1
|
Shah S, Nag A, Lucke-Wold B. Autologous tumor lysate-loaded dendritic cell vaccination in glioblastoma patients: a systematic review of literature. Clin Transl Oncol 2024:10.1007/s12094-024-03830-9. [PMID: 39714754 DOI: 10.1007/s12094-024-03830-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024]
Abstract
Glioblastoma (GBM) is one of the most common primary malignant brain tumors. Annually, there are about six instances recorded per 100,000 inhabitants. Treatment for GB has not advanced all that much. Novel medications have been investigated recently for the management of newly diagnosed and recurring instances of GBM. For GBM, surgery, radiation therapy, and alkylating chemotherapy are often used therapies. Immunotherapies, which use the patient's immune reaction against tumors, have long been seen as a potential cancer treatment. One such treatment is the dendritic cell (DC) vaccine. This cell-based vaccination works by stimulating the patient's own dendritic cells' antigenic repertoire, therefore inducing a polyclonal T-cell response. Systematic retrieval of information was performed on PubMed, Embase, and Google Scholar. Specified keywords were used to search, and the articles published in peer-reviewed scientific journals were associated with brain GBM, cancer, and Autologous Tumor Lysate-Loaded Dendritic Cell Vaccination. Selected 90 articles were used in this manuscript, of which 30 articles were clinical trials. Compared to shared tumor antigen peptide vaccines, autologous cancer DCs have a greater ability to stimulate the immune system, which is why dendritic cell fusion vaccines have shown early promise in several clinical studies. Survival rates for vaccinated patients were notably better compared to matched or historical controls. For newly diagnosed patients, the median overall survival (mOS) ranged from 15 to 41.4 months, while the progression-free survival (PFS) ranged from 6 to 25.3 months. We discovered through this analysis that autologous multiomics analysis of DC vaccines showed enhanced antitumor immunity with a focus on using activated, antigen-loaded donor DCs to trigger T-cell responses against cancer, particularly in glioblastoma. It also showed improved patient survival, especially when combined with standard chemoradiotherapy. DC vaccines show promise in treating GBM by enhancing survival and reducing tumor recurrence. However, challenges in vaccine production, antigen selection, and tumor heterogeneity highlight the need for continued research and optimization to improve efficacy and patient outcomes.
Collapse
Affiliation(s)
- Siddharth Shah
- Lillian S Wells Department of Neurosurgery at the University of Florida: University of Florida Lillian S Wells Department of Neurosurgery, Gainesville, FL, USA.
| | - Aiswarya Nag
- Sri Ramachandra University Medical College: Sri Ramachandra Medical College and Research Institute, Chennai, India
| | - Brandon Lucke-Wold
- Lillian S Wells Department of Neurosurgery at the University of Florida: University of Florida Lillian S Wells Department of Neurosurgery, Gainesville, FL, USA
| |
Collapse
|
2
|
Granato AM, Pancisi E, Piccinini C, Stefanelli M, Pignatta S, Soldati V, Carloni S, Fanini F, Arienti C, Bulgarelli J, Tazzari M, Scarpi E, Passardi A, Tauceri F, La Barba G, Maimone G, Baravelli S, de Rosa F, Ridolfi L, Petrini M. Dendritic cell vaccines as cancer treatment: focus on 13 years of manufacturing and quality control experience in advanced therapy medicinal products. Cytotherapy 2024; 26:1547-1555. [PMID: 39046388 DOI: 10.1016/j.jcyt.2024.07.005] [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: 04/12/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND AIMS Dendritic cells (DCs) are professional antigen-presenting cells of the mammalian immune system. Ex vivo differentiated DCs represent a unique Advanced Therapy Medicinal Product (ATMP), used in several clinical trials as personalized cancer immunotherapy. The therapy's reliability depends on its capacity to produce high-quality mature DCs (mDCs) in compliance with Good Manufacturing Practices. AIMS From March 2010 to December 2023, 103 patients were enrolled in multiple clinical trials at the Immuno-Gene Therapy Factory at IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori". Six hundred forty-two doses were produced, and the manufacturing process was implemented to optimize production. Our study is a retrospective analysis focusing on the quality control results. METHODS We retrospectively analyzed the results of the quality control tests carried out on each produced batch, evaluating viability, purity and phenotype of mDCs and their quality in terms of microbiological safety. The data obtained are given with median and interquartile range. RESULTS The batches were found to be microbiologically safe in terms of sterility, mycoplasma, and endotoxins. An increase in DC maturation markers was found. The release criteria checks showed a high percentage of viability and purity was maintained during the production process. CONCLUSIONS Our findings have confirmed that the measures implemented have ensured the safety of the products and have contributed to the establishing a robust "Pharmaceutical Quality System." This has enabled many safe mDCs to be produced for clinical trials.
