1
|
Volpe A, Lyashchenko SK, Ponomarev V. Nuclear-Based Labeling of Cellular Immunotherapies: A Simple Protocol for Preclinical Use. Mol Imaging Biol 2024; 26:555-568. [PMID: 38958882 PMCID: PMC11281953 DOI: 10.1007/s11307-024-01923-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/10/2024] [Accepted: 05/18/2024] [Indexed: 07/04/2024]
Abstract
Labeling and tracking existing and emerging cell-based immunotherapies using nuclear imaging is widely used to guide the preclinical phases of development and testing of existing and new emerging off-the-shelf cell-based immunotherapies. In fact, advancing our knowledge about their mechanism of action and limitations could provide preclinical support and justification for moving towards clinical experimentation of newly generated products and expedite their approval by the Food and Drug Administration (FDA).Here we provide the reader with a ready to use protocol describing the labeling methodologies and practical procedures to render different candidate cell therapies in vivo traceable by nuclear-based imaging. The protocol includes sufficient practical details to aid researchers at all career stages and from different fields in familiarizing with the described concepts and incorporating them into their work.
Collapse
Affiliation(s)
- Alessia Volpe
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Serge K Lyashchenko
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
2
|
Tai Y, Chen M, Wang F, Fan Y, Zhang J, Cai B, Yan L, Luo Y, Li Y. The role of dendritic cells in cancer immunity and therapeutic strategies. Int Immunopharmacol 2024; 128:111548. [PMID: 38244518 DOI: 10.1016/j.intimp.2024.111548] [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: 11/07/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Dendritic cells (DCs) are asserted as the most potent antigen-presenting cells (APCs) that orchestrate both innate and adaptive immunity, being extremely effective in the induction of robust anti-cancer T cell responses. Hence, the modulation of DCs function represents an attractive target for improving cancer immunotherapy efficacy. A better understanding of the immunobiology of DCs, the interaction among DCs, immune effector cells and tumor cells in tumor microenvironment (TME) and the latest advances in biomedical engineering technology would be required for the design of optimal DC-based immunotherapy. In this review, we focus on elaborating the immunobiology of DCs in healthy and cancer environments, the recent advances in the development of enhancing endogenous DCs immunocompetence via immunomodulators as well as DC-based vaccines. The rapidly developing field of applying nanotechnology to improve DC-based immunotherapy is also highlighted.
Collapse
Affiliation(s)
- Yunze Tai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Man Chen
- Hebei Yanda Lu Daopei Hospital, Langfang 065201, China
| | - Fang Wang
- Department of Medical Laboratory, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou 556000, China
| | - Yu Fan
- Department of Urology, National Clinical Research Center for Geriatrics and Organ Transplantation Center, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu 610041, China
| | - Junlong Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bei Cai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Yan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yao Luo
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Mateus D, Sebastião AI, Frasco MF, Carrascal MA, Falcão A, Gomes CM, Neves B, Sales MGF, Cruz MT. Artificial Dendritic Cells: A New Era of Promising Antitumor Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303940. [PMID: 37469192 DOI: 10.1002/smll.202303940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/03/2023] [Indexed: 07/21/2023]
Abstract
The accelerated development of antitumor immunotherapies in recent years has brought immunomodulation into the spotlight. These include immunotherapeutic treatments with dendritic cell (DC)-based vaccines which can elicit tumor-specific immune responses and prolong survival. However, this personalized treatment has several drawbacks, including being costly, labor-intensive, and time consuming. This has sparked interest in producing artificial dendritic cells (aDCs) to open up the possibility of standardized "off-the-shelf" protocols and circumvent the cumbersome and expensive personalized medicine. aDCs take advantage of materials that can be designed and tailored for specific clinical applications. Here, an overview of the immunobiology underlying antigen presentation by DCs is provided in an attempt to select the key features to be mimicked and/or improved through the development of aDCs. The inherent properties of aDCs that greatly impact their performance in vivo and, consequently, the fate of the triggered immune response are also outlined.
Collapse
Affiliation(s)
- Daniela Mateus
- Faculty of Pharmacy of the University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Neuroscience and Cell Biology-CNC, University of Coimbra, Coimbra, 3004-504, Portugal
- BioMark@UC/CEB - LABBELS Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3030-790, Portugal
| | - Ana I Sebastião
- Faculty of Pharmacy of the University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Neuroscience and Cell Biology-CNC, University of Coimbra, Coimbra, 3004-504, Portugal
| | - Manuela F Frasco
- BioMark@UC/CEB - LABBELS Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3030-790, Portugal
| | | | - Amílcar Falcão
- Faculty of Pharmacy of the University of Coimbra, Coimbra, 3000-548, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, CIBIT, University of Coimbra, Coimbra, 3000-548, Portugal
| | - Célia M Gomes
- Coimbra Institute for Clinical and Biomedical Research, iCBR, Faculty of Medicine, University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Innovation in Biomedicine and Biotechnology, CIBB, University of Coimbra, Coimbra, 3000-548, Portugal
| | - Bruno Neves
- Department of Medical Sciences and Institute of Biomedicine, iBiMED, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Maria G F Sales
- BioMark@UC/CEB - LABBELS Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3030-790, Portugal
| | - Maria T Cruz
- Faculty of Pharmacy of the University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Neuroscience and Cell Biology-CNC, University of Coimbra, Coimbra, 3004-504, Portugal
| |
Collapse
|
4
|
Subtirelu RC, Teichner EM, Ashok A, Parikh C, Talasila S, Matache IM, Alnemri AG, Anderson V, Shahid O, Mannam S, Lee A, Werner T, Revheim ME, Alavi A. Advancements in dendritic cell vaccination: enhancing efficacy and optimizing combinatorial strategies for the treatment of glioblastoma. Front Neurol 2023; 14:1271822. [PMID: 38020665 PMCID: PMC10644823 DOI: 10.3389/fneur.2023.1271822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Glioblastomas (GBM) are highly invasive, malignant primary brain tumors. The overall prognosis is poor, and management of GBMs remains a formidable challenge, necessitating novel therapeutic strategies such as dendritic cell vaccinations (DCVs). While many early clinical trials demonstrate an induction of an antitumoral immune response, outcomes are mixed and dependent on numerous factors that vary between trials. Optimization of DCVs is essential; the selection of GBM-specific antigens and the utilization of 18F-fludeoxyglucose Positron Emission Tomography (FDG-PET) may add significant value and ultimately improve outcomes for patients undergoing treatment for glioblastoma. This review provides an overview of the mechanism of DCV, assesses previous clinical trials, and discusses future strategies for the integration of DCV into glioblastoma treatment protocols. To conclude, the review discusses challenges associated with the use of DCVs and highlights the potential of integrating DCV with standard therapies.
Collapse
Affiliation(s)
- Robert C. Subtirelu
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Eric M. Teichner
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Arjun Ashok
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Chitra Parikh
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sahithi Talasila
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Irina-Mihaela Matache
- Department of Physiology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ahab G. Alnemri
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Victoria Anderson
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Osmaan Shahid
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Sricharvi Mannam
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Andrew Lee
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Thomas Werner
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Mona-Elisabeth Revheim
- Division of Technology and Innovation, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
5
|
Dillman RO, Nistor GI, Keirstead HS. Autologous dendritic cells loaded with antigens from self-renewing autologous tumor cells as patient-specific therapeutic cancer vaccines. Hum Vaccin Immunother 2023:2198467. [PMID: 37133853 DOI: 10.1080/21645515.2023.2198467] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023] Open
Abstract
A promising personal immunotherapy is autologous dendritic cells (DC) loaded ex vivo with autologous tumor antigens (ATA) derived from self-renewing autologous cancer cells. DC-ATA are suspended in granulocyte-macrophage colony stimulating factor at the time of each subcutaneous injection. Previously, irradiated autologous tumor cell vaccines have produced encouraging results in 150 cancer patients, but the DC-ATA vaccine demonstrated superiority in single-arm and randomized trials in metastatic melanoma. DC-ATA have been injected into more than 200 patients with melanoma, glioblastoma, and ovarian, hepatocellular, and renal cell cancers. Key observations include: [1] greater than 95% success rates for tumor cell cultures and monocyte collection for dendritic cell production; [2] injections are well-tolerated; [3] the immune response is rapid and includes primarily TH1/TH17 cellular responses; [4] efficacy has been suggested by delayed but durable complete tumor regressions in patients with measurable disease, by progression-free survival in glioblastoma, and by overall survival in melanoma.
Collapse
Affiliation(s)
| | - Gabriel I Nistor
- Research and Development, AIVITA Biomedical Inc, Irvine, CA, USA
| | | |
Collapse
|
6
|
Jin R, Fu X, Pu Y, Fu S, Liang H, Yang L, Nie Y, Ai H. Clinical translational barriers against nanoparticle-based imaging agents. Adv Drug Deliv Rev 2022; 191:114587. [PMID: 36309148 DOI: 10.1016/j.addr.2022.114587] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/22/2022] [Accepted: 10/20/2022] [Indexed: 01/24/2023]
Abstract
Nanoparticle based imaging agents (NIAs) have been intensively explored in bench studies. Unfortunately, only a few cases have made their ways to clinical translation. In this review, clinical trials of NIAs were investigated for understanding possible barriers behind that. First, the complexity of multifunctional NIAs is considered a main barrier because it brings uncertainty to batch-to-batch fabrication, and results in sophisticated in vivo behaviors. Second, inadequate biosafety studies slow down the translational work. Third, NIA uptake at disease sites is highly heterogeneous, and often exhibits poor targeting efficiency. Focusing on the aforementioned problems, key design parameters were analyzed including NIAs' size, composition, surface characteristics, dosage, administration route, toxicity, whole-body distribution and clearance in clinical trials. Possible strategies were suggested to overcome these barriers. Besides, regulatory guidelines as well as scale-up and reproducibility during manufacturing process were covered as they are also key factors to consider during clinical translation of NIAs.
Collapse
Affiliation(s)
- Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xiaomin Fu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yiyao Pu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Shengxiang Fu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Hong Liang
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| | - Hua Ai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
7
|
Volpe A, Adusumilli PS, Schöder H, Ponomarev V. Imaging cellular immunotherapies and immune cell biomarkers: from preclinical studies to patients. J Immunother Cancer 2022; 10:jitc-2022-004902. [PMID: 36137649 PMCID: PMC9511655 DOI: 10.1136/jitc-2022-004902] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2022] [Indexed: 01/26/2023] Open
Abstract
Cellular immunotherapies have emerged as a successful therapeutic approach to fight a wide range of human diseases, including cancer. However, responses are limited to few patients and tumor types. An in-depth understanding of the complexity and dynamics of cellular immunotherapeutics, including what is behind their success and failure in a patient, the role of other immune cell types and molecular biomarkers in determining a response, is now paramount. As the cellular immunotherapy arsenal expands, whole-body non-invasive molecular imaging can shed a light on their in vivo fate and contribute to the reliable assessment of treatment outcome and prediction of therapeutic response. In this review, we outline the non-invasive strategies that can be tailored toward the molecular imaging of cellular immunotherapies and immune-related components, with a focus on those that have been extensively tested preclinically and are currently under clinical development or have already entered the clinical trial phase. We also provide a critical appraisal on the current role and consolidation of molecular imaging into clinical practice.
