81e1c944723cfa0bc4117416b558dad0Antigen-presentation properties of plasmacytoid dendritic cellsVilladangos, Jose A.; Young, LouiseImmunity (2008), 29 (3), 352-361CODEN: IUNIEH; ISSN:1074-7613. (Cell Press)A review. One of the remaining enigmas of the dendritic cell (DC) network is the potential contribution of plasmacytoid DCs (pDCs) to antigen presentation. Although the antigen-presentation capacity of conventional DCs (cDCs) is clearly defined, pDCs are generally attributed as having little, if any, antigen-presentation function. Instead, pDCs are regarded as immunomodulating cells, capable of directing the immune response through their secretion of large amts. of type I interferons. Here, the authors examine the evidence for a potential role of pDC in antigen capture, processing, and presentation to T cells at sites of infection and in the lymph nodes.
- 74Hildner, K.; Edelson, B. T.; Purtha, W. E.; Diamond, M.; Matsushita, H.; Kohyama, M.; Calderon, B.; Schraml, B. U.; Unanue, E. R.; Diamond, M. S.; Schreiber, R. D.; Murphy, T. L.; Murphy, K. M. Batf3 Deficiency Reveals a Critical Role for CD8α+ Dendritic Cells in Cytotoxic T Cell Immunity. Science 2008, 322, 1097– 1100, DOI: 10.1126/science.1164206Google Scholar74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlGhu7fF&md5=5edf74e0adedb0e092d59a241a711784Batf3 Deficiency Reveals a Critical Role for CD8α+ Dendritic Cells in Cytotoxic T Cell ImmunityHildner, Kai; Edelson, Brian T.; Purtha, Whitney E.; Diamond, Mark; Matsushita, Hirokazu; Kohyama, Masako; Calderon, Boris; Schraml, Barbara U.; Unanue, Emil R.; Diamond, Michael S.; Schreiber, Robert D.; Murphy, Theresa L.; Murphy, Kenneth M.Science (Washington, DC, United States) (2008), 322 (5904), 1097-1100CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Although in vitro observations suggest that cross-presentation of antigens is mediated primarily by CD8α+ dendritic cells, in vivo anal. has been hampered by the lack of systems that selectively eliminate this cell lineage. We show that deletion of the transcription factor Batf3 ablated development of CD8α+ dendritic cells, allowing us to examine their role in immunity in vivo. Dendritic cells from Batf3-/- mice were defective in cross-presentation, and Batf3-/- mice lacked virus-specific CD8+ T cell responses to West Nile virus. Importantly, rejection of highly immunogenic syngeneic tumors was impaired in Batf3-/- mice. These results suggest an important role for CD8α+ dendritic cells and cross-presentation in responses to viruses and in tumor rejection.
- 75Kaech, S. M.; Wherry, E. J.; Ahmed, R. Effector and Memory T-Cell Differentiation: Implications for Vaccine Development. Nat. Rev. Immunol 2002, 2, 251– 262, DOI: 10.1038/nri778Google Scholar75https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XjtFKhtrc%253D&md5=7a63c84ffcfeae8b4362befd059980e3Effector and memory T-cell differentiation: implications for vaccine developmentKaech, Susan M.; Wherry, E. John; Ahmed, RafiNature Reviews Immunology (2002), 2 (4), 251-262CODEN: NRIABX; ISSN:1474-1733. (Nature Publishing Group)A review. Recent work shows that after stimulation with antigen, CD4+ and CD8+ T cells embark on a program of proliferation that is closely linked with the acquisition of effector functions and leads ultimately to memory-cell formation. Here, we discuss the signals required for commitment to this program of development and the factors that might influence its progression. Models of the pathways of effector and memory T-cell differentiation are discussed, and we highlight the implications of this new understanding for the optimization of vaccine strategies.
- 76Taylor, M. A.; Hughes, A. M.; Walton, J.; Coenen-Stass, A. M. L.; Magiera, L.; Mooney, L.; Bell, S.; Staniszewska, A. D.; Sandin, L. C.; Barry, S. T. Longitudinal Immune Characterization of Syngeneic Tumor Models to Enable Model Selection for Immune Oncology Drug Discovery. J. Immunother Cancer 2019, 7, 328, DOI: 10.1186/s40425-019-0794-7Google Scholar76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MfksVSntA%253D%253D&md5=7b3308ccad3e1cbb161737db47069240Longitudinal immune characterization of syngeneic tumor models to enable model selection for immune oncology drug discoveryTaylor Molly A; Hughes Adina M; Walton Josephine; Coenen-Stass Anna M L; Magiera Lukasz; Mooney Lorraine; Bell Sigourney; Staniszewska Anna D; Sandin Linda C; Barry Simon T; Watkins Amanda; Carnevalli Larissa S; Hardaker Elizabeth L; Mooney LorraineJournal for immunotherapy of cancer (2019), 7 (1), 328 ISSN:.BACKGROUND: The ability to modulate immune-inhibitory pathways using checkpoint blockade antibodies such as αPD-1, αPD-L1, and αCTLA-4 represents a significant breakthrough in cancer therapy in recent years. This has driven interest in identifying small-molecule-immunotherapy combinations to increase the proportion of responses. Murine syngeneic models, which have a functional immune system, represent an essential tool for pre-clinical evaluation of new immunotherapies. However, immune response varies widely between models and the translational relevance of each model is not fully understood, making selection of an appropriate pre-clinical model for drug target validation challenging. METHODS: Using flow cytometry, O-link protein analysis, RT-PCR, and RNAseq we have characterized kinetic changes in immune-cell populations over the course of tumor development in commonly used syngeneic models. RESULTS: This longitudinal profiling of syngeneic models enables pharmacodynamic time point selection within each model, dependent on the immune population of interest. Additionally, we have characterized the changes in immune populations in each of these models after treatment with the combination of α-PD-L1 and α-CTLA-4 antibodies, enabling benchmarking to known immune modulating treatments within each model. CONCLUSIONS: Taken together, this dataset will provide a framework for characterization and enable the selection of the optimal models for immunotherapy combinations and generate potential biomarkers for clinical evaluation in identifying responders and non-responders to immunotherapy combinations.
- 77Wang-Bishop, L.; Wehbe, M.; Shae, D.; James, J.; Hacker, B. C.; Garland, K.; Chistov, P. P.; Rafat, M.; Balko, J. M.; Wilson, J. T. Potent Sting Activation Stimulates Immunogenic Cell Death to Enhance Antitumor Immunity in Neuroblastoma. J. Immunother Cancer 2020, 8, e000282, DOI: 10.1136/jitc-2019-000282Google ScholarThere is no corresponding record for this reference.
- 78Kilchrist, K. V.; Dimobi, S. C.; Jackson, M. A.; Evans, B. C.; Werfel, T. A.; Dailing, E. A.; Bedingfield, S. K.; Kelly, I. B.; Duvall, C. L. Gal8 Visualization of Endosome Disruption Predicts Carrier-Mediated Biologic Drug Intracellular Bioavailability. ACS Nano 2019, 13, 1136– 1152, DOI: 10.1021/acsnano.8b05482Google Scholar78https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXht12qs7w%253D&md5=35d2d7494c1933249faf76094de990cfGal8 Visualization of Endosome Disruption Predicts Carrier-Mediated Biologic Drug Intracellular BioavailabilityKilchrist, Kameron V.; Dimobi, Somtochukwu C.; Jackson, Meredith A.; Evans, Brian C.; Werfel, Thomas A.; Dailing, Eric A.; Bedingfield, Sean K.; Kelly, Isom B.; Duvall, Craig L.ACS Nano (2019), 13 (2), 1136-1152CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Endolysosome entrapment is one of the key barriers to the therapeutic use of biol. drugs that act intracellularly. The screening of prospective nanoscale endosome-disrupting delivery technologies is currently limited by methods that are indirect and cumbersome. Here, we statistically validate Galectin 8 (Gal8) intracellular tracking as a superior approach that is direct, quant., and predictive of therapeutic cargo intracellular bioactivity through in vitro high-throughput screening and in vivo validation. Gal8 is a cytosolically dispersed protein that, when endosomes are disrupted, redistributes by binding to glycosylation moieties selectively located on the inner face of endosomal membranes. The quant. redistribution of a Gal8 fluorescent fusion protein from the cytosol into endosomes is demonstrated as a real-time, live-cell assessment of endosomal integrity that does not require labeling or modification of either the carrier or the biol. drug and that allows quant. distinction between closely related, endosome-disruptive drug carriers. Through screening two families of siRNA polymeric carrier compns. at varying dosages, we show that Gal8 endosomal recruitment correlates strongly (r = 0.95 and p < 10-4) with intracellular siRNA bioactivity. Through this screen, we gathered insights into how compn. and mol. wt. affect endosome disruption activity of poly[(ethylene glycol)-b-[(2-(dimethylamino)ethyl methacrylate)-co-(Bu methacrylate)]] [PEG-(DMAEMA-co-BMA)] siRNA delivery systems. Addnl. studies showed that Gal8 recruitment predicts intracellular bioactivity better than current std. methods such as Lysotracker colocalization (r = 0.35, not significant), pH-dependent hemolysis (not significant), or cellular uptake (r = 0.73 and p < 10-3). Importantly, the Gal8 recruitment method is also amenable to fully objective high-throughput screening using automated image acquisition and quant. image anal., with a robust estd. Z' of 0.6 (whereas assays with Z' > 0 have high-throughput screening utility). Finally, we also provide measurements of in vivo endosomal disruption based on Gal8 visualization (p < 0.03) of a nanocarrier formulation confirmed to produce significant cytosolic delivery and bioactivity of siRNA within tumors (p < 0.02). In sum, this report establishes the utility of Gal8 subcellular tracking for the rapid optimization and high-throughput screening of the endosome disruption potency of intracellular delivery technologies.
Cited By
This article is cited by 1 publications.
- Dehua Lu, Wenhua Li, Jingyun Tan, Ying Li, Wei Mao, Yuanyuan Zheng, Muwen Yang, Jin Wang, Weihu Wang, Shubin Wang, Jing Gao, Yajie Liu. STING Agonist Delivered by Neutrophil Membrane-Coated Gold Nanoparticles Exerts Synergistic Tumor Inhibition with Radiotherapy. ACS Applied Materials & Interfaces 2024, 16
(40)
, 53474-53488. https://doi.org/10.1021/acsami.4c09825
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 78 other publications.
- 1Korman, A. J.; Garrett-Thomson, S. C.; Lonberg, N. The Foundations of Immune Checkpoint Blockade and the Ipilimumab Approval Decennial. Nat. Rev. Drug Discov 2022, 21, 509– 528, DOI: 10.1038/s41573-021-00345-81https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXivValtr%252FJ&md5=4d70b7f025350759038f63b701a6c494The foundations of immune checkpoint blockade and the ipilimumab approval decennialKorman, Alan J.; Garrett-Thomson, Sarah C.; Lonberg, NilsNature Reviews Drug Discovery (2022), 21 (7), 509-528CODEN: NRDDAG; ISSN:1474-1776. (Nature Portfolio)Cancer immunity, and the potential for cancer immunotherapy, have been topics of scientific discussion and experimentation for over a hundred years. Several successful cancer immunotherapies - such as IL-2 and interferon-a (IFNa) - have appeared over the past 30 years. However, it is only in the past decade that immunotherapy has made a broad impact on patient survival in multiple high-incidence cancer indications. The emergence of immunotherapy as a new pillar of cancer treatment (adding to surgery, radiation, chemotherapy and targeted therapies) is due to the success of immune checkpoint blockade (ICB) drugs, the first of which - ipilimumab - was approved in 2011. ICB drugs block receptors and ligands involved in pathways that attenuate T cell activation - such as cytotoxic T lymphocyte antigen 4 (CTLA4), programed cell death 1 (PD1) and its ligand, PDL1 - and prevent, or reverse, acquired peripheral tolerance to tumor antigens. In this Review we mark the tenth anniversary of the approval of ipilimumab and discuss the foundational scientific history of ICB, together with the history of the discovery, development and elucidation of the mechanism of action of the first generation of drugs targeting the CTLA4 and PD1 pathways.
- 2Sharma, P.; Allison, J. P. The Future of Immune Checkpoint Therapy. Science 2015, 348, 56– 61, DOI: 10.1126/science.aaa81722https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXls1Wmurg%253D&md5=d63ae85b9c651ec64a3b5b002c609e35The future of immune checkpoint therapySharma, Padmanee; Allison, James P.Science (Washington, DC, United States) (2015), 348 (6230), 56-61CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)A review. Immune checkpoint therapy, which targets regulatory pathways in T cells to enhance antitumor immune responses, has led to important clin. advances and provided a new weapon against cancer. This therapy has elicited durable clin. responses and, in a fraction of patients, long-term remissions where patients exhibit no clin. signs of cancer for many years. The way forward for this class of novel agents lies in our ability to understand human immune responses in the tumor microenvironment. This will provide valuable information regarding the dynamic nature of the immune response and regulation of addnl. pathways that will need to be targeted through combination therapies to provide survival benefit for greater nos. of patients.
- 3Shum, B.; Larkin, J.; Turajlic, S. Predictive Biomarkers for Response to Immune Checkpoint Inhibition. Semin Cancer Biol. 2022, 79, 4– 17, DOI: 10.1016/j.semcancer.2021.03.0363https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtlWqt7jK&md5=17802852fefce3fedc8ce6b596fd7510Predictive biomarkers for response to immune checkpoint inhibitionShum, Benjamin; Larkin, James; Turajlic, SamraSeminars in Cancer Biology (2022), 79 (), 4-17CODEN: SECBE7; ISSN:1044-579X. (Elsevier Ltd.)A review. Immune checkpoint inhibitors have transformed the prognosis and treatment paradigm of many cancer types, through the potential for durable responses. However, the majority of patients still do not benefit. Response to checkpoint inhibition is detd. by dynamic host, tumor and tumor microenvironment factors that display spatial and temporal variability, but our understanding of these interactions is incomplete. Through investigating biomarkers of resistance and response, opportunities arise to discover new therapeutic targets and shape personalised treatment strategies. Here we review approved and emerging biomarkers of response to immune checkpoint inhibitors, in particular the recent rapid progress in host and tumor genomics. It is unlikely that a single biomarker will precisely predict response, but multivariate multiomic markers may provide a balanced assessment of these factors and more accurately identify patients who will benefit. Further efforts are required to translate these groundbreaking discoveries into novel therapeutics and biomarker driven clin. trials, to provide durable treatment response to a greater population of patients.
- 4Jenkins, R. W.; Barbie, D. A.; Flaherty, K. T. Mechanisms of Resistance to Immune Checkpoint Inhibitors. Br. J. Cancer 2018, 118, 9– 16, DOI: 10.1038/bjc.2017.4344https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlGmsQ%253D%253D&md5=98d6734fe4172247aa9402fd388159dfMechanisms of resistance to immune checkpoint inhibitorsJenkins, Russell W.; Barbie, David A.; Flaherty, Keith T.British Journal of Cancer (2018), 118 (1), 9-16CODEN: BJCAAI; ISSN:0007-0920. (Nature Research)Immune checkpoint inhibitors (ICI) targeting CTLA-4 and the PD-1/PD-L1 axis have shown unprecedented clin. activity in several types of cancer and are rapidly transforming the practice of medical oncol. Whereas cytotoxic chemotherapy and small mol. inhibitors ('targeted therapies') largely act on cancer cells directly, immune checkpoint inhibitors reinvigorate anti-tumor immune responses by disrupting co-inhibitory T-cell signalling. While resistance routinely develops in patients treated with conventional cancer therapies and targeted therapies, durable responses suggestive of long-lasting immunol. memory are commonly seen in large subsets of patients treated with ICI. However, initial response appears to be a binary event, with most non-responders to single-agent ICI therapy progressing at a rate consistent with the natural history of disease. In addn., late relapses are now emerging with longer follow-up of clin. trial populations, suggesting the emergence of acquired resistance. As robust biomarkers to predict clin. response and/or resistance remain elusive, the mechanisms underlying innate (primary) and acquired (secondary) resistance are largely inferred from pre-clin. studies and correlative clin. data. Improved understanding of mol. and immunol. mechanisms of ICI response (and resistance) may not only identify novel predictive and/or prognostic biomarkers, but also ultimately guide optimal combination/sequencing of ICI therapy in the clinic. Here we review the emerging clin. and pre-clin. data identifying novel mechanisms of innate and acquired resistance to immune checkpoint inhibition.
- 5Ye, Z.; Qian, Q.; Jin, H.; Qian, Q. Cancer Vaccine: Learning Lessons from Immune Checkpoint Inhibitors. J. Cancer 2018, 9, 263– 268, DOI: 10.7150/jca.200595https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MvhslGhtQ%253D%253D&md5=c878c5e7ffce77d94296fce9ae06908aCancer vaccine: learning lessons from immune checkpoint inhibitorsYe ZhenLong; Qian Qiming; Jin HuaJun; Qian QiJunJournal of Cancer (2018), 9 (2), 263-268 ISSN:1837-9664.Cancer vaccines have been exclusively studied all through the past decades, and have made exceptional achievements in cancer treatment. Few cancer vaccines have been approved by the US Food and Drug Administration (FDA), for instance, Provenge, which was approved for the treatment of prostate carcinoma in 2012. Moreover, more recently, T-VEC got approval for the treatment of melanoma. While, the overall therapeutic effects of cancer vaccines have been taken into consideration as below expectations, low antigenicity of targeting antigen and tumor heterogeneity are the two key limiting barriers encountered by the cancer vaccines. Nonetheless, recent developments in cancer immune-therapies together with associated technologies, for instance the unparalleled achievements bagged by immune checkpoint inhibitor based therapies and neo-antigen identification tools, envisage potential improvements in cancer vaccines in respect to the treatments of malignancies. This review brings forth measures for the purpose of refining therapeutic cancer vaccines by learning lessons from the success of PD-1 inhibitor based immune-therapies.