Collapse
Affiliation(s)
- Anna Maria Granato
- Immuno-Gene Therapy Factory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Elena Pancisi
- Immuno-Gene Therapy Factory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Claudia Piccinini
- Immuno-Gene Therapy Factory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Monica Stefanelli
- Immuno-Gene Therapy Factory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Pignatta
- Immuno-Gene Therapy Factory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Valentina Soldati
- Immuno-Gene Therapy Factory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Silvia Carloni
- Immuno-Gene Therapy Factory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Francesca Fanini
- Immuno-Gene Therapy Factory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Arienti
- Immuno-Gene Therapy Factory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Jenny Bulgarelli
- Experimental and Clinical Oncology Unit of Immunotherapy and Rare Cancers, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Marcella Tazzari
- Experimental and Clinical Oncology Unit of Immunotherapy and Rare Cancers, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Emanuela Scarpi
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandro Passardi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Francesca Tauceri
- Department of General and Oncologic Surgery, Morgagni-Pierantoni Hospital, AUSL Romagna, Forlì, Italy
| | - Giuliano La Barba
- Department of General and Oncologic Surgery, Morgagni-Pierantoni Hospital, AUSL Romagna, Forlì, Italy
| | | | - Stefano Baravelli
- Unit of Immunohematology and Transfusion Medicine, GB Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Francesco de Rosa
- Experimental and Clinical Oncology Unit of Immunotherapy and Rare Cancers, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Laura Ridolfi
- Experimental and Clinical Oncology Unit of Immunotherapy and Rare Cancers, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Massimiliano Petrini
- Immuno-Gene Therapy Factory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
3
|
Capelli C, Cuofano C, Pavoni C, Frigerio S, Lisini D, Nava S, Quaroni M, Colombo V, Galli F, Bezukladova S, Panina-Bordignon P, Gaipa G, Comoli P, Cossu G, Martino G, Biondi A, Introna M, Golay J. Potency assays and biomarkers for cell-based advanced therapy medicinal products. Front Immunol 2023; 14:1186224. [PMID: 37359560 PMCID: PMC10288881 DOI: 10.3389/fimmu.2023.1186224] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
Advanced Therapy Medicinal Products (ATMPs) based on somatic cells expanded in vitro, with or without genetic modification, is a rapidly growing area of drug development, even more so following the marketing approval of several such products. ATMPs are produced according to Good Manufacturing Practice (GMP) in authorized laboratories. Potency assays are a fundamental aspect of the quality control of the end cell products and ideally could become useful biomarkers of efficacy in vivo. Here we summarize the state of the art with regard to potency assays used for the assessment of the quality of the major ATMPs used clinic settings. We also review the data available on biomarkers that may substitute more complex functional potency tests and predict the efficacy in vivo of these cell-based drugs.