Collapse
Affiliation(s)
- Alessia Volpe
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA,Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York, USA,Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
8
|
Wang D, Cui Q, Yang YJ, Liu AQ, Zhang G, Yu JC. Application of dendritic cells in tumor immunotherapy and progress in the mechanism of anti-tumor effect of Astragalus polysaccharide (APS) modulating dendritic cells: a review. Biomed Pharmacother 2022; 155:113541. [PMID: 36127221 DOI: 10.1016/j.biopha.2022.113541] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells (APCs) that are essential in mediating the body's natural and adaptive immune responses. The body can regulate the function of DCs in various ways to enhance their antitumor effects. In the tumour microenvironment (TME), antigen-specific T cell responses are initiated through DC processing and delivery of tumour-associated antigens (TAAs); conversely, tumour cells inhibit DC recruitment by releasing metabolites, cytokines and other regulatory TME and function. Different subpopulations of DCs exist in tumour tissues, and their functions vary. Insight into DC subgroups in TME allows assessment of the effectiveness of tumour immunotherapy. Astragalus polysaccharide (APS) is the main component of the Chinese herb Astragalus membranaceus. The study found that the antitumor effects of APS are closely related to DCs. APS can promote the expression of surface molecules CD80 and CD86, promote the maturation of DCs, and activate CTL to exert antitumor effects. We reviewed the application of DCs in tumor immunotherapy and the mechanism of modulation of DCs by Astragalus polysaccharide to provide new directions and strategies for tumor therapy and new drug development.
Collapse
Affiliation(s)
- Dong Wang
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - Qian Cui
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - Yan Jie Yang
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - A Qing Liu
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - Guan Zhang
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Graduate School of Tianjin University of traditional Chinese Medicine, Tianjin, China
| | - Jian Chun Yu
- Department of Oncology, First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China.
| |
Collapse
|
9
|
Activation of Cellular Players in Adaptive Immunity via Exogenous Delivery of Tumor Cell Lysates. Pharmaceutics 2022; 14:pharmaceutics14071358. [PMID: 35890254 PMCID: PMC9316852 DOI: 10.3390/pharmaceutics14071358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 12/04/2022] Open
Abstract
Tumor cell lysates (TCLs) are a good immunogenic source of tumor-associated antigens. Since whole necrotic TCLs can enhance the maturation and antigen-presenting ability of dendritic cells (DCs), multiple strategies for the exogenous delivery of TCLs have been investigated as novel cancer immunotherapeutic solutions. The TCL-mediated induction of DC maturation and the subsequent immunological response could be improved by utilizing various material-based carriers. Enhanced antitumor immunity and cancer vaccination efficacy could be eventually achieved through the in vivo administration of TCLs. Therefore, (1) important engineering methodologies to prepare antigen-containing TCLs, (2) current therapeutic approaches using TCL-mediated DC activation, and (3) the significant sequential mechanism of DC-based signaling and stimulation in adaptive immunity are summarized in this review. More importantly, the recently reported developments in biomaterial-based exogenous TCL delivery platforms and co-delivery strategies with adjuvants for effective cancer vaccination and antitumor effects are emphasized.
Collapse
|
10
|
Brown LV, Wagg J, Darley R, van Hateren A, Elliott T, Gaffney EA, Coles MC. De-risking clinical trial failure through mechanistic simulation. IMMUNOTHERAPY ADVANCES 2022; 2:ltac017. [PMID: 36176591 PMCID: PMC9514113 DOI: 10.1093/immadv/ltac017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 07/04/2022] [Indexed: 11/19/2022] Open
Abstract
Drug development typically comprises a combination of pre-clinical experimentation, clinical trials, and statistical data-driven analyses. Therapeutic failure in late-stage clinical development costs the pharmaceutical industry billions of USD per year. Clinical trial simulation represents a key derisking strategy and combining them with mechanistic models allows one to test hypotheses for mechanisms of failure and to improve trial designs. This is illustrated with a T-cell activation model, used to simulate the clinical trials of IMA901, a short-peptide cancer vaccine. Simulation results were consistent with observed outcomes and predicted that responses are limited by peptide off-rates, peptide competition for dendritic cell (DC) binding, and DC migration times. These insights were used to hypothesise alternate trial designs predicted to improve efficacy outcomes. This framework illustrates how mechanistic models can complement clinical, experimental, and data-driven studies to understand, test, and improve trial designs, and how results may differ between humans and mice.
Collapse
Affiliation(s)
- Liam V Brown
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford , Oxford , UK
- Kennedy Institute of Rheumatology, University of Oxford , Oxford , UK
| | - Jonathan Wagg
- Pharmaceutical Sciences–Clinical Pharmacology, Roche Innovation Center Basel , Basel , Switzerland
| | - Rachel Darley
- Centre for Cancer Immunology, Institute for Life Sciences, University of Southampton , Southampton , UK
| | - Andy van Hateren
- Centre for Cancer Immunology, Institute for Life Sciences, University of Southampton , Southampton , UK
| | - Tim Elliott
- Centre for Immuno-oncology, Nuffield Department of Medicine, University of Oxford , Oxford , UK
| | - Eamonn A Gaffney
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford , Oxford , UK
| | - Mark C Coles
- Kennedy Institute of Rheumatology, University of Oxford , Oxford , UK
| |
Collapse
|
11
|
Wang LL, Janes ME, Kumbhojkar N, Kapate N, Clegg JR, Prakash S, Heavey MK, Zhao Z, Anselmo AC, Mitragotri S. Cell therapies in the clinic. Bioeng Transl Med 2021; 6:e10214. [PMID: 34027097 PMCID: PMC8126820 DOI: 10.1002/btm2.10214] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/30/2021] [Accepted: 02/09/2021] [Indexed: 12/16/2022] Open
Abstract
Cell therapies have emerged as a promising therapeutic modality with the potential to treat and even cure a diverse array of diseases. Cell therapies offer unique clinical and therapeutic advantages over conventional small molecules and the growing number of biologics. Particularly, living cells can simultaneously and dynamically perform complex biological functions in ways that conventional drugs cannot; cell therapies have expanded the spectrum of available therapeutic options to include key cellular functions and processes. As such, cell therapies are currently one of the most investigated therapeutic modalities in both preclinical and clinical settings, with many products having been approved and many more under active clinical investigation. Here, we highlight the diversity and key advantages of cell therapies and discuss their current clinical advances. In particular, we review 28 globally approved cell therapy products and their clinical use. We also analyze >1700 current active clinical trials of cell therapies, with an emphasis on discussing their therapeutic applications. Finally, we critically discuss the major biological, manufacturing, and regulatory challenges associated with the clinical translation of cell therapies.
Collapse
Affiliation(s)
- Lily Li‐Wen Wang
- John A. Paulson School of Engineering & Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringBostonMassachusettsUSA
- Harvard‐MIT Division of Health Sciences and Technology, Massachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Morgan E. Janes
- John A. Paulson School of Engineering & Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringBostonMassachusettsUSA
- Harvard‐MIT Division of Health Sciences and Technology, Massachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Ninad Kumbhojkar
- John A. Paulson School of Engineering & Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Neha Kapate
- John A. Paulson School of Engineering & Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringBostonMassachusettsUSA
- Harvard‐MIT Division of Health Sciences and Technology, Massachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - John R. Clegg
- John A. Paulson School of Engineering & Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Supriya Prakash
- John A. Paulson School of Engineering & Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Mairead K. Heavey
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Zongmin Zhao
- John A. Paulson School of Engineering & Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Aaron C. Anselmo
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Samir Mitragotri
- John A. Paulson School of Engineering & Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringBostonMassachusettsUSA
| |
Collapse
|
12
|
Harari A, Graciotti M, Bassani-Sternberg M, Kandalaft LE. Antitumour dendritic cell vaccination in a priming and boosting approach. Nat Rev Drug Discov 2020; 19:635-652. [PMID: 32764681 DOI: 10.1038/s41573-020-0074-8] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
Abstract
Mobilizing antitumour immunity through vaccination potentially constitutes a powerful anticancer strategy but has not yet provided robust clinical benefits in large patient populations. Although major hurdles still exist, we believe that currently available strategies for vaccines that target dendritic cells or use them to present antitumour antigens could be integrated into existing clinical practice using prime-boost approaches. In the priming phase, these approaches capitalize on either standard treatment modalities to trigger in situ vaccination and release tumour antigens or vaccination with dendritic cells loaded with tumour lysates or patient-specific neoantigens. In a second boost phase, personalized synthetic vaccines specifically boost T cells that were triggered during the priming phase. This immunotherapy approach has been enabled by the substantial recent improvements in dendritic cell vaccines. In this Perspective, we discuss these improvements, highlight how the prime-boost approach can be translated into clinical practice and provide solutions for various anticipated hurdles.
Collapse
Affiliation(s)
- Alexandre Harari
- Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Michele Graciotti
- Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Lana E Kandalaft
- Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland. .,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
13
|
De Vleeschouwer S. Vaccines against glioblastoma: reflections on the ICT-107 phase IIb trial. Transl Cancer Res 2020; 9:4473-4475. [PMID: 35117812 PMCID: PMC8799264 DOI: 10.21037/tcr-2020-004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/01/2020] [Indexed: 11/06/2022]
Affiliation(s)
- Steven De Vleeschouwer
- Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Leuven Brain Institute (LBI), Leuven, Belgium
| |
Collapse
|
14
|
Lechermann LM, Manavaki R, Attili B, Lau D, Jarvis LB, Fryer TD, Bird N, Aloj L, Patel N, Basu B, Cleveland M, Aigbirhio FI, Jones JL, Gallagher FA. Detection limit of 89Zr-labeled T cells for cellular tracking: an in vitro imaging approach using clinical PET/CT and PET/MRI. EJNMMI Res 2020; 10:82. [PMID: 32666311 PMCID: PMC7360010 DOI: 10.1186/s13550-020-00667-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Tracking cells in vivo using imaging can provide non-invasive information to understand the pharmacology, efficacy, and safety of novel cell therapies. Zirconium-89 (t1/2 = 78.4 h) has recently been used to synthesize [89Zr]Zr(oxinate)4 for cell tracking using positron emission tomography (PET). This work presents an in vitro approach to estimate the detection limit for in vivo PET imaging of Jurkat T cells directly labeled with [89Zr]Zr(oxinate)4 utilizing clinical PET/CT and PET/MRI. METHODS Jurkat T cells were labeled with varying concentrations of [89Zr]Zr(oxinate)4 to generate different cell-specific activities (0.43-31.91 kBq/106 cells). Different concentrations of labeled cell suspensions (104, 105, and 106 cells) were seeded on 6-well plates and into a 3 × 3 cubic-well plate with 1 cm3 cubic wells as a gel matrix. Plates were imaged on clinical PET/CT and PET/MRI scanners for 30 min. The total activity in each well was determined by drawing volumes of interest over each well on PET images. The total cell-associated activity was measured using a well counter and correlated with imaging data. Simulations for non-specific signal were performed to model the effect of non-specific radioactivity on detection. RESULTS Using this in vitro model, the lowest cell number that could be visualized on 6-well plate images was 6.8 × 104, when the specific activity was 27.8 kBq/106 cells. For the 3 × 3 cubic-well, a plate of 3.3 × 104 cells could be detected on images with a specific activity of 15.4 kBq/106 cells. CONCLUSION The results show the feasibility of detecting [89Zr]Zr(oxinate)4-labeled Jurkat T cells on clinical PET systems. The results provide a best-case scenario, as in vivo detection using PET/CT or PET/MRI will be affected by cell number, specific activity per cell, the density of cells within the target volume, and non-specific signal. This work has important implications for cell labeling studies in patients, particularly when using radiosensitive cells (e.g., T cells), which require detection of low cell numbers while minimizing radiation dose per cell.