- 6Lin, M. J.; Svensson-Arvelund, J.; Lubitz, G. S.; Marabelle, A.; Melero, I.; Brown, B. D.; Brody, J. D. Cancer Vaccines: The Next Immunotherapy Frontier. Nat. Cancer 2022, 3, 911– 926, DOI: 10.1038/s43018-022-00418-66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitlCit7jE&md5=47262a65ec90f090054e453a5bae8b98Cancer vaccines: the next immunotherapy frontierLin, Matthew J.; Svensson-Arvelund, Judit; Lubitz, Gabrielle S.; Marabelle, Aurelien; Melero, Ignacio; Brown, Brian D.; Brody, Joshua D.Nature Cancer (2022), 3 (8), 911-926CODEN: NCAADQ; ISSN:2662-1347. (Springer International Publishing AG)A review. After several decades, therapeutic cancer vaccines now show signs of efficacy and potential to help patients resistant to other std.-of-care immunotherapies, but they have yet to realize their full potential and expand the oncol. armamentarium. Here, we classify cancer vaccines by what is known of the included antigens, which tumors express those antigens and where the antigens colocalize with antigen-presenting cells, thus delineating predefined vaccines (shared or personalized) and anonymous vaccines (ex vivo or in situ). To expedite clin. development, we highlight the need for accurate immune monitoring of early trials to acknowledge failures and advance the most promising vaccines.
- 7Curran, M. A.; Glisson, B. S. New Hope for Therapeutic Cancer Vaccines in the Era of Immune Checkpoint Modulation. Annu. Rev. Med. 2019, 70, 409– 424, DOI: 10.1146/annurev-med-050217-1219007https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitVGksLzI&md5=58f81e23d229c445006c8672cf6b338bNew Hope for Therapeutic Cancer Vaccines in the Era of Immune Checkpoint ModulationCurran, Michael A.; Glisson, Bonnie S.Annual Review of Medicine (2019), 70 (), 409-424CODEN: ARMCAH; ISSN:0066-4219. (Annual Reviews)A review. The driver and passenger mutations accumulated in the process of malignant transformation offer an adequate spectrum of immune visible alterations to the cellular proteome and resulting peptidome to render these cancers targetable-and, in theory, rejectable-by the host T cell immune response. In addn., cancers often overexpress tissue-specific and developmental antigens to which immune tolerance is incomplete. Sometimes, virally transferred oncogenes drive malignant transformation and remain expressed throughout the cancer. Despite this state of antigenic sufficiency, cancer grows progressively and overcomes all efforts of the host immune system to contain it. While therapeutic cancer vaccination can mobilize high frequencies of tumor-specific T cells, these responses remain subject to intratumoral attenuation. Antibody modulation of T cell function through checkpoint blockade or costimulatory activation can restore survival, proliferation, and effector function to these tumor-infiltrating T cells and convert otherwise subtherapeutic vaccines into potentially curative cancer immunotherapeutics.
- 8Shae, D.; Baljon, J. J.; Wehbe, M.; Becker, K. W.; Sheehy, T. L.; Wilson, J. T. At the Bench: Engineering the Next Generation of Cancer Vaccines. J. Leukoc Biol. 2020, 108, 1435– 1453, DOI: 10.1002/JLB.5BT0119-016R8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1ShsrbN&md5=cd67858ef425a0f35c2b527fcf950ea3At the bench: Engineering the next generation of cancer vaccinesShae, Daniel; Baljon, Jessalyn J.; Wehbe, Mohamed; Becker, Kyle W.; Sheehy, Taylor L.; Wilson, John TannerJournal of Leukocyte Biology (2020), 108 (4), 1435-1453CODEN: JLBIE7; ISSN:1938-3673. (John Wiley & Sons, Inc.)A review. Cancer vaccines hold promise as an immunotherapeutic modality based on their potential to generate tumor antigen-specific T cell responses and long-lived antitumor responses capable of combating metastatic disease and recurrence. However, cancer vaccines have historically failed to deliver significant therapeutic benefit in the clinic, which we maintain is due in part to drug delivery challenges that have limited vaccine immunogenicity and efficacy. In this review, we examine some of the known and putative failure mechanisms of common first-generation clin. cancer vaccines, and describe how the rational design of materials engineered for vaccine delivery and immunomodulation can address these shortcomings. First, we outline vaccine design principles for augmenting cellular immunity to tumor antigens and describe how well-engineered materials can improve vaccine efficacy, highlighting recent innovations in vaccine delivery technol. that are primed for integration into neoantigen vaccine development pipelines. We also discuss the importance of sequencing, timing, and kinetics in mounting effective immune responses to cancer vaccines, and highlight examples of materials that potentiate antitumor immunity through spatiotemporal control of immunomodulation. Furthermore, we describe several engineering strategies for improving outcomes of in situ cancer vaccines, which leverage local, intratumoral delivery to stimulate systemic immunity. Finally, we highlight recent innovations leveraging nanotechnol. for increasing the immunogenicity of the tumor microenvironment (TME), which is crit. to enhancing tumor infiltration and function of T cells elicited in response to cancer vaccines. These immunoengineering strategies and tools complement ongoing advances in cancer vaccines as they reemerge as an important component of the immunotherapeutic armamentarium.
- 9Kleponis, J.; Skelton, R.; Zheng, L. Fueling the Engine and Releasing the Break: Combinational Therapy of Cancer Vaccines and Immune Checkpoint Inhibitors. Cancer Biol. Med. 2015, 12, 201– 208, DOI: 10.7497/j.issn.2095-3941.2015.00469https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXlslGjsbY%253D&md5=98c84619ccf7224c4d30882d091a89a5Fueling the engine and releasing the break: combinational therapy of cancer vaccines and immune checkpoint inhibitorsKleponis, Jennifer; Skelton, Richard; Zheng, LeiCancer Biology & Medicine (2015), 12 (3Spec.Iss.), 201-208CODEN: CBMADQ ISSN:. (Tianjin Medical University Cancer Institute and Hospital)Immune checkpoint inhibitors are increasingly drawing much attention in the therapeutic development for cancer treatment. However, many cancer patients do not respond to treatments with immune checkpoint inhibitors, partly because of the lack of tumor-infiltrating effector T cells. Cancer vaccines may prime patients for treatments with immune checkpoint inhibitors by inducing effector T-cell infiltration into the tumors and immune checkpoint signals. The combination of cancer vaccine and an immune checkpoint inhibitor may function synergistically to induce more effective antitumor immune responses, and clin. trials to test the combination are currently ongoing.
- 10Shemesh, C. S.; Hsu, J. C.; Hosseini, I.; Shen, B. Q.; Rotte, A.; Twomey, P.; Girish, S.; Wu, B. Personalized Cancer Vaccines: Clinical Landscape, Challenges, and Opportunities. Mol. Ther 2021, 29, 555– 570, DOI: 10.1016/j.ymthe.2020.09.03810https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjsVCmsLY%253D&md5=020291133534cc7a2697ae7101562a6cPersonalized Cancer Vaccines: Clinical Landscape, Challenges, and OpportunitiesShemesh, Colby S.; Hsu, Joy C.; Hosseini, Iraj; Shen, Ben-Quan; Rotte, Anand; Twomey, Patrick; Girish, Sandhya; Wu, BenjaminMolecular Therapy (2021), 29 (2), 555-570CODEN: MTOHCK; ISSN:1525-0024. (Cell Press)A review. Tremendous innovation is underway among a rapidly expanding repertoire of promising personalized immune-based treatments. Therapeutic cancer vaccines (TCVs) are attractive systemic immunotherapies that activate and expand antigen-specific CD8+ and CD4+ T cells to enhance anti-tumor immunity. Our review highlights key issues impacting TCVs in clin. practice and reports on progress in development. We review the mechanism of action, immune-monitoring, dosing strategies, combinations, obstacles, and regulation of cancer vaccines. Most trials of personalized TCVs are ongoing and represent diverse platforms with predominantly early investigations of mRNA, DNA, or peptide-based targeting strategies against neoantigens in solid tumors, with many in combination immunotherapies. Multiple delivery systems, routes of administration, and dosing strategies are used. I.v. or i.m. administration is common, including delivery by lipid nanoparticles. Absorption and biodistribution impact antigen uptake, expression, and presentation, affecting the strength, speed, and duration of immune response. The emerging trials illustrate the complexity of developing this class of innovative immunotherapies. Methodical testing of the multiple potential factors influencing immune responses, as well as refined quant. methodologies to facilitate optimal dosing strategies, could help resolve uncertainty of therapeutic approaches. To increase the likelihood of success in bringing these medicines to patients, several unique development challenges must be overcome.
- 11Zhao, J.; Chen, Y.; Ding, Z. Y.; Liu, J. Y. Safety and Efficacy of Therapeutic Cancer Vaccines Alone or in Combination with Immune Checkpoint Inhibitors in Cancer Treatment. Front Pharmacol 2019, 10, 1184, DOI: 10.3389/fphar.2019.0118411https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXpvVeqtLk%253D&md5=b9137c6f5bd286e8db5eb610319bab4bSafety and efficacy of therapeutic cancer vaccines alone or in combination with immune checkpoint inhibitors in cancer treatmentZhao, Jing; Chen, Ye; Ding, Zhen-Yu.; Liu, Ji-YanFrontiers in Pharmacology (2019), 10 (), 1184CODEN: FPRHAU; ISSN:1663-9812. (Frontiers Media S.A.)A review. Therapeutic cancer vaccines have proven to seldom induce dramatic clin. response when used alone, and therefore, they are being studied in combination with addnl. treatment modalities to achieve optimal treatment activities. Growing preclin. data show that combining vaccines and immune checkpoint inhibitors (ICIs) can prime intensified immunogenicity and modulate immunosuppressive tumor microenvironment. Herein, we focus on the safety and efficacy of approved and promising cancer vaccines alone or combined with ICIs in the treatment of several malignancies. Generally, the majority of clin. trials support the concept of synergy that combination therapy of vaccines and ICIs holds maximized potential to improve clin. outcomes. Importantly, the combination has acceptable safety and minimal addnl. toxicity compared with single-agent vaccines or ICIs. Addnl., the potential strategies of combining personalized tumor vaccines with ICIs will become priority option and future direction of vaccine development and application and the urgent need to develop effective biomarkers to screen appropriate patient populations and predict response to combination therapy.
- 12Blass, E.; Ott, P. A. Advances in the Development of Personalized Neoantigen-Based Therapeutic Cancer Vaccines. Nat. Rev. Clin Oncol 2021, 18, 215– 229, DOI: 10.1038/s41571-020-00460-212https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3srivFOqtQ%253D%253D&md5=82c721ffe87a85367bc1081598d18a5dAdvances in the development of personalized neoantigen-based therapeutic cancer vaccinesBlass Eryn; Ott Patrick A; Ott Patrick A; Ott Patrick A; Ott Patrick ANature reviews. Clinical oncology (2021), 18 (4), 215-229 ISSN:.Within the past decade, the field of immunotherapy has revolutionized the treatment of many cancers with the development and regulatory approval of various immune-checkpoint inhibitors and chimeric antigen receptor T cell therapies in diverse indications. Another promising approach to cancer immunotherapy involves the use of personalized vaccines designed to trigger de novo T cell responses against neoantigens, which are highly specific to tumours of individual patients, in order to amplify and broaden the endogenous repertoire of tumour-specific T cells. Results from initial clinical studies of personalized neoantigen-based vaccines, enabled by the availability of rapid and cost-effective sequencing and bioinformatics technologies, have demonstrated robust tumour-specific immunogenicity and preliminary evidence of antitumour activity in patients with melanoma and other cancers. Herein, we provide an overview of the complex process that is necessary to generate a personalized neoantigen vaccine, review the types of vaccine-induced T cells that are found within tumours and outline strategies to enhance the T cell responses. In addition, we discuss the current status of clinical studies testing personalized neoantigen vaccines in patients with cancer and considerations for future clinical investigation of this novel, individualized approach to immunotherapy.
- 13Schumacher, T. N.; Scheper, W.; Kvistborg, P. Cancer Neoantigens. Annu. Rev. Immunol. 2019, 37, 173– 200, DOI: 10.1146/annurev-immunol-042617-05340213https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFejsLfP&md5=21b8e70354a6e489f6f56f526f2f08baCancer NeoantigensSchumacher, Ton N.; Scheper, Wouter; Kvistborg, PiaAnnual Review of Immunology (2019), 37 (), 173-200CODEN: ARIMDU; ISSN:0732-0582. (Annual Reviews)A review. Malignant transformation of cells depends on accumulation of DNA damage. Over the past years we have learned that the T cell-based immune system frequently responds to the neoantigens that arise as a consequence of this DNA damage. Furthermore, recognition of neoantigens appears an important driver of the clin. activity of both T cell checkpoint blockade and adoptive T cell therapy as cancer immunotherapies. Here we review the evidence for the relevance of cancer neoantigens in tumor control and the biol. properties of these antigens. We discuss recent technol. advances utilized to identify neoantigens, and the T cells that recognize them, in individual patients. Finally, we discuss strategies that can be employed to exploit cancer neoantigens in clin. interventions.
- 14Janes, M. E.; Gottlieb, A. P.; Park, K. S.; Zhao, Z.; Mitragotri, S. Cancer Vaccines in the Clinic. Bioeng Transl Med. 2024, 9, e10588 DOI: 10.1002/btm2.10588There is no corresponding record for this reference.
- 15Baljon, J. J.; Wilson, J. T. Bioinspired Vaccines to Enhance Mhc Class-I Antigen Cross-Presentation. Curr. Opin Immunol 2022, 77, 102215, DOI: 10.1016/j.coi.2022.10221515https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xhtl2jtr7M&md5=66ac8dde3eed78c8f43ac96caae336feBioinspired vaccines to enhance MHC class-I antigen cross-presentationBaljon, Jessalyn J.; Wilson, John T.Current Opinion in Immunology (2022), 77 (), 102215CODEN: COPIEL; ISSN:0952-7915. (Elsevier Ltd.)A review. Here, we briefly summarize known cross-presentation pathways and highlight how synthetic vaccines can be engineered to enhance MHC-I presentation of exogenous peptide and protein antigens by professional antigen-presenting cells (APCs). In particular, we summarize how mol. engineering and nanotechnol. are being harnessed to enhance antigen delivery to lymph nodes and to cross-presenting dendritic cells, to bypass endosomal trafficking of exogenous antigen to promote delivery of antigen to the cytosol of APCs, and to coordinate the delivery of antigen with immune-stimulating adjuvants that can act synergistically to augment antigen cross-presentation. Cross-presentation of exogenous antigen on MHC class-I is a crucial process for generating a CD8+ T cell response, and is therefore an important design consideration in the development of T-cell-engaging vaccines against viruses, intracellular bacteria, and cancers.
- 16Liu, W.; Tang, H.; Li, L.; Wang, X.; Yu, Z.; Li, J. Peptide-Based Therapeutic Cancer Vaccine: Current Trends in Clinical Application. Cell Prolif 2021, 54, e13025 DOI: 10.1111/cpr.1302516https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVWrtLfK&md5=00a6a94c14b8864a77e1fcce50f5f785Peptide-based therapeutic cancer vaccine: Current trends in clinical applicationLiu, Wensi; Tang, Haichao; Li, Luanfeng; Wang, Xiangyi; Yu, Zhaojin; Li, JianpingCell Proliferation (2021), 54 (5), e13025CODEN: CPROEM; ISSN:0960-7722. (John Wiley & Sons Ltd.)A review. The peptide-based therapeutic cancer vaccines have attracted enormous attention in recent years as one of the effective treatments of tumor immunotherapy. Most of peptide-based vaccines are based on epitope peptides stimulating CD8+ T cells or CD4+ T helper cells to target tumor-assocd. antigens (TAAs) or tumor-specific antigens (TSAs). Some adjuvants and nanomaterials have been exploited to optimize the efficiency of immune response of the epitope peptide to improve its clin. application. At present, numerous peptide-based therapeutic cancer vaccines have been developed and achieved significant clin. benefits. Similarly, the combination of peptide-based vaccines and other therapies has demonstrated a superior efficacy in improving anti-cancer activity. We delve deeper into the choices of targets, design and screening of epitope peptides, clin. efficacy and adverse events of peptide-based vaccines, and strategies combination of peptide-based therapeutic cancer vaccines and other therapies. The review will provide a detailed overview and basis for future clin. application of peptide-based therapeutic cancer vaccines.
- 17Stephens, A. J.; Burgess-Brown, N. A.; Jiang, S. Beyond Just Peptide Antigens: The Complex World of Peptide-Based Cancer Vaccines. Front Immunol 2021, 12, 696791, DOI: 10.3389/fimmu.2021.69679117https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFOitrvK&md5=d642fe90d7d555cd18312a800d8be80fBeyond just peptide antigens: the complex world of peptide-based cancer vaccinesStephens, Alexander J.; Burgess-Brown, Nicola A.; Jiang, ShisongFrontiers in Immunology (2021), 12 (), 696791CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)A review. Peptide-based cancer vaccines rely upon the strong activation of the adaptive immune response to elicit its effector function. They have shown to be highly specific and safe, but have yet to prove themselves as an efficacious treatment for cancer in the clinic. This is for a variety of reasons, including tumor heterogeneity, self-tolerance, and immune suppression. Importance has been placed on the overall design of peptide-based cancer vaccines, which have evolved from simple peptide derivs. of a cancer antigen, to complex drugs; incorporating overlapping regions, conjugates, and delivery systems to target and stimulate different components of antigen presenting cells, and to bolster antigen cross-presentation. Peptide-based cancer vaccines are increasingly becoming more personalised to an individuals tumor antigen repertoire and are often combined with existing cancer treatments. This strategy ultimately aids in combating the shortcomings of a more generalised vaccine strategy and provides a comprehensive treatment, taking into consideration cancer cell variability and its ability to avoid immune interrogation.
- 18Yewdell, J. W. Designing CD8+ T Cell Vaccines: It’s Not Rocket Science (Yet). Curr. Opin Immunol 2010, 22, 402– 410, DOI: 10.1016/j.coi.2010.04.00218https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXntV2jsbk%253D&md5=f6f6853cd382fab6b98cb14088f25b2fDesigning CD8+ T cell vaccines: it's not rocket science (yet)Yewdell, Jonathan W.Current Opinion in Immunology (2010), 22 (3), 402-410CODEN: COPIEL; ISSN:0952-7915. (Elsevier B.V.)A review. CD8+ T cells play important roles in clearing viral infections and eradicating tumors. Designing vaccines that elicit effective CD8+ T cell responses requires a thorough knowledge of the pathways of antigen presentation in vivo. Here, I review recent progress in understanding the activation of naive CD8+ T cells in vivo, with particular emphasis on cross-priming, the presentation of protein antigens acquired by dendritic cells from their environment. With the rapid advances in this area of research, the dawn of rational vaccine design is at hand.
