1
|
Vidal-Manrique M, Nieuwenstein T, Hooijmaijers L, de Jonge P, Djojoatmo M, Jansen J, van der Waart A, Brock R, Dolstra H. IL-15 transpresentation by ovarian cancer cells improves CD34 + progenitor-derived NK cell's anti-tumor functionality. Oncoimmunology 2025; 14:2465010. [PMID: 39960378 PMCID: PMC11834524 DOI: 10.1080/2162402x.2025.2465010] [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: 10/24/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy. As high numbers of Natural Killer (NK) cells in ascites associate with improved survival, the adoptive transfer of allogeneic NK cells is an attractive therapeutic strategy. An approach to further improve NK cell expansion and anti-tumor functionality post-infusion includes IL-15 transpresentation (transIL-15), which involves surface expression of the IL-15 cytokine bound to IL-15Rα. However, others have substantiated that systemic administration of ALT/N-803, a soluble molecule mimicking transIL-15, leads to T cell-mediated rejection of the infused allogeneic NK cell product. In addition, whether transIL-15 induce superior expansion and functionality of our hematopoietic progenitor cell-derived NK cells (HPC-NK) remains understudied. Here, we propose to transfect OC cells with IL-15 and IL-15Rα mRNA and evaluate HPC-NK cell stimulation in vitro. Co-transfection of both mRNAs resulted in surface co-expression of both components, thus mimicking the transIL-15. Importantly, co-culture of HPC-NK cells with transIL-15 OC cells resulted in superior proliferation, IFNγ production, cytotoxicity and granzyme B secretion. Furthermore, we observed uptake of IL-15Rα by HPC-NK cells when co-cultured with transIL-15 OC cells, which associates with NK cell long-term proliferation and survival. Superior killing and granzyme B secretion were also observed in transIL-15 OC spheroids. Our results demonstrate that local delivery of IL-15 and IL-15Rα mRNA to OC tumors may be a safer strategy to boost HPC-NK cell therapy of OC through IL-15 transpresentation.
Collapse
Affiliation(s)
- M. Vidal-Manrique
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - T. Nieuwenstein
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - L. Hooijmaijers
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - P.K.J.D. de Jonge
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M. Djojoatmo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J. Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A.B. van der Waart
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - R. Brock
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - H. Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
2
|
Shariati A, Khani P, Nasri F, Afkhami H, Khezrpour A, Kamrani S, Shariati F, Alavimanesh S, Modarressi MH. mRNA cancer vaccines from bench to bedside: a new era in cancer immunotherapy. Biomark Res 2024; 12:157. [PMID: 39696625 DOI: 10.1186/s40364-024-00692-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/15/2024] [Indexed: 12/20/2024] Open
Abstract
Harnessing the power of the immune system to target cancer cells is one of the most appealing approaches for cancer therapy. Among these immunotherapies, messenger ribonucleic acid (mRNA) cancer vaccines are worthy of consideration, as they have demonstrated promising results in clinical trials. These vaccines have proven to be safe and well-tolerated. They can be easily mass-produced in a relatively short time and induce a systemic immune response effective against both the primary tumor and metastases. Transcripts encoding immunomodulatory molecules can also be incorporated into the mRNA, enhancing its efficacy. On the other hand, there are some challenges associated with their application, including mRNA instability, insufficient uptake by immune cells, and intrinsic immunogenicity, which can block mRNA translation. Many innovations have been suggested to overcome these obstacles, including structural modification (such as 5' cap modification), optimizing delivery vehicles (especially dendritic cells (DCs) and nanoparticles), and using antigens that can enhance immunogenicity by circumventing tolerance mechanisms. A popular approach is to combine mRNA cancer vaccines with traditional and novel cancer treatments like chemotherapy, radiotherapy, and immune checkpoint blockade (ICB). They are most efficacious when combined with other therapies like ICBs. There is still a long way to go before these vaccines enter the standard of care for cancer patients, but with the incredible pace of development in this field, their clinical application will soon be witnessed. This review highlights the recent advances and challenges of mRNA cancer vaccines. Finally, some of the most prominent clinical applications of these vaccines will be reviewed.
Collapse
Affiliation(s)
- Alireza Shariati
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Farzad Nasri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Arya Khezrpour
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Sina Kamrani
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Shariati
- Department of Genetics, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran.
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Mohammad Hossein Modarressi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
3
|
Han J, Wang H. Cytokine-overexpressing dendritic cells for cancer immunotherapy. Exp Mol Med 2024; 56:2559-2568. [PMID: 39617785 DOI: 10.1038/s12276-024-01353-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/11/2024] [Indexed: 12/28/2024] Open
Abstract
Dendritic cells (DCs), the main type of antigen-presenting cells in the body, act as key mediators of adaptive immunity by sampling antigens from diseased cells for the subsequent priming of antigen-specific T and B cells. While DCs can secrete a diverse array of cytokines that profoundly shape the immune milieu, exogenous cytokines are often needed to maintain the survival, proliferation, and differentiation of DCs, T cells, and B cells. However, conventional cytokine therapies for cancer treatment are limited by their low therapeutic benefit and severe side effects. The overexpression of cytokines in DCs, followed by paracrine release or membrane display, has emerged as a viable approach for controlling the exposure of cytokines to interacting DCs and T/B cells. This approach can potentially reduce the necessary dose of cytokines and associated side effects to achieve comparable or enhanced antitumor efficacy. Various strategies have been developed to enable the overexpression or chemical conjugation of cytokines on DCs for the subsequent modulation of DC-T/B-cell interactions. This review provides a brief overview of strategies that enable the overexpression of cytokines in or on DCs via genetic engineering or chemical modification methods and discusses the promise of cytokine-overexpressing DCs for the development of new-generation cancer immunotherapy.
Collapse
Affiliation(s)
- Joonsu Han
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois (CCIL), Urbana, IL, USA.
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
4
|
Chen P, Li S, Nagaoka K, Kakimi K, Kataoka K, Cabral H. Nanoenabled IL-15 Superagonist via Conditionally Stabilized Protein-Protein Interactions Eradicates Solid Tumors by Precise Immunomodulation. J Am Chem Soc 2024; 146:32431-32444. [PMID: 39356776 PMCID: PMC11613988 DOI: 10.1021/jacs.4c08327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Protein complexes are crucial structures that control many biological processes. Harnessing these structures could be valuable for therapeutic therapy. However, their instability and short lifespans need to be addressed for effective use. Here, we propose an innovative approach based on a functional polymeric cloak that coordinately anchors different domains of protein complexes and assembles them into a stabilized nanoformulation. As the polymer-protein association in the cloak is pH sensitive, the nanoformulation also allows targeting the release of the protein complexes to the acidic microenvironment of tumors for aiding their therapeutic performance. Building on this strategy, we developed an IL-15 nanosuperagonist (Nano-SA) by encapsulating the interleukin-15 (IL-15)/IL-15 Receptor α (IL-15Rα) complex (IL-15cx) for fostering synergistic transpresentation in tumors. Upon intravenous administration, Nano-SA stably circulated in the bloodstream, safeguarding the integrity of IL-15cx until reaching the tumor site, where it selectively released the active complex. Thus, Nano-SA significantly amplified the antitumor immune signals while diminishing systemic off-target effects. In murine colon cancer models, Nano-SA achieved potent immunotherapeutic effects, eradicating tumors without adverse side effects. These findings highlight the transformative potential of nanotechnology for advancing protein complex-based therapies.
Collapse
Affiliation(s)
- Pengwen Chen
- Department
of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shangwei Li
- Department
of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Koji Nagaoka
- Department
of Immunotherapeutics, The University of
Tokyo Hospital, 7-3-1
Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazuhiro Kakimi
- Department
of Immunotherapeutics, The University of
Tokyo Hospital, 7-3-1
Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazunori Kataoka
- Innovation
Center of NanoMedicine (iCONM), Kawasaki
Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Horacio Cabral
- Department
of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
5
|
Baharom F, Hermans D, Delamarre L, Seder RA. Vax-Innate: improving therapeutic cancer vaccines by modulating T cells and the tumour microenvironment. Nat Rev Immunol 2024:10.1038/s41577-024-01091-9. [PMID: 39433884 DOI: 10.1038/s41577-024-01091-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 10/23/2024]
Abstract
T cells have a critical role in mediating antitumour immunity. The success of immune checkpoint inhibitors (ICIs) for cancer treatment highlights how enhancing endogenous T cell responses can mediate tumour regression. However, mortality remains high for many cancers, especially in the metastatic setting. Based on advances in the genetic characterization of tumours and identification of tumour-specific antigens, individualized therapeutic cancer vaccines targeting mutated tumour antigens (neoantigens) are being developed to generate tumour-specific T cells for improved therapeutic responses. Early clinical trials using individualized neoantigen vaccines for patients with advanced disease had limited clinical efficacy despite demonstrated induction of T cell responses. Therefore, enhancing T cell activity by improving the magnitude, quality and breadth of T cell responses following vaccination is one current goal for improving outcome against metastatic tumours. Another major consideration is how T cells can be further optimized to function within the tumour microenvironment (TME). In this Perspective, we focus on neoantigen vaccines and propose a new approach, termed Vax-Innate, in which vaccination through intravenous delivery or in combination with tumour-targeting immune modulators may improve antitumour efficacy by simultaneously increasing the magnitude, quality and breadth of T cells while transforming the TME into a largely immunostimulatory environment for T cells.