Collapse
Affiliation(s)
- Laura M Lechermann
- Department of Radiology, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
| | - Roido Manavaki
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Bala Attili
- Department of Radiology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Doreen Lau
- Department of Radiology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Lorna B Jarvis
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Nick Bird
- Department of Nuclear Medicine, Addenbrooke's Hospital, Cambridge, UK
| | - Luigi Aloj
- Department of Radiology, University of Cambridge, Cambridge, UK
- Department of Nuclear Medicine, Addenbrooke's Hospital, Cambridge, UK
| | - Neel Patel
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Bristi Basu
- Department of Oncology, University of Cambridge, Cambridge, UK
| | | | - Franklin I Aigbirhio
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Joanne L Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
15
|
Piao YJ, Kim HS, Moon WK. Noninvasive Photoacoustic Imaging of Dendritic Cell Stimulated with Tumor Cell-Derived Exosome. Mol Imaging Biol 2020; 22:612-622. [PMID: 31385127 DOI: 10.1007/s11307-019-01410-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE The tools to trigger dendritic cell (DC) activation and to verify DC migration in vivo are important for directing DC immunotherapy toward successful treatment. We evaluated whether tumor cell-derived exosome (TEX)-stimulated DC migration into lymph node (LN) in mouse could be tracked using gold nanoparticle (GN)-labeling and ultrasound (US)-guided photoacoustic imaging (PAI). PROCEDURES GFP-transduced DC2.4 cells were used. RFP-tagged TEXs were purified from a stable 4T1 cell line overexpressing the exosomal CD63-RFP fusion protein. The TEX uptake by DCs was visualized using confocal laser scanning microscopy. GNs with surface plasmon resonance at 808 nm were used for DC-labeling. DCs that migrated into axillary LN were longitudinally monitored by US-guided PAI and analyzed by silver staining and immunohistochemistry. RESULTS TEXs were easily internalized in DCs, increased proliferation and migration capacities, and upregulated co-stimulatory molecules, CCR7 and TNF-α without cytotoxicity. The GN-labeling exerted no adverse effects on the biological functions of DCs. US-guided PAI and DC-labeling allowed for sensitive and longitudinal monitoring of TEX-stimulated DC migration toaxillary LN. CONCLUSIONS TEXs efficiently activated DCs and GN-labeled DC migration into LN was successfully monitored using US-guided PAI, suggesting that TEXs are a good source for DC activation and US-guided PAI is a cost-effective and easy-to-use imaging modality for noninvasive tracking of DCs.
Collapse
Affiliation(s)
- Yin Ji Piao
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Hoe Suk Kim
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
| | - Woo Kyung Moon
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
| |
Collapse
|
16
|
Gardner A, de Mingo Pulido Á, Ruffell B. Dendritic Cells and Their Role in Immunotherapy. Front Immunol 2020; 11:924. [PMID: 32508825 PMCID: PMC7253577 DOI: 10.3389/fimmu.2020.00924] [Citation(s) in RCA: 274] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Despite significant advances in the field of cancer immunotherapy, the majority of patients still do not benefit from treatment and must rely on traditional therapies. Dendritic cells have long been a focus of cancer immunotherapy due to their role in inducing protective adaptive immunity, but cancer vaccines have shown limited efficacy in the past. With the advent of immune checkpoint blockade and the ability to identify patient-specific neoantigens, new vaccines, and combinatorial therapies are being evaluated in the clinic. Dendritic cells are also emerging as critical regulators of the immune response within tumors. Understanding how to augment the function of these intratumoral dendritic cells could offer new approaches to enhance immunotherapy, in addition to improving the cytotoxic and targeted therapies that are partially dependent upon a robust immune response for their efficacy. Here we will discuss the role of specific dendritic cell subsets in regulating the anti-tumor immune response, as well as the current status of dendritic cell-based immunotherapies, in order to provide an overview for future lines of research and clinical trials.
Collapse
Affiliation(s)
- Alycia Gardner
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States.,Cancer Biology PhD Program, University of South Florida, Tampa, FL, United States
| | - Álvaro de Mingo Pulido
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States.,Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| |
Collapse
|
17
|
van Willigen WW, Bloemendal M, Boers-Sonderen MJ, de Groot JWB, Koornstra RHT, van der Veldt AAM, Haanen JBAG, Boudewijns S, Schreibelt G, Gerritsen WR, de Vries IJM, Bol KF. Response and survival of metastatic melanoma patients treated with immune checkpoint inhibition for recurrent disease on adjuvant dendritic cell vaccination. Oncoimmunology 2020; 9:1738814. [PMID: 33457087 PMCID: PMC7790511 DOI: 10.1080/2162402x.2020.1738814] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Vaccination with autologous dendritic cells (DC) loaded ex vivo with melanoma-associated antigens is currently being tested as an adjuvant treatment modality for resected locoregional metastatic (stage III) melanoma. Based on its mechanism of action, DC vaccination might potentiate the clinical efficacy of concurrent or sequential immune checkpoint inhibition (ICI). The purpose of this study was to determine the efficacy of ICI administered following recurrent disease during, or after, adjuvant DC vaccination. To this end, we retrospectively analyzed clinical responses of 51 melanoma patients with either irresectable stage III or stage IV disease treated with first- or second-line ICI following recurrence on adjuvant DC vaccination. Patients were analyzed according to the form of ICI administered: PD-1 inhibition monotherapy (nivolumab or pembrolizumab), ipilimumab monotherapy or combined treatment with ipilimumab and nivolumab. Treatment with first- or second-line PD-1 inhibition monotherapy after recurrence on adjuvant DC vaccination resulted in a response rate of 52%. In patients treated with ipilimumab monotherapy and ipilimumab-nivolumab response rates were 35% and 75%, respectively. In conclusion, ICI is effective in melanoma patients with recurrent disease on adjuvant DC vaccination.
Collapse
Affiliation(s)
- Wouter W van Willigen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | - Martine Bloemendal
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | | | | | - Rutger H T Koornstra
- Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands.,Department of Internal Medicine, Hospital Rijnstate, Arnhem, The Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands.,Department of Radiology & Nuclear Medicine, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - John B A G Haanen
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Steve Boudewijns
- Department of Medical Oncology, Bravis Hospital, Roosendaal, The Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | | | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | - Kalijn F Bol
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
18
|
Gierlich P, Lex V, Technau A, Keupp A, Morper L, Glunz A, Sennholz H, Rachor J, Sauer S, Marcu A, Grigoleit GU, Wölfl M, Schlegel PG, Eyrich M. Prostaglandin E 2 in a TLR3- and 7/8-agonist-based DC maturation cocktail generates mature, cytokine-producing, migratory DCs but impairs antigen cross-presentation to CD8 + T cells. Cancer Immunol Immunother 2020; 69:1029-1042. [PMID: 32100075 PMCID: PMC7223547 DOI: 10.1007/s00262-019-02470-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 12/31/2019] [Indexed: 12/21/2022]
Abstract
Mature dendritic cells (DCs) represent cellular adjuvants for optimal antigen presentation in cancer vaccines. Recently, a combination of prostaglandin E2 (PGE2) with Toll-like receptor agonists (TLR-P) was proposed as a new standard to generate superior cytokine-producing DCs with high migratory capacity. Here, we compare TLR-P DCs with conventional DCs matured only with the proinflammatory cytokines TNFα and IL-1ß (CDCs), focussing on the interaction of resulting DCs with CD8+ T-cells. TLR-P matured DCs showed elevated expression of activation markers such as CD80 and CD83 compared to CDCs, together with a significantly higher migration capacity. Secretion of IL-6, IL-8, IL-10, and IL-12 was highest after 16 h in TLR-P DCs, and only TLR-P DCs secreted active IL-12p70. TLR-P DCs as well as CDCs successfully primed multifunctional CD8+ T-cells from naïve precursors specific for the peptide antigens Melan-A, NLGN4X, and PTP with comparable priming efficacy and T-cell receptor avidity. CD8+ T-cells primed by TLR-P DCs showed significantly elevated expression of the integrin VLA-4 and a trend for higher T-cell numbers after expansion. In contrast, TLR-P DCs displayed a substantially reduced capability to cross-present CMVpp65 protein antigen to pp65-specific T cells, an effect that was dose-dependent on PGE2 during DC maturation and reproducible with several responder T-cell lines. In conclusion, TLR-P matured DCs might be optimal presenters of antigens not requiring processing such as short peptides. However, PGE2 seems less favorable for maturation of DCs intended to process and cross-present more complex vaccine antigens such as lysates, proteins or long peptides.
Collapse
Affiliation(s)
- Philipp Gierlich
- Laboratory for Stem Cell Processing and Cellular TherapyUniversity Medical Center, Children's Hospital, Würzburg, Germany
| | - Veronika Lex
- Laboratory for Stem Cell Processing and Cellular TherapyUniversity Medical Center, Children's Hospital, Würzburg, Germany
| | - Antje Technau
- Laboratory for Stem Cell Processing and Cellular TherapyUniversity Medical Center, Children's Hospital, Würzburg, Germany
| | - Anne Keupp
- Laboratory for Stem Cell Processing and Cellular TherapyUniversity Medical Center, Children's Hospital, Würzburg, Germany
| | - Lorenz Morper
- Laboratory for Stem Cell Processing and Cellular TherapyUniversity Medical Center, Children's Hospital, Würzburg, Germany
| | - Amelie Glunz
- Laboratory for Stem Cell Processing and Cellular TherapyUniversity Medical Center, Children's Hospital, Würzburg, Germany
| | - Hanno Sennholz
- Laboratory for Stem Cell Processing and Cellular TherapyUniversity Medical Center, Children's Hospital, Würzburg, Germany
| | - Johannes Rachor
- Laboratory for Stem Cell Processing and Cellular TherapyUniversity Medical Center, Children's Hospital, Würzburg, Germany
| | - Sascha Sauer
- CU Systems Medicine, University of Würzburg, Würzburg, Germany.,Max Delbrück Center for Molecular Medicine (BIMSB/BIH), Berlin, Germany
| | - Ana Marcu
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | | | - Matthias Wölfl
- Laboratory for Stem Cell Processing and Cellular TherapyUniversity Medical Center, Children's Hospital, Würzburg, Germany
| | - Paul G Schlegel
- Laboratory for Stem Cell Processing and Cellular TherapyUniversity Medical Center, Children's Hospital, Würzburg, Germany
| | - Matthias Eyrich
- Laboratory for Stem Cell Processing and Cellular TherapyUniversity Medical Center, Children's Hospital, Würzburg, Germany. .,University Children's Hospital Würzburg, Josef-Schneider-Straße 3, Building D30, 97080, Würzburg, Germany.
| |
Collapse
|
19
|
Westdorp H, Creemers JHA, van Oort IM, Schreibelt G, Gorris MAJ, Mehra N, Simons M, de Goede AL, van Rossum MM, Croockewit AJ, Figdor CG, Witjes JA, Aarntzen EHJG, Mus RDM, Brüning M, Petry K, Gotthardt M, Barentsz JO, de Vries IJM, Gerritsen WR. Blood-derived dendritic cell vaccinations induce immune responses that correlate with clinical outcome in patients with chemo-naive castration-resistant prostate cancer. J Immunother Cancer 2019; 7:302. [PMID: 31727154 PMCID: PMC6854814 DOI: 10.1186/s40425-019-0787-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Clinical benefit of cellular immunotherapy has been shown in patients with castration-resistant prostate cancer (CRPC). We investigated the immunological response and clinical outcome of vaccination with blood-derived CD1c+ myeloid dendritic cells (mDCs; cDC2) and plasmacytoid DCs (pDCs). METHODS In this randomized phase IIa trial, 21 chemo-naive CRPC patients received maximally 9 vaccinations with mature mDCs, pDCs or a combination of mDCs plus pDCs. DCs were stimulated with protamine/mRNA and loaded with tumor-associated antigens NY-ESO-1, MAGE-C2 and MUC1. Primary endpoint was the immunological response after DC vaccination, which was monitored in peripheral blood and in T cell cultures of biopsies of post-treatment delayed-type hypersensitivity-skin tests. Main secondary endpoints were safety, feasibility, radiological PFS (rPFS) and overall survival. Radiological responses were assessed by MRIs and contrast-enhanced 68Ga-prostate-specific membrane antigen PET/CT, according to RECIST 1.1, PCWG2 criteria and immune-related response criteria. RESULTS Both tetramer/dextramer-positive (dm+) and IFN-γ-producing (IFN-γ+) antigen specific T cells were detected more frequently in skin biopsies of patients with radiological non-progressive disease (5/13 patients; 38%) compared to patients with progressive disease (0/8 patients; 0%). In these patients with vaccination enhanced dm+ and IFN-γ+ antigen-specific T cells median rPFS was 18.8 months (n = 5) vs. 5.1 months (n = 16) in patients without IFN-γ-producing antigen-specific T cells (p = 0.02). The overall median rPFS was 9.5 months. All DC vaccines were well tolerated with grade 1-2 toxicity. CONCLUSIONS Immunotherapy with blood-derived DC subsets was feasible and safe and induced functional antigen-specific T cells. The presence of functional antigen-specific T cells correlated with an improved clinical outcome. TRIAL REGISTRATION ClinicalTrials.gov identifier NCT02692976, registered 26 February 2016, retrospectively registered.