- 19Khong, H.; Overwijk, W. W. Adjuvants for Peptide-Based Cancer Vaccines. J. Immunother Cancer 2016, 4, 56, DOI: 10.1186/s40425-016-0160-y19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2svjtl2ntw%253D%253D&md5=caffd368c197f407b12a8b1264be8038Adjuvants for peptide-based cancer vaccinesKhong Hiep; Overwijk Willem WJournal for immunotherapy of cancer (2016), 4 (), 56 ISSN:2051-1426.Cancer therapies based on T cells have shown impressive clinical benefit. In particular, immune checkpoint blockade therapies with anti-CTLA-4 and anti-PD-1/PD-L1 are causing dramatic tumor shrinkage and prolonged patient survival in a variety of cancers. However, many patients do not benefit, possibly due to insufficient spontaneous T cell reactivity against their tumors and/or lacking immune cell infiltration to tumor site. Such tumor-specific T cell responses could be induced through anti-cancer vaccination; but despite great success in animal models, only a few of many cancer vaccine trials have demonstrated robust clinical benefit. One reason for this difference may be the use of potent, effective vaccine adjuvants in animal models, vs. the use of safe, but very weak, vaccine adjuvants in clinical trials. As vaccine adjuvants dictate the type and magnitude of the T cell response after vaccination, it is critical to understand how they work to design safe, but also effective, cancer vaccines for clinical use. Here we discuss current insights into the mechanism of action and practical application of vaccine adjuvants, with a focus on peptide-based cancer vaccines.
- 20Gouttefangeas, C.; Rammensee, H. G. Personalized Cancer Vaccines: Adjuvants Are Important, Too. Cancer Immunol Immunother 2018, 67, 1911– 1918, DOI: 10.1007/s00262-018-2158-420https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXnsVGqsrc%253D&md5=f0bb1792ca78fcf2e11b427377a3cd44Personalized cancer vaccines: adjuvants are important, tooGouttefangeas, Cecile; Rammensee, Hans-GeorgCancer Immunology Immunotherapy (2018), 67 (12), 1911-1918CODEN: CIIMDN; ISSN:0340-7004. (Springer)Therapeutic cancer vaccines have shown limited clin. efficacy so far. Nevertheless, in the meantime, our understanding of immune cell function and the interactions of immune cells with growing tumors has advanced considerably. We are now in a position to invest this knowledge into the design of more powerful vaccines and therapy combinations aimed at increasing immunogenicity and decreasing tumor-induced immunosuppression. This review focuses essentially on peptide-based human vaccines. We will discuss two aspects that are crit. for increasing their intrinsic immunogenicity: the selection of the antigen(s) to be targeted, and the as yet unmet need for strong adjuvants.
- 21Temizoz, B.; Kuroda, E.; Ishii, K. J. Vaccine Adjuvants as Potential Cancer Immunotherapeutics. Int. Immunol. 2016, 28, 329– 338, DOI: 10.1093/intimm/dxw01521https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFOru7fE&md5=92aa03134406c55673017cb24da45eb4Vaccine adjuvants as potential cancer immunotherapeuticsTemizoz, Burcu; Kuroda, Etsushi; Ishii, Ken J.International Immunology (2016), 28 (7), 329-338CODEN: INIMEN; ISSN:0953-8178. (Oxford University Press)Accumulated evidence obtained from various clin. trials and animal studies suggested that cancer vaccines need better adjuvants than those that are currently licensed, which include the most commonly used alum and incomplete Freund's adjuvant, because of either a lack of potent anti-tumor immunity or the induction of undesired immunity. Several clin. trials using immunostimulatory adjuvants, particularly agonistic as well as non-agonistic ligands for TLRs, C-type lectin receptors, retinoic acid-inducible gene I-like receptors and stimulator of interferon genes, have revealed their therapeutic potential not only as vaccine adjuvants but also as anti-tumor agents. Recently, combinations of such immunostimulatory or immunomodulatory adjuvants have shown superior efficacy over their singular use, suggesting that seeking optimal combinations of the currently available or well-characterized adjuvants may provide a better chance for the development of novel adjuvants for cancer immunotherapy.
- 22Kaur, A.; Baldwin, J.; Brar, D.; Salunke, D. B.; Petrovsky, N. Toll-Like Receptor (TLR) Agonists as a Driving Force Behind Next-Generation Vaccine Adjuvants and Cancer Therapeutics. Curr. Opin Chem. Biol. 2022, 70, 102172, DOI: 10.1016/j.cbpa.2022.10217222https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xhs1yitbnL&md5=3150d579bdb7406e044934c74a7af2cdToll-like receptor (TLR) agonists as a driving force behind next-generation vaccine adjuvants and cancer therapeuticsKaur, Arshpreet; Baldwin, Jeremy; Brar, Deshkanwar; Salunke, Deepak B.; Petrovsky, NikolaiCurrent Opinion in Chemical Biology (2022), 70 (), 102172CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review until recently, the development of new human adjuvants was held back by a poor understanding of their mechanisms of action. The field was revolutionized by the discovery of the toll-like receptors (TLRs), innate immune receptors that directly or indirectly are responsible for detecting pathogen-assocd. mol. patterns (PAMPs) and respond to them by activating innate and adaptive immune pathways. Hundreds of ligands targeting various TLRs have since been identified and characterized as vaccine adjuvants. This work has important implications not only for the development of vaccines against infectious diseases but also for immuno-therapies against cancer, allergy, Alzheimer's disease, drug addiction and other diseases. Each TLR has its own specific tissue localization and downstream gene pathways, providing researchers the opportunity to precisely tailor adjuvants with specific immune effects. TLR agonists can be combined with other TLR or alternative adjuvants to create combination adjuvants with synergistic or modulatory effects. This review provides an introduction to the various classes of TLR adjuvants and their resp. pathways. It provides an overview of recent advancements in the TLR field in the past 2-3 years and discusses criteria for selecting specific TLR adjuvants based on considerations, such as disease mechanisms and correlates of protection, TLR immune biasing capabilities, route of administration, antigen compatibility, new vaccine technol. platforms, and age- and species-specific effects.
- 23Hu, H. G.; Li, Y. M. Emerging Adjuvants for Cancer Immunotherapy. Front Chem. 2020, 8, 601, DOI: 10.3389/fchem.2020.0060123https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitFWit7vF&md5=03aa3cb231a4e3245efde5c3e6959b61Emerging adjuvants for cancer immunotherapyHu, Hong-Guo; Li, Yan-MeiFrontiers in Chemistry (Lausanne, Switzerland) (2020), 8 (), 601CODEN: FCLSAA; ISSN:2296-2646. (Frontiers Media S.A.)A review. Cancer is a life-threatening disease, and immunotherapies have been developed as a novel, potent treatment for cancer. Adjuvants, used alone or in combination with other agents, play crucial roles in immune activation. This is necessary for cancer immunotherapy, particularly in the construction of therapeutic cancer vaccines. Adjuvants activate antigen-presenting cells and promote the presentation of antigen epitopes on major histocompatibility complex mols., further enhancing adaptive immune responses, including cytotoxic T lymphocytes, to elicit cancer-cell death. However, the applications of adjuvants are limited by their poor efficacy or insufficient safety. In recent studies, researchers attempted to develop safe, efficacious adjuvants for cancer immunotherapy, and many compds. (including inorg. compds., org. mols., polymers, and colloids) have been identified and optimized as agonists of various pathways. In this , we focus on the discovery and structural design of emerging adjuvants and discuss how these findings benefit healthcare.
- 24Paston, S. J.; Brentville, V. A.; Symonds, P.; Durrant, L. G. Cancer Vaccines, Adjuvants, and Delivery Systems. Front Immunol 2021, 12, 627932, DOI: 10.3389/fimmu.2021.62793224https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXpvVyhtrg%253D&md5=3d9e90696dda37b5ab117b67a282262dCancer vaccines, adjuvants, and delivery systemsPaston, Samantha j.; Brentville, Victoria a.; Symonds, Peter; Durrant, Lindy g.Frontiers in Immunology (2021), 12 (), 627932CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)A review. Vaccination was first pioneered in the 18th century by Edward Jenner and eventually led to the development of the smallpox vaccine and subsequently the eradication of smallpox. The impact of vaccination to prevent infectious diseases has been outstanding with many infections being prevented and a significant decrease in mortality worldwide. Cancer vaccines aim to clear active disease instead of aiming to prevent disease, the only exception being the recently approved vaccine that prevents cancers caused by the Human Papillomavirus. The development of therapeutic cancer vaccines has been disappointing with many early cancer vaccines that showed promise in preclin. models often failing to translate into efficacy in the clinic. In this review we provide an overview of the current vaccine platforms, adjuvants and delivery systems that are currently being investigated or have been approved. With the advent of immune checkpoint inhibitors, we also review the potential of these to be used with cancer vaccines to improve efficacy and help to overcome the immune suppressive tumor microenvironment.
- 25Keskin, D. B.; Anandappa, A. J.; Sun, J.; Tirosh, I.; Mathewson, N. D.; Li, S.; Oliveira, G.; Giobbie-Hurder, A.; Felt, K.; Gjini, E. Neoantigen Vaccine Generates Intratumoral T Cell Responses in Phase Ib Glioblastoma Trial. Nature 2019, 565, 234– 239, DOI: 10.1038/s41586-018-0792-925https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFyksLvE&md5=665425d605411ebfd4764314e7d1f220Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trialKeskin, Derin B.; Anandappa, Annabelle J.; Sun, Jing; Tirosh, Itay; Mathewson, Nathan D.; Li, Shuqiang; Oliveira, Giacomo; Giobbie-Hurder, Anita; Felt, Kristen; Gjini, Evisa; Shukla, Sachet A.; Hu, Zhuting; Li, Letitia; Le, Phuong M.; Allesoee, Rosa L.; Richman, Alyssa R.; Kowalczyk, Monika S.; Abdelrahman, Sara; Geduldig, Jack E.; Charbonneau, Sarah; Pelton, Kristine; Iorgulescu, J. Bryan; Elagina, Liudmila; Zhang, Wandi; Olive, Oriol; McCluskey, Christine; Olsen, Lars R.; Stevens, Jonathan; Lane, William J.; Salazar, Andres M.; Daley, Heather; Wen, Patrick Y.; Chiocca, E. Antonio; Harden, Maegan; Lennon, Niall J.; Gabriel, Stacey; Getz, Gad; Lander, Eric S.; Regev, Aviv; Ritz, Jerome; Neuberg, Donna; Rodig, Scott J.; Ligon, Keith L.; Suva, Mario L.; Wucherpfennig, Kai W.; Hacohen, Nir; Fritsch, Edward F.; Livak, Kenneth J.; Ott, Patrick A.; Wu, Catherine J.; Reardon, David A.Nature (London, United Kingdom) (2019), 565 (7738), 234-239CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Neoantigens, which are derived from tumor-specific protein-coding mutations, are exempt from central tolerance, can generate robust immune responses1,2 and can function as bona fide antigens that facilitate tumor rejection3. Here we demonstrate that a strategy that uses multi-epitope, personalized neoantigen vaccination, which has previously been tested in patients with high-risk melanoma4-6, is feasible for tumors such as glioblastoma, which typically have a relatively low mutation load1,7 and an immunol. 'cold' tumor microenvironment8. We used personalized neoantigen-targeting vaccines to immunize patients newly diagnosed with glioblastoma following surgical resection and conventional radiotherapy in a phase I/Ib study. Patients who did not receive dexamethasone-a highly potent corticosteroid that is frequently prescribed to treat cerebral edema in patients with glioblastoma-generated circulating polyfunctional neoantigen-specific CD4+ and CD8+ T cell responses that were enriched in a memory phenotype and showed an increase in the no. of tumor-infiltrating T cells. Using single-cell T cell receptor anal., we provide evidence that neoantigen-specific T cells from the peripheral blood can migrate into an intracranial glioblastoma tumor. Neoantigen-targeting vaccines thus have the potential to favorably alter the immune milieu of glioblastoma.
- 26Ott, P. A.; Hu, Z.; Keskin, D. B.; Shukla, S. A.; Sun, J.; Bozym, D. J.; Zhang, W.; Luoma, A.; Giobbie-Hurder, A.; Peter, L.; Chen, C.; Olive, O.; Carter, T. A.; Li, S.; Lieb, D. J.; Eisenhaure, T.; Gjini, E.; Stevens, J.; Lane, W. J.; Javeri, I. An Immunogenic Personal Neoantigen Vaccine for Patients with Melanoma. Nature 2017, 547, 217– 221, DOI: 10.1038/nature2299126https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFaqt7rO&md5=e235523b95d4f6ae4aeceb1047460ab3An immunogenic personal neoantigen vaccine for patients with melanomaOtt, Patrick A.; Hu, Zhuting; Keskin, Derin B.; Shukla, Sachet A.; Sun, Jing; Bozym, David J.; Zhang, Wandi; Luoma, Adrienne; Giobbie-Hurder, Anita; Peter, Lauren; Chen, Christina; Olive, Oriol; Carter, Todd A.; Li, Shuqiang; Lieb, David J.; Eisenhaure, Thomas; Gjini, Evisa; Stevens, Jonathan; Lane, William J.; Javeri, Indu; Nellaiappan, Kaliappanadar; Salazar, Andres M.; Daley, Heather; Seaman, Michael; Buchbinder, Elizabeth I.; Yoon, Charles H.; Harden, Maegan; Lennon, Niall; Gabriel, Stacey; Rodig, Scott J.; Barouch, Dan H.; Aster, Jon C.; Getz, Gad; Wucherpfennig, Kai; Neuberg, Donna; Ritz, Jerome; Lander, Eric S.; Fritsch, Edward F.; Hacohen, Nir; Wu, Catherine J.Nature (London, United Kingdom) (2017), 547 (7662), 217-221CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Effective anti-tumor immunity in humans has been assocd. with the presence of T cells directed at cancer neoantigens, a class of HLA-bound peptides that arise from tumor-specific mutations. They are highly immunogenic because they are not present in normal tissues and hence bypass central thymic tolerance. Although neoantigens were long-envisioned as optimal targets for an anti-tumor immune response, their systematic discovery and evaluation only became feasible with the recent availability of massively parallel sequencing for detection of all coding mutations within tumors, and of machine learning approaches to reliably predict those mutated peptides with high-affinity binding of autologous human leukocyte antigen (HLA) mols. We hypothesized that vaccination with neoantigens can both expand pre-existing neoantigen-specific T-cell populations and induce a broader repertoire of new T-cell specificities in cancer patients, tipping the intra-tumoral balance in favor of enhanced tumor control. Here we demonstrate the feasibility, safety, and immunogenicity of a vaccine that targets up to 20 predicted personal tumor neoantigens. Vaccine-induced polyfunctional CD4+ and CD8+ T cells targeted 58 (60%) and 15 (16%) of the 97 unique neoantigens used across patients, resp. These T cells discriminated mutated from wild-type antigens, and in some cases directly recognized autologous tumor. Of six vaccinated patients, four had no recurrence at 25 mo after vaccination, while two with recurrent disease were subsequently treated with anti-PD-1 (anti-programmed cell death-1) therapy and experienced complete tumor regression, with expansion of the repertoire of neoantigen-specific T cells. These data provide a strong rationale for further development of this approach, alone and in combination with checkpoint blockade or other immunotherapies.
- 27Swartz, M. A.; Hirosue, S.; Hubbell, J. A. Engineering Approaches to Immunotherapy. Science Translational Medicine 2012, 4, 148rv149, DOI: 10.1126/scitranslmed.3003763There is no corresponding record for this reference.
- 28Knight, F. C.; Gilchuk, P.; Kumar, A.; Becker, K. W.; Sevimli, S.; Jacobson, M. E.; Suryadevara, N.; Wang-Bishop, L.; Boyd, K. L.; Crowe, J. E., Jr.; Joyce, S.; Wilson, J. T. Mucosal Immunization with a pH-Responsive Nanoparticle Vaccine Induces Protective CD8(+) Lung-Resident Memory T Cells. ACS Nano 2019, 13, 10939– 10960, DOI: 10.1021/acsnano.9b0032628https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvVCnt7%252FO&md5=44662b0a177490dcb700c98b7fd56e5eMucosal Immunization with a pH-Responsive Nanoparticle Vaccine Induces Protective CD8+ Lung-Resident Memory T CellsKnight, Frances C.; Gilchuk, Pavlo; Kumar, Amrendra; Becker, Kyle W.; Sevimli, Sema; Jacobson, Max E.; Suryadevara, Naveenchandra; Wang-Bishop, Lihong; Boyd, Kelli L.; Crowe, James E.; Joyce, Sebastian; Wilson, John T.ACS Nano (2019), 13 (10), 10939-10960CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Tissue-resident memory T cells (TRM) patrol nonlymphoid organs and provide superior protection against pathogens that commonly infect mucosal and barrier tissues, such as the lungs, intestine, liver, and skin. Thus, there is a need for vaccine technologies that can induce a robust, protective TRM response in these tissues. Nanoparticle (NP) vaccines offer important advantages over conventional vaccines; however, there has been minimal investigation into the design of NP-based vaccines for eliciting TRM responses. Here, we describe a pH-responsive polymeric nanoparticle vaccine for generating antigen-specific CD8+ TRM cells in the lungs. With a single intranasal dose, the NP vaccine elicited airway- and lung-resident CD8+ TRM cells and protected against respiratory virus challenge in both sublethal (vaccinia) and lethal (influenza) infection models for up to 9 wk after immunization. In elucidating the contribution of material properties to the resulting TRM response, we found that the pH-responsive activity of the carrier was important, as a structurally analogous non-pH-responsive control carrier elicited significantly fewer lung-resident CD8+ T cells. We also demonstrated that dual-delivery of protein antigen and nucleic acid adjuvant on the same NP substantially enhanced the magnitude, functionality, and longevity of the antigen-specific CD8+ TRM response in the lungs. Compared to administration of sol. antigen and adjuvant, the NP also mediated retention of vaccine cargo in pulmonary antigen-presenting cells (APCs), enhanced APC activation, and increased prodn. of TRM-related cytokines. Overall, these data suggest a promising vaccine platform technol. for rapid generation of protective CD8+ TRM cells in the lungs.
