1
|
Giram P, Md Mahabubur Rahman K, Aqel O, You Y. In Situ Cancer Vaccines: Redefining Immune Activation in the Tumor Microenvironment. ACS Biomater Sci Eng 2025; 11:2550-2583. [PMID: 40223683 DOI: 10.1021/acsbiomaterials.5c00121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Cancer is one of the leading causes of mortality worldwide. Nanomedicines have significantly improved life expectancy and survival rates for cancer patients in current standard care. However, recurrence of cancer due to metastasis remains a significant challenge. Vaccines can provide long-term protection and are ideal for preventing bacterial and viral infections. Cancer vaccines, however, have shown limited therapeutic efficacy and raised safety concerns despite extensive research. Cancer vaccines target and stimulate responses against tumor-specific antigens and have demonstrated great potential for cancer treatment in preclinical studies. However, tumor-associated immunosuppression and immune tolerance driven by immunoediting pose significant challenges for vaccine design. In situ vaccination represents an alternative approach to traditional cancer vaccines. This strategy involves the intratumoral administration of immunostimulants to modulate the growth and differentiation of innate immune cells, such as dendritic cells, macrophages, and neutrophils, and restore T-cell activity. Currently approved in situ vaccines, such as T-VEC, have demonstrated clinical promise, while ongoing clinical trials continue to explore novel strategies for broader efficacy. Despite these advancements, failures in vaccine research highlight the need to address tumor-associated immune suppression and immune escape mechanisms. In situ vaccination strategies combine innate and adaptive immune stimulation, leveraging tumor-associated antigens to activate dendritic cells and cross-prime CD8+ T cells. Various vaccine modalities, such as nucleotide-based vaccines (e.g., RNA and DNA vaccines), peptide-based vaccines, and cell-based vaccines (including dendritic, T-cell, and B-cell approaches), show significant potential. Plant-based viral approaches, including cowpea mosaic virus and Newcastle disease virus, further expand the toolkit for in situ vaccination. Therapeutic modalities such as chemotherapy, radiation, photodynamic therapy, photothermal therapy, and Checkpoint blockade inhibitors contribute to enhanced antigen presentation and immune activation. Adjuvants like CpG-ODN and PRR agonists further enhance immune modulation and vaccine efficacy. The advantages of in situ vaccination include patient specificity, personalization, minimized antigen immune escape, and reduced logistical costs. However, significant barriers such as tumor heterogeneity, immune evasion, and logistical challenges remain. This review explores strategies for developing potent cancer vaccines, examines ongoing clinical trials, evaluates immune stimulation methods, and discusses prospects for advancing in situ cancer vaccination.
Collapse
Affiliation(s)
- Prabhanjan Giram
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Kazi Md Mahabubur Rahman
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Osama Aqel
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Youngjae You
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| |
Collapse
|
2
|
Huis In 't Veld LG, Cornelissen LA, van den Bogaard L, Ansems M, Ho NI, Adema GJ. Saponin-based adjuvant uptake and induction of antigen cross-presentation by CD11b+ dendritic cells and macrophages. NPJ Vaccines 2025; 10:15. [PMID: 39843492 PMCID: PMC11754886 DOI: 10.1038/s41541-024-01056-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Saponin-based adjuvants (SBAs) distinguish themselves as vaccine adjuvants by instigating a potent activation of CD8+ T cells. Previously, we discovered SBA's ability to induce cross-presentation in dendritic cells (DCs) leading to CD8+ T cell activation. Moreover, the MHCIIloCD11bhi bone marrow-derived DC (BMDC) subset was identified to be the most responsive DC subset to SBA treatment. To further investigate SBA's mode of action, labeling of SBAs was optimized with the fluorescent dye SP-DiIC18(3). Efficient uptake of SBAs occurs specifically by MHCIIloCD11bhi BMDCs and bone marrow-derived macrophages (BMDMs) in vitro and cDC2s and macrophages ex vivo. Furthermore, SBAs are primarily taken up by clathrin-mediated endocytosis and uptake induces lipid bodies and antigen translocation to the cytosol in MHCIIloCD11bhi BMDCs and BMDMs. Importantly, BMDMs treated with SBAs exhibit cross-presentation leading to potent CD8+ T cells activation. Our findings explain the potency of SBAs as vaccine adjuvants and contribute to vaccine development.
Collapse
Affiliation(s)
- Lisa Gm Huis In 't Veld
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lenneke Am Cornelissen
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lune van den Bogaard
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marleen Ansems
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nataschja I Ho
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
3
|
Bai D, Kim H, Wang P. Development of semisynthetic saponin immunostimulants. Med Chem Res 2024; 33:1292-1306. [PMID: 39132259 PMCID: PMC11315725 DOI: 10.1007/s00044-024-03227-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 04/20/2024] [Indexed: 08/13/2024]
Abstract
Many natural saponins demonstrate immunostimulatory adjuvant activities, but they also have some inherent drawbacks that limit their clinical use. To overcome these limitations, extensive structure-activity-relationship (SAR) studies have been conducted. The SAR studies of QS-21 and related saponins reveal that their respective fatty side chains are crucial for potentiating a strong cellular immune response. Replacing the hydrolytically unstable ester side chain in the C28 oligosaccharide domain with an amide side chain in the same domain or in the C3 branched trisaccharide domain is a viable approach for generating robust semisynthetic saponin immunostimulants. Given the striking resemblance of natural momordica saponins (MS) I and II to the deacylated Quillaja Saponaria (QS) saponins (e.g., QS-17, QS-18, and QS-21), incorporating an amide side chain into the more sustainable MS, instead of deacylated QS saponins, led to the discovery of MS-derived semisynthetic immunostimulatory adjuvants VSA-1 and VSA-2. This review focuses on the authors' previous work on SAR studies of QS and MS saponins.
Collapse
Affiliation(s)
- Di Bai
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL AL35294 USA
| | - Hyunjung Kim
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL AL35294 USA
| | - Pengfei Wang
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL AL35294 USA
| |
Collapse
|
4
|
Séguier D, Adams ES, Kotamarti S, D'Anniballe V, Michael ZD, Deivasigamani S, Olivier J, Villers A, Hoimes C, Polascik TJ. Intratumoural immunotherapy plus focal thermal ablation for localized prostate cancer. Nat Rev Urol 2024; 21:290-302. [PMID: 38114768 DOI: 10.1038/s41585-023-00834-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 12/21/2023]
Abstract
Major advances have been made in the use of immunotherapy for the treatment of solid tumours, including the use of intratumourally injected immunotherapy instead of systemically delivered immunotherapy. The success of immunotherapy in prostate cancer treatment has been limited to specific populations with advanced disease, which is thought to be a result of prostate cancer being an immunologically 'cold' cancer. Accordingly, combining intratumoural immunotherapy with other treatments that would increase the immunological heat of prostate cancer is of interest. Thermal ablation therapy is currently one of the main strategies used for the treatment of localized prostate cancer and it causes immunological activation against prostate tissue. The use of intratumoural immunotherapy as an adjunct to thermal ablation offers the potential to elicit a systemic and lasting adaptive immune response to cancer-specific antigens, leading to a synergistic effect of combination therapy. The combination of thermal ablation and immunotherapy is currently in the early stages of investigation for the treatment of multiple solid tumour types, and the potential for this combination therapy to also offer benefit to prostate cancer patients is exciting.
Collapse
Affiliation(s)
- Denis Séguier
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA.
- Department of Urology, Lille University, Lille, France.