Collapse
Affiliation(s)
| | - Dalton Hermans
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
| | | | - Robert A Seder
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Eslami SM, Lu X. Recent advances in mRNA-based cancer vaccines encoding immunostimulants and their delivery strategies. J Control Release 2024; 376:S0168-3659(24)00708-9. [PMID: 39437963 DOI: 10.1016/j.jconrel.2024.10.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/01/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
The high prevalence of drug resistance, relapse, and unfavorable response rate of conventional cancer therapies necessitate the development of more efficient treatment modalities. Immunotherapy represents a novel therapeutic approach to cancer treatment in which the immune system's potential is harnessed to recognize and eliminate tumor cells. mRNA cancer vaccines, as a burgeoning field of immunotherapy, have recently drawn particular attention, and among mRNAs encoding tumor-associated antigens, tumor-specific antigens, and immune stimulatory factors, the latter has been relatively less explored. These immunostimulatory mRNAs encode a range of proteins, including stimulatory ligands, receptors, enzymes, pro-inflammatory cytokines, and inhibitory binding proteins, which collectively augment the host immune system's ability against cancerous cells. In this review, we aimed to provide a comprehensive account of mRNA-based cancer vaccines encoding immune stimulants, encompassing their current status, mechanisms of action, delivery strategies employed, as well as recent advances in preclinical and clinical studies. The potential challenges, strategies and future perspectives have also been discussed.
Collapse
Affiliation(s)
- Seyyed Majid Eslami
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Storrs, CT 06269, USA.
| | - Xiuling Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Storrs, CT 06269, USA.
| |
Collapse
|
7
|
Bento R, Scheller J, Parekkadan B. Intratumoral Delivery of Genetically Engineered Anti-IL-6 Trans-signaling Therapeutics. Mol Biotechnol 2024:10.1007/s12033-024-01230-6. [PMID: 38980514 DOI: 10.1007/s12033-024-01230-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024]
Abstract
Interleukin-6 (IL-6) is a highly pro-inflammatory cytokine involved in the etiopathology of several inflammatory diseases and cancer. As so, the inhibition of IL-6 signaling pathways has emerged as an attractive therapeutic avenue for the treatment of several chronic diseases. Since IL-6 trans-signaling was described as the pathological branch of IL-6, selective inhibitors were developed. Next-generation variants with increased trans-signaling specificity and potency emerged as great candidates for the treatment of several diseases, with reduced off-target effects. The highly time-consuming and costly processes involving recombinant protein production, however, have hampered the progress of anti-cytokine pharmaceuticals in clinic so far. Herein, we developed gene therapeutic modalities of IL-6-trans-signaling inhibitors as alternatives for sustained recombinant protein secretion. By using an IL-6-dependent lymphoma cell line and xenograft tumor model, we demonstrated the superior inhibitory potential of second-generation anti-IL-6 trans-signaling therapeutic. We compared the efficiency of distinct gene delivery modalities using a bioluminescent biomarker probe and observed consistent protein production via cell-based delivery. When delivered intratumorally, genetically engineered sgp130FlyRFc-secreting cells significantly reduced tumor burden and increased animal survival, representing a promising therapeutic avenue to be explored in clinically relevant gene delivery applications.
Collapse
Affiliation(s)
- Raphaela Bento
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
8
|
Balar PC, Apostolopoulos V, Chavda VP. A new era of immune therapeutics for pancreatic cancer: Monoclonal antibodies paving the way. Eur J Pharmacol 2024; 969:176451. [PMID: 38408598 DOI: 10.1016/j.ejphar.2024.176451] [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: 11/19/2023] [Revised: 02/06/2024] [Accepted: 02/20/2024] [Indexed: 02/28/2024]
Abstract
Pancreatic cancer, particularly pancreatic ductal adenocarcinoma, remains a devastating disease with a dismal prognosis and limited survival rates. Despite various drug treatments and regimens showing promise in managing the disease, the clinical outcomes have not significantly improved. Immunotherapy however, has become a forefront area in pancreatic cancer treatment. This approach comprises a range of agents, including small molecule drugs, antibodies, combination therapies, and vaccines. In the last 5-8 years, there has been an upsurge of research into the use of monoclonal antibodies to block receptors on cancer or immune cells, revolutionising cancer treatment and management. Several targets have been identified and studied, with the most encouraging noted in relation to checkpoint markers, namely, antibodies targeting anti-programmed cell death 1 (PD-1) and its receptor PD-L1. Herein, we present the clinical developments in immunotherapy in the last 5 years especially those which have been tested in humans against pancreatic cancer.
Collapse
Affiliation(s)
- Pankti C Balar
- Pharmacy Section, L.M. College of Pharmacy, Ahmedabad, India
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Werribee Campus, Melbourne, VIC, 3030, Australia
| | - Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, India.
| |
Collapse
|
9
|
Haldar SD, Vilar E, Maitra A, Zaidi N. Worth a Pound of Cure? Emerging Strategies and Challenges in Cancer Immunoprevention. Cancer Prev Res (Phila) 2023; 16:483-495. [PMID: 37001882 PMCID: PMC10548442 DOI: 10.1158/1940-6207.capr-22-0478] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/06/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
Cancer immunoprevention applies immunologic approaches such as vaccines to prevent, rather than to treat or cure, cancer. Despite limited success in the treatment of advanced disease, the development of cancer vaccines to intercept premalignant states is a promising area of current research. These efforts are supported by the rationale that vaccination in the premalignant setting is less susceptible to mechanisms of immune evasion compared with established cancer. Prophylactic vaccines have already been developed for a minority of cancers mediated by oncogenic viruses (e.g., hepatitis B and human papillomavirus). Extending the use of preventive vaccines to non-virally driven malignancies remains an unmet need to address the rising global burden of cancer. This review provides a broad overview of clinical trials in cancer immunoprevention with an emphasis on emerging vaccine targets and delivery platforms, translational challenges, and future directions.
Collapse
Affiliation(s)
- Saurav D. Haldar
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anirban Maitra
- Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neeha Zaidi
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
10
|
Lee KW, Yam JWP, Mao X. Dendritic Cell Vaccines: A Shift from Conventional Approach to New Generations. Cells 2023; 12:2147. [PMID: 37681880 PMCID: PMC10486560 DOI: 10.3390/cells12172147] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
In the emerging era of cancer immunotherapy, immune checkpoint blockades (ICBs) and adoptive cell transfer therapies (ACTs) have gained significant attention. However, their therapeutic efficacies are limited due to the presence of cold type tumors, immunosuppressive tumor microenvironment, and immune-related side effects. On the other hand, dendritic cell (DC)-based vaccines have been suggested as a new cancer immunotherapy regimen that can address the limitations encountered by ICBs and ACTs. Despite the success of the first generation of DC-based vaccines, represented by the first FDA-approved DC-based therapeutic cancer vaccine Provenge, several challenges remain unsolved. Therefore, new DC vaccine strategies have been actively investigated. This review addresses the limitations of the currently most adopted classical DC vaccine and evaluates new generations of DC vaccines in detail, including biomaterial-based, immunogenic cell death-inducing, mRNA-pulsed, DC small extracellular vesicle (sEV)-based, and tumor sEV-based DC vaccines. These innovative DC vaccines are envisioned to provide a significant breakthrough in cancer immunotherapy landscape and are expected to be supported by further preclinical and clinical studies.
Collapse
Affiliation(s)
- Kyu-Won Lee
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; (K.-W.L.); (J.W.P.Y.)
| | - Judy Wai Ping Yam
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; (K.-W.L.); (J.W.P.Y.)
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| |
Collapse
|
11
|
Trinkner P, Günther S, Monsef I, Kerschbaum E, von Bergwelt-Baildon M, Cordas Dos Santos DM, Theurich S. Survival and immunotoxicities in association with sex-specific body composition patterns of cancer patients undergoing immune-checkpoint inhibitor therapy - A systematic review and meta-analysis. Eur J Cancer 2023; 184:151-171. [PMID: 36931074 DOI: 10.1016/j.ejca.2023.01.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/19/2023]
Abstract
BACKGROUND Imbalanced body composition is mechanistically connected to dysregulated immune activities. Whether overweight/obesity or sarcopenia has an impact on treatment results in cancer patients undergoing immune checkpoint inhibitor (ICI) therapy is currently under debate. We aimed to answer if survival rates and occurrence of immune-related adverse events (irAEs) were different in obese or sarcopenic patients. METHODS A systematic search was conducted in PubMed, Embase and CENTRAL for all records published until July 2022 using specific search terms for body composition in combination with terms for ICI regimens. Two authors screened independently. All studies that reported on body mass index or sarcopenia measures were selected for further analysis. RESULTS 48 studies reporting on overweight/obesity comprising of 19,767 patients, and 32 studies reporting on sarcopenia comprising of 3193 patients fulfilled the inclusion criteria. In the entire cohort, overweight/obesity was significantly associated with better progression-free survival (PFS; p = 0.009) and overall survival (OS; p <0.00001). Subgroup analyses stratified by sex revealed that overweight/obese males had the strongest survival benefit (PFS: p = 0.05; OS: p = 0.0005), and overweight/obese female patients did not show any. However, overweight/obese patients of both sexes had a higher risk to develop irAEs grade ≥3 (p = 0.0009). Sarcopenic patients showed significantly shorter PFS (p <0.0001) and OS (p <0.0001). The frequency of irAEs did not differ between sarcopenic and non-sarcopenic patients. CONCLUSION This meta-analysis suggests that body composition is associated in a sex-specific manner with survival and irAEs in cancer patients undergoing ICI treatment.
Collapse
Affiliation(s)
- Paul Trinkner
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; Cancer- and Immunometabolism Research Group, Gene Center, LMU Munich, Munich, Germany
| | - Sophie Günther
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; Cancer- and Immunometabolism Research Group, Gene Center, LMU Munich, Munich, Germany
| | - Ina Monsef
- Evidence-based Medicine, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Eva Kerschbaum
- Comprehensive Cancer Center Munich (CCCM), Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; Comprehensive Cancer Center Munich (CCCM), Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David M Cordas Dos Santos
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; Cancer- and Immunometabolism Research Group, Gene Center, LMU Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Theurich
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; Cancer- and Immunometabolism Research Group, Gene Center, LMU Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
12
|
Deckers J, Anbergen T, Hokke AM, de Dreu A, Schrijver DP, de Bruin K, Toner YC, Beldman TJ, Spangler JB, de Greef TFA, Grisoni F, van der Meel R, Joosten LAB, Merkx M, Netea MG, Mulder WJM. Engineering cytokine therapeutics. NATURE REVIEWS BIOENGINEERING 2023; 1:286-303. [PMID: 37064653 PMCID: PMC9933837 DOI: 10.1038/s44222-023-00030-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Cytokines have pivotal roles in immunity, making them attractive as therapeutics for a variety of immune-related disorders. However, the widespread clinical use of cytokines has been limited by their short blood half-lives and severe side effects caused by low specificity and unfavourable biodistribution. Innovations in bioengineering have aided in advancing our knowledge of cytokine biology and yielded new technologies for cytokine engineering. In this Review, we discuss how the development of bioanalytical methods, such as sequencing and high-resolution imaging combined with genetic techniques, have facilitated a better understanding of cytokine biology. We then present an overview of therapeutics arising from cytokine re-engineering, targeting and delivery, mRNA therapeutics and cell therapy. We also highlight the application of these strategies to adjust the immunological imbalance in different immune-mediated disorders, including cancer, infection and autoimmune diseases. Finally, we look ahead to the hurdles that must be overcome before cytokine therapeutics can live up to their full potential.