Collapse
Affiliation(s)
- Harm Westdorp
- Department of Tumor Immunology and Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Geert Grooteplein 26, 6525 GA, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | - Jeroen H A Creemers
- Department of Tumor Immunology and Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Geert Grooteplein 26, 6525 GA, Nijmegen, The Netherlands
| | - Inge M van Oort
- Department of Urology, Radboudumc, Nijmegen, The Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology and Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Geert Grooteplein 26, 6525 GA, Nijmegen, The Netherlands
| | - Mark A J Gorris
- Department of Tumor Immunology and Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Geert Grooteplein 26, 6525 GA, Nijmegen, The Netherlands
| | - Niven Mehra
- Department of Tumor Immunology and Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Geert Grooteplein 26, 6525 GA, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | - Michiel Simons
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Anna L de Goede
- Department of Pharmacy, Radboudumc, Nijmegen, The Netherlands
| | | | | | - Carl G Figdor
- Department of Tumor Immunology and Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Geert Grooteplein 26, 6525 GA, Nijmegen, The Netherlands
| | - J Alfred Witjes
- Department of Urology, Radboudumc, Nijmegen, The Netherlands
| | - Erik H J G Aarntzen
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Roel D M Mus
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, The Netherlands
| | | | - Katja Petry
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Martin Gotthardt
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Jelle O Barentsz
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology and Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Geert Grooteplein 26, 6525 GA, Nijmegen, The Netherlands. .,Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands.
| | - Winald R Gerritsen
- Department of Tumor Immunology and Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Geert Grooteplein 26, 6525 GA, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
20
|
Fedorova L, Mudry P, Pilatova K, Selingerova I, Merhautova J, Rehak Z, Valik D, Hlavackova E, Cerna D, Faberova L, Mazanek P, Pavelka Z, Demlova R, Sterba J, Zdrazilova-Dubska L. Assessment of Immune Response Following Dendritic Cell-Based Immunotherapy in Pediatric Patients With Relapsing Sarcoma. Front Oncol 2019; 9:1169. [PMID: 31799177 PMCID: PMC6868036 DOI: 10.3389/fonc.2019.01169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/18/2019] [Indexed: 12/15/2022] Open
Abstract
Monocyte-derived dendritic cell (DC)-based vaccines loaded with tumor self-antigens represent a novel approach in anticancer therapy. We evaluated DC-based anticancer immunotherapy (ITx) in an academic Phase I/II clinical trial for children, adolescent, and young adults with progressive, recurrent, or primarily metastatic high-risk tumors. The primary endpoint was safety of intradermal administration of manufactured DCs. Here, we focused on relapsing high-risk sarcoma subgroup representing a major diagnosis in DC clinical trial. As a part of peripheral blood immunomonitoring, we evaluated quantitative association between basic cell-based immune parameters. Furthermore, we describe the pattern of these parameters and their time-dependent variations during the DC vaccination in the peripheral blood immunograms. The peripheral blood immunograms revealed distinct patterns in particular patients in the study group. As a functional testing, we evaluated immune response of patient T-cells to the tumor antigens presented by DCs in the autoMLR proliferation assay. This analysis was performed with T-cells obtained prior to DC ITx initiation and with T-cells collected after the fifth dose of DCs, demonstrating that the anticancer DC-based vaccine stimulates a preexisting immune response against self-tumor antigens. Finally, we present clinical and immunological findings in a Ewing's sarcoma patient with an interesting clinical course. Prior to DC therapy, we observed prevailing CD8+ T-cell stimulation and low immunosuppressive monocytic myeloid-derived suppressor cells (M-MDSC) and regulatory T-cells (Tregs). This patient was subsequently treated with 19 doses of DCs and experienced substantial regression of metastatic lesions after second disease relapse and was further rechallenged with DCs. In this patient, functional ex vivo testing of autologous T-cell activation by manufactured DC medicinal product during the course of DC ITx revealed that personalized anticancer DC-based vaccine stimulates a preexisting immune response against self-tumor antigens and that the T-cell reactivity persisted for the period without DC treatment and was further boosted by DC rechallenge. Trial Registration Number: EudraCT 2014-003388-39.
Collapse
Affiliation(s)
- Lenka Fedorova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Laboratory Medicine, Masaryk Memorial Cancer Institute, Brno, Czechia.,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Peter Mudry
- Department of Pediatric Oncology, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Katerina Pilatova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Laboratory Medicine, Masaryk Memorial Cancer Institute, Brno, Czechia.,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Iveta Selingerova
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Jana Merhautova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Zdenek Rehak
- Department of Laboratory Medicine, Masaryk Memorial Cancer Institute, Brno, Czechia.,Department of Nuclear Medicine, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Dalibor Valik
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Laboratory Medicine, Masaryk Memorial Cancer Institute, Brno, Czechia.,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Eva Hlavackova
- Department of Pediatric Oncology, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Dasa Cerna
- Department of Pediatric Oncology, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Lucie Faberova
- Department of Pediatric Oncology, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Pavel Mazanek
- Department of Pediatric Oncology, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Zdenek Pavelka
- Department of Pediatric Oncology, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Regina Demlova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Jaroslav Sterba
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Pediatric Oncology, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Lenka Zdrazilova-Dubska
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Laboratory Medicine, Masaryk Memorial Cancer Institute, Brno, Czechia.,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| |
Collapse
|
21
|
Wculek SK, Cueto FJ, Mujal AM, Melero I, Krummel MF, Sancho D. Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol 2019; 20:7-24. [PMID: 31467405 DOI: 10.1038/s41577-019-0210-z] [Citation(s) in RCA: 1444] [Impact Index Per Article: 288.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2019] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DCs) are a diverse group of specialized antigen-presenting cells with key roles in the initiation and regulation of innate and adaptive immune responses. As such, there is currently much interest in modulating DC function to improve cancer immunotherapy. Many strategies have been developed to target DCs in cancer, such as the administration of antigens with immunomodulators that mobilize and activate endogenous DCs, as well as the generation of DC-based vaccines. A better understanding of the diversity and functions of DC subsets and of how these are shaped by the tumour microenvironment could lead to improved therapies for cancer. Here we will outline how different DC subsets influence immunity and tolerance in cancer settings and discuss the implications for both established cancer treatments and novel immunotherapy strategies.
Collapse
Affiliation(s)
- Stefanie K Wculek
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Francisco J Cueto
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Adriana M Mujal
- Department of Pathology, University of California, San Francisco, CA, USA.,Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ignacio Melero
- Division of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.,University Clinic, University of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, CA, USA
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
22
|
Brown LV, Gaffney EA, Wagg J, Coles MC. An in silico model of cytotoxic T-lymphocyte activation in the lymph node following short peptide vaccination. J R Soc Interface 2019. [PMID: 29540543 DOI: 10.1098/rsif.2018.0041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Tumour immunotherapy is dependent upon activation and expansion of tumour-targetting immune cells, known as cytotoxic T-lymphocytes (CTLs). Cancer vaccines developed in the past have had limited success and the mechanisms resulting in failure are not well characterized. To elucidate these mechanisms, we developed a human-parametrized, in silico, agent-based model of vaccination-driven CTL activation within a clinical short-peptide vaccination context. The simulations predict a sharp transition in the probability of CTL activation, which occurs with variation in the separation rate (or off-rate) of tumour-specific immune response-inducing peptides (cognate antigen) from the major histocompatibility class I (MHC-I) receptors of dendritic cells (DCs) originally at the vaccination site. For peptides with MHC-I off-rates beyond this transition, it is predicted that no vaccination strategy will lead to successful expansion of CTLs. For slower off-rates, below the transition, the probability of CTL activation becomes sensitive to the numbers of DCs and T cells that interact subsequent to DC migration to the draining lymph node of the vaccination site. Thus, the off-rate is a key determinant of vaccine design.
Collapse
Affiliation(s)
- Liam V Brown
- Wolfson Centre For Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Eamonn A Gaffney
- Wolfson Centre For Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Jonathan Wagg
- Clinical Pharmacology, Roche Innovation Center Basel, Basel, Switzerland
| | - Mark C Coles
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
23
|
Koshkina O, Lajoinie G, Bombelli FB, Swider E, Cruz LJ, White PB, Schweins R, Dolen Y, van Dinther EAW, van Riessen NK, Rogers SE, Fokkink R, Voets IK, van Eck ERH, Heerschap A, Versluis M, de Korte CL, Figdor CG, de Vries IJM, Srinivas M. Multicore Liquid Perfluorocarbon-Loaded Multimodal Nanoparticles for Stable Ultrasound and 19F MRI Applied to In Vivo Cell Tracking. ADVANCED FUNCTIONAL MATERIALS 2019; 29:1806485. [PMID: 32132881 PMCID: PMC7056356 DOI: 10.1002/adfm.201806485] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Indexed: 05/22/2023]
Abstract
Ultrasound is the most commonly used clinical imaging modality. However, in applications requiring cell-labeling, the large size and short active lifetime of ultrasound contrast agents limit their longitudinal use. Here, 100 nm radius, clinically applicable, polymeric nanoparticles containing a liquid perfluorocarbon, which enhance ultrasound contrast during repeated ultrasound imaging over the course of at least 48 h, are described. The perfluorocarbon enables monitoring the nanoparticles with quantitative 19F magnetic resonance imaging, making these particles effective multimodal imaging agents. Unlike typical core-shell perfluorocarbon-based ultrasound contrast agents, these nanoparticles have an atypical fractal internal structure. The nonvaporizing highly hydrophobic perfluorocarbon forms multiple cores within the polymeric matrix and is, surprisingly, hydrated with water, as determined from small-angle neutron scattering and nuclear magnetic resonance spectroscopy. Finally, the nanoparticles are used to image therapeutic dendritic cells with ultrasound in vivo, as well as with 19F MRI and fluorescence imaging, demonstrating their potential for long-term in vivo multimodal imaging.