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER; UMR9020-U1277), Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France.
| | - Eric S Adams
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Srinath Kotamarti
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Vincent D'Anniballe
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Zoe D Michael
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Sriram Deivasigamani
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Jonathan Olivier
- Department of Urology, Lille University, Lille, France
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER; UMR9020-U1277), Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Arnauld Villers
- Department of Urology, Lille University, Lille, France
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER; UMR9020-U1277), Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Christopher Hoimes
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, North Carolina, 27708, USA
| | - Thomas J Polascik
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| |
Collapse
|
5
|
Oladejo M, Tijani AO, Puri A, Chablani L. Adjuvants in cutaneous vaccination: A comprehensive analysis. J Control Release 2024; 369:475-492. [PMID: 38569943 DOI: 10.1016/j.jconrel.2024.03.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024]
Abstract
Skin is the body's largest organ and serves as a protective barrier from physical, thermal, and mechanical environmental challenges. Alongside, the skin hosts key immune system players, such as the professional antigen-presenting cells (APCs) like the Langerhans cells in the epidermis and circulating macrophages in the blood. Further, the literature supports that the APCs can be activated by antigen or vaccine delivery via multiple routes of administration through the skin. Once activated, the stimulated APCs drain to the associated lymph nodes and gain access to the lymphatic system. This further allows the APCs to engage with the adaptive immune system and activate cellular and humoral immune responses. Thus, vaccine delivery via skin offers advantages such as reliable antigen delivery, superior immunogenicity, and convenient delivery. Several preclinical and clinical studies have demonstrated the significance of vaccine delivery using various routes of administration via skin. However, such vaccines often employ adjuvant/(s), along with the antigen of interest. Adjuvants augment the immune response to a vaccine antigen and improve the therapeutic efficacy. Due to these reasons, adjuvants have been successfully used with infectious disease vaccines, cancer immunotherapy, and immune-mediated diseases. To capture these developments, this review will summarize preclinical and clinical study results of vaccine delivery via skin in the presence of adjuvants. A focused discussion regarding the FDA-approved adjuvants will address the experiences of using such adjuvant-containing vaccines. In addition, the challenges and regulatory concerns with these adjuvants will be discussed. Finally, the review will share the prospects of adjuvant-containing vaccines delivered via skin.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Akeemat O Tijani
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA
| | - Ashana Puri
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA.
| | - Lipika Chablani
- Wegmans School of Pharmacy, St. John Fisher University, 3690 East Ave, Rochester, NY 14618, USA.
| |
Collapse
|
6
|
Ho NI, Huis In 't Veld LGM, van Eck van der Sluijs J, Heuts BMH, Looman MWG, Kers-Rebel ED, van den Dries K, Dolstra H, Martens JHA, Hobo W, Adema GJ. Saponin-based adjuvants enhance antigen cross-presentation in human CD11c + CD1c + CD5 - CD163 + conventional type 2 dendritic cells. J Immunother Cancer 2023; 11:e007082. [PMID: 37612044 PMCID: PMC10450066 DOI: 10.1136/jitc-2023-007082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Adjuvants are key for effective vaccination against cancer and chronic infectious diseases. Saponin-based adjuvants (SBAs) are unique among adjuvants in their ability to induce robust cell-mediated immune responses in addition to antibody responses. Recent preclinical studies revealed that SBAs induced cross-presentation and lipid bodies in otherwise poorly cross-presenting CD11b+ murine dendritic cells (DCs). METHOD Here, we investigated the response of human DC subsets to SBAs with RNA sequencing and pathway analyses, lipid body induction visualized by laser scanning microscopy, antigen translocation to the cytosol, and antigen cross-presentation to CD8+ T cells. RESULTS RNA sequencing of SBA-treated conventional type 1 DC (cDC1) and type 2 DC (cDC2) subsets uncovered that SBAs upregulated lipid-related pathways in CD11c+ CD1c+ cDC2s, especially in the CD5- CD163+ CD14+ cDC2 subset. Moreover, SBAs induced lipid bodies and enhanced endosomal antigen translocation into the cytosol in this particular cDC2 subset. Finally, SBAs enhanced cross-presentation only in cDC2s, which requires the CD163+ CD14+ cDC2 subset. CONCLUSIONS These data thus identify the CD163+ CD14+ cDC2 subset as the main SBA-responsive DC subset in humans and imply new strategies to optimize the application of saponin-based adjuvants in a potent cancer vaccine.
Collapse
Affiliation(s)
- Nataschja I Ho
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Lisa G M Huis In 't Veld
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Jesper van Eck van der Sluijs
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Branco M H Heuts
- Department of Molecular Biology, Faculty of Science, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Maaike W G Looman
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Esther D Kers-Rebel
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Koen van den Dries
- Radboud Technology Center Microscopy, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Willemijn Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
7
|
Jiang M, Fiering S, Shao Q. Combining energy-based focal ablation and immune checkpoint inhibitors: preclinical research and clinical trials. Front Oncol 2023; 13:1153066. [PMID: 37251920 PMCID: PMC10211342 DOI: 10.3389/fonc.2023.1153066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/12/2023] [Indexed: 05/31/2023] Open
Abstract
Energy-based focal therapy (FT) uses targeted, minimally invasive procedures to destroy tumors while preserving normal tissue and function. There is strong emerging interest in understanding how systemic immunity against the tumor can occur with cancer immunotherapy, most notably immune checkpoint inhibitors (ICI). The motivation for combining FT and ICI in cancer management relies on the synergy between the two different therapies: FT complements ICI by reducing tumor burden, increasing objective response rate, and reducing side effects of ICI; ICI supplements FT by reducing local recurrence, controlling distal metastases, and providing long-term protection. This combinatorial strategy has shown promising results in preclinical study (since 2004) and the clinical trials (since 2011). Understanding the synergy calls for understanding the physics and biology behind the two different therapies with distinctive mechanisms of action. In this review, we introduce different types of energy-based FT by covering the biophysics of tissue-energy interaction and present the immunomodulatory properties of FT. We discuss the basis of cancer immunotherapy with the emphasis on ICI. We examine the approaches researchers have been using and the results from both preclinical models and clinical trials from our exhaustive literature research. Finally, the challenges of the combinatory strategy and opportunities of future research is discussed extensively.
Collapse
Affiliation(s)
- Minhan Jiang
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
- Dartmouth Cancer Center, Dartmouth Geisel School of Medicine and Dartmouth Health, Lebanon, NH, United States
| | - Qi Shao
- Department of Radiology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
8
|
van den Bijgaart RJE, Mekers VE, Schuurmans F, Raaijmakers TK, Wassink M, Veltien A, Dumont E, Heerschap A, Fütterer JJ, Adema GJ. Mechanical high-intensity focused ultrasound creates unique tumor debris enhancing dendritic cell-induced T cell activation. Front Immunol 2022; 13:1038347. [PMID: 36569907 PMCID: PMC9768443 DOI: 10.3389/fimmu.2022.1038347] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction In situ tumor ablation releases a unique repertoire of antigens from a heterogeneous population of tumor cells. High-intensity focused ultrasound (HIFU) is a completely noninvasive ablation therapy that can be used to ablate tumors either by heating (thermal (T)-HIFU) or by mechanical disruption (mechanical (M)-HIFU). How different HIFU ablation techniques compare with respect to their antigen release profile, their activation of responder T cells, and their ability to synergize with immune stimuli remains to be elucidated. Methods and results Here, we compare the immunomodulatory effects of T-HIFU and M-HIFU ablation with or without the TLR9 agonist CpG in the ovalbumin-expressing lymphoma model EG7. M-HIFU ablation alone, but much less so T-HIFU, significantly increased dendritic cell (DC) activation in draining lymph nodes (LNs). Administration of CpG following T- or M-HIFU ablation increased DC activation in draining LNs to a similar extend. Interestingly, ex vivo co-cultures of draining LN suspensions from HIFU plus CpG treated mice with CD8+ OT-I T cells demonstrate that LN cells from M-HIFU treated mice most potently induced OT-I proliferation. To delineate the mechanism for the enhanced anti-tumor immune response induced by M-HIFU, we characterized the RNA, DNA and protein content of tumor debris generated by both HIFU methods. M-HIFU induced a uniquely altered RNA, DNA and protein profile, all showing clear signs of fragmentation, whereas T-HIFU did not. Moreover, western blot analysis showed decreased levels of the immunosuppressive cytokines IL-10 and TGF-β in M-HIFU generated tumor debris compared to untreated tumor tissue or T-HIFU. Conclusion Collectively, these results imply that M-HIFU induces a unique context of the ablated tumor material, enhancing DC-mediated T cell responses when combined with CpG.