Collapse
Affiliation(s)
- Jeroen Deckers
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
| | - Tom Anbergen
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
| | - Ayla M. Hokke
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Anne de Dreu
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - David P. Schrijver
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Koen de Bruin
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Yohana C. Toner
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
| | - Thijs J. Beldman
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
| | - Jamie B. Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Tom F. A. de Greef
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
- Centre for Living Technologies, Alliance Eindhoven University of Technology, Wageningen University & Research, Utrecht University and University Medical Center Utrecht (EWUU), Utrecht, Netherlands
| | - Francesca Grisoni
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- Centre for Living Technologies, Alliance Eindhoven University of Technology, Wageningen University & Research, Utrecht University and University Medical Center Utrecht (EWUU), Utrecht, Netherlands
| | - Roy van der Meel
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Present Address: Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Leo A. B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
- Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, Netherlands
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Maarten Merkx
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Present Address: Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
- Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, Netherlands
- Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Willem J. M. Mulder
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Medical Centre, Nijmegen, Netherlands
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Present Address: Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
13
|
Qureischi M, Mohr J, Arellano-Viera E, Knudsen SE, Vohidov F, Garitano-Trojaola A. mRNA-based therapies: Preclinical and clinical applications. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 372:1-54. [PMID: 36064262 DOI: 10.1016/bs.ircmb.2022.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
At the fundamental level, messenger RNA (mRNA)-based therapeutics involves the delivery of in vitro-transcribed (IVT) mRNA into the cytoplasm of a target cell, where it is translated into the desired protein. IVT mRNA presents various advantages compared to DNA and recombinant protein-based approaches that make it ideal for a broad range of therapeutic applications. IVT mRNA, which is translated in the cytoplasm after transfection into cells, can encode virtually any target protein. Notably, it does not enter the nucleus, which avoids its integration into the genome and the risk of insertional mutagenesis. The large-scale production of IVT mRNA is less complex than production of recombinant proteins, and Good Manufacturing Practice-compliant mRNA production is easily scalable, ideally poising mRNA for not only off-the-shelf, but more personalized treatment approaches. IVT mRNA's safety profile, pharmacokinetics, and pharmacodynamics, including its inherent immunostimulatory capacity, can be optimized for different therapeutic applications by harnessing a wide array of optimized sequence elements, chemical modifications, purification techniques, and delivery methods. The value of IVT mRNA was recently proved during the COVID-19 pandemic when mRNA-based vaccines outperformed the efficacy of established technologies, and millions of doses were rapidly deployed. In this review, we will discuss chemical modifications of IVT mRNA and highlight numerous preclinical and clinical applications including vaccines for cancer and infectious diseases, cancer immunotherapy, protein replacement, gene editing, and cell reprogramming.
Collapse
|
14
|
Huff AL, Jaffee EM, Zaidi N. Messenger RNA vaccines for cancer immunotherapy: progress promotes promise. J Clin Invest 2022; 132:e156211. [PMID: 35289317 PMCID: PMC8920340 DOI: 10.1172/jci156211] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The COVID-19 pandemic has elevated mRNA vaccines to global recognition due to their unprecedented success rate in protecting against a deadly virus. This international success is underscored by the remarkable versatility, favorable immunogenicity, and overall safety of the mRNA platform in diverse populations. Although mRNA vaccines have been studied in preclinical models and patients with cancer for almost three decades, development has been slow. The recent technological advances responsible for the COVID-19 vaccines have potential implications for successfully adapting this vaccine platform for cancer therapeutics. Here we discuss the lessons learned along with the chemical, biologic, and immunologic adaptations needed to optimize mRNA technology to successfully treat cancers.
Collapse
Affiliation(s)
- Amanda L. Huff
- Department of Oncology
- The Sidney Kimmel Comprehensive Cancer Center
- The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, and
- The Cancer Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth M. Jaffee
- Department of Oncology
- The Sidney Kimmel Comprehensive Cancer Center
- The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, and
- The Cancer Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Neeha Zaidi
- Department of Oncology
- The Sidney Kimmel Comprehensive Cancer Center
- The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, and
- The Cancer Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Bosch NC, Martin LM, Voskens CJ, Berking C, Seliger B, Schuler G, Schaft N, Dörrie J. A Chimeric IL-15/IL-15Rα Molecule Expressed on NFκB-Activated Dendritic Cells Supports Their Capability to Activate Natural Killer Cells. Int J Mol Sci 2021; 22:ijms221910227. [PMID: 34638566 PMCID: PMC8508776 DOI: 10.3390/ijms221910227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/02/2021] [Accepted: 09/18/2021] [Indexed: 12/31/2022] Open
Abstract
Natural killer (NK) cells, members of the innate immune system, play an important role in the rejection of HLA class I negative tumor cells. Hence, a therapeutic vaccine, which can activate NK cells in addition to cells of the adaptive immune system might induce a more comprehensive cellular response, which could lead to increased tumor elimination. Dendritic cells (DCs) are capable of activating and expanding NK cells, especially when the NFκB pathway is activated in the DCs thereby leading to the secretion of the cytokine IL-12. Another prominent NK cell activator is IL-15, which can be bound by the IL-15 receptor alpha-chain (IL-15Rα) to be transpresented to the NK cells. However, monocyte-derived DCs do neither secrete IL-15, nor express the IL-15Rα. Hence, we designed a chimeric protein consisting of IL-15 and the IL-15Rα. Upon mRNA electroporation, the fusion protein was detectable on the surface of the DCs, and increased the potential of NFκB-activated, IL-12-producing DC to activate NK cells in an autologous cell culture system with ex vivo-generated cells from healthy donors. These data show that a chimeric IL-15/IL-15Rα molecule can be expressed by monocyte-derived DCs, is trafficked to the cell surface, and is functional regarding the activation of NK cells. These data represent an initial proof-of-concept for an additional possibility of further improving cellular DC-based immunotherapies of cancer.
Collapse
Affiliation(s)
- Naomi C. Bosch
- Institute of Medical Immunology, Martin-Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (N.C.B.); (B.S.)
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
- Comprehensive Cancer Center Erlangen–EMN, NCT WERA, 91054 Erlangen, Germany
| | - Lena-Marie Martin
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
| | - Caroline J. Voskens
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
- Comprehensive Cancer Center Erlangen–EMN, NCT WERA, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
- Comprehensive Cancer Center Erlangen–EMN, NCT WERA, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin-Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (N.C.B.); (B.S.)
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), 04103 Leipzig, Germany
| | - Gerold Schuler
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
- Correspondence: ; Tel.: +49-9131-8531127
| |
Collapse
|
16
|
Castleman MJ, Dillon SM, Thompson TA, Santiago ML, McCarter MD, Barker E, Wilson CC. Gut Bacteria Induce Granzyme B Expression in Human Colonic ILC3s In Vitro in an IL-15-Dependent Manner. THE JOURNAL OF IMMUNOLOGY 2021; 206:3043-3052. [PMID: 34117105 DOI: 10.4049/jimmunol.2000239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/13/2021] [Indexed: 12/13/2022]
Abstract
Group 3 innate lymphoid cells (ILC3s) in the gut mucosa have long been thought to be noncytotoxic lymphocytes that are critical for homeostasis of intestinal epithelial cells through secretion of IL-22. Recent work using human tonsillar cells demonstrated that ILC3s exposed to exogenous inflammatory cytokines for a long period of time acquired expression of granzyme B, suggesting that under pathological conditions ILC3s may become cytotoxic. We hypothesized that inflammation associated with bacterial exposure might trigger granzyme B expression in gut ILC3s. To test this, we exposed human colon lamina propria mononuclear cells to a panel of enteric bacteria. We found that the Gram-negative commensal and pathogenic bacteria induced granzyme B expression in a subset of ILC3s that were distinct from IL-22-producing ILC3s. A fraction of granzyme B+ ILC3s coexpressed the cytolytic protein perforin. Granzyme B expression was mediated, in part, by IL-15 produced upon exposure to bacteria. ILC3s coexpressing all three IL-15R subunits (IL15Rα/β/γ) increased following bacterial stimulation, potentially allowing for cis presentation of IL-15 during bacterial exposure. Additionally, a large frequency of colonic myeloid dendritic cells expressed IL-15Rα, implicating myeloid dendritic cells in trans presentation of IL-15 to ILC3s. Tonsillar ILC3s minimally expressed granzyme B when exposed to the same bacteria or to rIL-15. Overall, these data establish the novel, to our knowledge, finding that human colonic ILC3s can express granzyme B in response to a subset of enteric bacteria through a process mediated by IL-15. These observations raise new questions about the multifunctional role of human gut ILC3s.