- 29Kuai, R.; Ochyl, L. J.; Bahjat, K. S.; Schwendeman, A.; Moon, J. J. Designer Vaccine Nanodiscs for Personalized Cancer Immunotherapy. Nat. Mater. 2017, 16, 489– 496, DOI: 10.1038/nmat482229https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFGitLvO&md5=247bc6ef1a21f3a1bce8addcbb7a6a0dDesigner vaccine nanodiscs for personalized cancer immunotherapyKuai, Rui; Ochyl, Lukasz J.; Bahjat, Keith S.; Schwendeman, Anna; Moon, James J.Nature Materials (2017), 16 (4), 489-496CODEN: NMAACR; ISSN:1476-1122. (Nature Publishing Group)Despite the tremendous potential of peptide-based cancer vaccines, their efficacy has been limited in humans. Recent innovations in tumor exome sequencing have signalled the new era of personalized immunotherapy with patient-specific neoantigens, but a general methodol. for stimulating strong CD8α+ cytotoxic T-lymphocyte (CTL) responses remains lacking. Here we demonstrate that high-d. lipoprotein-mimicking nanodiscs coupled with antigen (Ag) peptides and adjuvants can markedly improve Ag/adjuvant co-delivery to lymphoid organs and sustain Ag presentation on dendritic cells. Strikingly, nanodiscs elicited up to 47-fold greater frequencies of neoantigen-specific CTLs than sol. vaccines and even 31-fold greater than perhaps the strongest adjuvant in clin. trials (i.e., CpG in Montanide). Moreover, multi-epitope vaccination generated broad-spectrum T-cell responses that potently inhibited tumor growth. Nanodiscs eliminated established MC-38 and B16F10 tumors when combined with anti-PD-1 and anti-CTLA-4 therapy. These findings represent a new powerful approach for cancer immunotherapy and suggest a general strategy for personalized nanomedicine.
- 30Li, A. W.; Sobral, M. C.; Badrinath, S.; Choi, Y.; Graveline, A.; Stafford, A. G.; Weaver, J. C.; Dellacherie, M. O.; Shih, T. Y.; Ali, O. A.; Kim, J.; Wucherpfennig, K. W.; Mooney, D. J. A Facile Approach to Enhance Antigen Response for Personalized Cancer Vaccination. Nat. Mater. 2018, 17, 528– 534, DOI: 10.1038/s41563-018-0028-230https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXltFOrsrs%253D&md5=3723b6388b35a6a546eb78ace8db9e9fA facile approach to enhance antigen response for personalized cancer vaccinationLi, Aileen Weiwei; Sobral, Miguel C.; Badrinath, Soumya; Choi, Youngjin; Graveline, Amanda; Stafford, Alexander G.; Weaver, James C.; Dellacherie, Maxence O.; Shih, Ting-Yu; Ali, Omar A.; Kim, Jaeyun; Wucherpfennig, Kai W.; Mooney, David J.Nature Materials (2018), 17 (6), 528-534CODEN: NMAACR; ISSN:1476-1122. (Nature Research)Existing strategies to enhance peptide immunogenicity for cancer vaccination generally require direct peptide alteration, which, beyond practical issues, may impact peptide presentation and result in vaccine variability. Here, we report a simple adsorption approach using polyethyleneimine (PEI) in a mesoporous silica microrod (MSR) vaccine to enhance antigen immunogenicity. The MSR-PEI vaccine significantly enhanced host dendritic cell activation and T-cell response over the existing MSR vaccine and bolus vaccine formulations. Impressively, a single injection of the MSR-PEI vaccine using an E7 peptide completely eradicated large, established TC-1 tumors in about 80% of mice and generated immunol. memory. When immunized with a pool of B16F10 or CT26 neoantigens, the MSR-PEI vaccine eradicated established lung metastases, controlled tumor growth and synergized with anti-CTLA4 therapy. Our findings from three independent tumor models suggest that the MSR-PEI vaccine approach may serve as a facile and powerful multi-antigen platform to enable robust personalized cancer vaccination.
- 31Liu, H.; Moynihan, K. D.; Zheng, Y.; Szeto, G. L.; Li, A. V.; Huang, B.; Van Egeren, D. S.; Park, C.; Irvine, D. J. Structure-Based Programming of Lymph-Node Targeting in Molecular Vaccines. Nature 2014, 507, 519– 522, DOI: 10.1038/nature1297831https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXkvV2ru70%253D&md5=2890c3d27d8b989930052ba6bbed3294Structure-based programming of lymph-node targeting in molecular vaccinesLiu, Haipeng; Moynihan, Kelly D.; Zheng, Yiran; Szeto, Gregory L.; Li, Adrienne V.; Huang, Bonnie; Van Egeren, Debra S.; Park, Clara; Irvine, Darrell J.Nature (London, United Kingdom) (2014), 507 (7493), 519-522CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)In cancer patients, visual identification of sentinel lymph nodes (LNs) is achieved by the injection of dyes that bind avidly to endogenous albumin, targeting these compds. to LNs, where they are efficiently filtered by resident phagocytes. Here we translate this 'albumin hitchhiking' approach to mol. vaccines, through the synthesis of amphiphiles (amph-vaccines) comprising an antigen or adjuvant cargo linked to a lipophilic albumin-binding tail by a soly.-promoting polar polymer chain. Administration of structurally optimized CpG-DNA/peptide amph-vaccines in mice resulted in marked increases in LN accumulation and decreased systemic dissemination relative to their parent compds., leading to 30-fold increases in T-cell priming and enhanced anti-tumor efficacy while greatly reducing systemic toxicity. Amph-vaccines provide a simple, broadly applicable strategy to simultaneously increase the potency and safety of subunit vaccines.
- 32Lynn, G. M.; Sedlik, C.; Baharom, F.; Zhu, Y.; Ramirez-Valdez, R. A.; Coble, V. L.; Tobin, K.; Nichols, S. R.; Itzkowitz, Y.; Zaidi, N.; Gammon, J. M.; Blobel, N. J.; Denizeau, J.; de la Rochere, P.; Francica, B. J.; Decker, B.; Maciejewski, M.; Cheung, J.; Yamane, H.; Smelkinson, M. G. Peptide-TLR-7/8a Conjugate Vaccines Chemically Programmed for Nanoparticle Self-Assembly Enhance CD8 T-Cell Immunity to Tumor Antigens. Nat. Biotechnol. 2020, 38, 320– 332, DOI: 10.1038/s41587-019-0390-x32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXotFGltw%253D%253D&md5=2b4321aa1d0eec8e7a4b2d5aca33a648Peptide-TLR-7/8a conjugate vaccines chemically programmed for nanoparticle self-assembly enhance CD8 T-cell immunity to tumor antigensLynn, Geoffrey M.; Sedlik, Christine; Baharom, Faezzah; Zhu, Yaling; Ramirez-Valdez, Ramiro A.; Coble, Vincent L.; Tobin, Kennedy; Nichols, Sarah R.; Itzkowitz, Yaakov; Zaidi, Neeha; Gammon, Joshua M.; Blobel, Nicolas J.; Denizeau, Jordan; de la Rochere, Philippe; Francica, Brian J.; Decker, Brennan; Maciejewski, Mateusz; Cheung, Justin; Yamane, Hidehiro; Smelkinson, Margery G.; Francica, Joseph R.; Laga, Richard; Bernstock, Joshua D.; Seymour, Leonard W.; Drake, Charles G.; Jewell, Christopher M.; Lantz, Olivier; Piaggio, Eliane; Ishizuka, Andrew S.; Seder, Robert A.Nature Biotechnology (2020), 38 (3), 320-332CODEN: NABIF9; ISSN:1087-0156. (Nature Research)Personalized cancer vaccines targeting patient-specific neoantigens are a promising cancer treatment modality; however, neoantigen physicochem. variability can present challenges to manufg. personalized cancer vaccines in an optimal format for inducing anticancer T cells. Here, we developed a vaccine platform (SNP-7/8a) based on charge-modified peptide-TLR-7/8a conjugates that are chem. programmed to self-assemble into nanoparticles of uniform size (∼20 nm) irresp. of the peptide antigen compn. This approach provided precise loading of diverse peptide neoantigens linked to TLR-7/8a (adjuvant) in nanoparticles, which increased uptake by and activation of antigen-presenting cells that promote T-cell immunity. Vaccination of mice with SNP-7/8a using predicted neoantigens (n = 179) from three tumor models induced CD8 T cells against ∼50% of neoantigens with high predicted MHC-I binding affinity and led to enhanced tumor clearance. SNP-7/8a delivering in silico-designed mock neoantigens also induced CD8 T cells in nonhuman primates. Altogether, SNP-7/8a is a generalizable approach for codelivering peptide antigens and adjuvants in nanoparticles for inducing anticancer T-cell immunity.
- 33Shae, D.; Baljon, J. J.; Wehbe, M.; Christov, P. P.; Becker, K. W.; Kumar, A.; Suryadevara, N.; Carson, C. S.; Palmer, C. R.; Knight, F. C.; Joyce, S.; Wilson, J. T. Co-Delivery of Peptide Neoantigens and Stimulator of Interferon Genes Agonists Enhances Response to Cancer Vaccines. ACS Nano 2020, 14, 9904– 9916, DOI: 10.1021/acsnano.0c0276533https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVCmsLjL&md5=e52e9db129f9916ccbb51b96ead541fcCo-delivery of Peptide Neoantigens and Stimulator of Interferon Genes Agonists Enhances Response to Cancer VaccinesShae, Daniel; Baljon, Jessalyn J.; Wehbe, Mohamed; Christov, Plamen P.; Becker, Kyle W.; Kumar, Amrendra; Suryadevara, Naveenchandra; Carson, Carcia S.; Palmer, Christian R.; Knight, Frances C.; Joyce, Sebastian; Wilson, John T.ACS Nano (2020), 14 (8), 9904-9916CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Cancer vaccines targeting patient-specific neoantigens have emerged as a promising strategy for improving responses to immune checkpoint blockade. However, neoantigenic peptides are poorly immunogenic and inept at stimulating CD8+ T cell responses, motivating a need for new vaccine technologies that enhance their immunogenicity. The stimulator of interferon genes (STING) pathway is an endogenous mechanism by which the innate immune system generates an immunol. context for priming and mobilizing neoantigen-specific T cells. Owing to this crit. role in tumor immune surveillance, a synthetic cancer nanovaccine platform (nanoSTING-vax) was developed that mimics immunogenic cancer cells in its capacity to efficiently promote co-delivery of peptide antigens and the STING agonist, cGAMP. The co-loading of cGAMP and peptides into pH-responsive, endosomolytic polymersomes promoted the coordinated delivery of both cGAMP and peptide antigens to the cytosol, thereby eliciting inflammatory cytokine prodn., co-stimulatory marker expression, and antigen cross-presentation. Consequently, nanoSTING-vax significantly enhanced CD8+ T cell responses to a range of peptide antigens. Therapeutic immunization with nanoSTING-vax, in combination with immune checkpoint blockade, inhibited tumor growth in multiple murine tumor models, even leading to complete tumor rejection and generation of durable antitumor immune memory. Collectively, this work establishes nanoSTING-vax as a versatile platform for enhancing immune responses to neoantigen-targeted cancer vaccines.
- 34Tornesello, A. L.; Tagliamonte, M.; Tornesello, M. L.; Buonaguro, F. M.; Buonaguro, L. Nanoparticles to Improve the Efficacy of Peptide-Based Cancer Vaccines. Cancers (Basel) 2020, 12, 1049, DOI: 10.3390/cancers12041049There is no corresponding record for this reference.
- 35Viswanath, D. I.; Liu, H. C.; Huston, D. P.; Chua, C. Y. X.; Grattoni, A. Emerging Biomaterial-Based Strategies for Personalized Therapeutic in Situ Cancer Vaccines. Biomaterials 2022, 280, 121297, DOI: 10.1016/j.biomaterials.2021.12129735https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXislWkurfI&md5=6c16a058af7c31ba986ce1a85033ee82Emerging biomaterial-based strategies for personalized therapeutic in situ cancer vaccinesViswanath, Dixita Ishani; Liu, Hsuan-Chen; Huston, David P.; Chua, Corrine Ying Xuan; Grattoni, AlessandroBiomaterials (2022), 280 (), 121297CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)A review. Landmark successes in oncoimmunol. have led to development of therapeutics boosting the host immune system to eradicate local and distant tumors with impactful tumor redn. in a subset of patients. However, current immunotherapy modalities often demonstrate limited success when involving immunol. cold tumors and solid tumors. Here, we describe the role of various biomaterials to formulate cancer vaccines as a form of cancer immunotherapy, seeking to utilize the host immune system to activate and expand tumor-specific T cells. Biomaterial-based cancer vaccines enhance the cancer-immunity cycle by harnessing cellular recruitment and activation against tumor-specific antigens. In this review, we discuss biomaterial-based vaccine strategies to induce lymphocytic responses necessary to mediate anti-tumor immunity. We focus on strategies that selectively attract dendritic cells via immunostimulatory gradients, activate them against presented tumor-specific antigens, and induce effective cross-presentation to T cells in secondary lymphoid organs, thereby generating immunity. We posit that personalized cancer vaccines are promising targets to generate long-term systemic immunity against patient- and tumor-specific antigens to ensure long-term cancer remission.
- 36Wu, S.; Xia, Y.; Hu, Y.; Ma, G. Bio-Mimic Particles for the Enhanced Vaccinations: Lessons Learnt from the Natural Traits and Pathogenic Invasion. Adv. Drug Deliv Rev. 2021, 176, 113871, DOI: 10.1016/j.addr.2021.11387136https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhs1KgsL7O&md5=106fa4408f1ebb0c26ae41f4a4c8df36Bio-mimic particles for the enhanced vaccinations: Lessons learnt from the natural traits and pathogenic invasionWu, Sihua; Xia, Yufei; Hu, Yuning; Ma, GuanghuiAdvanced Drug Delivery Reviews (2021), 176 (), 113871CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)A review. In the combat against pathogens, the immune systems were evolved with the immune recognitions against the various danger signals, which responded vigorously upon the pathogen invasions and elicited potent antibodies or T cell engagement against the re-infections. Envisage with the prevailing pandemics and increasing demands for cancer vaccines, bio-mimic particles were developed to imitate the natural traits of the pathogens, which conferred the optimal strategies to stimulate the immune engagement and let to the increased vaccine efficacy. Here, the recent development in bio-mimic particles, as well as the natural cues from the pathogens were discussed. As such, the designing principles that adapted from the physiochem. properties of the pathogens were unfolded as the surface characteristics (hydrophobic, nano-pattern, antigen display, charge), properties (size, shape, softness) and the delivered components (peptide, protein, nuclear acids, toll-like receptor (TLR) agonist, antibody). Addnl., the strategies for the efficient delivery, regarding the biodistribution, internalization and presentation of the antigens were also illustrated. Through reviewing the state-of-art in biomimetic particles, the lesson learnt from the natural traits and pathogenic invasion may shed light on the rational design for the enhanced vaccinations.
- 37Rosenthal, J. A.; Chen, L.; Baker, J. L.; Putnam, D.; DeLisa, M. P. Pathogen-Like Particles: Biomimetic Vaccine Carriers Engineered at the Nanoscale. Curr. Opin Biotechnol 2014, 28, 51– 58, DOI: 10.1016/j.copbio.2013.11.00537https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtF2ktLfK&md5=e2b314a29050e3a7f370dcf0137aa250Pathogen-like particles: biomimetic vaccine carriers engineered at the nanoscaleRosenthal, Joseph A.; Chen, Linxiao; Baker, Jenny L.; Putnam, David; DeLisa, Matthew P.Current Opinion in Biotechnology (2014), 28 (), 51-58CODEN: CUOBE3; ISSN:0958-1669. (Elsevier B.V.)Vaccine adjuvants are an essential component of vaccine design, helping to generate immunity to pathogen antigens in the absence of infection. Recent advances in nanoscale engineering have created a new class of particulate bionanotechnol. that uses biomimicry to better integrate adjuvant and antigen. These pathogen-like particles, or PLPs, can come from a variety of sources, ranging from fully synthetic platforms to biol. derived, self-assembling systems. By employing molecularly engineered targeting and stimulation of key immune cells, recent studies utilizing PLPs as vaccine delivery platforms have shown great promise against high-impact, unsolved vaccine targets ranging from bacterial and viral pathogens to cancer and addiction.
- 38Gutjahr, A.; Papagno, L.; Nicoli, F.; Kanuma, T.; Kuse, N.; Cabral-Piccin, M. P.; Rochereau, N.; Gostick, E.; Lioux, T.; Perouzel, E. The STING Ligand cGamp Potentiates the Efficacy of Vaccine-Induced CD8+ T Cells. JCI Insight 2019, 4 (7), e125107, DOI: 10.1172/jci.insight.125107There is no corresponding record for this reference.
- 39He, Y.; Hong, C.; Fletcher, S. J.; Berger, A. G.; Sun, X.; Yang, M.; Huang, S.; Belcher, A. M.; Irvine, D. J.; Li, J.; Hammond, P. T. Peptide-Based Cancer Vaccine Delivery Via the STINGΔTM-cGamp Complex. Adv. Healthc Mater. 2022, 11, e2200905 DOI: 10.1002/adhm.20220090539https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsFGqt7jK&md5=48caf8a595d73d5693dcfc610067baf7Peptide-Based Cancer Vaccine Delivery via the STINGΔTM-cGAMP ComplexHe, Yanpu; Hong, Celestine; Fletcher, Samantha J.; Berger, Adam G.; Sun, Xin; Yang, Mengdi; Huang, Shengnan; Belcher, Angela M.; Irvine, Darrell J.; Li, Jiahe; Hammond, Paula T.Advanced Healthcare Materials (2022), 11 (15), 2200905CODEN: AHMDBJ; ISSN:2192-2640. (Wiley-VCH Verlag GmbH & Co. KGaA)With the advent of bioinformatic tools in efficiently predicting neo-antigens, peptide vaccines have gained tremendous attention in cancer immunotherapy. However, the delivery of peptide vaccines remains a major challenge, primarily due to ineffective transport to lymph nodes and low immunogenicity. Here, a strategy for peptide vaccine delivery is reported by first fusing the peptide to the cytosolic domain of the stimulator of interferon genes protein (STINGΔTM), then complexing the peptide-STINGΔTM protein with STING agonist 2'3' cyclic guanosine monophosphate-adenosine monophosphate (cGAMP). The process results in the formation of self-assembled cGAMP-peptide-STINGΔTM tetramers, which enables efficient lymphatic trafficking of the peptide. Moreover, the cGAMP-STINGΔTM complex acts not only as a protein carrier for the peptide, but also as a potent adjuvant capable of triggering STING signaling independent of endogenous STING protein-an esp. important attribute considering that certain cancer cells epigenetically silence their endogenous STING expression. With model antigen SIINFEKL, it is demonstrated that the platform elicits effective STING signaling in vitro, draining lymph node targeting in vivo, effective T cell priming in vivo as well as antitumoral immune response in a mouse colon carcinoma model, providing a versatile soln. to the challenges faced in peptide vaccine delivery.