Collapse
Affiliation(s)
- Renske J. E. van den Bijgaart
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Vera E. Mekers
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Fabian Schuurmans
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Tonke K. Raaijmakers
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Melissa Wassink
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Andor Veltien
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Arend Heerschap
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jurgen J. Fütterer
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, Netherlands,Department of Robotics and Mechatronics, University of Twente, Enschede, Netherlands
| | - Gosse J. Adema
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands,*Correspondence: Gosse J. Adema,
| |
Collapse
|
9
|
Saponin-based adjuvant-induced dendritic cell cross-presentation is dependent on PERK activation. Cell Mol Life Sci 2022; 79:231. [PMID: 35396971 PMCID: PMC8994093 DOI: 10.1007/s00018-022-04253-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 12/22/2022]
Abstract
Saponin-based adjuvants (SBAs) are promising new adjuvants that stand out as they not only enforce CD4 + T cell-mediated immunity and antibody responses, but also induce an unprecedented level of antigen cross-presentation by dendritic cells (DC) and subsequent CD8 + T cell activation. We discovered that SBA’s ability to boost cross-presentation depends on the induction of lipid bodies (LBs). Moreover, the MHCIIloCD11bhi DC subset was identified to be most responsive to SBA-induced cross-presentation. The aim is to further unravel the mechanisms behind the induction of DC cross-presentation by SBAs. Here we show that SBAs specifically induce the PKR-like Endoplasmic Reticulum kinase (PERK) pathway and that SBA-induced DC cross-presentation is dependent on activation of the PERK pathway. PERK activation and LB formation are both crucial for SBA-induced cross-presentation and PERK inhibition has little or no effect on SBA-induced LB formation. SBA’s responsiveness, LB formation and PERK activation are specific for the MHCIIloCD11bhi DCs. These findings contribute to understanding the pathways involved in SBA-induced cross-presentation and immune activation which will ultimately lead to the development of vaccines with improved efficiency and safety.
Collapse
|
10
|
van den Bijgaart RJE, Schuurmans F, Fütterer JJ, Verheij M, Cornelissen LAM, Adema GJ. Immune Modulation Plus Tumor Ablation: Adjuvants and Antibodies to Prime and Boost Anti-Tumor Immunity In Situ. Front Immunol 2021; 12:617365. [PMID: 33936033 PMCID: PMC8079760 DOI: 10.3389/fimmu.2021.617365] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
In situ tumor ablation techniques, like radiotherapy, cryo- and heat-based thermal ablation are successfully applied in oncology for local destruction of tumor masses. Although diverse in technology and mechanism of inducing cell death, ablative techniques share one key feature: they generate tumor debris which remains in situ. This tumor debris functions as an unbiased source of tumor antigens available to the immune system and has led to the concept of in situ cancer vaccination. Most studies, however, report generally modest tumor-directed immune responses following local tumor ablation as stand-alone treatment. Tumors have evolved mechanisms to create an immunosuppressive tumor microenvironment (TME), parts of which may admix with the antigen depot. Provision of immune stimuli, as well as approaches that counteract the immunosuppressive TME, have shown to be key to boost ablation-induced anti-tumor immunity. Recent advances in protein engineering have yielded novel multifunctional antibody formats. These multifunctional antibodies can provide a combination of distinct effector functions or allow for delivery of immunomodulators specifically to the relevant locations, thereby mitigating potential toxic side effects. This review provides an update on immune activation strategies that have been tested to act in concert with tumor debris to achieve in situ cancer vaccination. We further provide a rationale for multifunctional antibody formats to be applied together with in situ ablation to boost anti-tumor immunity for local and systemic tumor control.
Collapse
Affiliation(s)
- Renske J E van den Bijgaart
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Fabian Schuurmans
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jurgen J Fütterer
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Robotics and Mechatronics, University of Twente, Enschede, Netherlands
| | - Marcel Verheij
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Lenneke A M Cornelissen
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Gosse J Adema
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
11
|
Raaijmakers TK, van den Bijgaart RJE, den Brok MH, Wassink M, de Graaf A, Wagenaars JA, Nierkens S, Ansems M, Scheffer GJ, Adema GJ. Tumor ablation plus co-administration of CpG and saponin adjuvants affects IL-1 production and multifunctional T cell numbers in tumor draining lymph nodes. J Immunother Cancer 2021; 8:jitc-2020-000649. [PMID: 32461350 PMCID: PMC7254152 DOI: 10.1136/jitc-2020-000649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2020] [Indexed: 12/25/2022] Open
Abstract
Background Tumor ablation techniques, like cryoablation, are successfully used in the clinic to treat tumors. The tumor debris remaining in situ after ablation is a major antigen depot, including neoantigens, which are presented by dendritic cells (DCs) in the draining lymph nodes to induce tumor-specific CD8+ T cells. We have previously shown that co-administration of adjuvants is essential to evoke strong in vivo antitumor immunity and the induction of long-term memory. However, which adjuvants most effectively combine with in situ tumor ablation remains unclear. Methods and results Here, we show that simultaneous administration of cytidyl guanosyl (CpG) with saponin-based adjuvants following cryoablation affects multifunctional T-cell numbers and interleukin (IL)-1 induced polymorphonuclear neutrophil recruitment in the tumor draining lymph nodes, relative to either adjuvant alone. The combination of CpG and saponin-based adjuvants induces potent DC maturation (mainly CpG-mediated), antigen cross-presentation (mainly saponin-based adjuvant mediated), while excretion of IL-1β by DCs in vitro depends on the presence of both adjuvants. Most strikingly, CpG/saponin-based adjuvant exposed DCs potentiate antigen-specific T-cell proliferation resulting in multipotent T cells with increased capacity to produce interferon (IFN)γ, IL-2 and tumor necrosis factor-α in vitro. Also in vivo the CpG/saponin-based adjuvant combination plus cryoablation increased the numbers of tumor-specific CD8+ T cells showing enhanced IFNγ production as compared with single adjuvant treatments. Conclusions Collectively, these data indicate that co-injection of CpG with saponin-based adjuvants after cryoablation induces an increased amount of tumor-specific multifunctional T cells. The combination of saponin-based adjuvants with toll-like receptor 9 adjuvant CpG in a cryoablative setting therefore represents a promising in situ vaccination strategy.