Collapse
Affiliation(s)
- Olga Koshkina
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands; Physical Chemistry of Polymers, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Guillaume Lajoinie
- Physics of Fluids Group, Technical Medical (TechMed) Centre and MESA+ Institute for, Nanotechnology, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Francesca Baldelli Bombelli
- Laboratory of Supramolecular and BioNano Materials, (SupraBioNanoLab), Department of Chemistry, Materials, and Chemical Engineering, "Giulio Natta,", Politecnico di Milano, Via Luigi Mancinelli 7, 20131 Milan, Italy
| | - Edyta Swider
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Paul B White
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Ralf Schweins
- Institut Laue - Langevin, DS/LSS, 71 Avenue des Martyrs, CS 20 156, 38042 Grenoble CEDEX 9, France
| | - Yusuf Dolen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Eric A W van Dinther
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - N Koen van Riessen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Sarah E Rogers
- ISIS Pulsed Neutron and Muon Source, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Harwell, Oxford OX11 0QX, UK
| | - Remco Fokkink
- Department of Agrotechnology and Food Sciences, Physical Chemistry and Soft Matter, Wageningen University, 6708 WE, Wageningen, Netherlands
| | - Ilja K Voets
- Laboratory of Self-Organizing Soft Matter, Laboratory of Macromolecular and Organic Chemistry, Department of Chemical Engineering and Chemistry and Institute for Complex Molecular Systems, Eindhoven University of Technology, De Rondom 70, 5612 AP, Eindhoven, The Netherlands
| | - Ernst R H van Eck
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Michel Versluis
- Physics of Fluids Group, Technical Medical (TechMed) Centre and MESA+ Institute for, Nanotechnology, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Chris L de Korte
- Physics of Fluids Group, Technical Medical (TechMed) Centre and MESA+ Institute for, Nanotechnology, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands; Department of Radiology and Nuclear Medicine, Radboudumc, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Lee H, Lee HW, La Lee Y, Jeon YH, Jeong SY, Lee SW, Lee J, Ahn BC. Optimization of Dendritic Cell-Mediated Cytotoxic T-Cell Activation by Tracking of Dendritic Cell Migration Using Reporter Gene Imaging. Mol Imaging Biol 2019; 20:398-406. [PMID: 29027077 DOI: 10.1007/s11307-017-1127-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE The aim of this study is to optimize the dendritic cell (DC)-mediated T-cell activation using reporter gene imaging and flow cytometric analysis in living mice. PROCEDURES A murine dendritic cell line (DC2.4) co-expressing effluc and Thy1.1 genes were established by transfection with retroviral vectors. Thy1.1 positive cells were sorted by magnetic bead separation system (DC2.4/effluc). Cell proliferation assay and phenotype analysis to determine the effects of gene transduction on the function of dendritic cells between parental DC2.4 and DC2.4/effluc were performed. To optimize the DC-mediated immune response by cell number or frequency, different cell numbers (5 × 105, 1 × 106, and 2 × 106 DC2.4/effluc) or different frequencies of DC2.4/effluc (first, second, and third injections) were injected in the right footpad of mice. The migration of the DC2.4/effluc into the draining popliteal lymph node of mice was monitored by bioluminescence imaging (BLI). Flow cytometric analysis was performed with splenocytes to determine the cytotoxic T-cell population after injection of DC2.4/effluc. RESULTS Parental DC2.4 and DC2.4/effluc exhibit no significant differences in their proliferation and phenotype. BLI signals were observed in the draining popliteal lymph node at day 1 after injection of DC2.4/effluc in 1 × 106 and 2 × 106 cells-injected groups. The highest BLI signal intensity was detected in 2 × 106 cells-injected mice. On day 11, the BLI signal was detected in only 2 × 106 cell-injected group but not in other groups. Optimized cell numbers (2 × 106) were injected in three animal groups with a different frequency (first, second, and third injection groups). The BLI signal was detected at day 1 and maintained until day 7 in the first injection group, but there is low signal intensity in the second and the third injection groups. Although the expression levels of Thy1.1 gene in the first injection group were very high, there reveals no expression of Thy1.1 gene in the second and the third injection groups. The number of tumor-specific CD8+ T-cells in the spleen significantly increased, as the number of DC injections increases. CONCLUSIONS Successful optimization of DC-mediated cytotoxic T-cell activation in living mice using reporter gene imaging and flow cytometric analysis was achieved. The optimization of DC-mediated cytotoxic T-cell activation could be applied for the future DC-based immunotherapy.
Collapse
Affiliation(s)
- Hongje Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea.,Department of Nuclear Medicine, Dongnam Institution of Radiological & Medical Sciences (DIRAMS), Busan, South Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea
| | - You La Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea
| | - Yong Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea.,Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50, Samduk-dong 2-ga, Jung Gu, Daegu, 700-721, South Korea.
| |
Collapse
|
25
|
Martin Lluesma S, Graciotti M, Chiang CLL, Kandalaft LE. Does the Immunocompetent Status of Cancer Patients Have an Impact on Therapeutic DC Vaccination Strategies? Vaccines (Basel) 2018; 6:E79. [PMID: 30477198 PMCID: PMC6313858 DOI: 10.3390/vaccines6040079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/09/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022] Open
Abstract
Although different types of therapeutic vaccines against established cancerous lesions in various indications have been developed since the 1990s, their clinical benefit is still very limited. This observed lack of effectiveness in cancer eradication may be partially due to the often deficient immunocompetent status of cancer patients, which may facilitate tumor development by different mechanisms, including immune evasion. The most frequently used cellular vehicle in clinical trials are dendritic cells (DCs), thanks to their crucial role in initiating and directing immune responses. Viable vaccination options using DCs are available, with a positive toxicity profile. For these reasons, despite their limited therapeutic outcomes, DC vaccination is currently considered an additional immunotherapeutic option that still needs to be further explored. In this review, we propose potential actions aimed at improving DC vaccine efficacy by counteracting the detrimental mechanisms recognized to date and implicated in establishing a poor immunocompetent status in cancer patients.
Collapse
Affiliation(s)
- Silvia Martin Lluesma
- Center of Experimental Therapeutics, Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
| | - Michele Graciotti
- Vaccine development laboratory, Ludwig Center for Cancer Research, Lausanne 1011, Switzerland.
| | - Cheryl Lai-Lai Chiang
- Vaccine development laboratory, Ludwig Center for Cancer Research, Lausanne 1011, Switzerland.
| | - Lana E Kandalaft
- Center of Experimental Therapeutics, Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
- Vaccine development laboratory, Ludwig Center for Cancer Research, Lausanne 1011, Switzerland.
| |
Collapse
|
26
|
Fruhwirth GO, Kneilling M, de Vries IJM, Weigelin B, Srinivas M, Aarntzen EHJG. The Potential of In Vivo Imaging for Optimization of Molecular and Cellular Anti-cancer Immunotherapies. Mol Imaging Biol 2018; 20:696-704. [PMID: 30030697 PMCID: PMC6153672 DOI: 10.1007/s11307-018-1254-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This review aims to emphasize the potential of in vivo imaging to optimize current and upcoming anti-cancer immunotherapies: spanning from preclinical to clinical applications. Immunotherapies are an emerging class of treatments for a variety of diseases. The agents include molecular and cellular therapeutics, which aim to treat the disease through re-education of the host immune system, often via complex mechanisms of action. In vivo imaging has the potential to contribute in several different ways: (1) as a drug development tool to improve our understanding of their complex mechanisms of action, (2) as a tool to predict efficacy, for example, to stratify patients into probable responders and likely non-responders, and (3) as a non-invasive treatment response biomarker to guide efficient immunotherapy use and to recognize early signs of potential loss of efficacy or resistance in patients. Areas where in vivo imaging is already successfully implemented in onco-immunology research will be discussed and domains where its use offers great potential will be highlighted. The focus of this article is on anti-cancer immunotherapy as it currently is the most advanced immunotherapy area. However, the described concepts can also be paralleled in other immune-mediated disorders and for conditions requiring immunotherapeutic intervention. Importantly, we introduce a new study group within the European Society of Molecular Imaging with the goal to facilitate and enhance immunotherapy development through the use of in vivo imaging.
Collapse
Affiliation(s)
- Gilbert O Fruhwirth
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Kings' College London, London, UK
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, Tuebingen, Germany
- Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Bettina Weigelin
- Genitourinary Medical Oncology and Koch Center, MD Anderson Cancer Center, Houston, USA
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Erik H J G Aarntzen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein 10, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
27
|
Schineis P, Runge P, Halin C. Cellular traffic through afferent lymphatic vessels. Vascul Pharmacol 2018; 112:31-41. [PMID: 30092362 DOI: 10.1016/j.vph.2018.08.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/26/2018] [Accepted: 08/01/2018] [Indexed: 12/15/2022]
Abstract
The lymphatic system has long been known to serve as a highway for migrating leukocytes from peripheral tissue to draining lymph nodes (dLNs) and back to circulation, thereby contributing to the induction of adaptive immunity and immunesurveillance. Lymphatic vessels (LVs) present in peripheral tissues upstream of a first dLN are generally referred to as afferent LVs. In contrast to migration through blood vessels (BVs), the detailed molecular and cellular requirements of cellular traffic through afferent LVs have only recently started to be unraveled. Progress in our ability to track the migration of lymph-borne cell populations, in combination with cutting-edge imaging technologies, nowadays allows the investigation and visualization of lymphatic migration of endogenous leukocytes, both at the population and at the single-cell level. These studies have revealed that leukocyte trafficking through afferent LVs generally follows a step-wise migration pattern, relying on the active interplay of numerous molecules. In this review, we will summarize and discuss current knowledge of cellular traffic through afferent LVs. We will first outline how the structure of the afferent LV network supports leukocyte migration and highlight important molecules involved in the migration of dendritic cells (DCs), T cells and neutrophils, i.e. the most prominent cell types trafficking through afferent LVs. Additionally, we will describe how tumor cells hijack the lymphatic system for their dissemination to draining LNs. Finally, we will summarize and discuss our current understanding of the functional significance as well as the therapeutic implications of cell traffic through afferent LVs.
Collapse
Affiliation(s)
| | - Peter Runge
- Institute of Pharmaceutical Sciences, ETH Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, Switzerland.
| |
Collapse
|
28
|
Subbiah V, Murthy R, Hong DS, Prins RM, Hosing C, Hendricks K, Kolli D, Noffsinger L, Brown R, McGuire M, Fu S, Piha-Paul S, Naing A, Conley AP, Benjamin RS, Kaur I, Bosch ML. Cytokines Produced by Dendritic Cells Administered Intratumorally Correlate with Clinical Outcome in Patients with Diverse Cancers. Clin Cancer Res 2018; 24:3845-3856. [PMID: 30018119 DOI: 10.1158/1078-0432.ccr-17-2707] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/05/2018] [Accepted: 05/07/2018] [Indexed: 01/08/2023]
Abstract
Purpose: Dendritic cells (DC) initiate adaptive immune responses through the uptake and presentation of antigenic material. In preclinical studies, intratumorally injected activated DCs (aDCs; DCVax-Direct) were superior to immature DCs in rejecting tumors from mice.Experimental Design: This single-arm, open-label phase I clinical trial evaluated the safety and efficacy of aDCs, administered intratumorally, in patients with solid tumors. Three dose levels (2 million, 6 million, and 15 million aDCs per injection) were tested using a standard 3 + 3 dose-escalation trial design. Feasibility, immunogenicity, changes to the tumor microenvironment after direct injection, and survival were evaluated. We also investigated cytokine production of aDCs prior to injection.Results: In total, 39 of the 40 enrolled patients were evaluable. The injections of aDCs were well tolerated with no dose-limiting toxicities. Increased lymphocyte infiltration was observed in 54% of assessed patients. Stable disease (SD; best response) at week 8 was associated with increased overall survival. Increased secretion of interleukin (IL)-8 and IL12p40 by aDCs was significantly associated with survival (P = 0.023 and 0.024, respectively). Increased TNFα levels correlated positively with SD at week 8 (P < 0.01).Conclusions: Intratumoral aDC injections were feasible and safe. Increased production of specific cytokines was correlated with SD and prolonged survival, demonstrating a link between the functional profile of aDCs prior to injection and patient outcomes. Clin Cancer Res; 24(16); 3845-56. ©2018 AACR.