- 40Li, X. D.; Wu, J.; Gao, D.; Wang, H.; Sun, L.; Chen, Z. J. Pivotal Roles of cGAS-cGAMP Signaling in Antiviral Defense and Immune Adjuvant Effects. Science 2013, 341, 1390– 1394, DOI: 10.1126/science.124404040https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVCmu7vK&md5=46c48534439d7b6442911d63f7c3fa51Pivotal Roles of cGAS-cGAMP Signaling in Antiviral Defense and Immune Adjuvant EffectsLi, Xiao-Dong; Wu, Jiaxi; Gao, Daxing; Wang, Hua; Sun, Lijun; Chen, Zhijian J.Science (Washington, DC, United States) (2013), 341 (6152), 1390-1394CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Invasion of microbial DNA into the cytoplasm of animal cells triggers a cascade of host immune reactions that help clear the infection; however, self DNA in the cytoplasm can cause autoimmune diseases. Biochem. approaches led to the identification of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) as a cytosolic DNA sensor that triggers innate immune responses. Here, we show that cells from cGAS-deficient (cGas-/-) mice, including fibroblasts, macrophages, and dendritic cells, failed to produce type I interferons and other cytokines in response to DNA transfection or DNA virus infection. cGas-/- mice were more susceptible to lethal infection with herpes simplex virus 1 (HSV1) than wild-type mice. We also show that cGAMP is an adjuvant that boosts antigen-specific T cell activation and antibody prodn. in mice.
- 41Van Herck, S.; Feng, B.; Tang, L. Delivery of STING Agonists for Adjuvanting Subunit Vaccines. Adv. Drug Deliv Rev. 2021, 179, 114020, DOI: 10.1016/j.addr.2021.11402041https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVyrsrnM&md5=991bac587ff443dcb8ebbbdef18d31aeDelivery of STING agonists for adjuvanting subunit vaccinesVan Herck, Simon; Feng, Bing; Tang, LiAdvanced Drug Delivery Reviews (2021), 179 (), 114020CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)A review. Adjuvant is an essential component in subunit vaccines. Many agonists of pathogen recognition receptors have been developed as potent adjuvants to optimize the immunogenicity and efficacy of vaccines. Recently discovered cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway has attracted much attention as it is a key mediator for modulating immune responses. Vaccines adjuvanted with STING agonists are found to mediate a robust immune defense against infections and cancer. In this review, we first discuss the mechanisms of STING agonists in the context of vaccination. Next, we present recent progress in novel STING agonist discovery and the delivery strategies. We next highlight recent work in optimizing the efficacy while minimizing toxicity of STING agonist-assisted subunit vaccines for protection against infectious diseases or treatment of cancer. Finally, we share our perspectives of current issues and future directions in further developing STING agonists for adjuvanting subunit vaccines.
- 42Embgenbroich, M.; Burgdorf, S. Current Concepts of Antigen Cross-Presentation. Front Immunol 2018, 9, 1643, DOI: 10.3389/fimmu.2018.0164342https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlSisLvP&md5=c6d5b139383717e9d5ed31dafc8a55d3Current concepts of antigen cross-presentationEmbgenbroich, Maria; Burgdorf, SvenFrontiers in Immunology (2018), 9 (), 1643/1-1643/10CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)Dendritic cells have the ability to efficiently present internalized antigens on major histocompatibility complex (MHC) I mols. This process is termed cross-presentation and is important role in the generation of an immune response against viruses and tumors, after vaccinations or in the induction of immune tolerance. The mol. mechanisms enabling cross-presentation have been topic of intense debate since many years. However, a clear view on these mechanisms remains difficult, partially due to important remaining questions, controversial results and discussions. Here, we give an overview of the current concepts of antigen cross-presentation and focus on a description of the major cross-presentation pathways, the role of retarded antigen degrdn. for efficient cross-presentation, the dislocation of antigens from endosomal compartment into the cytosol, the reverse transport of proteasome-derived peptides for loading on MHC I and the translocation of the cross-presentation machinery from the ER to endosomes. We try to highlight recent advances, discuss some of the controversial data and point out some of the major open questions in the field.
- 43Tom, J. K.; Albin, T. J.; Manna, S.; Moser, B. A.; Steinhardt, R. C.; Esser-Kahn, A. P. Applications of Immunomodulatory Immune Synergies to Adjuvant Discovery and Vaccine Development. Trends Biotechnol 2019, 37, 373– 388, DOI: 10.1016/j.tibtech.2018.10.00443https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitV2itbrN&md5=c5510e87652758ab52910904679d93f2Applications of Immunomodulatory Immune Synergies to Adjuvant Discovery and Vaccine DevelopmentTom, Janine K.; Albin, Tyler J.; Manna, Saikat; Moser, Brittany A.; Steinhardt, Rachel C.; Esser-Kahn, Aaron P.Trends in Biotechnology (2019), 37 (4), 373-388CODEN: TRBIDM; ISSN:0167-7799. (Elsevier Ltd.)A review. Pathogens comprise a diverse set of immunostimulatory mols. that activate the innate immune system during infection. The immune system recognizes distinct combinations of pathogenic mols. leading to multiple immune activation events that cooperate to produce enhanced immune responses, known as 'immune synergies'. Effective immune synergies are essential for the clearance of pathogens, thus inspiring novel adjuvant design to improve vaccines. We highlight current vaccine adjuvants and the importance of immune synergies to adjuvant and vaccine design. The focus is on new technologies used to study and apply immune synergies to adjuvant and vaccine development. Finally, we discuss how recent findings can be applied to the future design and characterization of synergistic adjuvants and vaccines.
- 44Collier, M. A.; Junkins, R. D.; Gallovic, M. D.; Johnson, B. M.; Johnson, M. M.; Macintyre, A. N.; Sempowski, G. D.; Bachelder, E. M.; Ting, J. P.; Ainslie, K. M. Acetalated Dextran Microparticles for Codelivery of STING and TLR7/8 Agonists. Mol. Pharmaceutics 2018, 15, 4933– 4946, DOI: 10.1021/acs.molpharmaceut.8b0057944https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvVertLvM&md5=72cfae51f29f59ae7c4a6b11a36d7acbAcetalated Dextran Microparticles for Codelivery of STING and TLR7/8 AgonistsCollier, Michael A.; Junkins, Robert D.; Gallovic, Matthew D.; Johnson, Brandon M.; Johnson, Monica M.; Macintyre, Andrew N.; Sempowski, Gregory D.; Bachelder, Eric M.; Ting, Jenny P.-Y.; Ainslie, Kristy M.Molecular Pharmaceutics (2018), 15 (11), 4933-4946CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Vaccines are the most effective tool for preventing infectious diseases; however, subunit vaccines, considered the safest type, suffer from poor immunogenicity and require adjuvants to create a strong and sustained immune response. As adjuvants, pathogen-assocd. mol. patterns (PAMPs) offer potent immunostimulatory properties and defined mechanisms of action through their cognate pattern recognition receptors (PRRs). Their activity can be further enhanced through combining two or more PAMPs, particularly those that activate multiple immune signaling pathways. However, the cytosolic localization of many PRRs requires intracellular delivery of PAMPs for optimal biol. activity, which is particularly true of the stimulator of interferon genes (STING) PRR. Using acetalated dextran (Ace-DEX) microparticles (MPs) encapsulating STING agonist 3'3'-cyclic GMP-AMP (cGAMP) combined with sol. PAMPS, we screened the effect of codelivery of adjuvants using primary mouse bone marrow derived dendritic cells (BMDCs). We identified that codelivery of cGAMP MPs and sol. Toll-like receptor 7/8 (TLR7/8) agonist resiquimod (R848) elicited the broadest cytokine response. CGAMP and R848 were then coencapsulated within Ace-DEX MPs via electrospray. Using the model antigen ovalbumin, we obsd. that Ace-DEX MPs coencapsulating cGAMP and R848 (cGAMP/R848 Ace-DEX MPs) induced antigen-specific cellular immunity, and a balanced Th1/Th2 humoral response that was greater than cGAMP Ace-DEX MPs alone and PAMPs delivered in sep. MPs. These data indicate that polymeric Ace-DEX MPs loaded with STING and TLR7/8 agonists represent a potent cellular and humoral vaccine adjuvant.
- 45Barman, S.; Borriello, F.; Brook, B.; Pietrasanta, C.; De Leon, M.; Sweitzer, C.; Menon, M.; van Haren, S. D.; Soni, D.; Saito, Y.; Nanishi, E.; Yi, S.; Bobbala, S.; Levy, O.; Scott, E. A.; Dowling, D. J. Shaping Neonatal Immunization by Tuning the Delivery of Synergistic Adjuvants Via Nanocarriers. ACS Chem. Biol. 2022, 17, 2559– 2571, DOI: 10.1021/acschembio.2c0049745https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xit1alsb3K&md5=779648f1160a984f3c8b7b98026bf77cShaping Neonatal Immunization by Tuning the Delivery of Synergistic Adjuvants via NanocarriersBarman, Soumik; Borriello, Francesco; Brook, Byron; Pietrasanta, Carlo; De Leon, Maria; Sweitzer, Cali; Menon, Manisha; van Haren, Simon D.; Soni, Dheeraj; Saito, Yoshine; Nanishi, Etsuro; Yi, Sijia; Bobbala, Sharan; Levy, Ofer; Scott, Evan A.; Dowling, David J.ACS Chemical Biology (2022), 17 (9), 2559-2571CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Adjuvanted nanocarrier-based vaccines hold substantial potential for applications in novel early-life immunization strategies. Here, via mouse and human age-specific in vitro modeling, we identified the combination of a small-mol. STING agonist (2'3'-cyclic GMP-AMP, cGAMP) and a TLR7/8 agonist (CL075) to drive the synergistic activation of neonatal dendritic cells and precision CD4 T-helper (Th) cell expansion via the IL-12/IFNγ axis. We further demonstrate that the vaccination of neonatal mice with quadrivalent influenza recombinant hemagglutinin (rHA) and an admixt. of two polymersome (PS) nanocarriers sep. encapsulating cGAMP (cGAMP-PS) and CL075 (CL075-PS) drove robust Th1 bias, high frequency of T follicular helper (TFH) cells, and germinal center (GC) B cells along with the IgG2c-skewed humoral response in vivo. Dual-loaded cGAMP/CL075-PSs did not outperform admixed cGAMP-PS and CL075-PS in vivo. These data validate an optimally designed adjuvantation system via age-selected small-mol. synergy and a multicomponent nanocarrier formulation as an effective approach to induce type 1 immune responses in early life.
- 46Kuai, R.; Sun, X.; Yuan, W.; Ochyl, L. J.; Xu, Y.; Hassani Najafabadi, A.; Scheetz, L.; Yu, M. Z.; Balwani, I.; Schwendeman, A.; Moon, J. J. Dual TLR Agonist Nanodiscs as a Strong Adjuvant System for Vaccines and Immunotherapy. J. Controlled Release 2018, 282, 131– 139, DOI: 10.1016/j.jconrel.2018.04.04146https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXosFWrtr0%253D&md5=00b84399e940517a6e19c016bd9fc318Dual TLR agonist nanodiscs as a strong adjuvant system for vaccines and immunotherapyKuai, Rui; Sun, Xiaoqi; Yuan, Wenmin; Ochyl, Lukasz J.; Xu, Yao; Hassani Najafabadi, Alireza; Scheetz, Lindsay; Yu, Min-Zhi; Balwani, Ishina; Schwendeman, Anna; Moon, James J.Journal of Controlled Release (2018), 282 (), 131-139CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Recent studies have shown that certain combinations of Toll-like receptor (TLR) agonists can induce synergistic immune activation. However, it remains challenging to achieve such robust responses in vivo in a manner that is effective, facile, and amenable for clin. translation. Here, we show that MPLA, a TLR4 agonist, and CpG, a TLR9 agonist, can be efficiently co-loaded into synthetic high-d. lipoprotein nanodiscs, forming a potent adjuvant system (ND-MPLA/CpG) that can be readily combined with a variety of subunit antigens, including proteins and peptides. ND-MPLA/CpG significantly enhanced activation of dendritic cells, compared with free dual adjuvants or nanodiscs delivering a single TLR agonist. Importantly, mice immunized with phys. mixts. of protein antigens ND-MPLA/CpG generated strong humoral responses, including induction of IgG responses against protein convertase subtilisin/kexin 9 (PCSK9), leading to 17-30% redn. of the total plasma cholesterol levels. Moreover, ND-MPLA/CpG exerted strong anti-tumor efficacy in multiple murine tumor models. Compared with free adjuvants, ND-MPLA/CpG admixed with ovalbumin markedly improved antigen-specific CD8+ T cell responses by 8-fold and promoted regression of B16F10-OVA melanoma (P<0.0001). Furthermore, ND-MPLA/CpG admixed with E7 peptide antigen elicited ∼20% E7-specific CD8+ T cell responses and achieved complete regression of established TC-1 tumors in all treated animals. Taken together, our work highlights the simplicity, versatility, and potency of dual TLR agonist nanodiscs for applications in vaccines and cancer immunotherapy.
- 47Zhang, B. D.; Wu, J. J.; Li, W. H.; Hu, H. G.; Zhao, L.; He, P. Y.; Zhao, Y. F.; Li, Y. M. STING and TLR7/8 Agonists-Based Nanovaccines for Synergistic Antitumor Immune Activation. Nano Res. 2022, 15, 6328– 6339, DOI: 10.1007/s12274-022-4282-x47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtVeitLbO&md5=0103b8d2efebe7669675a9d6ee1e51b5STING and TLR7/8 agonists-based nanovaccines for synergistic antitumor immune activationZhang, Bo-Dou; Wu, Jun-Jun; Li, Wen-Hao; Hu, Hong-Guo; Zhao, Lang; He, Pei-Yang; Zhao, Yu-Fen; Li, Yan-MeiNano Research (2022), 15 (7), 6328-6339CODEN: NRAEB5; ISSN:1998-0000. (Springer GmbH)Immunostimulatory therapies based on pattern recognition receptors (PRRs) have emerged as an effective approach in the fight against cancer, with the ability to recruit tumor-specific lymphocytes in a low-immunogenicity tumor environment. The agonist cyclic dinucleotides (CDNs) of the stimulator of interferon gene (STING) are a group of very promising anticancer mols. that increase tumor immunogenicity by activating innate immunity. However, the tumor immune efficacy of CDNs is limited by several factors, including relatively narrow cytokine prodn., inefficient delivery to STING, and rapid clearance. In addn., a single adjuvant mol. is unable to elicit a broad cytokine response and thus cannot further amplify the anticancer effect. To address this problem, two or more agonist mols. are often used together to synergistically enhance immune efficacy. In this work, we found that a combination of the STING agonist CDGSF and the Toll-like receptor 7/8 (TLR7/8) agonist 522 produced a broader cytokine response. Subsequently, we developed multicomponent nanovaccines (MCNVs) consisting of a PC7A polymer as a nanocarrier encapsulating the antigen OVA and adjuvant mols. These MCNVs activate bone marrow-derived dendritic cells (BMDCs) to produce multiple proinflammatory factors that promote antigen cross-presentation to stimulate specific antitumor T-cell responses. In in vivo expts., we obsd. that MCNVs triggered a strong T-cell response in tumor-infiltrating lymphocytes, resulting in significant tumor regression and, notably, a 100% survival rate in mice through 25 days without other partnering therapies. These data suggest that our nanovaccines have great potential to advance cancer immunotherapy with increased durability and potency.
- 48Pradhan, P.; Toy, R.; Jhita, N.; Atalis, A.; Pandey, B.; Beach, A.; Blanchard, E. L.; Moore, S. G.; Gaul, D. A.; Santangelo, P. J. TRAF6-IRF5 Kinetics, TRIF, and Biophysical Factors Drive Synergistic Innate Responses to Particle-Mediated MPLA-CpG Co-Presentation. Sci. Adv. 2021, 7, abd4235, DOI: 10.1126/sciadv.abd4235There is no corresponding record for this reference.
- 49Temizoz, B.; Kuroda, E.; Ohata, K.; Jounai, N.; Ozasa, K.; Kobiyama, K.; Aoshi, T.; Ishii, K. J. TLR9 and STING Agonists Synergistically Induce Innate and Adaptive Type-II IFN. Eur. J. Immunol. 2015, 45, 1159– 1169, DOI: 10.1002/eji.20144513249https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXks12gsr4%253D&md5=01e7129b3bc00d09165a64e1fa103859TLR9 and STING agonists synergistically induce innate and adaptive type-II IFNTemizoz, Burcu; Kuroda, Etsushi; Ohata, Keiichi; Jounai, Nao; Ozasa, Koji; Kobiyama, Kouji; Aoshi, Taiki; Ishii, Ken J.European Journal of Immunology (2015), 45 (4), 1159-1169CODEN: EJIMAF; ISSN:0014-2980. (Wiley-VCH Verlag GmbH & Co. KGaA)Agonists for TLR9 and Stimulator of IFN Gene (STING) act as vaccine adjuvants that induce type-1 immune responses. However, currently available CpG oligodeoxynucleotide (ODN) (K-type) induces IFNs only weakly and STING ligands rather induce type-2 immune responses, limiting their potential therapeutic applications. Here, we show a potent synergism between TLR9 and STING agonists. Together, they make an effective type-1 adjuvant and an anticancer agent. The synergistic effect between CpG ODN (K3) and STING-ligand cyclic GMP-AMP (cGAMP), culminating in NK cell IFN-γ (type-II IFN) prodn., is due to the concurrent effects of IL-12 and type-I IFNs, which are differentially regulated by IRF3/7, STING, and MyD88. The combination of CpG ODN with cGAMP is a potent type-1 adjuvant, capable of inducing strong Th1-type responses, as demonstrated by enhanced antigen-specific IgG2c and IFN-γ prodn., as well as cytotoxic CD8+ T-cell responses. In our murine tumor models, intratumoral injection of CpG ODN and cGAMP together reduced tumor size significantly compared with the singular treatments, acting as an antigen-free anticancer agent. Thus, the combination of CpG ODN and a STING ligand may offer therapeutic application as a potent type-II IFN inducer.