Collapse
Affiliation(s)
- Tonke K Raaijmakers
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Renske J E van den Bijgaart
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martijn H den Brok
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Melissa Wassink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Annemarie de Graaf
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jori A Wagenaars
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Stefan Nierkens
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.,Center for Translational Immunology, Utrecht University, Utrecht, The Netherlands
| | - Marleen Ansems
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gert Jan Scheffer
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
12
|
Natural and Synthetic Saponins as Vaccine Adjuvants. Vaccines (Basel) 2021; 9:vaccines9030222. [PMID: 33807582 PMCID: PMC8001307 DOI: 10.3390/vaccines9030222] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/20/2022] Open
Abstract
Saponin adjuvants have been extensively studied for their use in veterinary and human vaccines. Among them, QS-21 stands out owing to its unique profile of immunostimulating activity, inducing a balanced Th1/Th2 immunity, which is valuable to a broad scope of applications in combating various microbial pathogens, cancers, and other diseases. It has recently been approved for use in human vaccines as a key component of combination adjuvants, e.g., AS01b in Shingrix® for herpes zoster. Despite its usefulness in research and clinic, the cellular and molecular mechanisms of QS-21 and other saponin adjuvants are poorly understood. Extensive efforts have been devoted to studies for understanding the mechanisms of QS-21 in different formulations and in different combinations with other adjuvants, and to medicinal chemistry studies for gaining mechanistic insights and development of practical alternatives to QS-21 that can circumvent its inherent drawbacks. In this review, we briefly summarize the current understandings of the mechanism underlying QS-21’s adjuvanticity and the encouraging results from recent structure-activity-relationship (SAR) studies.
Collapse
|
13
|
The Impact of Focused Ultrasound in Two Tumor Models: Temporal Alterations in the Natural History on Tumor Microenvironment and Immune Cell Response. Cancers (Basel) 2020; 12:cancers12020350. [PMID: 32033171 PMCID: PMC7072338 DOI: 10.3390/cancers12020350] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 02/06/2023] Open
Abstract
Image-guided focused ultrasound (FUS) has been successfully employed as an ablative treatment for solid malignancies by exposing immune cells to tumor debris/antigens, consequently inducing an immune response within the tumor microenvironment (TME). To date, immunomodulation effects of non-ablative pulsed-FUS (pFUS) on the TME are poorly understood. In this study, the temporal differences of cytokines, chemokines, and trophic factors (CCTFs) and immune cell populations induced by pFUS were interrogated in murine B16 melanoma or 4T1 breast cancer cells subcutaneously inoculated into C57BL/6 or BALB/c mice. Natural history growth characteristics during the course of 11 days showed a progressive increase in size for both tumors, and proteomic analysis revealed a shift toward an immunosuppressive TME. With respect to tumor natural growth, pFUS applied to tumors on days 1, 5, or 9 demonstrated a decrease in the growth rate 24 h post-sonication. Flow cytometry analysis of tumors, LNs, and Sp, as well as CCTF profiles, relative DNA damage, and adaptive T-cell localization within tumors, demonstrated dynamic innate and adaptive immune-modulation following pFUS in early time points of B16 tumors and in advanced 4T1 tumors. These results provide insight into the temporal dynamics in the treatment-associated TME, which could be used to evaluate an immunomodulatory approach in different tumor types.
Collapse
|
14
|
Kemeny N, Kurilova I, Li J, Camacho JC, Sofocleous CT. Liver-Directed and Systemic Therapies for Colorectal Cancer Liver Metastases. Cardiovasc Intervent Radiol 2019; 42:1240-1254. [PMID: 31312902 DOI: 10.1007/s00270-019-02284-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/03/2019] [Indexed: 02/07/2023]
|
15
|
Ojiako CM, Okoye EI, Oli AN, Ike CJ, Esimone CO, Attama AA. Preliminary studies on the formulation of immune stimulating complexes using saponin from Carica papaya leaves. Heliyon 2019; 5:e01962. [PMID: 31294113 PMCID: PMC6595190 DOI: 10.1016/j.heliyon.2019.e01962] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/12/2019] [Accepted: 06/12/2019] [Indexed: 11/25/2022] Open
Abstract
There have been several modifications in the use of immune stimulating complexes as adjuvants, such as the replacement of phospholipids with saponin content. Not much research has been done on the use of local alternatives. This actually instigated the use of a local alternative saponin source from Carica papaya leaves to formulate Iscomatrix adjuvant. The Iscomatrix samples used in this study were formulated using different methods (the rapid injection, the reversed rapid injection, the slow/dropwise injection and the reversed slow/dropwise injection methods). Furthermore, the quantity of the components was also varied. These formulated samples were compared with other adjuvants and analysed for their ability to induce antibody and cell mediated immune responses using animal model i.e. mice. The results showed that the Iscomatrix samples formulated, were able to induce significant humoral and antibody mediated immune response (ranging from 16.7 % - 38.88 %) and they also elicited cell mediated immune response (ranging from 8.33 % - 16.7 %) when compared to the models that were administered with antigen only. Further characterizations were made, such as pH, UV scanning, Scanning Electron Microscopy. The analysis revealed that the samples were slightly soluble in distilled water with a neutral pH ranging from 7.26 - 7.43. The UV analysis also indicated that they all had a close range of absorption peaks (between 266.8-269.37 nm). Saponin from Carica papaya leaves can be used to formulate Iscomatrix adjuvant capable of stimulating cell mediated and antibody mediated immune responses.
Collapse
Affiliation(s)
- Chioma Miracle Ojiako
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - Ebere Innocent Okoye
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - Angus Nnamdi Oli
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - Chibueze Jeremiah Ike
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - Charles O Esimone
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - Anthony A Attama
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| |
Collapse
|
16
|
Unleashing Tumour-Dendritic Cells to Fight Cancer by Tackling Their Three A's: Abundance, Activation and Antigen-Delivery. Cancers (Basel) 2019; 11:cancers11050670. [PMID: 31091774 PMCID: PMC6562396 DOI: 10.3390/cancers11050670] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/22/2019] [Accepted: 05/10/2019] [Indexed: 12/24/2022] Open
Abstract
Recent advances in cancer immunotherapy have mainly focused on re-activating T-cell responses against cancer cells. However, both priming and activation of effector T-cell responses against cancer-specific antigens require cross-talk with dendritic cells (DCs), which are responsible for the capturing, processing and presentation of tumour-(neo)antigens to T cells. DCs consequently constitute an essential target in efforts to generate therapeutic immunity against cancer. This review will discuss recent research that is unlocking the cancer-fighting potential of tumour-infiltrating DCs. First, the complexity of DCs in the tumour microenvironment regarding the different subsets and the difficulty of translating mouse data into equivalent human data will be briefly touched upon. Mainly, possible solutions to problems currently faced in DC-based cancer treatments will be discussed, including their infiltration into tumours, activation strategies, and antigen delivery methods. In this way, we hope to put together a broad picture of potential synergistic therapies that could be implemented to harness the full capacity of tumour-infiltrating DCs to stimulate anti-tumour immune responses in patients.
Collapse
|
17
|
Koshkina O, Lajoinie G, Bombelli FB, Swider E, Cruz LJ, White PB, Schweins R, Dolen Y, van Dinther EAW, van Riessen NK, Rogers SE, Fokkink R, Voets IK, van Eck ERH, Heerschap A, Versluis M, de Korte CL, Figdor CG, de Vries IJM, Srinivas M. Multicore Liquid Perfluorocarbon-Loaded Multimodal Nanoparticles for Stable Ultrasound and 19F MRI Applied to In Vivo Cell Tracking. ADVANCED FUNCTIONAL MATERIALS 2019; 29:1806485. [PMID: 32132881 PMCID: PMC7056356 DOI: 10.1002/adfm.201806485] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Indexed: 05/22/2023]
Abstract
Ultrasound is the most commonly used clinical imaging modality. However, in applications requiring cell-labeling, the large size and short active lifetime of ultrasound contrast agents limit their longitudinal use. Here, 100 nm radius, clinically applicable, polymeric nanoparticles containing a liquid perfluorocarbon, which enhance ultrasound contrast during repeated ultrasound imaging over the course of at least 48 h, are described. The perfluorocarbon enables monitoring the nanoparticles with quantitative 19F magnetic resonance imaging, making these particles effective multimodal imaging agents. Unlike typical core-shell perfluorocarbon-based ultrasound contrast agents, these nanoparticles have an atypical fractal internal structure. The nonvaporizing highly hydrophobic perfluorocarbon forms multiple cores within the polymeric matrix and is, surprisingly, hydrated with water, as determined from small-angle neutron scattering and nuclear magnetic resonance spectroscopy. Finally, the nanoparticles are used to image therapeutic dendritic cells with ultrasound in vivo, as well as with 19F MRI and fluorescence imaging, demonstrating their potential for long-term in vivo multimodal imaging.