Collapse
Affiliation(s)
- Moriah J Castleman
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Stephanie M Dillon
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Tezha A Thompson
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Mario L Santiago
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Martin D McCarter
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO; and
| | - Edward Barker
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL
| | - Cara C Wilson
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO;
| |
Collapse
|
17
|
Jacobs B, Gebel V, Heger L, Grèze V, Schild H, Dudziak D, Ullrich E. Characterization and Manipulation of the Crosstalk Between Dendritic and Natural Killer Cells Within the Tumor Microenvironment. Front Immunol 2021; 12:670540. [PMID: 34054844 PMCID: PMC8160470 DOI: 10.3389/fimmu.2021.670540] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 04/19/2021] [Indexed: 01/22/2023] Open
Abstract
Cellular therapy has entered the daily clinical life with the approval of CAR T cell therapeutics and dendritic cell (DCs) vaccines in the US and the EU. In addition, numerous other adoptive cellular products, including natural killer (NK) cells, are currently evaluated in early phase I/ II clinical trials for the treatment of cancer patients. Despite these promising accomplishments, various challenges remain to be mastered in order to ensure sustained therapeutic success. These include the identification of strategies by which tumor cells escape the immune system or establish an immunosuppressive tumor microenvironment (TME). As part of the innate immune system, DCs and NK cells are both present within the TME of various tumor entities. While NK cells are well known for their intrinsic anti-tumor activity by their cytotoxicity capacities and the secretion of pro-inflammatory cytokines, the role of DCs within the TME is a double-edged sword as different DC subsets have been described with either tumor-promoting or -inhibiting characteristics. In this review, we will discuss recent findings on the interaction of DCs and NK cells under physiological conditions and within the TME. One focus is the crosstalk of various DC subsets with NK cells and their impact on the progression or inhibition of tumor growth. In addition, we will provide suggestions to overcome the immunosuppressive outcome of the interaction of DCs and NK cells within the TME.
Collapse
Affiliation(s)
- Benedikt Jacobs
- Department of Internal Medicine 5, Haematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Veronika Gebel
- Children's Hospital, Goethe-University Frankfurt, Frankfurt, Germany.,Experimental Immunology, Goethe University Frankfurt , Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - Lukas Heger
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Victoria Grèze
- Children's Hospital, Goethe-University Frankfurt, Frankfurt, Germany.,Experimental Immunology, Goethe University Frankfurt , Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - Hansjörg Schild
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany.,Research Centre for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Evelyn Ullrich
- Children's Hospital, Goethe-University Frankfurt, Frankfurt, Germany.,Experimental Immunology, Goethe University Frankfurt , Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| |
Collapse
|
18
|
Wu M, Wang S, Chen JY, Zhou LJ, Guo ZW, Li YH. Therapeutic cancer vaccine therapy for acute myeloid leukemia. Immunotherapy 2021; 13:863-877. [PMID: 33955237 DOI: 10.2217/imt-2020-0277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Antitumor function of the immune system has been harnessed to eradicate tumor cells as cancer therapy. Therapeutic cancer vaccines aim to help immune cells recognize tumor cells, which are difficult to target owing to immune escape. Many attempts at vaccine designs have been conducted throughout the last decades. In addition, as the advanced understanding of immunosuppressive mechanisms mediated by tumor cells, combining cancer vaccines with other immune therapies seems to be more efficient for cancer treatment. Acute myeloid leukemia (AML) is the most common acute leukemia in adults with poor prognosis. Evidence has shown T-cell-mediated immune responses in AML, which encourages the utility of immune therapies in AML. This review discusses cancer vaccines in AML from vaccine design as well as recent progress in vaccination combination with other immune therapies.
Collapse
Affiliation(s)
- Ming Wu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.,Department of Hematology, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Sheng Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jian-Yu Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Li-Juan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Zi-Wen Guo
- Department of Hematology, Zhongshan People's Hospital, Zhongshan 528400, China
| | - Yu-Hua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
19
|
Beck JD, Reidenbach D, Salomon N, Sahin U, Türeci Ö, Vormehr M, Kranz LM. mRNA therapeutics in cancer immunotherapy. Mol Cancer 2021; 20:69. [PMID: 33858437 PMCID: PMC8047518 DOI: 10.1186/s12943-021-01348-0] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/15/2021] [Indexed: 02/08/2023] Open
Abstract
Synthetic mRNA provides a template for the synthesis of any given protein, protein fragment or peptide and lends itself to a broad range of pharmaceutical applications, including different modalities of cancer immunotherapy. With the ease of rapid, large scale Good Manufacturing Practice-grade mRNA production, mRNA is ideally poised not only for off-the shelf cancer vaccines but also for personalized neoantigen vaccination. The ability to stimulate pattern recognition receptors and thus an anti-viral type of innate immune response equips mRNA-based vaccines with inherent adjuvanticity. Nucleoside modification and elimination of double-stranded RNA can reduce the immunomodulatory activity of mRNA and increase and prolong protein production. In combination with nanoparticle-based formulations that increase transfection efficiency and facilitate lymphatic system targeting, nucleoside-modified mRNA enables efficient delivery of cytokines, costimulatory receptors, or therapeutic antibodies. Steady but transient production of the encoded bioactive molecule from the mRNA template can improve the pharmacokinetic, pharmacodynamic and safety properties as compared to the respective recombinant proteins. This may be harnessed for applications that benefit from a higher level of expression control, such as chimeric antigen receptor (CAR)-modified adoptive T-cell therapies. This review highlights the advancements in the field of mRNA-based cancer therapeutics, providing insights into key preclinical developments and the evolving clinical landscape.
Collapse
Affiliation(s)
- Jan D Beck
- BioNTech SE, An der Goldgrube 12, 55131, Mainz, Germany
| | - Daniel Reidenbach
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University gGmbH, Freiligrathstraße 12, 55131, Mainz, Germany
| | - Nadja Salomon
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University gGmbH, Freiligrathstraße 12, 55131, Mainz, Germany
| | - Ugur Sahin
- BioNTech SE, An der Goldgrube 12, 55131, Mainz, Germany
| | - Özlem Türeci
- BioNTech SE, An der Goldgrube 12, 55131, Mainz, Germany
| | | | - Lena M Kranz
- BioNTech SE, An der Goldgrube 12, 55131, Mainz, Germany.
| |
Collapse
|
20
|
Giehl E, Kosaka H, Liu Z, Feist M, Kammula US, Lotze MT, Ma C, Guo ZS, Bartlett DL. In Vivo Priming of Peritoneal Tumor-Reactive Lymphocytes With a Potent Oncolytic Virus for Adoptive Cell Therapy. Front Immunol 2021; 12:610042. [PMID: 33679747 PMCID: PMC7930493 DOI: 10.3389/fimmu.2021.610042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/04/2021] [Indexed: 01/07/2023] Open
Abstract
Adoptive cell therapy (ACT) using autologous tumor infiltrating lymphocytes (TIL) achieves durable clinical benefit for patients from whom these cells can be derived in advanced metastatic melanoma but is limited in most solid tumors as a result of immune escape and exclusion. A tumor microenvironment (TME) priming strategy to improve the quantity and quality of TIL represents an important tactic to explore. Oncolytic viruses expressing immune stimulatory cytokines induce a potent inflammatory response that may enhance infiltration and activation of T cells. In this study, we examined the ability of an attenuated oncolytic vaccinia virus expressing IL15/IL15Rα (vvDD-IL15/Rα) to enhance recovery of lavage T cells in peritoneal carcinomatosis (PC). We found that intraperitoneal (IP) vvDD-IL15/Rα treatment of animals bearing PC resulted in a significant increase in cytotoxic function and memory formation in CD8+ T cells in peritoneal fluid. Using tetramers for vaccinia virus B8R antigen and tumor rejection antigen p15E, we found that the expanded population of peritoneal CD8+ T cells are specific for vaccinia or tumor with increased tumor-specificity over time, reinforced with viral clearance. Application of these vvDD-IL15/Rα induced CD8+ T cells in ACT of a lethal model of PC significantly increased survival. In addition, we found in patients with peritoneal metastases from various primary solid tumors that peritoneal T cells could be recovered but were exhausted with infrequent tumor-reactivity. If clinically translatable, vvDD-IL15/Rα in vivo priming would greatly expand the number of patients with advanced metastatic cancers responsive to T cell therapy.
Collapse
Affiliation(s)
- Esther Giehl
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Hiromichi Kosaka
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Oncology Research Laboratories Oncology R&D Unit, Kyowa Kirin Co., Ltd., Shizuoka, Japan
| | - Zuqiang Liu
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Mathilde Feist
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Department of Surgery, CCM/CVK, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Udai S. Kammula
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Michael T. Lotze
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Congrong Ma
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Zong Sheng Guo
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - David L. Bartlett
- Departments of Surgery, University of Pittsburgh School of Medicine, and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| |
Collapse
|
21
|
Khaddour K, Gomez-Perez SL, Jain N, Patel JD, Boumber Y. Obesity, Sarcopenia, and Outcomes in Non-Small Cell Lung Cancer Patients Treated With Immune Checkpoint Inhibitors and Tyrosine Kinase Inhibitors. Front Oncol 2020; 10:576314. [PMID: 33194687 PMCID: PMC7607047 DOI: 10.3389/fonc.2020.576314] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Body composition refers to the proportional content of body fat mass and lean body mass that can lead to a continuum of different phenotypes ranging from cachectic/sarcopenic state to obesity. The heterogenetic phenotypes of body composition can contribute to formation of some cancer types and can sometimes lead to disparate outcomes. Both of these extremes of the spectrum exist in patients with non-small cell lung carcinoma (NSCLC). The discovery of new pathways that drive tumorigenesis contributing to cancer progression and resistance have expanded our understanding of cancer biology leading to development of new targeted therapies including tyrosine kinase inhibitors (TKI) and immune checkpoint inhibitors (ICI) that have changed the landscape of NSCLC treatment. However, in the new era of precision medicine, the impact of body composition phenotypes on treatment outcomes and survival is now being elucidated. In this review, we will discuss the emerging evidence of a link between body composition and outcomes in patients with NSCLC treated with TKI and ICI. We will also discuss suggested mechanisms by which body composition can impact tumor behavior and anti-tumor immunological response.