- 50Kim, J. Y.; Rosenberger, M. G.; Chen, S.; Ip, C. K.; Bahmani, A.; Chen, Q.; Shen, J.; Tang, Y.; Wang, A.; Kenna, E.; Son, M.; Tay, S.; Ferguson, A. L.; Esser-Kahn, A. P. Discovery of New States of Immunomodulation for Vaccine Adjuvants Via High Throughput Screening: Expanding Innate Responses to PRRs. ACS Cent Sci. 2023, 9, 427– 439, DOI: 10.1021/acscentsci.2c0135150https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXjs1ygtb4%253D&md5=b2ef1701e544feba43084f5fc8b5d3adDiscovery of New States of Immunomodulation for Vaccine Adjuvants via High Throughput Screening: Expanding Innate Responses to PRRsKim, Jeremiah Y.; Rosenberger, Matthew G.; Chen, Siquan; IP, Carman KM; Bahmani, Azadeh; Chen, Qing; Shen, Jinjing; Tang, Yifeng; Wang, Andrew; Kenna, Emma; Son, Minjun; Tay, Savas; Ferguson, Andrew L.; Esser-Kahn, Aaron P.ACS Central Science (2023), 9 (3), 427-439CODEN: ACSCII; ISSN:2374-7951. (American Chemical Society)Stimulation of the innate immune system is crucial in both effective vaccinations and immunotherapies. This is often achieved through adjuvants, mols. that usually activate pattern recognition receptors (PRRs) and stimulate two innate immune signaling pathways: the nuclear factor kappa-light-chain-enhancer of activated B-cells pathway (NF-κB) and the interferon regulatory factors pathway (IRF). Here, we demonstrate the ability to alter and improve adjuvant activity via the addn. of small mol. "immunomodulators". By modulating signaling activity instead of receptor binding, these mols. allow the customization of select innate responses. We demonstrate both inhibition and enhancement of the products of the NF-κB and IRF pathways by several orders of magnitude. Some modulators apply generally across many receptors, while others focus specifically on individual receptors. Modulators boost correlates of a protective immune responses in a com. flu vaccine model and reduced correlates of reactogenicity in a typhoid vaccine model. These modulators have a range of applications: from adjuvanticity in prophylactics to enhancement of immunotherapy.
- 51Nihesh, N.; Manna, S.; Studnitzer, B.; Shen, J.; Esser-Kahn, A. P. A Synthetic Pathogen Mimetic Molecule Induces a Highly Amplified Synergistic Immune Response Via Activation of Multiple Signaling Pathways. Chem. Sci. 2021, 12, 6646– 6651, DOI: 10.1039/D1SC00964H51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXnvVGitr8%253D&md5=26cbbad0fab4671e7767affb9acdd81aA synthetic pathogen mimetic molecule induces a highly amplified synergistic immune response via activation of multiple signaling pathwaysNihesh, Naorem; Manna, Saikat; Studnitzer, Bradley; Shen, Jingjing; Esser-Kahn, Aaron P.Chemical Science (2021), 12 (19), 6646-6651CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)The current understanding of how the immune system processes complex information during natural infections is yet to be exploited for the mol. design of potent immune activators. Here, we address this challenge by design of a pathogen-mimetic mol. that simultaneously co-activates cell-surface active, endosomal and cytosolic immune receptors.
- 52Taylor, D.; Meyer, C. T.; Graves, D.; Sen, R.; Fu, J.; Tran, E.; Mirza, B.; Rodriguez, G.; Lang, C.; Feng, H.; Quaranta, V.; Wilson, J. T.; Kim, Y. J.; Korrer, M. J. MuSyC Dosing of Adjuvanted Cancer Vaccines Optimizes Antitumor Responses. Front Immunol 2022, 13, 936129, DOI: 10.3389/fimmu.2022.93612952https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitlChu73M&md5=4f4dfbaf0362bfb299001faff278e0f6MuSyC dosing of adjuvanted cancer vaccines optimizes antitumor responsesTaylor, David; Meyer, Christian T.; Graves, Diana; Sen, Rupashree; Fu, Juan; Tran, Emily; Mirza, Bilal; Rodriguez, Gabriel; Lang, Cara; Feng, Hanwen; Quaranta, Vito; Wilson, John T.; Kim, Young J.; Korrer, Michael J.Frontiers in Immunology (2022), 13 (), 936129CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)With the clin. approval of T-cell-dependent immune checkpoint inhibitors for many cancers, therapeutic cancer vaccines have re-emerged as a promising immunotherapy. Cancer vaccines require the addn. of immunostimulatory adjuvants to increase vaccine immunogenicity, and increasingly multiple adjuvants are used in combination to bolster further and shape cellular immunity to tumor antigens. However, rigorous quantification of adjuvants' synergistic interactions is challenging due to partial redundancy in costimulatory mols. and cytokine prodn., leading to the common assumption that combining both adjuvants at the max. tolerated dose results in optimal efficacy. Herein, we examine this max. dose assumption and find combinations of these doses are suboptimal. Instead, we optimized dendritic cell activation by extending the Multidimensional Synergy of Combinations (MuSyC) framework that measures the synergy of efficacy and potency between two vaccine adjuvants. Initially, we performed a preliminary in vitro screening of clin. translatable adjuvant receptor targets (TLR, STING, NLL, and RIG-I). We detd. that STING agonist (CDN) plus TLR4 agonist (MPL-A) or TLR7/8 agonist (R848) as the best pairwise combinations for dendritic cell activation. In addn., we found that the combination of R848 and CDN is synergistically efficacious and potent in activating both murine and human antigen-presenting cells (APCs) in vitro. These two selected adjuvants were then used to est. a MuSyC-dose optimized for in vivo T-cell priming using ovalbumin-based peptide vaccines. Finally, using B16 melanoma and MOC1 head and neck cancer models, MuSyC-dose-based adjuvating of cancer vaccines improved the antitumor response, increased tumor infiltrating lymphocytes, and induced novel myeloid tumor infiltration changes. Further, the MuSyC-dose-based adjuvants approach did not cause addnl. wt. changes or increased plasma cytokine levels compared to CDN alone. Collectively, our findings offer a proof of principle that our MuSyC-extended approach can be used to optimize cancer vaccine formulations for immunotherapy.
- 53Hanson, M. C.; Crespo, M. P.; Abraham, W.; Moynihan, K. D.; Szeto, G. L.; Chen, S. H.; Melo, M. B.; Mueller, S.; Irvine, D. J. Nanoparticulate STING Agonists Are Potent Lymph Node-Targeted Vaccine Adjuvants. J. Clin Invest 2015, 125, 2532– 2546, DOI: 10.1172/JCI7991553https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2MfgsVChtw%253D%253D&md5=b840cee3cc090db0b723072b6936b05cNanoparticulate STING agonists are potent lymph node-targeted vaccine adjuvantsHanson Melissa C; Crespo Monica P; Abraham Wuhbet; Moynihan Kelly D; Szeto Gregory L; Chen Stephanie H; Melo Mariane B; Mueller Stefanie; Irvine Darrell JThe Journal of clinical investigation (2015), 125 (6), 2532-46 ISSN:.Cyclic dinucleotides (CDNs) are agonists of stimulator of IFN genes (STING) and have potential as vaccine adjuvants. However, cyclic di-GMP (cdGMP) injected s.c. shows minimal uptake into lymphatics/draining lymph nodes (dLNs) and instead is rapidly distributed to the bloodstream, leading to systemic inflammation. Here, we encapsulated cdGMP within PEGylated lipid nanoparticles (NP-cdGMP) to redirect this adjuvant to dLNs. Compared with unformulated CDNs, encapsulation blocked systemic dissemination and markedly enhanced dLN accumulation in murine models. Delivery of NP-cdGMP increased CD8+ T cell responses primed by peptide vaccines and enhanced therapeutic antitumor immunity. A combination of a poorly immunogenic liposomal HIV gp41 peptide antigen and NP-cdGMP robustly induced type I IFN in dLNs, induced a greater expansion of vaccine-specific CD4+ T cells, and greatly increased germinal center B cell differentiation in dLNs compared with a combination of liposomal HIV gp41 and soluble CDN. Further, NP-cdGMP promoted durable antibody titers that were substantially higher than those promoted by the well-studied TLR agonist monophosphoryl lipid A and comparable to a much larger dose of unformulated cdGMP, without the systemic toxicity of the latter. These results demonstrate that nanoparticulate delivery safely targets CDNs to the dLNs and enhances the efficacy of this adjuvant. Moreover, this approach can be broadly applied to other small-molecule immunomodulators of interest for vaccines and immunotherapy.
- 54Atukorale, P. U.; Raghunathan, S. P.; Raguveer, V.; Moon, T. J.; Zheng, C.; Bielecki, P. A.; Wiese, M. L.; Goldberg, A. L.; Covarrubias, G.; Hoimes, C. J.; Karathanasis, E. Nanoparticle Encapsulation of Synergistic Immune Agonists Enables Systemic Codelivery to Tumor Sites and IFNβ-Driven Antitumor Immunity. Cancer Res. 2019, 79, 5394– 5406, DOI: 10.1158/0008-5472.CAN-19-038154https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtFSqtb0%253D&md5=469de56cb58341e82e13aa77c48c6515Nanoparticle encapsulation of synergistic immune agonists enables systemic codelivery to tumor sites and IFNβ-driven antitumor immunityAtukorale, Prabhani U.; Raghunathan, Shruti P.; Raguveer, Vanitha; Moon, Tayior J.; Zheng, Carolyn; Bielecki, Peter A.; Wiese, Michelle L.; Goldberg, Amy L.; Covarrubias, Gil; Hoimes, Christopher J.; Karathanasis, EfstathiosCancer Research (2019), 79 (20), 5394-5406CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Effective cancer immunotherapy depends on the robust activation of tumor-specific antigen-presenting cells (APC). Immune agonists encapsulated within nanoparticles (NP) can be delivered to tumor sites to generate powerful antitumor immune responses with minimal off-target dissemination. Systemic delivery enables widespread access to the microvasculature and draining to the APC-rich perivasculature. We developed an immuno-nanoparticle (immuno-NP) coloaded with cyclic diguanylate monophosphate, an agonist of the stimulator of interferon genes pathway, and monophosphoryl lipid A, and a Toll-like receptor 4 agonist, which synergize to produce high levels of type I ΙΡΝβ. Usinga murine model of metastatic triple-neg. breast cancer. systemic delivery of these immuno-NPs resulted in significant therapeutic outcomes due to extensive upregulation of APCs and natural killer cells in the blood and tumor compared with control treatments. These results indicate that NPs can facilitate systemic delivery of multiple immune potentiating cargoes for effective APC-driven local and systemic antitumor immunity.
- 55Pandey, S.; Gruenbaum, A.; Kanashova, T.; Mertins, P.; Cluzel, P.; Chevrier, N. Pairwise Stimulations of Pathogen-Sensing Pathways Predict Immune Responses to Multi-Adjuvant Combinations. Cell Syst 2020, 11, 495– 508, DOI: 10.1016/j.cels.2020.10.00155https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisVaqu7jK&md5=04738bf46febbbc04848669c0147be24Pairwise Stimulations of Pathogen-Sensing Pathways Predict Immune Responses to Multi-adjuvant CombinationsPandey, Surya; Gruenbaum, Adam; Kanashova, Tamara; Mertins, Philipp; Cluzel, Philippe; Chevrier, NicolasCell Systems (2020), 11 (5), 495-508.e10CODEN: CSEYA4; ISSN:2405-4712. (Cell Press)The immune system makes decisions in response to combinations of multiple microbial inputs. We do not understand the combinatorial logic governing how higher-order combinations of microbial signals shape immune responses. Here, using coculture expts. and statistical analyses, we discover a general property for the combinatorial sensing of microbial signals, whereby the effects of triplet combinations of microbial signals on immune responses can be predicted by combining the effects of single and pairs. Mechanistically, we find that singles and pairs dictate the information signaled by triplets in mouse and human DCs at the levels of transcription, chromatin, and protein secretion. We exploit this simplifying property to develop cell-based immunotherapies prepd. with adjuvant combinations that trigger protective responses in mouse models of cancer. We conclude that the processing of multiple input signals by innate immune cells is governed by pairwise effects, which will inform the rationale combination of adjuvants to manipulate immunity.
- 56Andrade, W. A.; Agarwal, S.; Mo, S.; Shaffer, S. A.; Dillard, J. P.; Schmidt, T.; Hornung, V.; Fitzgerald, K. A.; Kurt-Jones, E. A.; Golenbock, D. T. Type I Interferon Induction by Neisseria Gonorrhoeae: Dual Requirement of Cyclic GMP-AMP Synthase and Toll-Like Receptor 4. Cell Rep 2016, 15, 2438– 2448, DOI: 10.1016/j.celrep.2016.05.03056https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xpt1entbg%253D&md5=55421b21e0d3fce79cfbfe853151908dType I Interferon Induction by Neisseriagonorrhoeae: Dual Requirement of Cyclic GMP-AMP Synthase and Toll-like Receptor 4Andrade, Warrison A.; Agarwal, Sarika; Mo, Shunyan; Shaffer, Scott A.; Dillard, Joseph P.; Schmidt, Tobias; Hornung, Veit; Fitzgerald, Katherine A.; Kurt-Jones, Evelyn A.; Golenbock, Douglas T.Cell Reports (2016), 15 (11), 2438-2448CODEN: CREED8; ISSN:2211-1247. (Cell Press)The innate immune system is the first line of defense against Neisseria gonorrhoeae (GC). Exposure of cells to GC lipooligosaccharides induces a strong immune response, leading to type I interferon (IFN) prodn. via TLR4/MD-2. In addn. to living freely in the extracellular space, GC can invade the cytoplasm to evade detection and elimination. Double-stranded DNA introduced into the cytosol binds and activates the enzyme cyclic-GMP-AMP synthase (cGAS), which produces 2'3'-cGAMP and triggers STING/TBK-1/IRF3 activation, resulting in type I IFN expression. Here, we reveal a cytosolic response to GC DNA that also contributes to type I IFN induction. We demonstrate that complete IFN-β induction by live GC depends on both cGAS and TLR4. Type I IFN is detrimental to the host, and dysregulation of iron homeostasis genes may explain lower bacteria survival in cGAS-/- and TLR4-/- cells. Collectively, these observations reveal cooperation between TLRs and cGAS in immunity to GC infection.
- 57Kocabas, B. B.; Almacioglu, K.; Bulut, E. A.; Gucluler, G.; Tincer, G.; Bayik, D.; Gursel, M.; Gursel, I. Dual-Adjuvant Effect of pH-Sensitive Liposomes Loaded with STING and TLR9 Agonists Regress Tumor Development by Enhancing Th1 Immune Response. J. Controlled Release 2020, 328, 587– 595, DOI: 10.1016/j.jconrel.2020.09.04057https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitVCks7bE&md5=2eb22cda1e9a367c6e5e6757380a13efDual-adjuvant effect of pH-sensitive liposomes loaded with STING and TLR9 agonists regress tumor development by enhancing Th1 immune responseKocabas, Banu Bayyurt; Almacioglu, Kubra; Bulut, Esin Alpdundar; Gucluler, Gozde; Tincer, Gizem; Bayik, Defne; Gursel, Mayda; Gursel, IhsanJournal of Controlled Release (2020), 328 (), 587-595CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Nucleic acid-based pattern recognition receptor agonists are effective adjuvants and immunotherapeutic agents. Rather than single applications, ligand combinations could synergistically potentiate immune responses by elevating cytokine and chemokine prodn. via triggering multiple signaling pathways. However, short half-lives of such labile ligands due to nuclease attack and limited cellular uptake due to their structure significantly hamper their in vivo performances. More importantly, simultaneous delivery and activity presentation of protein antigen and nucleic acid ligands critically limit the clin. development of these constructs. In this work, we approached this problem by co-encapsulating a model antigen ovalbumin along with TLR9 and STING ligands within liposomes, a well-established drug delivery system that enables payload stability and enhanced cellular activity upon internalization. Moreover, by loading dual ligands we postulated to achieve heightened Th-1 immune response that would yield pronounced protective vaccine efficacy. We show that, pH-sensitive liposomes co-encapsulating CpG ODN and cGAMP induced synergistic innate immune response by elevating type I and type II interferon levels. Most importantly, this vaccine formulation led to ∼70% regression of established melanoma tumor. pH-sensitive liposomal vaccine administration elevated IgG2c/IgG1 antibody ratio, indicative of augmented OVA-specific Th1-biased immunity. Importantly, while the frequency of tumor-specific IFN-γ producing CD8+ T-cells was significantly increased, the M2-type anti-inflammatory macrophage levels were decreased in the tumor bed. In conclusion, our strategy induces reversal of immunosuppressive tumor microenvironment, while enhancing effective anti-tumor immune-response. We propose that this could be coupled with std. therapies during combating tumor eradication.
- 58Toy, R.; Keenum, M. C.; Pradhan, P.; Phang, K.; Chen, P.; Chukwu, C.; Nguyen, L. A. H.; Liu, J.; Jain, S.; Kozlowski, G.; Hosten, J.; Suthar, M. S.; Roy, K. TLR7 and RIG-I Dual-Adjuvant Loaded Nanoparticles Drive Broadened and Synergistic Responses in Dendritic Cells in Vitro and Generate Unique Cellular Immune Responses in Influenza Vaccination. J. Controlled Release 2021, 330, 866– 877, DOI: 10.1016/j.jconrel.2020.10.06058https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitlCku7rE&md5=4985b3db7e0bf52f50272a1d4e1238dcTLR7 and RIG-I dual-adjuvant loaded nanoparticles drive broadened and synergistic responses in dendritic cells in vitro and generate unique cellular immune responses in influenza vaccinationToy, Randall; Keenum, M. Cole; Pradhan, Pallab; Phang, Katelynn; Chen, Patrick; Chukwu, Chinwendu; Nguyen, Lily Anh H.; Liu, Jiaying; Jain, Sambhav; Kozlowski, Gabrielle; Hosten, Justin; Suthar, Mehul S.; Roy, KrishnenduJournal of Controlled Release (2021), 330 (), 866-877CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Although the existing flu vaccines elicit strong antigen-specific antibody responses, they fail to provide effective, long term protection - partly due to the absence of robust cellular memory immunity. We hypothesized that co-administration of combination adjuvants, mirroring the flu-virus related innate signaling pathways, could elicit strong cellular immunity. Here, we show that the small mol. adjuvant R848 and the RNA adjuvant PUUC, targeting endosomal TLR7s and cytoplasmic RLRs resp., when delivered together in polymer nanoparticles (NP), elicits a broadened immune responses in mouse bone marrow-derived dendritic cells (mBMDCs) and a synergistic response in both mouse and human plasmacytoid dendritic cells (pDCs). In mBMDCs, NP-R848-PUUC induced both NF-κB and interferon signaling. Interferon responses to co-delivered R848 and PUUC were additive in human peripheral blood mononuclear cells (PBMCs) and synergistic in human FLT3-differentiated mBMDCs and CAL-1 pDCs. Vaccination with NPs loaded with H1N1 Flu antigen, R848, and PUUC increased percentage of CD8+ T-cells in the lungs, percentage of antigen-specific CD4-T-cells in the spleen, and enhanced overall cytokine-secreting T cell percentages upon antigen restimulation. Also, in the spleen, T lymphopenia, esp. after in vitro restimulation with dual adjuvants, was obsd., indicating highly antigen-reactive T cells. Our results demonstrate that simultaneous engagement of TLR7 and RIG-I pathways using particulate carriers is a potential approach to improve cellular immunity in flu vaccination.