Collapse
Affiliation(s)
- Olga Koshkina
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands; Physical Chemistry of Polymers, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Guillaume Lajoinie
- Physics of Fluids Group, Technical Medical (TechMed) Centre and MESA+ Institute for, Nanotechnology, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Francesca Baldelli Bombelli
- Laboratory of Supramolecular and BioNano Materials, (SupraBioNanoLab), Department of Chemistry, Materials, and Chemical Engineering, "Giulio Natta,", Politecnico di Milano, Via Luigi Mancinelli 7, 20131 Milan, Italy
| | - Edyta Swider
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Paul B White
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Ralf Schweins
- Institut Laue - Langevin, DS/LSS, 71 Avenue des Martyrs, CS 20 156, 38042 Grenoble CEDEX 9, France
| | - Yusuf Dolen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Eric A W van Dinther
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - N Koen van Riessen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Sarah E Rogers
- ISIS Pulsed Neutron and Muon Source, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Harwell, Oxford OX11 0QX, UK
| | - Remco Fokkink
- Department of Agrotechnology and Food Sciences, Physical Chemistry and Soft Matter, Wageningen University, 6708 WE, Wageningen, Netherlands
| | - Ilja K Voets
- Laboratory of Self-Organizing Soft Matter, Laboratory of Macromolecular and Organic Chemistry, Department of Chemical Engineering and Chemistry and Institute for Complex Molecular Systems, Eindhoven University of Technology, De Rondom 70, 5612 AP, Eindhoven, The Netherlands
| | - Ernst R H van Eck
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Michel Versluis
- Physics of Fluids Group, Technical Medical (TechMed) Centre and MESA+ Institute for, Nanotechnology, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Chris L de Korte
- Physics of Fluids Group, Technical Medical (TechMed) Centre and MESA+ Institute for, Nanotechnology, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands; Department of Radiology and Nuclear Medicine, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
18
|
Erinjeri JP, Fine GC, Adema GJ, Ahmed M, Chapiro J, den Brok M, Duran R, Hunt SJ, Johnson DT, Ricke J, Sze DY, Toskich BB, Wood BJ, Woodrum D, Goldberg SN. Immunotherapy and the Interventional Oncologist: Challenges and Opportunities-A Society of Interventional Oncology White Paper. Radiology 2019; 292:25-34. [PMID: 31012818 DOI: 10.1148/radiol.2019182326] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Interventional oncology is a subspecialty field of interventional radiology that addresses the diagnosis and treatment of cancer and cancer-related problems by using targeted minimally invasive procedures performed with image guidance. Immuno-oncology is an innovative area of cancer research and practice that seeks to help the patient's own immune system fight cancer. Both interventional oncology and immuno-oncology can potentially play a pivotal role in cancer management plans when used alongside medical, surgical, and radiation oncology in the care of cancer patients.
Collapse
Affiliation(s)
- Joseph P Erinjeri
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - Gabriel C Fine
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - Gosse J Adema
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - Muneeb Ahmed
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - Julius Chapiro
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - Martijn den Brok
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - Rafael Duran
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - Stephen J Hunt
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - D Thor Johnson
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - Jens Ricke
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - Daniel Y Sze
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - Beau Bosko Toskich
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - Bradford J Wood
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - David Woodrum
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| | - S Nahum Goldberg
- From the Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-118, New York, NY 10065 (J.P.E.); Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, Utah (G.C.F.); Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands (G.J.A., M.d.B.); Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A.); Division of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Conn (J.C.); Department of Radiodiagnostic and Interventional Radiology, University of Lausanne, Lausanne, Switzerland (R.D.); Penn Image-Guided Interventions Laboratory and Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pa (S.J.H.); Department of Radiology, University of Colorado, Denver, Colo (D.T.J.); Department of Radiology, Ludwig-Maximilian University, Munich, Germany (J.R.); Division of Vascular and Interventional Radiology, Stanford University, Stanford, Calif (D.Y.S.); Division of Interventional Radiology, Mayo Clinic Florida, Jacksonville, Fla (B.B.T.); Center for Interventional Oncology, National Cancer Institute, Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Md (B.J.W.); Department of Radiology, Mayo Clinic, Rochester Minn (D.W.); and Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.)
| |
Collapse
|
19
|
Shao Q, O'Flanagan S, Lam T, Roy P, Pelaez F, Burbach BJ, Azarin SM, Shimizu Y, Bischof JC. Engineering T cell response to cancer antigens by choice of focal therapeutic conditions. Int J Hyperthermia 2019; 36:130-138. [DOI: 10.1080/02656736.2018.1539253] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Qi Shao
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Stephen O'Flanagan
- Department of Laboratory Medicine and Pathology and Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Tiffany Lam
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| | - Priyatanu Roy
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Francisco Pelaez
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| | - Brandon J Burbach
- Department of Laboratory Medicine and Pathology and Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| | - Yoji Shimizu
- Department of Laboratory Medicine and Pathology and Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - John C Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
20
|
Chua BY, Sekiya T, Jackson DC. Opinion: Making Inactivated and Subunit-Based Vaccines Work. Viral Immunol 2018; 31:150-158. [PMID: 29369750 DOI: 10.1089/vim.2017.0146] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Empirically derived vaccines have in the past relied on the isolation and growth of disease-causing microorganisms that are then inactivated or attenuated before being administered. This is often done without prior knowledge of the mechanisms involved in conferring protective immunity. Recent advances in scientific technologies and in our knowledge of how protective immune responses are induced enable us to rationally design novel and safer vaccination strategies. Such advances have accelerated the development of inactivated whole-organism- and subunit-based vaccines. In this review, we discuss ideal attributes and criteria that need to be considered for the development of vaccines and some existing vaccine platforms. We focus on inactivated vaccines against influenza virus and ways by which vaccine efficacy can be improved with the use of adjuvants and Toll-like receptor-2 signaling.