Collapse
Affiliation(s)
- Karam Khaddour
- Department of Medicine, Rosalind Franklin University of Medicine and Science, McHenry, IL, United States.,Department of Medicine, Division of Hematology and Oncology, University of Illinois at Chicago, Chicago, IL, United States
| | - Sandra L Gomez-Perez
- Department of Clinical Nutrition, Rush University Medical Center, Chicago, IL, United States
| | - Nikita Jain
- Department of Medicine, Rosalind Franklin University of Medicine and Science, McHenry, IL, United States
| | - Jyoti D Patel
- Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, United States
| | - Yanis Boumber
- Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, United States.,Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
22
|
Ahmed R, Crespo I, Tuyaerts S, Bekkar A, Graciotti M, Xenarios I, Kandalaft LE. Predicting combinations of immunomodulators to enhance dendritic cell-based vaccination based on a hybrid experimental and computational platform. Comput Struct Biotechnol J 2020; 18:2217-2227. [PMID: 32952936 PMCID: PMC7475195 DOI: 10.1016/j.csbj.2020.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/28/2020] [Accepted: 08/01/2020] [Indexed: 12/24/2022] Open
Abstract
Dendritic cell (DC)-based vaccines have been largely used in the adjuvant setting for the treatment of cancer, however, despite their proven safety, clinical outcomes still remain modest. In order to improve their efficacy, DC-based vaccines are often combined with one or multiple immunomodulatory agents. However, the selection of the most promising combinations is hampered by the plethora of agents available and the unknown interplay between these different agents. To address this point, we developed a hybrid experimental and computational platform to predict the effects and immunogenicity of dual combinations of stimuli once combined with DC vaccination, based on the experimental data of a variety of assays to monitor different aspects of the immune response after a single stimulus. To assess the stimuli behavior when used as single agents, we first developed an in vitro co-culture system of T cell priming using monocyte-derived DCs loaded with whole tumor lysate to prime autologous peripheral blood mononuclear cells in the presence of the chosen stimuli, as single adjuvants, and characterized the elicited response assessing 18 different phenotypic and functional traits important for an efficient anti-cancer response. We then developed and applied a prediction algorithm, generating a ranking for all possible dual combinations of the different single stimuli considered here. The ranking generated by the prediction tool was then validated with experimental data showing a strong correlation with the predicted scores, confirming that the top ranked conditions globally significantly outperformed the worst conditions. Thus, the method developed here constitutes an innovative tool for the selection of the best immunomodulatory agents to implement in future DC-based vaccines.
Collapse
Affiliation(s)
- Rita Ahmed
- Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Isaac Crespo
- Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
- Vital-IT group, SIB Swiss Institute of Bioinformatics, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Sandra Tuyaerts
- Department of Oncology, Leuven Cancer Institute (LKI), University of Leuven (KU Leuven), Leuven, Belgium
| | - Amel Bekkar
- Center for Integrative Genomics, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Michele Graciotti
- Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Ioannis Xenarios
- Center for Integrative Genomics, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Lana E. Kandalaft
- Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
23
|
Ahmed R, Sayegh N, Graciotti M, Kandalaft LE. Electroporation as a method of choice to generate genetically modified dendritic cell cancer vaccines. Curr Opin Biotechnol 2020; 65:142-155. [PMID: 32240923 DOI: 10.1016/j.copbio.2020.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/10/2019] [Accepted: 02/17/2020] [Indexed: 12/31/2022]
Abstract
In the last few decades, immunotherapy has emerged as an alternative therapeutic approach to treat cancer. Immunotherapy offers a plethora of different treatment possibilities. Among these, dendritic cell (DC)-based cancer vaccines constitute one of the most promising and valuable therapeutic options. DC-vaccines have been introduced into the clinics more than 15 years ago, and preclinical studies showed their general safety and low toxic effects on patients. However, their treatment efficacy is still rather limited, demanding for novel avenues to improve vaccine efficacy. One way to potentially achieve this is to focus on improving the DC-T cell interaction to further increase T cell priming and downstream activity. A successful DC-T cell interaction requires three different signals (Figure 1): (1) Major Histocompatibility Complex (MHC) and antigen complex interaction with T cell receptor (TCR) (2) interaction between co-stimulatory molecules and their cognate ligands at the cell surface and (3) secretion of cytokines to polarize the immune response toward a Type 1 helper (Th1) phenotype. In recent years, many studies attempted to improve the DC-T cell interaction and overall cancer vaccine therapeutic outcomes by increasing the expression of mediators of signal 1, 2 and/or 3, through genetic modifications of DCs. Transfection of genes of interest can be achieved through many different methods such as passive pulsing, lipofection, viral transfection, or electroporation (EP). However, EP is currently emerging as the method of choice thanks to its safety, versatility, and relatively easy clinical translation. In this review we will highlight the potential benefits of EP over other transfection methods as well as giving an overview of the available studies employing EP to gene-modify DCs in cancer vaccines. Crucial aspects such as safety, feasibility, and gene(s) of choice will be also discussed, together with future perspectives and opportunities for DC genetic engineering.
Collapse
Affiliation(s)
- Rita Ahmed
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland
| | - Naya Sayegh
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland
| | - Michele Graciotti
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland
| | - Lana E Kandalaft
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
| |
Collapse
|
24
|
Ribonucleic Acid Engineering of Dendritic Cells for Therapeutic Vaccination: Ready 'N Able to Improve Clinical Outcome? Cancers (Basel) 2020; 12:cancers12020299. [PMID: 32012714 PMCID: PMC7072269 DOI: 10.3390/cancers12020299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/06/2020] [Accepted: 01/19/2020] [Indexed: 02/06/2023] Open
Abstract
Targeting and exploiting the immune system has become a valid alternative to conventional options for treating cancer and infectious disease. Dendritic cells (DCs) take a central place given their role as key orchestrators of immunity. Therapeutic vaccination with autologous DCs aims to stimulate the patient's own immune system to specifically target his/her disease and has proven to be an effective form of immunotherapy with very little toxicity. A great amount of research in this field has concentrated on engineering these DCs through ribonucleic acid (RNA) to improve vaccine efficacy and thereby the historically low response rates. We reviewed in depth the 52 clinical trials that have been published on RNA-engineered DC vaccination, spanning from 2001 to date and reporting on 696 different vaccinated patients. While ambiguity prevents reliable quantification of effects, these trials do provide evidence that RNA-modified DC vaccination can induce objective clinical responses and survival benefit in cancer patients through stimulation of anti-cancer immunity, without significant toxicity. Succinct background knowledge of RNA engineering strategies and concise conclusions from available clinical and recent preclinical evidence will help guide future research in the larger domain of DC immunotherapy.
Collapse
|
25
|
Vanmeerbeek I, Sprooten J, De Ruysscher D, Tejpar S, Vandenberghe P, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L, Garg AD. Trial watch: chemotherapy-induced immunogenic cell death in immuno-oncology. Oncoimmunology 2020; 9:1703449. [PMID: 32002302 PMCID: PMC6959434 DOI: 10.1080/2162402x.2019.1703449] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022] Open
Abstract
The term ‘immunogenic cell death’ (ICD) denotes an immunologically unique type of regulated cell death that enables, rather than suppresses, T cell-driven immune responses that are specific for antigens derived from the dying cells. The ability of ICD to elicit adaptive immunity heavily relies on the immunogenicity of dying cells, implying that such cells must encode and present antigens not covered by central tolerance (antigenicity), and deliver immunostimulatory molecules such as damage-associated molecular patterns and cytokines (adjuvanticity). Moreover, the host immune system must be equipped to detect the antigenicity and adjuvanticity of dying cells. As cancer (but not normal) cells express several antigens not covered by central tolerance, they can be driven into ICD by some therapeutic agents, including (but not limited to) chemotherapeutics of the anthracycline family, oxaliplatin and bortezomib, as well as radiation therapy. In this Trial Watch, we describe current trends in the preclinical and clinical development of ICD-eliciting chemotherapy as partner for immunotherapy, with a focus on trials assessing efficacy in the context of immunomonitoring.
Collapse
Affiliation(s)
- Isaure Vanmeerbeek
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jenny Sprooten
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Dirk De Ruysscher
- Maastricht University Medical Center, Department of Radiation Oncology (MAASTRO Clinic), GROW-School for Oncology and Developmental Biology, Maastricht, Netherlands
| | - Sabine Tejpar
- Department of Oncology, KU Leuven, Leuven, Belgium.,UZ Leuven, Leuven, Belgium
| | - Peter Vandenberghe
- Department of Haematology, UZ Leuven, and Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Jitka Fucikova
- Sotio, Prague, Czech Republic.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio, Prague, Czech Republic.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, INSERM U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.,Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.,Université de Paris, Paris, France
| | - Abhishek D Garg
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
26
|
Węgierek K, Pajtasz-Piasecka E. Perspectives for the application of interleukin 15 in anti-cancer therapy. POSTEP HIG MED DOSW 2019. [DOI: 10.5604/01.3001.0013.7194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Interleukin (IL-) 15 plays a crucial role in the preservation of lymphoid cell homeostasis including maintaining a broad repertoire of naïve T, B and NK cells, eliminating effector cells and long-term survival of memory cells. It is an essential causative factor in generating CD8+ T cells of memory. In addition, it selectively promotes not only survival and proliferation, but also the effector function of antigen-specific cytotoxic T lymphocytes, even in the presence of regulatory T cells. Interleukin 15 can thus modulate immune suppression as well as promote an immune activation. All obtained data on the biology and function of IL-15 provide information essential to design the manners of its application in the fight against the solid cancers and myeloproliferative neoplasms and make it a promising therapeutic option provided that its potential is consciously used. In this paper we reviewed on the relationship between the biological properties of IL-15 and its IL-15/IL-15Rα complex and their antitumor potential in the light of recent reports about the possibilities of using these molecules in cancer therapy have been assessed.