Collapse
Affiliation(s)
- Chiara Capelli
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Carolina Cuofano
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Chiara Pavoni
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Simona Frigerio
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniela Lisini
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sara Nava
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Michele Quaroni
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Valentina Colombo
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Francesco Galli
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, Manchester, United Kingdom
| | - Svetlana Bezukladova
- Università Vita-Salute San Raffaele, Milan, Italy
- IRCCS San Raffaele Hospital, Neuroimmunology Unit, Division of Neuroscience, Milan, Italy
| | - Paola Panina-Bordignon
- Università Vita-Salute San Raffaele, Milan, Italy
- IRCCS San Raffaele Hospital, Neuroimmunology Unit, Division of Neuroscience, Milan, Italy
| | - Giuseppe Gaipa
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Patrizia Comoli
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giulio Cossu
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, Manchester, United Kingdom
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Gianvito Martino
- IRCCS San Raffaele Hospital, Neuroimmunology Unit, Division of Neuroscience, Milan, Italy
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Andrea Biondi
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Martino Introna
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Josée Golay
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
4
|
Stability Program in Dendritic Cell Vaccines: A “Real-World” Experience in the Immuno-Gene Therapy Factory of Romagna Cancer Center. Vaccines (Basel) 2022; 10:vaccines10070999. [PMID: 35891165 PMCID: PMC9323699 DOI: 10.3390/vaccines10070999] [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: 05/13/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 12/10/2022] Open
Abstract
Advanced therapy medical products (ATMPs) are rapidly growing as innovative medicines for the treatment of several diseases. Hence, the role of quality analytical tests to ensure consistent product safety and quality has become highly relevant. Several clinical trials involving dendritic cell (DC)-based vaccines for cancer treatment are ongoing at our institute. The DC-based vaccine is prepared via CD14+ monocyte differentiation. A fresh dose of 10 million DCs is administered to the patient, while the remaining DCs are aliquoted, frozen, and stored in nitrogen vapor for subsequent treatment doses. To evaluate the maintenance of quality parameters and to establish a shelf life of frozen vaccine aliquots, a stability program was developed. Several parameters of the DC final product at 0, 6, 12, 18, and 24 months were evaluated. Our results reveal that after 24 months of storage in nitrogen vapor, the cell viability is in a range between 82% and 99%, the expression of maturation markers remains inside the criteria for batch release, the sterility tests are compliant, and the cell costimulatory capacity unchanged. Thus, the data collected demonstrate that freezing and thawing do not perturb the DC vaccine product maintaining over time its functional and quality characteristics.
Collapse
|
5
|
Capelli C, Frigerio S, Lisini D, Nava S, Gaipa G, Belotti D, Cabiati B, Budelli S, Lazzari L, Bagnarino J, Tanzi M, Comoli P, Perico N, Introna M, Golay J. A comprehensive report of long-term stability data for a range ATMPs: A need to develop guidelines for safe and harmonized stability studies. Cytotherapy 2022; 24:544-556. [PMID: 35177338 DOI: 10.1016/j.jcyt.2021.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND AIMS Advanced therapy medicinal products (ATMPs) are novel drugs based on genes, cells or tissues developed to treat many different diseases. Stability studies of each new ATMP need to be performed to define its shelf life and guarantee efficacy and safety upon infusion, and these are presently based on guidelines originally drafted for standard pharmaceutical drugs, which have properties and are stored in conditions quite different from cell products. The aim of this report is to provide evidence-based information for stability studies on ATMPs that will facilitate the interlaboratory harmonization of practices in this area. METHODS We have collected and analyzed the results of stability studies on 19 different cell-based experimental ATMPs, produced by five authorized cell factories forming the Lombardy "Plagencell network" for use in 36 approved phase I/II clinical trials; most were cryopreserved and stored in liquid nitrogen vapors for 1 to 13 years. RESULTS The cell attributes collected in stability studies included cell viability, immunophenotype and potency assays, in particular immunosuppression, cytotoxicity, cytokine release and proliferation/differentiation capacity. Microbiological attributes including sterility, endotoxin levels and mycoplasma contamination were also analyzed. All drug products (DPs), cryopreserved in various excipients containing 10% DMSO and in different primary containers, were very stable long term at <-150°C and did not show any tendency for diminished viability or efficacy for up to 13.5 years. CONCLUSIONS Our data indicate that new guidelines for stability studies, specific for ATMPs and based on risk analyses, should be drafted to harmonize practices, significantly reduce the costs of stability studies without diminishing safety. Some specific suggestions are presented in the discussion.