Collapse
Affiliation(s)
- Brendon Y Chua
- 1 Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne, Australia .,2 Research Center for Zoonosis Control, Hokkaido University , Sapporo, Japan .,3 Global Institution for Collaborative Research and Education, Hokkaido University , Sapporo, Japan
| | - Toshiki Sekiya
- 2 Research Center for Zoonosis Control, Hokkaido University , Sapporo, Japan .,3 Global Institution for Collaborative Research and Education, Hokkaido University , Sapporo, Japan
| | - David C Jackson
- 1 Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne, Australia .,2 Research Center for Zoonosis Control, Hokkaido University , Sapporo, Japan .,3 Global Institution for Collaborative Research and Education, Hokkaido University , Sapporo, Japan
| |
Collapse
|
21
|
Chen M, Balhara V, Jaimes Castillo AM, Balsevich J, Johnston LJ. Interaction of saponin 1688 with phase separated lipid bilayers. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1263-1272. [PMID: 28389202 DOI: 10.1016/j.bbamem.2017.03.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/14/2017] [Accepted: 03/31/2017] [Indexed: 12/12/2022]
Abstract
Saponins are a diverse family of naturally occurring plant triterpene or steroid glycosides that have a wide range of biological activities. They have been shown to permeabilize membranes and in some cases membrane disruption has been hypothesized to involve saponin/cholesterol complexes. We have examined the interaction of steroidal saponin 1688-1 with lipid membranes that contain cholesterol and have a mixture of liquid-ordered (Lo) and liquid-disordered (Ld) phases as a model for lipid rafts in cellular membranes. A combination of atomic force microscopy (AFM) and fluorescence was used to probe the effect of saponin on the bilayer. The results demonstrate that saponin forms defects in the membrane and also leads to formation of small aggregates on the membrane surface. Although most of the membrane damage occurs in the liquid-disordered phase, fluorescence results demonstrate that saponin localizes in both ordered and disordered membrane phases, with a modest preference for the disordered regions. Similar effects are observed for both direct incorporation of saponin in the lipid mixture used to make vesicles/bilayers and for incubation of saponin with preformed bilayers. The results suggest that the initial sites of interaction are at the interface between the domains and surrounding disordered phase. The preference for saponin localization in the disordered phase may reflect the ease of penetration of saponin into a less ordered membrane, rather than the actual cholesterol concentration in the membrane. Dye leakage assays indicate that a high concentration of saponin is required for membrane permeabilization consistent with the supported lipid bilayer experiments.
Collapse
Affiliation(s)
- Maohui Chen
- Measurement Science and Standards, National Research Council Canada, Ottawa, ON K1A 0R6, Canada
| | - Vinod Balhara
- Measurement Science and Standards, National Research Council Canada, Ottawa, ON K1A 0R6, Canada
| | | | - John Balsevich
- Aquatic and Crop Resource Development, National Research Council Canada, Saskatoon, SK S7N 0W9, Canada
| | - Linda J Johnston
- Measurement Science and Standards, National Research Council Canada, Ottawa, ON K1A 0R6, Canada.
| |
Collapse
|
22
|
den Brok MH, Büll C, Wassink M, de Graaf AM, Wagenaars JA, Minderman M, Thakur M, Amigorena S, Rijke EO, Schrier CC, Adema GJ. Saponin-based adjuvants induce cross-presentation in dendritic cells by intracellular lipid body formation. Nat Commun 2016; 7:13324. [PMID: 27819292 PMCID: PMC5103066 DOI: 10.1038/ncomms13324] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/22/2016] [Indexed: 12/23/2022] Open
Abstract
Saponin-based adjuvants (SBAs) are being used in animal and human (cancer) vaccines, as they induce protective cellular immunity. Their adjuvant potency is a factor of inflammasome activation and enhanced antigen cross-presentation by dendritic cells (DCs), but how antigen cross-presentation is induced is not clear. Here we show that SBAs uniquely induce intracellular lipid bodies (LBs) in the CD11b+ DC subset in vitro and in vivo. Using genetic and pharmacological interference in models for vaccination and in situ tumour ablation, we demonstrate that LB induction is causally related to the saponin-dependent increase in cross-presentation and T-cell activation. These findings link adjuvant activity to LB formation, aid the application of SBAs as a cancer vaccine component, and will stimulate development of new adjuvants enhancing T-cell-mediated immunity. Saponin-based adjuvants are being explored as vaccine components as they induce high levels of antigen cross-presentation, but it is unknown how. Here the authors show that these adjuvants enhance cross-presentation by driving production of lipid bodies inside CD11b dendritic cells.
Collapse
Affiliation(s)
- Martijn H den Brok
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands.,Department of Anesthesiology, Pain and Palliative Medicine, Radboud UMC, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands
| | - Christian Büll
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
| | - Melissa Wassink
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
| | - Annemarie M de Graaf
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
| | - Jori A Wagenaars
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
| | - Marthe Minderman
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
| | - Mayank Thakur
- Institute for Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité Universitätsmedizin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Sebastian Amigorena
- INSERM, Institut Curie, Section Recherche, Rue d'Ulm 26, 75005 Paris, France
| | - Eric O Rijke
- MSD Animal Health, Wim de Korverstraat 35, 5831 AN Boxmeer, The Netherlands
| | - Carla C Schrier
- MSD Animal Health, Wim de Korverstraat 35, 5831 AN Boxmeer, The Netherlands
| | - Gosse J Adema
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
23
|
Thermal and mechanical high-intensity focused ultrasound: perspectives on tumor ablation, immune effects and combination strategies. Cancer Immunol Immunother 2016; 66:247-258. [PMID: 27585790 PMCID: PMC5281669 DOI: 10.1007/s00262-016-1891-9] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/18/2016] [Indexed: 12/21/2022]
Abstract
Tumor ablation technologies, such as radiofrequency-, cryo- or high-intensity focused ultrasound (HIFU) ablation will destroy tumor tissue in a minimally invasive manner. Ablation generates large volumes of tumor debris in situ, releasing multiple bio-molecules like tumor antigens and damage-associated molecular patterns. To initiate an adaptive antitumor immune response, antigen-presenting cells need to take up tumor antigens and, following activation, present them to immune effector cells. The impact of the type of tumor ablation on the precise nature, availability and suitability of the tumor debris for immune response induction, however, is poorly understood. In this review, we focus on immune effects after HIFU-mediated ablation and compare these to findings using other ablation technologies. HIFU can be used both for thermal and mechanical destruction of tissue, inducing coagulative necrosis or subcellular fragmentation, respectively. Preclinical and clinical results of HIFU tumor ablation show increased infiltration and activation of CD4+ and CD8+ T cells. As previously observed for other types of tumor ablation technologies, however, this ablation-induced enhanced infiltration alone appears insufficient to generate consistent protective antitumor immunity. Therapies combining ablation with immune stimulation are therefore expected to be key to boost HIFU-induced immune effects and to achieve systemic, long-lasting, antitumor immunity.
Collapse
|
24
|
Liu Y, Tian X, Jiang S, Ren X, Liu F, Yang J, Chen Y, Jiang Y. Umbilical cord blood-derived dendritic cells infected by adenovirus for SP17 expression induce antigen-specific cytotoxic T cells against NSCLC cells. Cell Immunol 2015; 298:18-24. [DOI: 10.1016/j.cellimm.2015.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/09/2015] [Accepted: 08/17/2015] [Indexed: 12/25/2022]
|
25
|
Kheirolomoom A, Ingham ES, Mahakian LM, Tam SM, Silvestrini MT, Tumbale SK, Foiret J, Hubbard NE, Borowsky AD, Murphy WJ, Ferrara KW. CpG expedites regression of local and systemic tumors when combined with activatable nanodelivery. J Control Release 2015; 220:253-264. [PMID: 26471394 DOI: 10.1016/j.jconrel.2015.10.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/01/2015] [Accepted: 10/09/2015] [Indexed: 12/22/2022]
Abstract
Ultrasonic activation of nanoparticles provides the opportunity to deliver a large fraction of the injected dose to insonified tumors and produce a complete local response. Here, we evaluate whether the local and systemic response to chemotherapy can be enhanced by combining such a therapy with locally-administered CpG as an immune adjuvant. In order to create stable, activatable particles, a complex between copper and doxorubicin (CuDox) was created within temperature-sensitive liposomes. Whereas insonation of the CuDox liposomes alone has been shown to produce a complete response in murine breast cancer after 8 treatments of 6 mg/kg delivered over 4 weeks, combining this treatment with CpG resolved local cancers within 3 treatments delivered over 7 days. Further, contralateral tumors regressed as a result of the combined treatment, and survival was extended in systemic disease. In both the treated and contralateral tumor site, the combined treatment increased leukocytes and CD4+ and CD8+ T-effector cells and reduced myeloid-derived suppressor cells (MDSCs). Taken together, the results suggest that this combinatorial treatment significantly enhances the systemic efficacy of locally-activated nanotherapy.