Collapse
Affiliation(s)
- Vivek Subbiah
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ravi Murthy
- Department of Interventional Radiology, Division of Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David S Hong
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert M Prins
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Chitra Hosing
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | - Robert Brown
- Department of Pathology and Laboratory Medicine, UT Health, University of Texas Health Science Center, Houston, Texas
| | - Mary McGuire
- Department of Pathology and Laboratory Medicine, UT Health, University of Texas Health Science Center, Houston, Texas
| | - Siquing Fu
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarina Piha-Paul
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anthony P Conley
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert S Benjamin
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Indreshpal Kaur
- Cell Therapy Labs, GMP Laboratory, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
29
|
Bryant CE, Sutherland S, Kong B, Papadimitrious MS, Fromm PD, Hart DNJ. Dendritic cells as cancer therapeutics. Semin Cell Dev Biol 2018; 86:77-88. [PMID: 29454038 DOI: 10.1016/j.semcdb.2018.02.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/14/2017] [Accepted: 02/10/2018] [Indexed: 02/06/2023]
Abstract
The ability of immune therapies to control cancer has recently generated intense interest. This therapeutic outcome is reliant on T cell recognition of tumour cells. The natural function of dendritic cells (DC) is to generate adaptive responses, by presenting antigen to T cells, hence they are a logical target to generate specific anti-tumour immunity. Our understanding of the biology of DC is expanding, and they are now known to be a family of related subsets with variable features and function. Most clinical experience to date with DC vaccination has been using monocyte-derived DC vaccines. There is now growing experience with alternative blood-derived DC derived vaccines, as well as with multiple forms of tumour antigen and its loading, a wide range of adjuvants and different modes of vaccine delivery. Key insights from pre-clinical studies, and lessons learned from early clinical testing drive progress towards improved vaccines. The potential to fortify responses with other modalities of immunotherapy makes clinically effective "second generation" DC vaccination strategies a priority for cancer immune therapists.
Collapse
Affiliation(s)
- Christian E Bryant
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW Australia; Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia.
| | - Sarah Sutherland
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia
| | - Benjamin Kong
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia
| | - Michael S Papadimitrious
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia
| | - Phillip D Fromm
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia
| | - Derek N J Hart
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW Australia; Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia.
| |
Collapse
|
30
|
De Laere M, Derdelinckx J, Hassi M, Kerosalo M, Oravamäki H, Van den Bergh J, Berneman Z, Cools N. Shuttling Tolerogenic Dendritic Cells across the Blood-Brain Barrier In Vitro via the Introduction of De Novo C-C Chemokine Receptor 5 Expression Using Messenger RNA Electroporation. Front Immunol 2018; 8:1964. [PMID: 29403473 PMCID: PMC5778265 DOI: 10.3389/fimmu.2017.01964] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/19/2017] [Indexed: 01/06/2023] Open
Abstract
The use of tolerance-inducing dendritic cells (tolDCs) has been proven to be safe and well tolerated in the treatment of autoimmune diseases. Nevertheless, several challenges remain, including finding ways to facilitate the migration of cell therapeutic products to lymph nodes, and the site of inflammation. In the treatment of neuroinflammatory diseases, such as multiple sclerosis (MS), the blood-brain barrier (BBB) represents a major obstacle to the delivery of therapeutic agents to the inflamed central nervous system (CNS). As it was previously demonstrated that C-C chemokine receptor 5 (CCR5) may be involved in inflammatory migration of DCs, the aim of this study was to investigate CCR5-driven migration of tolDCs. Only a minority of in vitro generated vitamin D3 (vitD3)-treated tolDCs expressed the inflammatory chemokine receptor CCR5. Thus, messenger RNA (mRNA) encoding CCR5 was introduced by means of electroporation (EP). After mRNA EP, tolDCs transiently displayed increased levels of CCR5 protein expression. Accordingly, the capacity of mRNA electroporated tolDCs to transmigrate toward a chemokine gradient in an in vitro model of the BBB improved significantly. Neither the tolerogenic phenotype nor the T cell-stimulatory function of tolDCs was affected by mRNA EP. EP of tolDCs with mRNA encoding CCR5 enabled these cells to migrate to inflammatory sites. The approach used herein has important implications for the treatment of MS. Using this approach, tolDCs actively shuttle across the BBB, allowing in situ down-modulation of autoimmune responses in the CNS.
Collapse
Affiliation(s)
- Maxime De Laere
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| | - Judith Derdelinckx
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium.,Department of Neurology, Antwerp University Hospital, Edegem, Belgium
| | - Mari Hassi
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| | - Mari Kerosalo
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| | - Heidi Oravamäki
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| | - Johan Van den Bergh
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
31
|
Berraondo P, Labiano S, Minute L, Etxeberria I, Vasquez M, Sanchez-Arraez A, Teijeira A, Melero I. Cellular immunotherapies for cancer. Oncoimmunology 2017. [PMID: 28638729 DOI: 10.1080/2162402x.2017.1306619] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Lessons learned over decades on the use of gene and cell therapies have found clinical applicability in the field of cancer immunotherapy. On December 16th, 2016 a symposium was held in Pamplona (Spain) to analyze and discuss the critical points for the clinical success of adoptive cell transfer strategies in cancer immunotherapy. Cellular immunotherapy is being currently exploited for the development of new cancer vaccines using ex vivo manipulated dendritic cells or to enhance the number of effector cells, transferring reinvigorated NK cells or T cells. In this meeting report, we summarize the main topics covered and provide an overview of the field of cellular immunotherapy.
Collapse
Affiliation(s)
- Pedro Berraondo
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | - Sara Labiano
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Luna Minute
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Iñaki Etxeberria
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Marcos Vasquez
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Alvaro Sanchez-Arraez
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Alvaro Teijeira
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain.,Servicio de Inmunología e Inmunoterapia, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
32
|
Abstract
Immunotherapy using dendritic cell (DC)-based vaccination is an approved approach for harnessing the potential of a patient's own immune system to eliminate tumor cells in metastatic hormone-refractory cancer. Overall, although many DC vaccines have been tested in the clinic and proven to be immunogenic, and in some cases associated with clinical outcome, there remains no consensus on how to manufacture DC vaccines. In this review we will discuss what has been learned thus far about human DC biology from clinical studies, and how current approaches to apply DC vaccines in the clinic could be improved to enhance anti-tumor immunity.
Collapse
|
33
|
Dillman RO, Nistor GI, Cornforth AN. Dendritic cell vaccines for melanoma: past, present and future. Melanoma Manag 2016; 3:273-289. [PMID: 30190899 PMCID: PMC6094661 DOI: 10.2217/mmt-2016-0014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 07/19/2016] [Indexed: 02/06/2023] Open
Abstract
Administering dendritic cells (DC) loaded with tumor-associated antigens (TAA) ex vivo is a promising strategy for therapeutic vaccines in advanced melanoma. To date the induction of immune responses to specific TAA has been more impressive than clinical benefit because of TAA limitations, suboptimal DC and possibly immune-checkpoint inhibition. Various products, antigen-loading techniques, treatment schedules, routes of administration and adjunctive agents continue to be explored. Biologic heterogeneity suggests autologous tumor as the optimal TAA source to induce immune responses to the entire repertoire of unique patient-specific neoantigens. Many questions remain regarding the optimal preparation of DC and strategies for antigen loading. Effective DC vaccines should result in additive or synergistic effects when combined with checkpoint inhibitors.
Collapse
Affiliation(s)
- Robert O Dillman
- AiVita Biomedical, Inc., 18301 Von Karman Avenue, Suite 130, Clinical, Research, and Manufacturing Departments, Irvine, CA 92612, USA
| | - Gabriel I Nistor
- AiVita Biomedical, Inc., 18301 Von Karman Avenue, Suite 130, Clinical, Research, and Manufacturing Departments, Irvine, CA 92612, USA
| | - Andrew N Cornforth
- AiVita Biomedical, Inc., 18301 Von Karman Avenue, Suite 130, Clinical, Research, and Manufacturing Departments, Irvine, CA 92612, USA
| |
Collapse
|
34
|
Seyfizadeh N, Muthuswamy R, Mitchell DA, Nierkens S, Seyfizadeh N. Migration of dendritic cells to the lymph nodes and its enhancement to drive anti-tumor responses. Crit Rev Oncol Hematol 2016; 107:100-110. [PMID: 27823637 DOI: 10.1016/j.critrevonc.2016.09.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 09/02/2016] [Accepted: 09/06/2016] [Indexed: 12/29/2022] Open
Abstract
Better prognoses associated with increased T cell infiltration of tumors, as seen with chimeric antigen receptor (CAR) T cell therapies and immune checkpoint inhibitors, portray the importance and potential of the immune system in controlling tumors. This has rejuvenated the field of cancer immunotherapy leading to an increasing number of immunotherapies developed for cancer patients. Dendritic Cells (DCs) vaccines represent an appealing option for cancer immunotherapy since DCs have the ability to circumvent tolerance to tumors by its adjuvant properties and to induce memory T cells that can become persistent after initial tumor clearance to engage potential metastatic tumors. In the past, DC-based cancer vaccines have elicited only poor clinical response in cancer patients, which can be attributed to complex and a multitude of issues associated with generation, implementing, delivery of DC vaccine and their potential interaction with effector cells. The current review mainly focuses on migration/trafficking of DCs, as one of the key issues that affect the success of DC-based cancer vaccines, and discusses strategies to enhance it for cancer immunotherapy. Additionally, impact of maturation, route of DC delivery and negative effects of tumor microenvironment (TME) on DC homing to LN are reviewed. Moreover, strategies to increase the expression of genes involved in Lymph node homing, preconditioning of the vaccination site, enhancing lymph node ability to attract and receive DCs, while limiting negative impact of TME on DC migration are discussed.
Collapse
Affiliation(s)
- Narges Seyfizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Duane A Mitchell
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Stefan Nierkens
- Laboratory of Translational Immunology, U-DAIR, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nayer Seyfizadeh
- Umbilical Cord Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Biochemistry and Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
35
|
Ku MC, Edes I, Bendix I, Pohlmann A, Waiczies H, Prozorovski T, Günther M, Martin C, Pagès G, Wolf SA, Kettenmann H, Uckert W, Niendorf T, Waiczies S. ERK1 as a Therapeutic Target for Dendritic Cell Vaccination against High-Grade Gliomas. Mol Cancer Ther 2016; 15:1975-87. [PMID: 27256374 DOI: 10.1158/1535-7163.mct-15-0850] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 05/23/2016] [Indexed: 11/16/2022]
Abstract
Glioma regression requires the recruitment of potent antitumor immune cells into the tumor microenvironment. Dendritic cells (DC) play a role in immune responses to these tumors. The fact that DC vaccines do not effectively combat high-grade gliomas, however, suggests that DCs need to be genetically modified specifically to promote their migration to tumor relevant sites. Previously, we identified extracellular signal-regulated kinase (ERK1) as a regulator of DC immunogenicity and brain autoimmunity. In the current study, we made use of modern magnetic resonance methods to study the role of ERK1 in regulating DC migration and tumor progression in a model of high-grade glioma. We found that ERK1-deficient mice are more resistant to the development of gliomas, and tumor growth in these mice is accompanied by a higher infiltration of leukocytes. ERK1-deficient DCs exhibit an increase in migration that is associated with sustained Cdc42 activation and increased expression of actin-associated cytoskeleton-organizing proteins. We also demonstrated that ERK1 deletion potentiates DC vaccination and provides a survival advantage in high-grade gliomas. Considering the therapeutic significance of these results, we propose ERK1-deleted DC vaccines as an additional means of eradicating resilient tumor cells and preventing tumor recurrence. Mol Cancer Ther; 15(8); 1975-87. ©2016 AACR.