Collapse
Affiliation(s)
- Chiara Capelli
- Center of Cellular Therapy "G. Lanzani", ASST Papa Giovanni XXIII, Bergamo, Italy; Fondazione per la Ricerca Ospedale di Bergamo, Bergamo, Italy
| | - Simona Frigerio
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniela Lisini
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sara Nava
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Gaipa
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Daniela Belotti
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Benedetta Cabiati
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Silvia Budelli
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Lorenza Lazzari
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Jessica Bagnarino
- UOSD Cell Factory, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Matteo Tanzi
- UOSD Cell Factory, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Patrizia Comoli
- UOSD Cell Factory, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Norberto Perico
- Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Istituto Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Martino Introna
- Center of Cellular Therapy "G. Lanzani", ASST Papa Giovanni XXIII, Bergamo, Italy.
| | - Josée Golay
- Center of Cellular Therapy "G. Lanzani", ASST Papa Giovanni XXIII, Bergamo, Italy; Fondazione per la Ricerca Ospedale di Bergamo, Bergamo, Italy
| |
Collapse
|
6
|
Kang X, Zheng Y, Hong W, Chen X, Li H, Huang B, Huang Z, Tang H, Geng W. Recent Advances in Immune Cell Therapy for Glioblastoma. Front Immunol 2020; 11:544563. [PMID: 33193310 PMCID: PMC7609403 DOI: 10.3389/fimmu.2020.544563] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is the most malignant form of astrocytoma with short survival and a high recurrence rate and remains a global problem. Currently, surgery, chemotherapy, radiotherapy, and other comprehensive treatments are the main treatment modalities, but patients still have a poor prognosis mainly due to the infiltrative growth of GBM and the protective effect of the blood–brain barrier on tumor cells. Therefore, immunotherapy is expected to be a good option for GBM. In the immune system, different cells play varying roles in the treatment of GBM, so understanding the roles played by various immune cells in treating GBM and considering how to combine these effects to maximize the efficacy of these cells is important for the selection of comprehensive and optimal treatment plans and improving GBM prognosis. Therefore, this study reviews the latest research progress on the role of various types of immune cells in the treatment of GBM.
Collapse
Affiliation(s)
- Xianhui Kang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yiyang Zheng
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Wandong Hong
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xixi Chen
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Huiting Li
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Baojun Huang
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zhenyang Huang
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Hongli Tang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wujun Geng
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Atkinson SP. A preview of selected articles. Stem Cells 2020. [DOI: 10.1002/stem.3138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
8
|
Eoli M, Corbetta C, Anghileri E, Di Ianni N, Milani M, Cuccarini V, Musio S, Paterra R, Frigerio S, Nava S, Lisini D, Pessina S, Maddaloni L, Lombardi R, Tardini M, Ferroli P, DiMeco F, Bruzzone MG, Antozzi C, Pollo B, Finocchiaro G, Pellegatta S. Expansion of effector and memory T cells is associated with increased survival in recurrent glioblastomas treated with dendritic cell immunotherapy. Neurooncol Adv 2019; 1:vdz022. [PMID: 32642658 PMCID: PMC7212883 DOI: 10.1093/noajnl/vdz022] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Background The efficacy of dendritic cell (DC) immunotherapy as a single therapeutic modality for the treatment of glioblastoma (GBM) patients remains limited. In this study, we evaluated in patients with GBM recurrence the immune-mediated effects of DC loaded with autologous tumor lysate combined with temozolomide (TMZ) or tetanus toxoid (TT). Methods In the phase I-II clinical study DENDR2, 12 patients were treated with 5 DC vaccinations combined with dose-dense TMZ. Subsequently, in eight patients, here defined as Variant (V)-DENDR2, the vaccine site was preconditioned with TT 24 hours before DC vaccination and TMZ was avoided. As a survival endpoint for these studies, we considered overall survival 9 months (OS9) after second surgery. Patients were analyzed for the generation of effector, memory, and T helper immune response. Results Four of 12 DENDR2 patients reached OS9, but all failed to show an immunological response. Five of eight V-DENDR2 patients (62%) reached OS9, and one patient is still alive (OS >30 months). A robust CD8+ T-cell activation and memory T-cell formation were observed in V-DENDR2 OS>9. Only in these patients, the vaccine-specific CD4+ T-cell activation (CD38+/HLA-DR+) was paralleled by an increase in TT-induced CD4+/CD38low/CD127high memory T cells. Only V-DENDR2 patients showed the formation of a nodule at the DC injection site infiltrated by CCL3-expressing CD4+ T cells. Conclusions TT preconditioning of the vaccine site and lack of TMZ could contribute to the efficacy of DC immunotherapy by inducing an effector response, memory, and helper T-cell generation.