Collapse
Affiliation(s)
- Katarzyna Węgierek
- Laboratorium Doświadczalnej Terapii Przeciwnowotworowej, Instytut Immunologii i Terapii Doświadczalnej Polskiej Akademii Nauk im. L. Hirszfelda we Wrocławiu
| | - Elżbieta Pajtasz-Piasecka
- Laboratorium Doświadczalnej Terapii Przeciwnowotworowej, Instytut Immunologii i Terapii Doświadczalnej Polskiej Akademii Nauk im. L. Hirszfelda we Wrocławiu
| |
Collapse
|
27
|
IL-15/IL-15Rα/CD80-expressing AML cell vaccines eradicate minimal residual disease in leukemic mice. Blood Adv 2019; 2:3177-3192. [PMID: 30482760 DOI: 10.1182/bloodadvances.2018019026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/14/2018] [Indexed: 12/17/2022] Open
Abstract
Engineered autologous acute myeloid leukemia (AML) cells present multiple leukemia-associated and patient-specific antigens and as such hold promise as immunotherapeutic vaccines. However, prior vaccines have not reliably induced effective antileukemic immunity, in part because AML blasts have immune inhibitory effects and lack expression of the critical costimulatory molecule CD80. To enhance induction of leukemia-specific cytolytic activity, 32Dp210 murine AML cells were engineered to express either CD80 alone, or the immunostimulatory cytokine interleukin-15 (IL-15) with its receptor α (IL-15Rα), or heterodimeric IL-15/IL-15Rα together with CD80 and tested as irradiated cell vaccines. IL-15 is a γc-chain cytokine, with unique properties suited to stimulating antitumor immunity, including stimulation of both natural killer and CD8+ memory T cells. Coexpression of IL-15 and IL-15Rα markedly increases IL-15 stability and secretion. Non-tumor-bearing mice vaccinated with irradiated 32Dp210-IL-15/IL-15Rα/CD80 and challenged with 32Dp210 leukemia had greater survival than did mice treated with 32Dp210-CD80 or 32Dp210-IL-15/IL-15Rα vaccines, whereas no unvaccinated mice inoculated with leukemia survived. In mice with established leukemia, treatment with 32Dp210-IL-15/IL-15Rα/CD80 vaccination stimulated unprecedented antileukemic immunity enabling 80% survival, an effect that was abrogated by anti-CD8 antibody-mediated depletion in vivo. Because, clinically, AML vaccines are administered as postremission therapy, we established a novel model in which mice with high leukemic burdens were treated with cytotoxic therapy to induce remission (<5% marrow blasts). Postremission vaccination with 32Dp210-IL-15/IL-15Rα/CD80 achieved 50% overall survival in these mice, whereas all unvaccinated mice achieving remission subsequently relapsed. These studies demonstrate that combined expression of IL-15/IL-15Rα and CD80 by syngeneic AML vaccines stimulates effective and long-lasting antileukemic immunity.
Collapse
|
28
|
Cortellini A, Verna L, Porzio G, Bozzetti F, Palumbo P, Masciocchi C, Cannita K, Parisi A, Brocco D, Tinari N, Ficorella C. Predictive value of skeletal muscle mass for immunotherapy with nivolumab in non-small cell lung cancer patients: A "hypothesis-generator" preliminary report. Thorac Cancer 2019; 10:347-351. [PMID: 30600905 PMCID: PMC6360197 DOI: 10.1111/1759-7714.12965] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/01/2022] Open
Abstract
Sarcopenia represents one of the hallmarks of all chronic disease, including non‐small cell lung cancer (NSCLC). A computed tomography scan is an easy modality to estimate the skeletal muscle mass through cross‐sectional image analysis at the level of the third lumbar vertebra (L3). Baseline skeletal muscle mass (SMM) was evaluated using gender‐specific cutoffs for skeletal muscle index in NSCLC patients administered immunotherapy with nivolumab to evaluate its possible correlations with clinical outcomes. From April 2015 to August 2018, 23 stage IV NSCLC patients were eligible for image analysis. Nine patients (39.1%) had low SMM. Among patients with baseline low and non‐low SMM, median progression free survival was 3.1 and 3.8 months, respectively (P = 0.0560), while median overall survival was 4.1 and 13 months, respectively (P = 0.2866). This hypothesis‐generating preliminary report offers the opportunity to speculate about the negative influence of sarcopenia on immune response. In our opinion, nutritional status could affect the clinical outcomes of immunotherapy, even if we cannot make definitive conclusions here. Further studies on the topic are required.
Collapse
Affiliation(s)
- Alessio Cortellini
- Medical Oncology, San Salvatore Hospital, University of L'Aquila, L'Aquila, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Lucilla Verna
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giampiero Porzio
- Medical Oncology, San Salvatore Hospital, University of L'Aquila, L'Aquila, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | | | - Pierpaolo Palumbo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Carlo Masciocchi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Katia Cannita
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alessandro Parisi
- Medical Oncology, San Salvatore Hospital, University of L'Aquila, L'Aquila, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Davide Brocco
- Clinical Oncology Unit, S.S. Annunziata Hospital, Chieti, Italy
| | - Nicola Tinari
- Department of Medical, Oral and Biotechnological Sciences, University G. D'Annunzio, Chieti, Italy
| | - Corrado Ficorella
- Medical Oncology, San Salvatore Hospital, University of L'Aquila, L'Aquila, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
29
|
Versteven M, Van den Bergh JMJ, Broos K, Fujiki F, Campillo-Davo D, De Reu H, Morimoto S, Lecocq Q, Keyaerts M, Berneman Z, Sugiyama H, Van Tendeloo VFI, Breckpot K, Lion E. A versatile T cell-based assay to assess therapeutic antigen-specific PD-1-targeted approaches. Oncotarget 2018; 9:27797-27808. [PMID: 29963238 PMCID: PMC6021243 DOI: 10.18632/oncotarget.25591] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/24/2018] [Indexed: 12/27/2022] Open
Abstract
Blockade of programmed cell death protein 1 (PD-1) immune checkpoint receptor signaling is an established standard treatment for many types of cancer and indications are expanding. Successful clinical trials using monoclonal antibodies targeting PD-1 signaling have boosted preclinical research, encouraging development of novel therapeutics. Standardized assays to evaluate their bioactivity, however, remain restricted. The robust bioassays available all lack antigen-specificity. Here, we developed an antigen-specific, short-term and high-throughput T cell assay with versatile readout possibilities. A genetically modified T cell receptor (TCR)-deficient T cell line was stably transduced with PD-1. Transfection with messenger RNA encoding a TCR of interest and subsequent overnight stimulation with antigen-presenting cells, results in eGFP-positive and granzyme B-producing T cells for single cell or bulk analysis. Control antigen-presenting cells induced reproducible high antigen-specific eGFP and granzyme B expression. Upon PD-1 interaction, ligand-positive antigen-presenting immune or tumor cells elicited significantly lower eGFP and granzyme B expression, which could be restored by anti-PD-(L)1 blocking antibodies. This convenient cell-based assay shows a valuable tool for translational and clinical research on antigen-specific checkpoint-targeted therapy approaches.
Collapse
Affiliation(s)
- Maarten Versteven
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Johan M J Van den Bergh
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Katrijn Broos
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fumihiro Fujiki
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Hans De Reu
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Soyoko Morimoto
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium.,Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Division of Hematology, University Hospital Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, University Hospital Antwerp, Antwerp, Belgium
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Viggo F I Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, University Hospital Antwerp, Antwerp, Belgium
| |
Collapse
|
30
|
Van Acker HH, Anguille S, De Reu H, Berneman ZN, Smits EL, Van Tendeloo VF. Interleukin-15-Cultured Dendritic Cells Enhance Anti-Tumor Gamma Delta T Cell Functions through IL-15 Secretion. Front Immunol 2018; 9:658. [PMID: 29692776 PMCID: PMC5902500 DOI: 10.3389/fimmu.2018.00658] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/16/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cell (DC) vaccination can be an effective post-remission therapy for acute myeloid leukemia (AML). Yet, current DC vaccines do not encompass the ideal stimulatory triggers for innate gamma delta (γδ) T cell anti-tumor activity. Promoting type 1 cytotoxic γδ T cells in patients with AML is, however, most interesting, considering these unconventional T cells are primed for rapid function and exert meaningful control over AML. In this work, we demonstrate that interleukin (IL)-15 DCs have the capacity to enhance the anti-tumoral functions of γδ T cells. IL-15 DCs of healthy donors and of AML patients in remission induce the upregulation of cytotoxicity-associated and co-stimulatory molecules on the γδ T cell surface, but not of co-inhibitory molecules, incite γδ T cell proliferation and stimulate their interferon-γ production in the presence of blood cancer cells and phosphoantigens. Moreover, the innate cytotoxic capacity of γδ T cells is significantly enhanced upon interaction with IL-15 DCs, both towards leukemic cell lines and allogeneic primary AML blasts. Finally, we address soluble IL-15 secreted by IL-15 DCs as the main mechanism behind the IL-15 DC-mediated γδ T cell activation. These results indicate that the application of IL-15-secreting DC subsets could render DC-based anti-cancer vaccines more effective through, among others, the involvement of γδ T cells in the anti-leukemic immune response.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Sébastien Anguille
- Division of Hematology, Antwerp University Hospital, Edegem, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Hans De Reu
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Division of Hematology, Antwerp University Hospital, Edegem, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium.,Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
31
|
Watson DC, Yung BC, Bergamaschi C, Chowdhury B, Bear J, Stellas D, Morales-Kastresana A, Jones JC, Felber BK, Chen X, Pavlakis GN. Scalable, cGMP-compatible purification of extracellular vesicles carrying bioactive human heterodimeric IL-15/lactadherin complexes. J Extracell Vesicles 2018; 7:1442088. [PMID: 29535850 PMCID: PMC5844027 DOI: 10.1080/20013078.2018.1442088] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 02/11/2018] [Indexed: 12/19/2022] Open
Abstract
The development of extracellular vesicles (EV) for therapeutic applications is contingent upon the establishment of reproducible, scalable, and high-throughput methods for the production and purification of clinical grade EV. Methods including ultracentrifugation (U/C), ultrafiltration, immunoprecipitation, and size-exclusion chromatography (SEC) have been employed to isolate EV, each facing limitations such as efficiency, particle purity, lengthy processing time, and/or sample volume. We developed a cGMP-compatible method for the scalable production, concentration, and isolation of EV through a strategy involving bioreactor culture, tangential flow filtration (TFF), and preparative SEC. We applied this purification method for the isolation of engineered EV carrying multiple complexes of a novel human immunostimulatory cytokine-fusion protein, heterodimeric IL-15 (hetIL-15)/lactadherin. HEK293 cells stably expressing the fusion cytokine were cultured in a hollow-fibre bioreactor. Conditioned medium was collected and EV were isolated comparing three procedures: U/C, SEC, or TFF + SEC. SEC demonstrated comparable particle recovery, size distribution, and hetIL-15 density as U/C purification. Relative to U/C, SEC preparations achieved a 100-fold reduction in ferritin concentration, a major protein-complex contaminant. Comparative proteomics suggested that SEC additionally decreased the abundance of cytoplasmic proteins not associated with EV. Combination of TFF and SEC allowed for bulk processing of large starting volumes, and resulted in bioactive EV, without significant loss in particle yield or changes in size, morphology, and hetIL-15/lactadherin density. Taken together, the combination of bioreactor culture with TFF + SEC comprises a scalable, efficient method for the production of highly purified, bioactive EV carrying hetIL-15/lactadherin, which may be useful in targeted cancer immunotherapy approaches.