- 59Hou, Y.; Wang, Y.; Tang, Y.; Zhou, Z.; Tan, L.; Gong, T.; Zhang, L.; Sun, X. Co-Delivery of Antigen and Dual Adjuvants by Aluminum Hydroxide Nanoparticles for Enhanced Immune Responses. J. Controlled Release 2020, 326, 120– 130, DOI: 10.1016/j.jconrel.2020.06.02159https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXht1Kgu7bL&md5=ec7771ba4a626526fd70d75d7fa0af14Co-delivery of antigen and dual adjuvants by aluminum hydroxide nanoparticles for enhanced immune responsesHou, Yingying; Wang, Ying; Tang, Yao; Zhou, Zixuan; Tan, Lu; Gong, Tao; Zhang, Ling; Sun, XunJournal of Controlled Release (2020), 326 (), 120-130CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Adjuvants that contain pathogen-assocd. mol. patterns (PAMPs) can enhance vaccination efficacy by binding to pattern recognition receptors (PRRs), thereby stimulating immune responses. Particularly effective may be the combination of multiple PAMPs that activate different PRRs, which occurs with natural pathogens. Here we hypothesized the enhanced effects would occur in two adjuvants that stimulate different PRRs: CpG oligodeoxynucleotide (CpG-ODN), which is Toll-like receptor 9 agonist; and 5'-triphosphate, short, double-stranded RNA (3pRNA), which activates retinoic acid-inducible gene I (RIG-I). The model antigen ovalbumin (OVA) was loaded and adjuvants were surface-adsorbed to aluminum hydroxide nanoparticles (hereafter NP-3pRNA-CpG) by electrostatic interaction with an av. size of 120 nm and a neg. surface charge for targeting lymph nodes. These nanoparticles were efficiently internalized by antigen-presenting cells (APCs) in the lymph nodes, and the resulting APC activation and antigen cross-presentation generated strong humoral immunity and cytotoxic T lymphocyte responses and IFN-γ secretion. NP-3pRNA-CpG significantly suppressed B16-OVA tumor growth and prolonged survival of tumor-bearing mice in therapeutic and prophylactic models, illustrating the enhanced effects of CpG-ODN and 3pRNA. Our study highlights the potential of combining multiple adjuvants for effective vaccine design.
- 60Pagendarm, H. M.; Stone, P. T.; Kimmel, B. R.; Baljon, J. J.; Aziz, M. H.; Pastora, L. E.; Hubert, L.; Roth, E. W.; Almunif, S.; Scott, E. A.; Wilson, J. T. Engineering Endosomolytic Nanocarriers of Diverse Morphologies Using Confined Impingement Jet Mixing. Nanoscale 2023, 15, 16016– 16029, DOI: 10.1039/D3NR02874G60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXhvF2qsLnK&md5=04a78b16072f0ff7e9436c3c62991260Engineering endosomolytic nanocarriers of diverse morphologies using confined impingement jet mixingPagendarm, Hayden M.; Stone, Payton T.; Kimmel, Blaise R.; Baljon, Jessalyn J.; Aziz, Mina H.; Pastora, Lucinda E.; Hubert, Lauren; Roth, Eric W.; Almunif, Sultan; Scott, Evan A.; Wilson, John T.Nanoscale (2023), 15 (39), 16016-16029CODEN: NANOHL; ISSN:2040-3372. (Royal Society of Chemistry)The clin. translation of many biomol. therapeutics has been hindered by undesirable pharmacokinetic (PK) properties, inadequate membrane permeability, poor endosomal escape and cytosolic delivery, and/or susceptibility to degrdn. Overcoming these challenges merits the development of nanoscale drug carriers (nanocarriers) to improve the delivery of therapeutic cargo. Herein, we implement a flash nanopptn. (FNP) approach to produce nanocarriers of diverse vesicular morphologies by using various mol. wt. PEG-bl-DEAEMA-co-BMA (PEG-DB) polymers. We demonstrated that FNP can produce uniform (PDI < 0.1) particles after 5 impingements, and that by varying the copolymer hydrophilic mass fraction, FNP enables access to a diverse variety of nanoarchitectures including micelles, unilamellar vesicles (polymersomes), and multi-compartment vesicles (MCVs). We synthesized a library of 2 kDa PEG block copolymers, with DEAEMA-co-BMA second block mol. wts. of 3, 6, 12, 15, 20, and 30 kDa. All formulations were both pH responsive, endosomolytic, and capable of loading and cytosolically delivering small neg. charged mols. - albeit to different degrees. Using a B16. F10 melanoma model, we showcased the therapeutic potential of a lead FNP formulated PEG-DB nanocarrier, encapsulating the cyclic dinucleotide (CDN) cGAMP to activate the stimulator of interferon genes (STING) pathway in a therapeutically relevant context. Collectively, these data demonstrate that an FNP process can be used to formulate pH-responsive nanocarriers of diverse morphologies using a PEG-DB polymer system. As FNP is an industrially scalable process, these data address the crit. translational challenge of producing PEG-DB nanoparticles at scale. Furthermore, the diverse morphologies produced may specialize in the delivery of distinct biomol. cargos for other therapeutic applications, implicating the therapeutic potential of this platform in an array of disease applications.
- 61Manganiello, M. J.; Cheng, C.; Convertine, A. J.; Bryers, J. D.; Stayton, P. S. Diblock Copolymers with Tunable pH Transitions for Gene Delivery. Biomaterials 2012, 33, 2301– 2309, DOI: 10.1016/j.biomaterials.2011.11.01961https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xnt1entw%253D%253D&md5=ec63b85f138fe9934ba5508bf04e594fDiblock copolymers with tunable pH transitions for gene deliveryManganiello, Matthew J.; Cheng, Connie; Convertine, Anthony J.; Bryers, James D.; Stayton, Patrick S.Biomaterials (2012), 33 (7), 2301-2309CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)A series of diblock copolymers contg. an endosomal-releasing segment composed of diethylaminoethyl methacrylate (DEAEMA) and Bu methacrylate (BMA) were synthesized via reversible addn.-fragmentation chain transfer (RAFT) polymn. The materials were designed to condense plasmid DNA (pDNA) through electrostatic interactions with a cationic poly(N,N-dimethylaminoethyl methacrylate) (DMAEMA) first block. The pDMAEMA was employed as a macro chain transfer agent (macroCTA) for the synthesis of a series in which the relative feed ratios of DEAEMA and BMA were systematically varied from 20% to 70% BMA. The resultant diblock copolymers exhibited low polydispersity (PDI ≤ 1.06) with similar mol. wts. (Mn = 19.3-23.1 kDa). Dynamic light scattering (DLS) measurements in combination with 1H NMR D2O studies demonstrated that the free copolymers assemble into core-shell micelles at physiol. pH. Redn. of the soln. pH to values representative of endosomal/lysosomal compartments induced an increase in the net cationic charge of the core through protonation of the DEAEMA residues. This protonation promotes micelle destabilization and exposure of the hydrophobic BMA residues that destabilize biol. membranes. The pH value at which this micelle-to-unimer transition occurred was dependent on the hydrophobic content of the copolymer, with higher BMA-contg. copolymer compns. exhibiting pH-induced transitions to the membrane-destabilizing state at successively lower pH values. The ability of the diblock copolymers to deliver pDNA was subsequently investigated using a GFP expression vector in two monocyte cell lines. High levels of DNA transfection were obsd. for the copolymer compns. exhibiting the sharpest pH transitions and membrane destabilizing activities, demonstrating the importance of tuning the endosomal-releasing segment compn.
- 62Allen, S.; Osorio, O.; Liu, Y. G.; Scott, E. Facile Assembly and Loading of Theranostic Polymersomes Via Multi-Impingement Flash Nanoprecipitation. J. Controlled Release 2017, 262, 91– 103, DOI: 10.1016/j.jconrel.2017.07.02662https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1egsbjK&md5=a52b3f1d74b3a23fe99693893e9eab06Facile assembly and loading of theranostic polymersomes via multi-impingement flash nanoprecipitationAllen, Sean; Osorio, Omar; Liu, Yu-Gang; Scott, EvanJournal of Controlled Release (2017), 262 (), 91-103CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Flash nanopptn. has proven to be a powerful tool for the rapid and scalable assembly of solid-core nanoparticles from block copolymers. To date, FNP has not been applied for the fabrication of complex or vesicular nanoarchitectures capable of encapsulating hydrophilic mols. or bioactive protein therapeutics. Here, we present FNP as a single customizable method for the assembly of bicontinuous nanospheres, filomicelles and vesicular, multilamellar and tubular polymersomes from poly(ethylene glycol)-bl-poly(propylene sulfide) block copolymers. Multiple impingements of polymersomes assembled via FNP were shown to decrease vesicle diam. and polydispersity, allowing gram-scale fabrication of monodisperse polymersomes within minutes. Furthermore, we demonstrate that FNP supports simultaneous loading of both hydrophobic and hydrophilic mols. resp. into the polymersome membrane and aq. lumen, and encapsulated enzymes were found to be released and remain active following vesicle lysis. As an example application, theranostic polymersomes were generated via FNP that were dual loaded with the immunosuppressant rapamycin and a fluorescent dye to link targeted immune cells with the elicited immunomodulation of T cells. By expanding the capabilities of FNP, we present a rapid, scalable and reproducible method of nanofabrication for a wide range of nanoarchitectures that are typically challenging to assemble and load with therapeutics for controlled delivery and theranostic strategies.
- 63Han, J.; Zhu, Z.; Qian, H.; Wohl, A. R.; Beaman, C. J.; Hoye, T. R.; Macosko, C. W. A Simple Confined Impingement Jets Mixer for Flash Nanoprecipitation. J. Pharm. Sci. 2012, 101, 4018– 4023, DOI: 10.1002/jps.2325963https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XpvVers70%253D&md5=c5e9605fc18c6dfbb6818cfe3a8888f6A simple confined impingement jets mixer for flash nanoprecipitationHan, Jing; Zhu, Zhengxi; Qian, Haitao; Wohl, Adam R.; Beaman, Charles J.; Hoye, Thomas R.; Macosko, Christopher W.Journal of Pharmaceutical Sciences (2012), 101 (10), 4018-4023CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)Johnson and Prud'homme (2003. AICHE J 49:2264-2282) introduced the confined impingement jets (CIJ) mixer to prep. nanoparticles loaded with hydrophobic compds. (e.g., drugs, inks, fragrances, or pheromones) via flash nanopptn. (FNP). We have modified the original CIJ design to allow hand operation, eliminating the need for a syringe pump, and we added a second antisolvent diln. stage. Impingement mixing requires equal flow momentum from two opposing jets, one contg. the drug in org. solvent and the other contg. an antisolvent, typically water. The subsequent diln. step in the new design allows rapid quenching with high antisolvent concn. that enhances nanoparticle stability. This new CIJ with diln. (CIJ-D) mixer is a simple, cheap, and efficient device to produce nanoparticles. We have made 55 nm diam. β-carotene nanoparticles using the CIJ-D mixer. They are stable and reproducible in terms of particle size and distribution. We have also compared the performance of our CIJ-D mixer with the vortex mixer, which can operate at unequal flow rates (Liu et al., 2008. Chem Eng Sci 63:2829-2842), to make β-carotene-contg. particles over a series of turbulent conditions. On the basis of dynamic light scattering measurements, the new CIJ-D mixer produces stable particles of a size similar to the vortex mixer. Our CIJ-D design requires less vol. and provides an easily operated and inexpensive tool to produce nanoparticles via FNP and to evaluate new nanoparticle formulation. © 2012 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 64Johnson, B. K.; Prud’homme, R. K. Mechanism for Rapid Self-Assembly of Block Copolymer Nanoparticles. Phys. Rev. Lett. 2003, 91, 118302, DOI: 10.1103/PhysRevLett.91.11830264https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXnt1Oqurw%253D&md5=465c945d7870908649f6f231a6677a98Mechanism for Rapid Self-Assembly of Block Copolymer NanoparticlesJohnson, Brian K.; Prud'homme, Robert K.Physical Review Letters (2003), 91 (11), 118302/1-118302/4CODEN: PRLTAO; ISSN:0031-9007. (American Physical Society)Amphiphilic block copolymers in soln. spontaneously self-assemble when the solvent quality for one block is selectively decreased. We demonstrate that, for supersatn. ratio changes [d(S)/dt] over 105 per s from equil., nanoparticles are obtained with a formation mechanism and size dependent on the jumping rate and magnitude. The threshold rate for homogeneous pptn. is detd. by the induction time of a particle, equiv. to the diffusion limited fusion of copolymer chains to form a corona of overlapping sol. brushes. Via detn. of the induction time with a novel confined impinging jets mixer and use of a scaling relation, the interfacial free energy of a block copolymer nanoparticle was measured for the first time.
- 65Daniel, S.; Kis, Z.; Kontoravdi, C.; Shah, N. Quality by Design for Enabling RNA Platform Production Processes. Trends Biotechnol 2022, 40, 1213– 1228, DOI: 10.1016/j.tibtech.2022.03.01265https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XpvVahurs%253D&md5=7b64c147111978920111452f652888dcQuality by Design for enabling RNA platform production processesDaniel, Simon; Kis, Zoltan; Kontoravdi, Cleo; Shah, NilayTrends in Biotechnology (2022), 40 (10), 1213-1228CODEN: TRBIDM; ISSN:0167-7799. (Elsevier Ltd.)A review. RNA-based products have emerged as one of the most promising and strategic technologies for global vaccination, infectious disease control, and future therapy development. The assessment of crit. quality attributes (CQAs), product-process interactions, relevant process anal. technologies, and process modeling capabilities can feed into a robust Quality by Design (QbD) framework for future development, design, and control of manufg. processes. QbD implementation will help the RNA technol. reach its full potential and will be central to the development, pre-qualification, and regulatory approval of rapid response, disease-agnostic RNA platform prodn. processes.
- 66Warne, N.; Ruesch, M.; Siwik, P.; Mensah, P.; Ludwig, J.; Hripcsak, M.; Godavarti, R.; Prigodich, A.; Dolsten, M. Delivering 3 Billion Doses of Comirnaty in 2021. Nat. Biotechnol. 2023, 41, 183– 188, DOI: 10.1038/s41587-022-01643-166https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXis1Sjs7o%253D&md5=c0125b7d1eb92de3d174e35c99a37aecDelivering 3 billion doses of Comirnaty in 2021Warne, Nicholas; Ruesch, Margaret; Siwik, Pamela; Mensah, Paul; Ludwig, John; Hripcsak, Michael; Godavarti, Ranga; Prigodich, Andrew; Dolsten, MikaelNature Biotechnology (2023), 41 (2), 183-188CODEN: NABIF9; ISSN:1087-0156. (Nature Portfolio)Pfizer created a 'light-speed' approach to meet the challenge of vaccinating the world against COVID-19. It involved developing new strategies for all aspects of vaccine development, from sourcing materials and scaling up manufg. to transportation and dosing.
- 67Chang, T. Z.; Stadmiller, S. S.; Staskevicius, E.; Champion, J. A. Effects of Ovalbumin Protein Nanoparticle Vaccine Size and Coating on Dendritic Cell Processing. Biomater Sci. 2017, 5, 223– 233, DOI: 10.1039/C6BM00500D67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvF2gurnK&md5=2847bc4357609887cd5580a2ae9eef75Effects of ovalbumin protein nanoparticle vaccine size and coating on dendritic cell processingChang, Timothy Z.; Stadmiller, Samantha S.; Staskevicius, Erika; Champion, Julie A.Biomaterials Science (2017), 5 (2), 223-233CODEN: BSICCH; ISSN:2047-4849. (Royal Society of Chemistry)Nanoparticle vaccine delivery platforms are a promising technol. for enhancing vaccine immunogenicity. Protein nanoparticles (PNPs), made entirely from antigen, have been shown to induce protective immune responses against influenza. However, the fundamental mechanisms by which PNPs enhance component protein immunogenicity are not understood. Here, we investigate the role of size and coating of model ovalbumin (OVA) PNPs on particle uptake and trafficking, as well as on inflammation and maturation factor expression in dendritic cells (DCs) in vitro. OVA PNPs enhance antigen uptake in a size-independent manner, and experience attenuated endosomal acidification as compared to sol. OVA. OVA PNPs also trigger Fc receptor upregulation. Expression of cytokines IL-1β and TNF-α were PNP size- and coating-dependent, with small (∼270 nm) nanoparticles triggering greater inflammatory cytokine prodn. than large (∼560 nm) particles. IL-1β expression by DCs in response to PNP stimulation implies activation of the inflammasome, a pathway known to be activated by certain types of nanoparticulate adjuvants. The attenuated acidification and pro-inflammatory profile generated by PNPs in DCs demonstrate that phys. biomaterial properties can modulate dendritic cell-mediated antigen processing and adjuvancy. In addn. to nanoparticles' enhancement of DC antigen uptake, our work suggests that vaccine nanoparticle size and coating are uptake-independent modulators of immunogenicity.
- 68Thomas, S. N.; Schudel, A. Overcoming Transport Barriers for Interstitial-, Lymphatic-, and Lymph Node-Targeted Drug Delivery. Curr. Opin Chem. Eng. 2015, 7, 65– 74, DOI: 10.1016/j.coche.2014.11.00368https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2srotF2rsQ%253D%253D&md5=3253ad4ef935260a93f650e74b980196Overcoming transport barriers for interstitial-, lymphatic-, and lymph node-targeted drug deliveryThomas Susan N; Schudel AlexCurrent opinion in chemical engineering (2015), 7 (), 65-74 ISSN:2211-3398.Despite drug formulation improving circulation times and targeting, efficacy is stymied by inadequate penetration into and retention within target tissues. This review highlights the barriers restricting delivery to the connective tissue interstitium, lymphatics, and lymph nodes as well as advances in engineering drug carriers to overcome these delivery challenges. Three-dimensional tissue physiology is discussed in the context of providing material design principles for delivery to these tissues; in particular the influence of interstitial and lymphatic flows as well as differential permeabilities of the blood and lymphatic capillaries. Key examples of materials with different characteristics developed to overcome these transport barriers are discussed as well as potential areas for further development.