Collapse
Affiliation(s)
- Marica Eoli
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Cristina Corbetta
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Laboratory of Brain Tumor Immunotherapy, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Elena Anghileri
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Natalia Di Ianni
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Laboratory of Brain Tumor Immunotherapy, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Micaela Milani
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Laboratory of Brain Tumor Immunotherapy, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Valeria Cuccarini
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Unit of Neuro-Radiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Silvia Musio
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Laboratory of Brain Tumor Immunotherapy, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Rosina Paterra
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Simona Frigerio
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Cell Therapy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sara Nava
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Cell Therapy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniela Lisini
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Cell Therapy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sara Pessina
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Laboratory of Brain Tumor Immunotherapy, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Luisa Maddaloni
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Raffaella Lombardi
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,3rd Neurology Unit and Skin Biopsy, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Maria Tardini
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paolo Ferroli
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Unit of Neurosurgery 2, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesco DiMeco
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Unit of Neurosurgery 1, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Department of Neurological Surgery, Johns Hopkins Medical School, Baltimore, Maryland
| | - Maria Grazia Bruzzone
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Unit of Neuro-Radiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Carlo Antozzi
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Unit of Neuro-Immunology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Bianca Pollo
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Unit of Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Gaetano Finocchiaro
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Serena Pellegatta
- Laboratory of Brain Tumor Immunotherapy, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
9
|
Role of Interferon (IFN)α in “Cocktails” for the Generation of (Leukemia-derived) Dendritic Cells (DCleu) From Blasts in Blood From Patients (pts) With Acute Myeloid Leukemia (AML) and the Induction of Antileukemic Reactions. J Immunother 2019; 42:143-161. [DOI: 10.1097/cji.0000000000000266] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
10
|
A Characterization of Dendritic Cells and Their Role in Immunotherapy in Glioblastoma: From Preclinical Studies to Clinical Trials. Cancers (Basel) 2019; 11:cancers11040537. [PMID: 30991681 PMCID: PMC6521200 DOI: 10.3390/cancers11040537] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM) is the most common and fatal primary central nervous system malignancy in adults with a median survival of less than 15 months. Surgery, radiation, and chemotherapy are the standard of care and provide modest benefits in survival, but tumor recurrence is inevitable. The poor prognosis of GBM has made the development of novel therapies targeting GBM of paramount importance. Immunotherapy via dendritic cells (DCs) has garnered attention and research as a potential strategy to boost anti-tumor immunity in recent years. As the “professional” antigen processing and presenting cells, DCs play a key role in the initiation of anti-tumor immune responses. Pre-clinical studies in GBM have shown long-term tumor survival and immunological memory in murine models with stimulation of DC activity with various antigens and costimulatory molecules. Phase I and II clinical trials of DC vaccines in GBM have demonstrated some efficacy in improving the median overall survival with minimal to no toxicity with promising initial results from the first Phase III trial. However, there remains no standardization of vaccines in terms of which antigens are used to pulse DCs ex vivo, sites of DC injection, and optimal adjuvant therapies. Future work with DC vaccines aims to elucidate the efficacy of DC-based therapy alone or in combination with other immunotherapy adjuvants in additional Phase III trials.