Collapse
Affiliation(s)
- Azadeh Kheirolomoom
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Elizabeth S Ingham
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Lisa M Mahakian
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Sarah M Tam
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Matthew T Silvestrini
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Spencer K Tumbale
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Josquin Foiret
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Neil E Hubbard
- University of California, Davis, Center for Comparative Medicine, Davis, CA 95616, USA
| | - Alexander D Borowsky
- University of California, Davis, Center for Comparative Medicine, Davis, CA 95616, USA
| | - William J Murphy
- University of California, Davis, Department of Dermatology, 2921 Stockton Blvd., Institute for Regenerative Cures, Suite 1630, Sacramento, CA 95817, USA
| | - Katherine W Ferrara
- University of California, Davis, Department of Biomedical Engineering, 451 East Health Sciences Drive, Davis, CA 95616, USA.
| |
Collapse
|
26
|
Baust JG, Bischof JC, Jiang-Hughes S, Polascik TJ, Rukstalis DB, Gage AA, Baust JM. Re-purposing cryoablation: a combinatorial 'therapy' for the destruction of tissue. Prostate Cancer Prostatic Dis 2015; 18:87-95. [PMID: 25622539 DOI: 10.1038/pcan.2014.54] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/18/2014] [Accepted: 12/10/2014] [Indexed: 11/09/2022]
Abstract
It is now recognized that the tumor microenvironment creates a protective neo-tissue that isolates the tumor from the various defense strategies of the body. Evidence demonstrates that, with successive therapeutic attempts, cancer cells acquire resistance to individual treatment modalities. For example, exposure to cytotoxic drugs results in the survival of approximately 20-30% of the cancer cells as only dividing cells succumb to each toxic exposure. With follow-up treatments, each additional dose results in tumor-associated fibroblasts secreting surface-protective proteins, which enhance cancer cell resistance. Similar outcomes are reported following radiotherapy. These defensive strategies are indicative of evolved capabilities of cancer to assure successful tumor growth through well-established anti-tumor-protective adaptations. As such, successful cancer management requires the activation of multiple cellular 'kill switches' to prevent initiation of diverse protective adaptations. Thermal therapies are unique treatment modalities typically applied as monotherapies (without repetition) thereby denying cancer cells the opportunity to express defensive mutations. Further, the destructive mechanisms of action involved with cryoablation (CA) include both physical and molecular insults resulting in the disruption of multiple defensive strategies that are not cell cycle dependent and adds a damaging structural (physical) element. This review discusses the application and clinical outcomes of CA with an emphasis on the mechanisms of cell death induced by structural, metabolic, vascular and immune processes. The induction of diverse cell death cascades, resulting in the activation of apoptosis and necrosis, allows CA to be characterized as a combinatorial treatment modality. Our understanding of these mechanisms now supports adjunctive therapies that can augment cell death pathways.
Collapse
Affiliation(s)
- J G Baust
- 1] Institute of Biomedical Technology, State University of New York at Binghamton, Binghamton, NY, USA [2] Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| | - J C Bischof
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - S Jiang-Hughes
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - T J Polascik
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - D B Rukstalis
- Department of Urology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - A A Gage
- Department of Surgery, State University of New York at Buffalo, Medical School, Buffalo, NY, USA
| | | |
Collapse
|
27
|
In-situ administration of dendritic cells following argon-helium cryosurgery enhances specific antiglioma immunity in mice. Neuroreport 2015; 25:900-8. [PMID: 24942351 DOI: 10.1097/wnr.0000000000000196] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dendritic cells (DCs) are highly specialized antigen-presenting cells that play a key role in the activation of naive T cells. With an aim to explore whether in-situ administration of DCs following argon-helium cryosurgery could enhance specific antiglioma immunity in mice, we evaluated the validity of this approach in a murine subcutaneous GL261 glioma model. C57BL/6 mice models bearing subcutaneous GL261 glioma were established and then divided into four groups, namely, no-treatment group (n=14), DC group (n=14), cryosurgery group (n=15), and cryosurgery+DC group (n=15). Compared with the other groups, cryosurgery combined with DCs injection reduced tumor sizes and significantly prolonged survival. In addition, the combined treatment resulted in significantly increasing percentages of CD3, CD3CD4 cells, the ratio of CD3CD4/CD3CD8, and the level of serum interleukin-12 10 days after treatments. Furthermore, in the combined treatment group, Th1 cells were significantly higher than those in the other groups, and the splenic cytotoxic T lymphocyte of mice showed significantly increasing specific cytotoxicity against GL261 cells. These results indicated that in addition to the destruction of tumor, cryosurgery combined with DCs injection enhanced systemic antitumor immunity, suggesting the potential usefulness of the combined treatment in the clinical management of gliomas.
Collapse
|
28
|
Lee JS, Cho MK, Hwang HS, Ko EJ, Lee YN, Kwon YM, Kim MC, Kim KH, Lee YT, Jung YJ, Kang SM. Ginseng diminishes lung disease in mice immunized with formalin-inactivated respiratory syncytial virus after challenge by modulating host immune responses. J Interferon Cytokine Res 2014; 34:902-14. [PMID: 25051168 PMCID: PMC4217040 DOI: 10.1089/jir.2013.0093] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 04/29/2014] [Indexed: 12/30/2022] Open
Abstract
Formalin-inactivated respiratory syncytial virus (FI-RSV) immunization is known to cause severe pulmonary inflammatory disease after subsequent RSV infection. Ginseng has been used in humans for thousands of years due to its potential health benefits. We investigated whether ginseng would have immune modulating effects on RSV infection in mice previously immunized with FI-RSV. Oral administration of mice with ginseng increased IgG2a isotype antibody responses to FI-RSV immunization, indicating T-helper type 1 (Th1) immune responses. Ginseng-treated mice that were nonimmunized or previously immunized with FI-RSV showed improved protection against RSV challenge compared with control mice without ginseng treatment. Ginseng-mediated improved clinical outcomes after live RSV infection were evidenced by diminished weight losses, decreased interleukin-4 cytokine production but increased interferon-γ production, modulation of CD3 T-cell populations toward a Th1 response, and reduced inflammatory response. Ginseng-mediated protective host immune modulation against RSV pulmonary inflammation was observed in different strains of wild-type and mutant mice. These results indicate that ginseng can modulate host immune responses to FI-RSV immunization and RSV infection, resulting in protective effects against pulmonary inflammatory disease.