Collapse
Affiliation(s)
- Min-Chi Ku
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Inan Edes
- Department of Molecular Cell Biology and Gene Therapy, Humboldt-University Berlin and Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ivo Bendix
- Department of Pediatrics I, Neonatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Andreas Pohlmann
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | | | - Tim Prozorovski
- Department of Neurology, Heinrich Heine University, Düsseldorf, Germany
| | - Martin Günther
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | | | - Gilles Pagès
- University Nice-Sophia Antipolis, Institute for Research on Cancer and Aging of Nice (IRCAN), Nice, France
| | - Susanne A Wolf
- Department of Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Helmut Kettenmann
- Department of Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Wolfgang Uckert
- Department of Molecular Cell Biology and Gene Therapy, Humboldt-University Berlin and Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sonia Waiczies
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
| |
Collapse
|
36
|
Stanton SE, Eary JF, Marzbani EA, Mankoff D, Salazar LG, Higgins D, Childs J, Reichow J, Dang Y, Disis ML. Concurrent SPECT/PET-CT imaging as a method for tracking adoptively transferred T-cells in vivo. J Immunother Cancer 2016; 4:27. [PMID: 27190628 PMCID: PMC4869363 DOI: 10.1186/s40425-016-0131-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/15/2016] [Indexed: 12/17/2022] Open
Abstract
Background The ability of T-cells to traffic to and penetrate tumors impacts the clinical efficacy of T-cell therapy therefore methods to track transferred T-cells in vivo are needed. In this preliminary report, we evaluated the use of concurrent SPECT/PET-CT imaging to monitor the egress of HER-2/neu specific T-cells in a breast cancer patient with extensive bone-only metastatic disease. Findings Indium (In-111) labeled T-cells demonstrated similar or greater viability than unlabeled T-cells at either a low or high dose of In-111 over a 24-h incubation period in vitro. The function of labeled or unlabeled T-cells was not significantly different (p > 0.05) at either dose. T-cells trafficked to all sites of metastatic disease and infiltrated the tumor as assessed by SPECT imaging. In-111 uptake at 24 h after infusion varied from 3.8 (right proximal humerus) to 6.3 (right sacrum) background corrected counts per pixel and remained elevated at 48 h. Concurrent PET-CT imaging demonstrated a fluorodeoxyglucose flare, measured by increase in tumor site uptake as high as 32 % and at most sites of disease at 48 h. This flare was associated with focal pain after T-cell infusion at metastatic sites. The patient had stable disease for 18 months after completion of T-cell therapy. Conclusion Concurrent SPECT/PET-CT imaging, over a 48-h period after T-cell infusion, provided evidence of T-cell homing to all disease sites as well as a tumor metabolism flare response. This technique may be useful for monitoring T-cell trafficking after autologous as well as chimeric antigen receptor T-cell infusion. Trial Registraion Trial registered at ClinicalTrials.gov registration number NCT00791037, registered 13 November 2008.
Collapse
Affiliation(s)
- Sasha E Stanton
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - Janet F Eary
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35249 USA
| | - Edmond A Marzbani
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - David Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Lupe G Salazar
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - Doreen Higgins
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - Jennifer Childs
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - Jessica Reichow
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - Yushe Dang
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| |
Collapse
|
37
|
Benham H, Nel HJ, Law SC, Mehdi AM, Street S, Ramnoruth N, Pahau H, Lee BT, Ng J, Brunck MEG, Hyde C, Trouw LA, Dudek NL, Purcell AW, O'Sullivan BJ, Connolly JE, Paul SK, Lê Cao KA, Thomas R. Citrullinated peptide dendritic cell immunotherapy in HLA risk genotype-positive rheumatoid arthritis patients. Sci Transl Med 2016; 7:290ra87. [PMID: 26041704 DOI: 10.1126/scitranslmed.aaa9301] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In animals, immunomodulatory dendritic cells (DCs) exposed to autoantigen can suppress experimental arthritis in an antigen-specific manner. In rheumatoid arthritis (RA), disease-specific anti-citrullinated peptide autoantibodies (ACPA or anti-CCP) are found in the serum of about 70% of RA patients and are strongly associated with HLA-DRB1 risk alleles. This study aimed to explore the safety and biological and clinical effects of autologous DCs modified with a nuclear factor κB (NF-κB) inhibitor exposed to four citrullinated peptide antigens, designated "Rheumavax," in a single-center, open-labeled, first-in-human phase 1 trial. Rheumavax was administered once intradermally at two progressive dose levels to 18 human leukocyte antigen (HLA) risk genotype-positive RA patients with citrullinated peptide-specific autoimmunity. Sixteen RA patients served as controls. Rheumavax was well tolerated: adverse events were grade 1 (of 4) severity. At 1 month after treatment, we observed a reduction in effector T cells and an increased ratio of regulatory to effector T cells; a reduction in serum interleukin-15 (IL-15), IL-29, CX3CL1, and CXCL11; and reduced T cell IL-6 responses to vimentin(447-455)-Cit450 relative to controls. Rheumavax did not induce disease flares in patients recruited with minimal disease activity, and DAS28 decreased within 1 month in Rheumavax-treated patients with active disease. This exploratory study demonstrates safety and biological activity of a single intradermal injection of autologous modified DCs exposed to citrullinated peptides, and provides rationale for further studies to assess clinical efficacy and antigen-specific effects of autoantigen immunomodulatory therapy in RA.
Collapse
Affiliation(s)
- Helen Benham
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia. University of Queensland School of Medicine, Brisbane, Queensland 4102, Australia
| | - Hendrik J Nel
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - Soi Cheng Law
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - Ahmed M Mehdi
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - Shayna Street
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - Nishta Ramnoruth
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - Helen Pahau
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - Bernett T Lee
- Singapore Immunology Network, Agency for Science, Technology and Research, 8A Biomedical Grove, Immunos Building, Level 3, Biopolis, 138673 Singapore, Singapore
| | - Jennifer Ng
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - Marion E G Brunck
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - Claire Hyde
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - Leendert A Trouw
- Department of Rheumatology, Leiden University Medical Center, Leiden 2333, Netherlands
| | - Nadine L Dudek
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Anthony W Purcell
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Brendan J O'Sullivan
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - John E Connolly
- Singapore Immunology Network, Agency for Science, Technology and Research, 8A Biomedical Grove, Immunos Building, Level 3, Biopolis, 138673 Singapore, Singapore
| | - Sanjoy K Paul
- Queensland Clinical Trials & Biostatistics Centre, School of Population Health, The University of Queensland, Brisbane, Queensland 4006, Australia
| | - Kim-Anh Lê Cao
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - Ranjeny Thomas
- University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia.
| |
Collapse
|
38
|
Bol KF, Aarntzen EHJG, Pots JM, Olde Nordkamp MAM, van de Rakt MWMM, Scharenborg NM, de Boer AJ, van Oorschot TGM, Croockewit SAJ, Blokx WAM, Oyen WJG, Boerman OC, Mus RDM, van Rossum MM, van der Graaf CAA, Punt CJA, Adema GJ, Figdor CG, de Vries IJM, Schreibelt G. Prophylactic vaccines are potent activators of monocyte-derived dendritic cells and drive effective anti-tumor responses in melanoma patients at the cost of toxicity. Cancer Immunol Immunother 2016; 65:327-39. [PMID: 26861670 PMCID: PMC4779136 DOI: 10.1007/s00262-016-1796-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/11/2016] [Indexed: 01/22/2023]
Abstract
Dendritic cell (DC)-based immunotherapy is explored worldwide in cancer patients, predominantly with DC matured with pro-inflammatory cytokines and prostaglandin E2. We studied the safety and efficacy of vaccination with monocyte-derived DC matured with a cocktail of prophylactic vaccines that contain clinical-grade Toll-like receptor ligands (BCG, Typhim, Act-HIB) and prostaglandin E2 (VAC-DC). Stage III and IV melanoma patients were vaccinated via intranodal injection (12 patients) or combined intradermal/intravenous injection (16 patients) with VAC-DC loaded with keyhole limpet hemocyanin (KLH) and mRNA encoding tumor antigens gp100 and tyrosinase. Tumor antigen-specific T cell responses were monitored in blood and skin-test infiltrating-lymphocyte cultures. Almost all patients mounted prophylactic vaccine- or KLH-specific immune responses. Both after intranodal injection and after intradermal/intravenous injection, tumor antigen-specific immune responses were detected, which coincide with longer overall survival in stage IV melanoma patients. VAC-DC induce local and systemic CTC grade 2 and 3 toxicity, which is most likely caused by BCG in the maturation cocktail. The side effects were self-limiting or resolved upon a short period of systemic steroid therapy. We conclude that VAC-DC can induce functional tumor-specific responses. Unfortunately, toxicity observed after vaccination precludes the general application of VAC-DC, since in DC maturated with prophylactic vaccines BCG appears to be essential in the maturation cocktail.
Collapse
Affiliation(s)
- Kalijn F Bol
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Erik H J G Aarntzen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jeanette M Pots
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Michel A M Olde Nordkamp
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Mandy W M M van de Rakt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Nicole M Scharenborg
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Annemiek J de Boer
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Tom G M van Oorschot
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Sandra A J Croockewit
- Department of Hematology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Willeke A M Blokx
- Department of Pathology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Wim J G Oyen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Roel D M Mus
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Michelle M van Rossum
- Department of Dermatology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | - Cornelis J A Punt
- Department of Medical Oncology, Academic Medical Centre, Amsterdam, The Netherlands
| | - Gosse J Adema
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
39
|
Constantino J, Gomes C, Falcão A, Cruz MT, Neves BM. Antitumor dendritic cell-based vaccines: lessons from 20 years of clinical trials and future perspectives. Transl Res 2016; 168:74-95. [PMID: 26297944 DOI: 10.1016/j.trsl.2015.07.008] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 07/25/2015] [Accepted: 07/28/2015] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are versatile elements of the immune system and are best known for their unparalleled ability to initiate and modulate adaptive immune responses. During the past few decades, DCs have been the subject of numerous studies seeking new immunotherapeutic strategies against cancer. Despite the initial enthusiasm, disappointing results from early studies raised some doubts regarding the true clinical value of these approaches. However, our expanding knowledge of DC immunobiology and the definition of the optimal characteristics for antitumor immune responses have allowed a more rational development of DC-based immunotherapies in recent years. Here, after a brief overview of DC immunobiology, we sought to systematize the knowledge provided by 20 years of clinical trials, with a special emphasis on the diversity of approaches used to manipulate DCs and their consequent impact on vaccine effectiveness. We also address how new therapeutic concepts, namely the combination of DC vaccines with other anticancer therapies, are being implemented and are leveraging clinical outcomes. Finally, optimization strategies, new insights, and future perspectives on the field are also highlighted.
Collapse
Affiliation(s)
- João Constantino
- Faculty of Pharmacy and Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Célia Gomes
- Faculty of Medicine, Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI) and Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy and Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Maria T Cruz
- Faculty of Pharmacy and Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Bruno M Neves
- Faculty of Pharmacy and Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal; Department of Chemistry and QOPNA, Mass Spectrometry Centre, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
40
|
Anguille S, Smits EL, Bryant C, Van Acker HH, Goossens H, Lion E, Fromm PD, Hart DN, Van Tendeloo VF, Berneman ZN. Dendritic Cells as Pharmacological Tools for Cancer Immunotherapy. Pharmacol Rev 2015; 67:731-53. [DOI: 10.1124/pr.114.009456] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
41
|
Pizzurro GA, Barrio MM. Dendritic cell-based vaccine efficacy: aiming for hot spots. Front Immunol 2015; 6:91. [PMID: 25784913 PMCID: PMC4347494 DOI: 10.3389/fimmu.2015.00091] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/16/2015] [Indexed: 12/18/2022] Open
Abstract
Many approaches for cancer immunotherapy have targeted dendritic cells (DCs), directly or indirectly, for the induction of antitumor immune responses. DC-based vaccines have been developed using a wide variety of ex vivo DC culture conditions, antigen (Ag) source and loading strategies, maturation agents, and routes of vaccination. Adjuvants are used to activate innate immune cells at the vaccine injection site, to promote Ag transport to the draining lymph nodes (LNs) and to model adaptive immune responses. Despite years of effort, the effective induction of strong and durable antitumor T-cell responses in vaccinated patients remains a challenge. The study of vaccine interactions with other immune cells in the LNs and, more recently, in the injection site has opened new doors for understanding antitumor effector T-cell licensing and function. In this review, we will briefly discuss the relevant sites and up-to-date facts regarding possible targets for antitumor vaccine refinement. We will focus on the processes taking place at the injection site, adjuvant combinations and their role in DC-based vaccines, LN homing, and modeling vaccine-induced immune responses capable of controlling tumor growth and generating immune memory.