Collapse
Affiliation(s)
- Dionysios C. Watson
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Bryant C. Yung
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Bhabadeb Chowdhury
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Dimitris Stellas
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | | | - Jennifer C. Jones
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| |
Collapse
|
32
|
Lin S, Huang G, Xiao Y, Sun W, Jiang Y, Deng Q, Peng M, Wei X, Ye W, Li B, Lin S, Wang S, Wu Q, Liang Q, Li Y, Zhang X, Wu Y, Liu P, Pei D, Yu F, Wen Z, Yao Y, Wu D, Li P. CD215+ Myeloid Cells Respond to Interleukin 15 Stimulation and Promote Tumor Progression. Front Immunol 2017; 8:1713. [PMID: 29255466 PMCID: PMC5722806 DOI: 10.3389/fimmu.2017.01713] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 11/20/2017] [Indexed: 12/16/2022] Open
Abstract
Interleukin 15 (IL-15) regulates the development, survival, and functions of multiple innate and adaptive immune cells and plays a dual role in promoting both tumor cell growth and antitumor immunity. Here, we demonstrated that the in vivo injection of recombinant human IL-15 (200 µg/kg) or murine IL-15 (3 µg/kg) to tumor-bearing NOD-SCID-IL2Rg−/− (NSI) mice resulted in increased tumor progression and CD45+ CD11b+ Gr-1+ CD215+ cell expansion in the tumors and spleen. In B16F10-bearing C57BL/6 mice model, we found that murine IL-15 has antitumoral effect since the activation and expansion of CD8+ T cells with murine IL-15 treatment. But no enhanced or reduced tumor growth was observed in mice when human IL-15 was used. However, both murine and human IL-15 promote CD45+ CD11b+ Gr-1+ CD215+ cells expansion. In xenograft tumor models, CD215+ myeloid cells, but not CD215− cells, responded to human IL-15 stimulation and promoted tumor growth. Furthermore, we found that human IL-15 mediated insulin-like growth factor-1 production in CD215+ myeloid cells and blocking IGF-1 reduced the tumor-promoting effect of IL-15. Finally, we observed that higher IGF-1 expression is an indicator of poor prognosis among lung adenocarcinoma patients. These findings provide evidence that IL-15 may promote tumor cell progression via CD215+ myeloid cells, and IGF-1 may be an important candidate that IL-15 facilitates tumor growth.
Collapse
Affiliation(s)
- Shouheng Lin
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Guohua Huang
- Department of Respiratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiren Xiao
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wei Sun
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuchuan Jiang
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiuhua Deng
- Department of Respiratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Muyun Peng
- Department of Thoracic Oncology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xinru Wei
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Wei Ye
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Baiheng Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Simiao Lin
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Suna Wang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qiting Wu
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qiubin Liang
- Guangdong Zhaotai InVivo Biomedicine Co. Ltd., Guangzhou, China
| | - Yangqiu Li
- Medical College, Institute of Hematology, Jinan University, Guangzhou, China
| | - Xuchao Zhang
- Guangdong Lung Cancer Institute, Medical Research Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yilong Wu
- Guangdong Lung Cancer Institute, Medical Research Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Pentao Liu
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Duanqing Pei
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Fenglei Yu
- Department of Thoracic Oncology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhesheng Wen
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yao Yao
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Donghai Wu
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Peng Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
33
|
Garg AD, More S, Rufo N, Mece O, Sassano ML, Agostinis P, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Immunogenic cell death induction by anticancer chemotherapeutics. Oncoimmunology 2017; 6:e1386829. [PMID: 29209573 DOI: 10.1080/2162402x.2017.1386829] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 09/26/2017] [Indexed: 12/21/2022] Open
Abstract
The expression "immunogenic cell death" (ICD) refers to a functionally unique form of cell death that facilitates (instead of suppressing) a T cell-dependent immune response specific for dead cell-derived antigens. ICD critically relies on the activation of adaptive responses in dying cells, culminating with the exposure or secretion of immunostimulatory molecules commonly referred to as "damage-associated molecular patterns". Only a few agents can elicit bona fide ICD, including some clinically established chemotherapeutics such as doxorubicin, epirubicin, idarubicin, mitoxantrone, bleomycin, bortezomib, cyclophosphamide and oxaliplatin. In this Trial Watch, we discuss recent progress on the development of ICD-inducing chemotherapeutic regimens, focusing on studies that evaluate clinical efficacy in conjunction with immunological biomarkers.
Collapse
Affiliation(s)
- Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Sanket More
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Nicole Rufo
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Odeta Mece
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Maria Livia Sassano
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Paris, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
34
|
Van Acker HH, Beretta O, Anguille S, De Caluwé L, Papagna A, Van den Bergh JM, Willemen Y, Goossens H, Berneman ZN, Van Tendeloo VF, Smits EL, Foti M, Lion E. Desirable cytolytic immune effector cell recruitment by interleukin-15 dendritic cells. Oncotarget 2017; 8:13652-13665. [PMID: 28099143 PMCID: PMC5355127 DOI: 10.18632/oncotarget.14622] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 01/03/2017] [Indexed: 11/25/2022] Open
Abstract
Success of dendritic cell (DC) therapy in treating malignancies is depending on the DC capacity to attract immune effector cells, considering their reciprocal crosstalk is partially regulated by cell-contact-dependent mechanisms. Although critical for therapeutic efficacy, immune cell recruitment is a largely overlooked aspect regarding optimization of DC vaccination. In this paper we have made a head-to-head comparison of interleukin (IL)-15-cultured DCs and conventional IL-4-cultured DCs with regard to their proficiency in the recruitment of (innate) immune effector cells. Here, we demonstrate that IL-4 DCs are suboptimal in attracting effector lymphocytes, while IL15 DCs provide a favorable chemokine milieu for recruiting CD8+ T cells, natural killer (NK) cells and gamma delta (γδ) T cells. Gene expression analysis revealed that IL-15 DCs exhibit a high expression of chemokines involved in antitumor immune effector cell attraction, while IL-4 DCs display a more immunoregulatory profile characterized by the expression of Th2 and regulatory T cell-attracting chemokines. This is confirmed by functional data indicating an enhanced recruitment of granzyme B+ effector lymphocytes by IL-15 DCs, as compared to IL-4 DCs, and subsequent superior killing of tumor cells by the migrated lymphocytes. Elevated CCL4 gene expression in IL-15 DCs and lowered CCR5 expression on both migrated γδ T cells and NK cells, led to validation of increased CCL4 secretion by IL15 DCs. Moreover, neutralization of CCR5 prior to migration resulted in an important inhibition of γδ T cell and NK cell recruitment by IL-15 DCs. These findings further underscore the strong immunotherapeutic potential of IL-15 DCs.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Ottavio Beretta
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Lien De Caluwé
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.,Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Angela Papagna
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Johan M Van den Bergh
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Yannick Willemen
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Herman Goossens
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium.,Center for Oncological Research (CORE), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Maria Foti
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Eva Lion
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
35
|
Guo Y, Luan L, Patil NK, Sherwood ER. Immunobiology of the IL-15/IL-15Rα complex as an antitumor and antiviral agent. Cytokine Growth Factor Rev 2017; 38:10-21. [PMID: 28888485 DOI: 10.1016/j.cytogfr.2017.08.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 08/29/2017] [Indexed: 12/20/2022]
Abstract
Interleukin (IL)-15 is essential for natural killer (NK), NKT and memory (m) CD8+ T cell development and function, and is currently under investigation as an immunotherapeutic agent for the treatment of cancer. Recently, the creation of IL-15 superagonist by complexing IL-15 and its high affinity receptor alpha (IL-15 Rα) in solution, inspired by the natural trans-presentation of IL-15, advances the potential of IL-15-based tumor immunotherapy. IL-15 superagonist shows promising advantages over monomeric IL-15 such as sustaining high circulating concentrations due to prolonged half-life and more potently stimulating NK and CD8+ T effector lymphocytes. So far, there are three different forms of recombinant IL-15 superagonist fusion protein based on configurational modifications. Gene therapy using engineered cells co-expressing IL-15/IL-15 Rα complex for cancer treatment is also emerging. All forms have demonstrated efficacy in causing tumor regression in animal studies, which provides strong rationale for advancing IL-15 superagonist through clinical trials. To date, there are fourteen phase I/II IL-15 superagonist trials in cancer patients and one phase I trial in HIV patients. Information generated by ongoing trials regarding the toxicity and efficacy of IL-15 superagonist is awaited. Finally, we elaborate on immunotoxicity caused by IL-15 superagonist in preclinical studies and discuss important safety considerations.