Collapse
|
11
|
Schiffer D, Annovazzi L, Casalone C, Corona C, Mellai M. Glioblastoma: Microenvironment and Niche Concept. Cancers (Basel) 2018; 11:cancers11010005. [PMID: 30577488 PMCID: PMC6357107 DOI: 10.3390/cancers11010005] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 01/11/2023] Open
Abstract
The niche concept was originally developed to describe the location of normal neural stem cells (NSCs) in the subependymal layer of the sub-ventricular zone. In this paper, its significance has been extended to the location of tumor stem cells in glioblastoma (GB) to discuss the relationship between GB stem cells (GSCs) and endothelial cells (ECs). Their interaction is basically conceived as responsible for tumor growth, invasion and recurrence. Niches are described as the points of utmost expression of the tumor microenvironment (TME), therefore including everything in the tumor except for tumor cells: NSCs, reactive astrocytes, ECs, glioma-associated microglia/macrophages (GAMs), myeloid cells, pericytes, fibroblasts, etc. and all intrinsic and extrinsic signaling pathways. Perivascular (PVNs), perinecrotic (PNNs) and invasive niches were described from the pathological point of view, highlighting the basic significance of the EC/tumor stem cell couple. PNN development was reinterpreted based on the concept that hyperproliferative areas of GB are composed of GSCs/progenitors. TME was depicted in its function as the main regulator of everything that happens in the tumor. A particular emphasis was given to GAMs, pericytes and reactive astrocytes as important elements affecting proliferation, growth, invasion and resistance to therapies of tumor cells.
Collapse
Affiliation(s)
- Davide Schiffer
- Professore Emerito di Neurologia, Università di Torino, Corso Bramante 88/90, 10126 Torino, Italy.
| | - Laura Annovazzi
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Via P. Micca 29, 13100 Vercelli, Italy.
| | - Cristina Casalone
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Via Bologna 148, 10154 Torino, Italy.
| | - Cristiano Corona
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Via Bologna 148, 10154 Torino, Italy.
| | - Marta Mellai
- Dipartimento di Scienze della Salute, Scuola di Medicina, Università del Piemonte Orientale "A. Avogadro", Corso Mazzini 18, 28100 Novara, Italy.
- Fondazione Edo ed Elvo Tempia Valenta-Onlus, Via Malta 3, 13900 Biella, Italy.
| |
Collapse
|
12
|
Shinde P, Khan N, Melinkeri S, Kale V, Limaye L. Freezing of dendritic cells with trehalose as an additive in the conventional freezing medium results in improved recovery after cryopreservation. Transfusion 2018; 59:686-696. [PMID: 30456902 DOI: 10.1111/trf.15028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/19/2018] [Accepted: 09/25/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Dendritic cell (DC) vaccination involves administration of multiple doses. Cryopreservation of tumor antigen-pulsed DCs can provide a ready to use vaccine source and eliminate the need of frequent withdrawal of the patient's blood for vaccine preparation. The aim of this study was to assess the effect of addition of trehalose in the freezing medium on the recovery of DCs after cryopreservation. STUDY DESIGN AND METHODS DCs were generated from mononuclear cells from apheresis samples of healthy donors. For long-term storage of 6 months, cells were frozen with a rate-controlled programmable freezer and stored in liquid nitrogen. For short-term storage of 1 month, cells were frozen and stored at -80°C. DCs frozen with Iscove's Modified Dulbecco's Medium + 10% dimethyl sulfoxide + 20% fetal bovine serum served as the control group, while the test group was additionally supplemented with 50 μg/mL of trehalose. After revival of control and test DCs, they were assessed for viability, morphology, phenotype, and functions. RESULTS The addition of trehalose to the conventional freezing medium helped to preserve the viability and functionality of DCs better than dimethyl sulfoxide alone in both long- and short-term cryopreservation. Trehalose also protected the mitochondrial membrane potential and cytoskeleton integrity of DCs, which are necessary for their functionality. Mediators of the intrinsic apoptotic pathway like Caspase-9 and Bim-1 were found to be low in the test. CONCLUSION Supplementation of conventional freezing medium with trehalose results in better quality of DCs revived after cryopreservation. This finding could help improve DC vaccine preparation for cancer immunotherapy.