- 69Munson, M. J.; O’Driscoll, G.; Silva, A. M.; Lazaro-Ibanez, E.; Gallud, A.; Wilson, J. T.; Collen, A.; Esbjorner, E. K.; Sabirsh, A. A High-Throughput Galectin-9 Imaging Assay for Quantifying Nanoparticle Uptake, Endosomal Escape and Functional RNA Delivery. Commun. Biol. 2021, 4, 211, DOI: 10.1038/s42003-021-01728-869https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtV2iurvI&md5=2f6c6521ab150ffdcc94a614266cd245A high-throughput Galectin-9 imaging assay for quantifying nanoparticle uptake, endosomal escape and functional RNA deliveryMunson, Michael J.; O'Driscoll, Gwen; Silva, Andreia M.; Lazaro-Ibanez, Elisa; Gallud, Audrey; Wilson, John T.; Collen, Anna; Esbjoerner, Elin K.; Sabirsh, AlanCommunications Biology (2021), 4 (1), 211CODEN: CBOIDQ; ISSN:2399-3642. (Nature Research)RNA-based therapies have great potential to treat many undruggable human diseases. However, their efficacy, in particular for mRNA, remains hampered by poor cellular delivery and limited endosomal escape. Development and optimization of delivery vectors, such as lipid nanoparticles (LNPs), are impeded by limited screening methods to probe the intracellular processing of LNPs in sufficient detail. We have developed a high-throughput imaging-based endosomal escape assay utilizing a Galectin-9 reporter and fluorescently labeled mRNA to probe correlations between nanoparticle-mediated uptake, endosomal escape frequency, and mRNA translation. Furthermore, this assay has been integrated within a screening platform for optimization of lipid nanoparticle formulations. We show that Galectin-9 recruitment is a robust, quant. reporter of endosomal escape events induced by different mRNA delivery nanoparticles and small mols. We identify nanoparticles with superior escape properties and demonstrate cell line variances in endosomal escape response, highlighting the need for fine-tuning of delivery formulations for specific applications.
- 70Shae, D.; Becker, K. W.; Christov, P.; Yun, D. S.; Lytton-Jean, A. K. R.; Sevimli, S.; Ascano, M.; Kelley, M.; Johnson, D. B.; Balko, J. M.; Wilson, J. T. Endosomolytic Polymersomes Increase the Activity of Cyclic Dinucleotide STING Agonists to Enhance Cancer Immunotherapy. Nat. Nanotechnol 2019, 14, 269– 278, DOI: 10.1038/s41565-018-0342-570https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXmtVGitbo%253D&md5=4b95f1d3ac44492fd975fc9e37bcec58Endosomolytic polymersomes increase the activity of cyclic dinucleotide STING agonists to enhance cancer immunotherapyShae, Daniel; Becker, Kyle W.; Christov, Plamen; Yun, Dong Soo; Lytton-Jean, Abigail K. R.; Sevimli, Sema; Ascano, Manuel; Kelley, Mark; Johnson, Douglas B.; Balko, Justin M.; Wilson, John T.Nature Nanotechnology (2019), 14 (3), 269-278CODEN: NNAABX; ISSN:1748-3387. (Nature Research)Cyclic dinucleotide (CDN) agonists of stimulator of interferon genes (STING) are a promising class of immunotherapeutics that activate innate immunity to increase tumor immunogenicity. However, the efficacy of CDNs is limited by drug delivery barriers, including poor cellular targeting, rapid clearance and inefficient transport to the cytosol where STING is localized. Here, we describe STING-activating nanoparticles (STING-NPs)-rationally designed polymersomes for enhanced cytosolic delivery of the endogenous CDN ligand for STING, 2'3' cGMP-adenosine monophosphate (cGAMP). STING-NPs increase the biol. potency of cGAMP, enhance STING signaling in the tumor microenvironment and sentinel lymph node, and convert immunosuppressive tumors to immunogenic, tumoricidal microenvironments. This leads to enhanced therapeutic efficacy of cGAMP, inhibition of tumor growth, increased rates of long-term survival, improved response to immune checkpoint blockade and induction of immunol. memory that protects against tumor rechallenge. We validate STING-NPs in freshly isolated human melanoma tissue, highlighting their potential to improve clin. outcomes of immunotherapy.
- 71Zheng, S.; Wang, W.; Aldahdooh, J.; Malyutina, A.; Shadbahr, T.; Tanoli, Z.; Pessia, A.; Tang, J. Synergyfinder Plus: Toward Better Interpretation and Annotation of Drug Combination Screening Datasets. Genomics Proteomics Bioinformatics 2022, 20, 587– 596, DOI: 10.1016/j.gpb.2022.01.00471https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2M7jslymtA%253D%253D&md5=586c7516cf1d71a67d4788d4e640754eSynergyFinder Plus: Toward Better Interpretation and Annotation of Drug Combination Screening DatasetsZheng Shuyu; Wang Wenyu; Aldahdooh Jehad; Malyutina Alina; Shadbahr Tolou; Tanoli Ziaurrehman; Pessia Alberto; Tang JingGenomics, proteomics & bioinformatics (2022), 20 (3), 587-596 ISSN:.Combinatorial therapies have been recently proposed to improve the efficacy of anticancer treatment. The SynergyFinder R package is a software used to analyze pre-clinical drug combination datasets. Here, we report the major updates to the SynergyFinder R package for improved interpretation and annotation of drug combination screening results. Unlike the existing implementations, the updated SynergyFinder R package includes five main innovations. 1) We extend the mathematical models to higher-order drug combination data analysis and implement dimension reduction techniques for visualizing the synergy landscape. 2) We provide a statistical analysis of drug combination synergy and sensitivity with confidence intervals and P values. 3) We incorporate a synergy barometer to harmonize multiple synergy scoring methods to provide a consensus metric for synergy. 4) We evaluate drug combination synergy and sensitivity to provide an unbiased interpretation of the clinical potential. 5) We enable fast annotation of drugs and cell lines, including their chemical and target information. These annotations will improve the interpretation of the mechanisms of action of drug combinations. To facilitate the use of the R package within the drug discovery community, we also provide a web server at www.synergyfinderplus.org as a user-friendly interface to enable a more flexible and versatile analysis of drug combination data.
- 72Mouries, J.; Moron, G.; Schlecht, G.; Escriou, N.; Dadaglio, G.; Leclerc, C. Plasmacytoid Dendritic Cells Efficiently Cross-Prime Naive T Cells in Vivo after TLR Activation. Blood 2008, 112, 3713– 3722, DOI: 10.1182/blood-2008-03-14629072https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlCntrvF&md5=6484bd424f61a2bcf69ee7076b403ea6Plasmacytoid dendritic cells efficiently cross-prime naive T cells in vivo after TLR activationMouries, Juliette; Moron, Gabriel; Schlecht, Geraldine; Escriou, Nicolas; Dadaglio, Gilles; Leclerc, ClaudeBlood (2008), 112 (9), 3713-3722CODEN: BLOOAW; ISSN:0006-4971. (American Society of Hematology)Cross-presentation is a crucial mechanism in tumoral and microbial immunity because it allows internalized cell assocd. or exogenous antigens (Ags) to be delivered into the major histocompatibility complex I pathway. This pathway is important for the development of CD8+ T-cell responses and for the induction of tolerance. In mice, cross-presentation is considered to be a unique property of CD8α+ conventional dendritic cells (DCs). Here we show that splenic plasmacytoid DCs (pDCs) efficiently capture exogenous Ags in vivo but are not able to cross-present these Ags at steady state. However, in vitro and in vivo stimulation by Toll-like receptor-7, or -9 or viruses licenses pDCs to cross-present sol. or particulate Ags by a transporter assocd. with antigen processing-dependent mechanism. Induction of cross-presentation confers to pDCs the ability to generate efficient effector CD8+ T-cell responses against exogenous Ags in vivo, showing that pDCs may play a crucial role in induction of adaptive immune responses against pathogens that do not infect tissues of hemopoietic origin. This study provides the first evidence for an in vivo role of splenic pDCs in Ag cross-presentation and T-cell cross-priming and suggests that pDCs may constitute an attractive target to boost the efficacy of vaccines based on cytotoxic T lymphocyte induction.
- 73Villadangos, J. A.; Young, L. Antigen-Presentation Properties of Plasmacytoid Dendritic Cells. Immunity 2008, 29, 352– 361, DOI: 10.1016/j.immuni.2008.09.00273https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtF2jtLvL&md5=81e1c944723cfa0bc4117416b558dad0Antigen-presentation properties of plasmacytoid dendritic cellsVilladangos, Jose A.; Young, LouiseImmunity (2008), 29 (3), 352-361CODEN: IUNIEH; ISSN:1074-7613. (Cell Press)A review. One of the remaining enigmas of the dendritic cell (DC) network is the potential contribution of plasmacytoid DCs (pDCs) to antigen presentation. Although the antigen-presentation capacity of conventional DCs (cDCs) is clearly defined, pDCs are generally attributed as having little, if any, antigen-presentation function. Instead, pDCs are regarded as immunomodulating cells, capable of directing the immune response through their secretion of large amts. of type I interferons. Here, the authors examine the evidence for a potential role of pDC in antigen capture, processing, and presentation to T cells at sites of infection and in the lymph nodes.
- 74Hildner, K.; Edelson, B. T.; Purtha, W. E.; Diamond, M.; Matsushita, H.; Kohyama, M.; Calderon, B.; Schraml, B. U.; Unanue, E. R.; Diamond, M. S.; Schreiber, R. D.; Murphy, T. L.; Murphy, K. M. Batf3 Deficiency Reveals a Critical Role for CD8α+ Dendritic Cells in Cytotoxic T Cell Immunity. Science 2008, 322, 1097– 1100, DOI: 10.1126/science.116420674https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlGhu7fF&md5=5edf74e0adedb0e092d59a241a711784Batf3 Deficiency Reveals a Critical Role for CD8α+ Dendritic Cells in Cytotoxic T Cell ImmunityHildner, Kai; Edelson, Brian T.; Purtha, Whitney E.; Diamond, Mark; Matsushita, Hirokazu; Kohyama, Masako; Calderon, Boris; Schraml, Barbara U.; Unanue, Emil R.; Diamond, Michael S.; Schreiber, Robert D.; Murphy, Theresa L.; Murphy, Kenneth M.Science (Washington, DC, United States) (2008), 322 (5904), 1097-1100CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Although in vitro observations suggest that cross-presentation of antigens is mediated primarily by CD8α+ dendritic cells, in vivo anal. has been hampered by the lack of systems that selectively eliminate this cell lineage. We show that deletion of the transcription factor Batf3 ablated development of CD8α+ dendritic cells, allowing us to examine their role in immunity in vivo. Dendritic cells from Batf3-/- mice were defective in cross-presentation, and Batf3-/- mice lacked virus-specific CD8+ T cell responses to West Nile virus. Importantly, rejection of highly immunogenic syngeneic tumors was impaired in Batf3-/- mice. These results suggest an important role for CD8α+ dendritic cells and cross-presentation in responses to viruses and in tumor rejection.
- 75Kaech, S. M.; Wherry, E. J.; Ahmed, R. Effector and Memory T-Cell Differentiation: Implications for Vaccine Development. Nat. Rev. Immunol 2002, 2, 251– 262, DOI: 10.1038/nri77875https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XjtFKhtrc%253D&md5=7a63c84ffcfeae8b4362befd059980e3Effector and memory T-cell differentiation: implications for vaccine developmentKaech, Susan M.; Wherry, E. John; Ahmed, RafiNature Reviews Immunology (2002), 2 (4), 251-262CODEN: NRIABX; ISSN:1474-1733. (Nature Publishing Group)A review. Recent work shows that after stimulation with antigen, CD4+ and CD8+ T cells embark on a program of proliferation that is closely linked with the acquisition of effector functions and leads ultimately to memory-cell formation. Here, we discuss the signals required for commitment to this program of development and the factors that might influence its progression. Models of the pathways of effector and memory T-cell differentiation are discussed, and we highlight the implications of this new understanding for the optimization of vaccine strategies.
- 76Taylor, M. A.; Hughes, A. M.; Walton, J.; Coenen-Stass, A. M. L.; Magiera, L.; Mooney, L.; Bell, S.; Staniszewska, A. D.; Sandin, L. C.; Barry, S. T. Longitudinal Immune Characterization of Syngeneic Tumor Models to Enable Model Selection for Immune Oncology Drug Discovery. J. Immunother Cancer 2019, 7, 328, DOI: 10.1186/s40425-019-0794-776https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MfksVSntA%253D%253D&md5=7b3308ccad3e1cbb161737db47069240Longitudinal immune characterization of syngeneic tumor models to enable model selection for immune oncology drug discoveryTaylor Molly A; Hughes Adina M; Walton Josephine; Coenen-Stass Anna M L; Magiera Lukasz; Mooney Lorraine; Bell Sigourney; Staniszewska Anna D; Sandin Linda C; Barry Simon T; Watkins Amanda; Carnevalli Larissa S; Hardaker Elizabeth L; Mooney LorraineJournal for immunotherapy of cancer (2019), 7 (1), 328 ISSN:.BACKGROUND: The ability to modulate immune-inhibitory pathways using checkpoint blockade antibodies such as αPD-1, αPD-L1, and αCTLA-4 represents a significant breakthrough in cancer therapy in recent years. This has driven interest in identifying small-molecule-immunotherapy combinations to increase the proportion of responses. Murine syngeneic models, which have a functional immune system, represent an essential tool for pre-clinical evaluation of new immunotherapies. However, immune response varies widely between models and the translational relevance of each model is not fully understood, making selection of an appropriate pre-clinical model for drug target validation challenging. METHODS: Using flow cytometry, O-link protein analysis, RT-PCR, and RNAseq we have characterized kinetic changes in immune-cell populations over the course of tumor development in commonly used syngeneic models. RESULTS: This longitudinal profiling of syngeneic models enables pharmacodynamic time point selection within each model, dependent on the immune population of interest. Additionally, we have characterized the changes in immune populations in each of these models after treatment with the combination of α-PD-L1 and α-CTLA-4 antibodies, enabling benchmarking to known immune modulating treatments within each model. CONCLUSIONS: Taken together, this dataset will provide a framework for characterization and enable the selection of the optimal models for immunotherapy combinations and generate potential biomarkers for clinical evaluation in identifying responders and non-responders to immunotherapy combinations.
- 77Wang-Bishop, L.; Wehbe, M.; Shae, D.; James, J.; Hacker, B. C.; Garland, K.; Chistov, P. P.; Rafat, M.; Balko, J. M.; Wilson, J. T. Potent Sting Activation Stimulates Immunogenic Cell Death to Enhance Antitumor Immunity in Neuroblastoma. J. Immunother Cancer 2020, 8, e000282, DOI: 10.1136/jitc-2019-000282There is no corresponding record for this reference.
- 78Kilchrist, K. V.; Dimobi, S. C.; Jackson, M. A.; Evans, B. C.; Werfel, T. A.; Dailing, E. A.; Bedingfield, S. K.; Kelly, I. B.; Duvall, C. L. Gal8 Visualization of Endosome Disruption Predicts Carrier-Mediated Biologic Drug Intracellular Bioavailability. ACS Nano 2019, 13, 1136– 1152, DOI: 10.1021/acsnano.8b0548278https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXht12qs7w%253D&md5=35d2d7494c1933249faf76094de990cfGal8 Visualization of Endosome Disruption Predicts Carrier-Mediated Biologic Drug Intracellular BioavailabilityKilchrist, Kameron V.; Dimobi, Somtochukwu C.; Jackson, Meredith A.; Evans, Brian C.; Werfel, Thomas A.; Dailing, Eric A.; Bedingfield, Sean K.; Kelly, Isom B.; Duvall, Craig L.ACS Nano (2019), 13 (2), 1136-1152CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Endolysosome entrapment is one of the key barriers to the therapeutic use of biol. drugs that act intracellularly. The screening of prospective nanoscale endosome-disrupting delivery technologies is currently limited by methods that are indirect and cumbersome. Here, we statistically validate Galectin 8 (Gal8) intracellular tracking as a superior approach that is direct, quant., and predictive of therapeutic cargo intracellular bioactivity through in vitro high-throughput screening and in vivo validation. Gal8 is a cytosolically dispersed protein that, when endosomes are disrupted, redistributes by binding to glycosylation moieties selectively located on the inner face of endosomal membranes. The quant. redistribution of a Gal8 fluorescent fusion protein from the cytosol into endosomes is demonstrated as a real-time, live-cell assessment of endosomal integrity that does not require labeling or modification of either the carrier or the biol. drug and that allows quant. distinction between closely related, endosome-disruptive drug carriers. Through screening two families of siRNA polymeric carrier compns. at varying dosages, we show that Gal8 endosomal recruitment correlates strongly (r = 0.95 and p < 10-4) with intracellular siRNA bioactivity. Through this screen, we gathered insights into how compn. and mol. wt. affect endosome disruption activity of poly[(ethylene glycol)-b-[(2-(dimethylamino)ethyl methacrylate)-co-(Bu methacrylate)]] [PEG-(DMAEMA-co-BMA)] siRNA delivery systems. Addnl. studies showed that Gal8 recruitment predicts intracellular bioactivity better than current std. methods such as Lysotracker colocalization (r = 0.35, not significant), pH-dependent hemolysis (not significant), or cellular uptake (r = 0.73 and p < 10-3). Importantly, the Gal8 recruitment method is also amenable to fully objective high-throughput screening using automated image acquisition and quant. image anal., with a robust estd. Z' of 0.6 (whereas assays with Z' > 0 have high-throughput screening utility). Finally, we also provide measurements of in vivo endosomal disruption based on Gal8 visualization (p < 0.03) of a nanocarrier formulation confirmed to produce significant cytosolic delivery and bioactivity of siRNA within tumors (p < 0.02). In sum, this report establishes the utility of Gal8 subcellular tracking for the rapid optimization and high-throughput screening of the endosome disruption potency of intracellular delivery technologies.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsnano.3c04471.
Supplementary data describing polymer characterization, nanovaccine drug loading and sizing properties, BMDC costimulatory molecule expression and pro-inflammatory cytokine secretion, Loewe synergy scores, cellular uptake, tumor growth curves, and flow cytometry gating strategies (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.