Collapse
Affiliation(s)
- Jong Seok Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
- Department of Medicinal and Industrial Crops, Korea National College of Agriculture and Fisheries, Hwaseong, Korea
| | - Min Kyoung Cho
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Hye Suk Hwang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Young-Man Kwon
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
- Animal and Plant Quarantine Agency, Anyang, Gyeonggi-do, Korea
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| |
Collapse
|
29
|
Pouliot K, Buglione-Corbett R, Marty-Roix R, Montminy-Paquette S, West K, Wang S, Lu S, Lien E. Contribution of TLR4 and MyD88 for adjuvant monophosphoryl lipid A (MPLA) activity in a DNA prime-protein boost HIV-1 vaccine. Vaccine 2014; 32:5049-56. [PMID: 25045815 PMCID: PMC10687719 DOI: 10.1016/j.vaccine.2014.07.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 06/17/2014] [Accepted: 07/08/2014] [Indexed: 12/19/2022]
Abstract
Recombinant protein vaccines are commonly formulated with an immune-stimulatory compound, or adjuvant, to boost immune responses to a particular antigen. Recent studies have shown that, through recognition of molecular motifs, receptors of the innate immune system are involved in the functions of adjuvants to generate and direct adaptive immune responses. However, it is not clear to which degree those receptors are also important when the adjuvant is used as part of a novel heterologous prime-boost immunization process in which the priming and boosting components are not the same type of vaccines. In the current study, we compared the immune responses elicited by a pentavalent HIV-1 DNA prime-protein boost vaccine in mice deficient in either Toll-like receptor 4 (TLR4) or myeloid differentiation primary response gene 88 (MyD88) to wildtype mice. HIV gp120 protein administered in the boost phase was formulated with either monophosphoryl lipid A (MPLA), QS-21, or Al(OH)3. Endpoint antibody titer, serum cytokine response and T-cell memory response were assessed. Neither TLR4 nor MyD88 deficiency had a significant effect on the immune response of mice given vaccine formulated with QS-21 or Al(OH)3. However, TLR4- and MyD88-deficiency decreased both the antibody and T-cell responses in mice administered HIV gp120 formulated with MPLA. These results further our understanding of the activation of TLR4 and MyD88 by MPLA in the context of a DNA prime/protein boost immunization strategy.
Collapse
Affiliation(s)
- Kimberly Pouliot
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States
| | - Rachel Buglione-Corbett
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Robyn Marty-Roix
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States
| | - Sara Montminy-Paquette
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States
| | - Kim West
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Shixia Wang
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Egil Lien
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States; Centre of Molecular Inflammation Research, Dept. of Cancer and Molecular Medicine, NTNU, 7491 Trondheim, Norway.
| |
Collapse
|
30
|
Abstract
Cryoablation is increasingly being used as a primary treatment for localized cancers and as a salvage therapy for metastatic cancers. Anecdotal clinical reports and animal experiments have confirmed an induction of systemic antitumor immune response by tumor cryoablation. To capitalize on the stimulatory effects of cryoablation for cancer immunotherapy, this response must be intensified using other immunomodulatory agents. This article reviews the preclinical and clinical evidence and discusses the mechanism of the antitumor immune response generated by cryoablation. The rationale and evidence behind several immunotherapy approaches that can be combined with cryoablation to devise a cryoimmunotherapeutic strategy with a potential to impact the progression of metastatic disease are described.
Collapse
Affiliation(s)
- Abhinav Sidana
- Division of Urology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Suite 2513, Cincinnati, OH 45229, USA.
| |
Collapse
|
31
|
Kim JJ, Hwang YH, Kang KY, Kim I, Kim JB, Park JH, Yoo YC, Yee ST. Enhanced dendritic cell maturation by the B-chain of Korean mistletoe lectin (KML-B), a novel TLR4 agonist. Int Immunopharmacol 2014; 21:309-19. [PMID: 24859056 DOI: 10.1016/j.intimp.2014.05.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 04/24/2014] [Accepted: 05/06/2014] [Indexed: 01/04/2023]
Abstract
Korean mistletoe lectin (KML) is composed of A and B sub-chains. The B-chain binds to cell surfaces, whereas the A-chain hinders translation because it is a RIP (ribosome inactivating protein) inducing apoptosis. Although KML has various biological and immunological activities, its potential use in cancer therapy or as an adjuvant therapy is limited by its toxicity to normal cells. This study was conducted to determine whether the B-chain of KML (KML-B) has immunoadjuvant activity and cytotoxicity activity. To evaluate the immunomodulatory activities of B chain KML, in vitro experiments employing bone marrow-derived dendritic cells (BMDCs) were performed. Dendritic cells (DCs) are a unique group of white blood cells that are able to capture and process antigens for presentation to T cells, which constitute primary immune response. In the present study, KML-B was found to be non-cytotoxic to BMDCs. Furthermore, the expressions of co-stimulatory molecules (CD40, CD80, CD86, and MHC II) and the secretions of cytokines (IL-1β, IL-6, IL-12p70, and TNF-α) were increased in BMDCs by KML-B. In addition, other indicators (antigen-uptake and CCR7 expression) of BMDC maturation were changed by KML-B, and the ability of KML-B to enhance various functions by BMDCs was found to be dependent on TLR4 expression. Moreover, BMDCs matured by KML-B induced naïve CD4(+) T cell differentiation toward Th1 cells directly and indirectly. These experiments confirm that KML-B exhibits potent immunomodulatory properties and suggest that KML-B be considered a potential dendritic cell-based cancer therapy and immunoadjuvant.
Collapse
Affiliation(s)
- Jong-Jin Kim
- Department of Biology, Sunchon National University, Suncheon, Republic of Korea
| | - Yun-Ho Hwang
- Department of Pharmacy, Sunchon National University, Suncheon, Republic of Korea
| | - Kyung-Yun Kang
- Department of Pharmacy, Sunchon National University, Suncheon, Republic of Korea
| | - Inbo Kim
- School of Life Science, Handong Global University, Pohang, Republic of Korea
| | - Jong-Bae Kim
- School of Life Science, Handong Global University, Pohang, Republic of Korea
| | - Jong-Hwan Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Yung-Choon Yoo
- Department of Microbiology, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Sung-Tae Yee
- Department of Biology, Sunchon National University, Suncheon, Republic of Korea; Department of Pharmacy, Sunchon National University, Suncheon, Republic of Korea.
| |
Collapse
|
32
|
Kim JJ, Nam JP, Nah JW, Jang MK, Yee ST. Immunoadjuvant Efficacy of N-Carboxymethyl Chitosan for Vaccination via Dendritic Cell Activation. J Med Food 2014; 17:268-77. [DOI: 10.1089/jmf.2013.2921] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jong-Jin Kim
- Department of Biology, Sunchon National University, Suncheon, Korea
| | - Joung-Pyo Nam
- Department of Polymer Science and Engineering, Sunchon National University, Suncheon, Korea
| | - Jae-Woon Nah
- Department of Polymer Science and Engineering, Sunchon National University, Suncheon, Korea
| | - Mi-Kyeong Jang
- Department of Polymer Science and Engineering, Sunchon National University, Suncheon, Korea
| | - Sung-Tae Yee
- Department of Biology, Sunchon National University, Suncheon, Korea
- Department of Pharmacy, Sunchon National University, Suncheon, Korea
| |
Collapse
|
33
|
Nierkens S, Tel J, Janssen E, Adema GJ. Antigen cross-presentation by dendritic cell subsets: one general or all sergeants? Trends Immunol 2013; 34:361-70. [PMID: 23540650 DOI: 10.1016/j.it.2013.02.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/17/2013] [Accepted: 02/21/2013] [Indexed: 12/27/2022]
Abstract
Antigen cross-presentation describes the process through which dendritic cells (DCs) acquire exogenous antigens for presentation on MHC class I molecules. The ability to cross-present has been thought of as a feature of specialized DC subsets. Emerging data, however, suggest that the cross-presenting ability of each DC subset is tuned by and dependent on several factors, such as DC location and activation status, and the type of antigen and inflammatory signals. Thus, we argue that capacity of cross-presentation is not an exclusive trait of one or several distinct DC subtypes, but rather a common feature of the DC family in both mice and humans. Understanding DC subset activation and antigen-presentation pathways might yield improved tools and targets to exploit the unique cross-presenting capacity of DCs in immunotherapy.
Collapse
Affiliation(s)
- Stefan Nierkens
- Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Tumor Immunology Laboratory, Geert Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|