Collapse
Affiliation(s)
- Yin Guo
- Departments of Pathology, Microbiology and Immunology. Vanderbilt University, Nashville, TN, USA; Department of Anesthesiology. Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Liming Luan
- Department of Anesthesiology. Vanderbilt University Medical Center, Nashville, TN, USA
| | - Naeem K Patil
- Department of Anesthesiology. Vanderbilt University Medical Center, Nashville, TN, USA
| | - Edward R Sherwood
- Departments of Pathology, Microbiology and Immunology. Vanderbilt University, Nashville, TN, USA; Department of Anesthesiology. Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
36
|
Abraham RS, Mitchell DA. Gene-modified dendritic cell vaccines for cancer. Cytotherapy 2017; 18:1446-1455. [PMID: 27745604 DOI: 10.1016/j.jcyt.2016.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/16/2016] [Indexed: 12/13/2022]
Abstract
Dendritic cell (DC) vaccines are an immunotherapeutic approach to cancer treatment that use the antigen-presentation machinery of DCs to activate an endogenous anti-tumor response. In this treatment strategy, DCs are cultured ex vivo, exposed to tumor antigens and administered to the patient. The ex vivo culturing provides a unique and powerful opportunity to modify and enhance the DCs. As such, a variety of genetic engineering approaches have been employed to optimize DC vaccines, including the introduction of messenger RNA and small interfering RNA, viral gene transduction, and even fusion with whole tumor cells. In general, these modifications aim to improve targeting, enhance immunogenicity, and reduce susceptibility to the immunosuppressive tumor microenvironment. It has been demonstrated that several of these modifications can be employed in tandem, allowing for fine-tuning and optimization of the DC vaccine across multiple metrics. Thus, the application of genetic engineering techniques to the dendritic cell vaccine platform has the potential to greatly enhance its efficacy in the clinic.
Collapse
Affiliation(s)
- Rebecca S Abraham
- UF Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Department of Neurosurgery, University of Florida, Gainesville, FL 32605
| | - Duane A Mitchell
- UF Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Department of Neurosurgery, University of Florida, Gainesville, FL 32605.
| |
Collapse
|
37
|
Lichtenegger FS, Krupka C, Haubner S, Köhnke T, Subklewe M. Recent developments in immunotherapy of acute myeloid leukemia. J Hematol Oncol 2017; 10:142. [PMID: 28743264 PMCID: PMC5526264 DOI: 10.1186/s13045-017-0505-0] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 06/26/2017] [Indexed: 02/07/2023] Open
Abstract
The advent of new immunotherapeutic agents in clinical practice has revolutionized cancer treatment in the past decade, both in oncology and hematology. The transfer of the immunotherapeutic concepts to the treatment of acute myeloid leukemia (AML) is hampered by various characteristics of the disease, including non-leukemia-restricted target antigen expression profile, low endogenous immune responses, and intrinsic resistance mechanisms of the leukemic blasts against immune responses. However, considerable progress has been made in this field in the past few years.Within this manuscript, we review the recent developments and the current status of the five currently most prominent immunotherapeutic concepts: (1) antibody-drug conjugates, (2) T cell-recruiting antibody constructs, (3) chimeric antigen receptor (CAR) T cells, (4) checkpoint inhibitors, and (5) dendritic cell vaccination. We focus on the clinical data that has been published so far, both for newly diagnosed and refractory/relapsed AML, but omitting immunotherapeutic concepts in conjunction with hematopoietic stem cell transplantation. Besides, we have included important clinical trials that are currently running or have recently been completed but are still lacking full publication of their results.While each of the concepts has its particular merits and inherent problems, the field of immunotherapy of AML seems to have taken some significant steps forward. Results of currently running trials will reveal the direction of further development including approaches combining two or more of these concepts.
Collapse
Affiliation(s)
- Felix S Lichtenegger
- Department of Medicine III, University Hospital, LMU Munich, Germany
- Laboratory of Translational Cancer Immunology, Gene Center, Munich, Germany
| | - Christina Krupka
- Department of Medicine III, University Hospital, LMU Munich, Germany
- Laboratory of Translational Cancer Immunology, Gene Center, Munich, Germany
| | - Sascha Haubner
- Department of Medicine III, University Hospital, LMU Munich, Germany
- Laboratory of Translational Cancer Immunology, Gene Center, Munich, Germany
| | - Thomas Köhnke
- Department of Medicine III, University Hospital, LMU Munich, Germany
- Laboratory of Translational Cancer Immunology, Gene Center, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, LMU Munich, Germany.
- Laboratory of Translational Cancer Immunology, Gene Center, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site, Munich, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
38
|
Van den Bergh JMJ, Smits ELJM, Versteven M, De Reu H, Berneman ZN, Van Tendeloo VFI, Lion E. Characterization of Interleukin-15-Transpresenting Dendritic Cells for Clinical Use. J Immunol Res 2017; 2017:1975902. [PMID: 28785596 PMCID: PMC5530419 DOI: 10.1155/2017/1975902] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/04/2017] [Indexed: 11/17/2022] Open
Abstract
Personalized dendritic cell- (DC-) based vaccination has proven to be safe and effective as second-line therapy against various cancer types. In terms of overall survival, there is still room for improvement of DC-based therapies, including the development of more immunostimulatory DC vaccines. In this context, we redesigned our currently clinically used DC vaccine generation protocol to enable transpresentation of interleukin- (IL-) 15 to IL-15Rβγ-expressing cells aiming at boosting the antitumor immune response. In this study, we demonstrate that upon electroporation with both IL-15 and IL-15Rα-encoding messenger RNA, mature DC become highly positive for surface IL-15, without influencing the expression of prototypic mature DC markers and with preservation of their cytokine-producing capacity and their migratory profile. Functionally, we show that IL-15-transpresenting DC are equal if not better inducers of T-cell proliferation and are superior in tumor antigen-specific T-cell activation compared with DC without IL-15 conditioning. In view of the clinical use of DC vaccines, we evidence with a time- and cost-effective manner that clinical grade DC can be safely engineered to transpresent IL-15, hereby gaining the ability to transfer the immune-stimulating IL-15 signal towards antitumor immune effector cells.
Collapse
Affiliation(s)
- J. M. J. Van den Bergh
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - E. L. J. M. Smits
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
- Center for Oncological Research Antwerp, Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - M. Versteven
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - H. De Reu
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Z. N. Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - V. F. I. Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - E. Lion
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
39
|
Van den Bergh JMJ, Smits ELJM, Berneman ZN, Hutten TJA, De Reu H, Van Tendeloo VFI, Dolstra H, Lion E, Hobo W. Monocyte-Derived Dendritic Cells with Silenced PD-1 Ligands and Transpresenting Interleukin-15 Stimulate Strong Tumor-Reactive T-cell Expansion. Cancer Immunol Res 2017. [PMID: 28637876 DOI: 10.1158/2326-6066.cir-16-0336] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although allogeneic stem cell transplantation (allo-SCT) can elicit graft-versus-tumor (GVT) immunity, patients often relapse due to residual tumor cells. As essential orchestrators of the immune system, vaccination with dendritic cells (DC) is an appealing strategy to boost the GVT response. Nevertheless, durable clinical responses after DC vaccination are still limited, stressing the need to improve current DC vaccines. Aiming to empower DC potency, we engineered monocyte-derived DCs to deprive them of ligands for the immune checkpoint regulated by programmed death 1 (PD-1). We also equipped them with interleukin (IL)-15 "transpresentation" skills. Transfection with short interfering (si)RNA targeting the PD-1 ligands PD-L1 and PD-L2, in combination with IL15 and IL15Rα mRNA, preserved their mature DC profile and rendered the DCs superior in inducing T-cell proliferation and IFNγ and TNFα production. Translated into an ex vivo hematological disease setting, DCs deprived of PD-1 ligands (PD-L), equipped with IL15/IL15Rα expression, or most effectively, both, induced superior expansion of minor histocompatibility antigen-specific CD8+ T cells from transplanted cancer patients. These data support the combinatorial approach of in situ suppression of the PD-L inhibitory checkpoints with DC-mediated IL15 transpresentation to promote antigen-specific T-cell responses and, ultimately, contribute to GVT immunity. Cancer Immunol Res; 5(8); 710-5. ©2017 AACR.
Collapse
Affiliation(s)
- Johan M J Van den Bergh
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.
| | - Evelien L J M Smits
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Center for Oncological Research Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Tim J A Hutten
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hans De Reu
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Viggo F I Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Harry Dolstra
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Willemijn Hobo
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
40
|
IL-15 receptor alpha as the magic wand to boost the success of IL-15 antitumor therapies: The upswing of IL-15 transpresentation. Pharmacol Ther 2016; 170:73-79. [PMID: 27777088 DOI: 10.1016/j.pharmthera.2016.10.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Interleukin (IL)-15 as a stand-alone therapy can activate the antitumor functions of immune effector cells resulting in significant tumor regression. Interestingly, combining IL-15 with the α-moiety of its receptor (IL-15Rα), also called IL-15 transpresentation, increases the in vivo half-life of IL-15 and enhances binding of IL-15 with cells expressing the IL-15Rβγ, such as NK cells and CD8+ T cells. These features enlarge the signal transmission of IL-15, resulting in improved proliferation and antitumor activities of both NK cells and CD8+ T cells, eventually leading to enhanced killing of tumor cells. In this review, we discuss the antitumor strategies in which this IL-15 transpresentation mechanism is implemented, that are currently under preclinical investigation. Furthermore, we give an overview of the studies in which the IL-15/IL-15Rα complexes are combined with other antitumor therapies. The promising results in these preclinical studies have incited several clinical trials to test the safety and efficacy of IL-15 transpresentation strategies to treat both hematological and advanced solid tumors.
Collapse
|
41
|
Lapenta C, Donati S, Spadaro F, Castaldo P, Belardelli F, Cox MC, Santini SM. NK Cell Activation in the Antitumor Response Induced by IFN-α Dendritic Cells Loaded with Apoptotic Cells from Follicular Lymphoma Patients. THE JOURNAL OF IMMUNOLOGY 2016; 197:795-806. [DOI: 10.4049/jimmunol.1600262] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/01/2016] [Indexed: 11/19/2022]
|