Collapse
Affiliation(s)
- Gabriela Andrea Pizzurro
- Centro de Investigaciones Oncológicas - Fundación Cáncer (CIO - FUCA) , Buenos Aires , Argentina
| | - María Marcela Barrio
- Centro de Investigaciones Oncológicas - Fundación Cáncer (CIO - FUCA) , Buenos Aires , Argentina
| |
Collapse
|
42
|
Induction of cytomegalovirus-specific T cell responses in healthy volunteers and allogeneic stem cell recipients using vaccination with messenger RNA-transfected dendritic cells. Transplantation 2015; 99:120-7. [PMID: 25050468 PMCID: PMC4281162 DOI: 10.1097/tp.0000000000000272] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background Infection with human cytomegalovirus (CMV) is a significant cause of morbidity and mortality in solid organ and hematopoietic stem cell transplant (HSCT) recipients. Methods The present study explored the safety, feasibility, and immunogenicity of CMV pp65 messenger RNA–loaded autologous monocyte-derived dendritic cells (DC) as a cellular vaccine for active immunization in healthy volunteers and allogeneic HSCT recipients. Four CMV-seronegative healthy volunteers and three allogeneic HSCT recipients were included in the study. Four clinical-grade autologous monocyte-derived DC vaccines were prepared after a single leukapheresis procedure and administered intradermally at a weekly interval. Results De novo induction of CMV-specific T-cell responses was detected in three of four healthy volunteers without serious adverse events. Of the HSCT recipients, none developed CMV disease and one of two patients displayed a remarkable threefold increase in CMV pp65-specific T cells on completion of the DC vaccination trial. Conclusion In conclusion, our DC vaccination strategy induced or expanded a CMV-specific cellular response in four of six efficacy-evaluable study subjects, providing a base for its further exploration in larger cohorts. Supplemental digital content is available in the text.
Collapse
|
43
|
Inoue K, Saegusa N, Omiya M, Ashizawa T, Miyata H, Komiyama M, Iizuka A, Kume A, Sugino T, Yamaguchi K, Kiyohara Y, Nakagawa M, Akiyama Y. Immunologically augmented skin flap as a novel dendritic cell vaccine against head and neck cancer in a rat model. Cancer Sci 2015; 106:143-50. [PMID: 25492592 PMCID: PMC4399025 DOI: 10.1111/cas.12586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Revised: 12/01/2014] [Accepted: 12/03/2014] [Indexed: 12/28/2022] Open
Abstract
Local recurrence is a major clinical issue following surgical resection in head and neck cancer, and the dissemination and lymph node metastasis of minimal residual disease is relatively difficult to treat due to the lack of suitable therapeutic approaches. In the present study, we developed and evaluated a novel immunotherapy using a skin flap transfer treated with sensitized dendritic cells (DC), termed the “immuno-flap,” in a rat tumor model. After the local round area of skin was resected, SCC-158 cells (a rat head and neck cancer cell line) were inoculated into the muscle surface; lastly, the groin skin flap injected with mature DC was overlaid. Two weeks after the second DC injection, systemic immunological reactions and tumor size were measured. The DC-treated group showed a significant reduction in tumor size compared with the control. Although the induction of CTL activity in spleen cells was marginal, Th1 cytokines such as interleukin-2 and interferon-γ were elevated in the DC-treated group. These results suggest that a novel immunotherapy based on the immuno-flap method has the potential for clinical application to prevent the local recurrence of head and neck cancer patients.
Collapse
Affiliation(s)
- Keita Inoue
- Division of Plastic and Reconstructive Surgery, Shizuoka Cancer Center Hospital, Nagaizumi, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Wimmers F, Schreibelt G, Sköld AE, Figdor CG, De Vries IJM. Paradigm Shift in Dendritic Cell-Based Immunotherapy: From in vitro Generated Monocyte-Derived DCs to Naturally Circulating DC Subsets. Front Immunol 2014; 5:165. [PMID: 24782868 PMCID: PMC3990057 DOI: 10.3389/fimmu.2014.00165] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 03/28/2014] [Indexed: 12/31/2022] Open
Abstract
Dendritic cell (DC)-based immunotherapy employs the patients’ immune system to fight neoplastic lesions spread over the entire body. This makes it an important therapy option for patients suffering from metastatic melanoma, which is often resistant to chemotherapy. However, conventional cellular vaccination approaches, based on monocyte-derived DCs (moDCs), only achieved modest response rates despite continued optimization of various vaccination parameters. In addition, the generation of moDCs requires extensive ex vivo culturing conceivably hampering the immunogenicity of the vaccine. Recent studies, thus, focused on vaccines that make use of primary DCs. Though rare in the blood, these naturally circulating DCs can be readily isolated and activated thereby circumventing lengthy ex vivo culture periods. The first clinical trials not only showed increased survival rates but also the induction of diversified anti-cancer immune responses. Upcoming treatment paradigms aim to include several primary DC subsets in a single vaccine as pre-clinical studies identified synergistic effects between various antigen-presenting cells.
Collapse
Affiliation(s)
- Florian Wimmers
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands
| | - Annette E Sköld
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands
| | - I Jolanda M De Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , Netherlands ; Department of Medical Oncology, Radboud University Medical Center , Nijmegen , Netherlands
| |
Collapse
|
45
|
Chen YC, Wen S, Shang SA, Cui Y, Luo B, Teng GJ. Magnetic resonance and near-infrared imaging using a novel dual-modality nano-probe for dendritic cell tracking in vivo. Cytotherapy 2013; 16:699-710. [PMID: 24219906 DOI: 10.1016/j.jcyt.2013.09.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 09/14/2013] [Accepted: 09/28/2013] [Indexed: 01/11/2023]
Abstract
BACKGROUND AIMS The effect of cellular-based immunotherapy is highly correlated with the success of dendritic cells (DCs) homing to the draining lymph nodes (LNs) and interacting with antigen-specific CD4(+) T cells. In this study, a novel magneto-fluorescent nano-probe was used to track the in vivo migration of DCs to the draining LNs. METHODS A dual-modality nano-probe composed of superparamagnetic iron oxide (SPIO) and near-infrared fluorescent (NIRF) dye (NIR797) was developed, and its magnetic and optical contrasting properties were characterized. DCs generated from mouse bone marrow were co-cultured with the probe at a lower concentration of 10 μg/mL. The cell phenotype and function of DCs were also investigated by fluorescence-activated cell sorting analysis and mixed leukocyte reactivity assay. Labeled DCs were injected into the footpad of C57BL/6 mice. Afterward, magnetic resonance imaging, NIRF imaging, Perls staining and CD11c immunofluorescence were used to observe the migration of the labeled DCs into draining LNs. RESULTS The synthetic SPIO-NIR797 nano-probe had a desirable superparamagnetic and near-infrared behavior. Perls staining showed perfect labeling efficiency. The cell phenotypes, including CD11c, CD80, CD86 and major histocompatibility complex class II, as well as the T-cell activation potential of the mature DCs were insignificantly affected after incubation (P > 0.05). Labeled DCs migrating into LNs could be detected by both magnetic resonance imaging and NIRF imaging simultaneously, which was further confirmed by Perls staining and immunofluorescence. CONCLUSIONS The novel dual-modality SPIO-NIR797 nano-probe has highly biocompatible characteristics for labeling and tracking DCs, which can be used to evaluate cancer immunotherapy in clinical applications.
Collapse
Affiliation(s)
- Yu-Chen Chen
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Song Wen
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Song-An Shang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Ying Cui
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Bing Luo
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Gao-Jun Teng
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China.
| |
Collapse
|
46
|
Affiliation(s)
- Rachel Lubong Sabado
- NYU Langone Medical Center Cancer Institute; New York University School of Medicine, New York; New York
| | - Nina Bhardwaj
- NYU Langone Medical Center Cancer Institute; New York University School of Medicine, New York; New York
| |
Collapse
|
47
|
Aarntzen EH, Srinivas M, Schreibelt G, Heerschap A, Punt CJ, Figdor CG, Oyen WJ, de Vries IJM. Reducing cell number improves the homing of dendritic cells to lymph nodes upon intradermal vaccination. Oncoimmunology 2013; 2:e24661. [PMID: 24073362 PMCID: PMC3782158 DOI: 10.4161/onci.24661] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 04/11/2013] [Indexed: 02/03/2023] Open
Abstract
Dendritic cell (DC)-based vaccines require the cells to relocate to lymph nodes (LNs). Unfortunately, however, DC migration rates are typically very poor. We investigated strategies to increase the migration efficacy of DC-based vaccines. Surprisingly, a reduction in DC number, but not the conditioning of the injection site, improved LN targeting.
Collapse
Affiliation(s)
- Erik Hjg Aarntzen
- Department of Tumor Immunology; Nijmegen Centre for Molecular Life Sciences; Radboud University Nijmegen Medical Centre; Nijmegen, The Netherlands ; Department of Medical Oncology; Radboud University Nijmegen Medical Centre; Nijmegen, The Netherlands ; Department of Nuclear Medicine; Radboud University Nijmegen Medical Centre; Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Dekaban GA, Hamilton AM, Fink CA, Au B, de Chickera SN, Ribot EJ, Foster PJ. Tracking and evaluation of dendritic cell migration by cellular magnetic resonance imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2013; 5:469-83. [PMID: 23633389 DOI: 10.1002/wnan.1227] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/28/2013] [Accepted: 03/19/2013] [Indexed: 01/15/2023]
Abstract
Cellular magnetic resonance imaging (MRI) is a means by which cells labeled ex vivo with a contrast agent can be detected and tracked over time in vivo. This technology provides a noninvasive method with which to assess cell-based therapies in vivo. Dendritic cell (DC)-based vaccines are a promising cancer immunotherapy, but its success is highly dependent on the injected DC migrating to a secondary lymphoid organ such as a nearby lymph node. There the DC can interact with T cells to elicit a tumor-specific immune response. It is important to verify DC migration in vivo using a noninvasive imaging modality, such as cellular MRI, so that important information regarding the anatomical location and persistence of the injected DC in a targeted lymph node can be provided. An understanding of DC biology is critical in ascertaining how to label DC with sufficient contrast agent to render them detectable by MRI. While iron oxide nanoparticles provide the best sensitivity for detection of DC in vivo, a clinical grade iron oxide agent is not currently available. A clinical grade (19) Fluorine-based perfluorcarbon nanoemulsion is available but is less sensitive, and its utility to detect DC migration in humans remains to be demonstrated using clinical scanners presently available. The ability to quantitatively track DC migration in vivo can provide important information as to whether different DC maturation and activation protocols result in improved DC migration efficiency which will determine the vaccine's immunogenicity and ultimately the tumor immunotherapy's outcome in humans.
Collapse
Affiliation(s)
- Gregory A Dekaban
- BioTherapeutics Research Laboratories, Robarts Research Institute and Department of Microbiology & Immunology, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|