Collapse
Affiliation(s)
| | - Nikhat Khan
- National Centre for Cell Science, Pune, India
| | - Sameer Melinkeri
- Blood and Marrow Transplant Unit, Deenanath Mangeshkar Hospital, Pune, India
| | | | | |
Collapse
|
13
|
DE Wolf C, VAN DE Bovenkamp M, Hoefnagel M. Regulatory perspective on in vitro potency assays for human dendritic cells used in anti-tumor immunotherapy. Cytotherapy 2018; 20:1289-1308. [PMID: 30327247 DOI: 10.1016/j.jcyt.2018.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/25/2018] [Accepted: 07/14/2018] [Indexed: 12/18/2022]
Abstract
Dendritic cells (DCs) are key connectors between the innate and adaptive immune system and have an important role in modulating other immune cells. Therefore, their therapeutic application to steer immune responses is considered in various disorders, including cancer. Due to differences in the cell source and manufacturing process, each DC medicinal product is unique. Consequently, release tests to ensure consistent quality need to be product-specific. Although general guidance concerning quality control testing of cell-based therapies is available, cell type-specific regulation is still limited. Especially guidance related to potency testing is needed, because developing an in vitro assay measuring cell properties relevant for in vivo functionality is challenging. In this review, we provide DC-specific guidance for development of in vitro potency assays for characterisation and release. We present a broad overview of in vitro potency assays suggested for DC products to determine their anti-tumor functionality. Several advantages and limitations of these assays are discussed. Also, we provide some points to consider for selection and design of a potency test. The ideal functionality assay for anti-tumor products evaluates the capacity of DCs to stimulate antigen-specific T cells. Because this approach may not be feasible for release, use of surrogate potency markers could be considered, provided that these markers are sufficiently linked to the in vivo DC biological activity and clinical response. Further elucidation of the involvement of specific DC subsets in anti-tumor responses will result in improved manufacturing processes for DC-based products and should be considered during potency assay development.
Collapse
Affiliation(s)
- Charlotte DE Wolf
- Medicines Evaluation Board College ter Beoordeling van Geneesmiddelen-Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands; Department of Infectious Diseases and Immunology, Utrecht University, The Netherlands
| | - Marja VAN DE Bovenkamp
- Medicines Evaluation Board College ter Beoordeling van Geneesmiddelen-Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands
| | - Marcel Hoefnagel
- Medicines Evaluation Board College ter Beoordeling van Geneesmiddelen-Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands.
| |
Collapse
|
14
|
Pellegatta S, Eoli M, Cuccarini V, Anghileri E, Pollo B, Pessina S, Frigerio S, Servida M, Cuppini L, Antozzi C, Cuzzubbo S, Corbetta C, Paterra R, Acerbi F, Ferroli P, DiMeco F, Fariselli L, Parati EA, Bruzzone MG, Finocchiaro G. Survival gain in glioblastoma patients treated with dendritic cell immunotherapy is associated with increased NK but not CD8 + T cell activation in the presence of adjuvant temozolomide. Oncoimmunology 2018; 7:e1412901. [PMID: 29632727 PMCID: PMC5889286 DOI: 10.1080/2162402x.2017.1412901] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 11/24/2017] [Accepted: 11/28/2017] [Indexed: 01/23/2023] Open
Abstract
In a two-stage phase II study, 24 patients with first diagnosis of glioblastoma (GBM) were treated with dendritic cell (DC) immunotherapy associated to standard radiochemotherapy with temozolomide (TMZ) followed by adjuvant TMZ. Three intradermal injections of mature DC loaded with autologous GBM lysate were administered before adjuvant TMZ, while 4 injections were performed during adjuvant TMZ. According to a two-stage Simon design, to proceed to the second stage progression-free survival (PFS) 12 months after surgery was expected in at least 8 cases enrolled in the first stage. Evidence of immune response and interaction with chemotherapy were investigated. After a median follow up of 17.4 months, 9 patients reached PFS12. In these patients (responders, 37.5%), DC vaccination induced a significant, persistent activation of NK cells, whose increased response was significantly associated with prolonged survival. CD8+ T cells underwent rapid expansion and priming but, after the first administration of adjuvant TMZ, failed to generate a memory status. Resistance to TMZ was associated with robust expression of the multidrug resistance protein ABCC3 in NK but not CD8+ T cells. The negative effect of TMZ on the formation of T cell-associated antitumor memory deserves consideration in future clinical trials including immunotherapy.
Collapse
Affiliation(s)
- Serena Pellegatta
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Marica Eoli
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Valeria Cuccarini
- Unit of Neuro-Radiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Elena Anghileri
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Bianca Pollo
- Unit of Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sara Pessina
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Simona Frigerio
- Cell Therapy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Maura Servida
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Lucia Cuppini
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Carlo Antozzi
- Unit of Neuro-Immunology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefania Cuzzubbo
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Cristina Corbetta
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Rosina Paterra
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesco Acerbi
- Unit of Neurosurgery 2, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paolo Ferroli
- Unit of Neurosurgery 2, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesco DiMeco
- Unit of Neurosurgery 1, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Laura Fariselli
- Unit of Radiotherapy, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Eugenio A Parati
- Cell Therapy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Maria Grazia Bruzzone
- Unit of Neuro-Radiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Gaetano Finocchiaro
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
15
|
Borchers S, Maβlo C, Müller CA, Tahedl A, Volkind J, Nowak Y, Umansky V, Esterlechner J, Frank MH, Ganss C, Kluth MA, Utikal J. Detection of ABCB5 tumour antigen-specific CD8 + T cells in melanoma patients and implications for immunotherapy. Clin Exp Immunol 2017; 191:74-83. [PMID: 28940439 DOI: 10.1111/cei.13053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2017] [Indexed: 01/09/2023] Open
Abstract
ATP binding cassette subfamily B member 5 (ABCB5) has been identified as a tumour-initiating cell marker and is expressed in various malignancies, including melanoma. Moreover, treatment with anti-ABCB5 monoclonal antibodies has been shown to inhibit tumour growth in xenotransplantation models. Therefore, ABCB5 represents a potential target for cancer immunotherapy. However, cellular immune responses against ABCB5 in humans have not been described so far. Here, we investigated whether ABCB5-reactive T cells are present in human melanoma patients and tested the applicability of ABCB5-derived peptides for experimental induction of human T cell responses. Peripheral blood mononuclear cells (PBMNC) isolated from blood samples of melanoma patients (n = 40) were stimulated with ABCB5 peptides, followed by intracellular cytokine staining (ICS) for interferon (IFN)-γ and tumour necrosis factor (TNF)-α. To evaluate immunogenicity of ABCB5 peptides in naive healthy donors, CD8 T cells were co-cultured with ABCB5 antigen-loaded autologous dendritic cells (DC). ABCB5 reactivity in expanded T cells was assessed similarly by ICS. ABCB5-reactive CD8+ T cells were detected ex vivo in 19 of 29 patients, melanoma antigen recognised by T cells (MART-1)-reactive CD8+ T cells in six of 21 patients. In this small, heterogeneous cohort, reactivity against ABCB5 was significantly higher than against MART-1. It occurred significantly more often and independently of clinical characteristics. Reactivity against ABCB5 could be induced in 14 of 16 healthy donors in vitro by repeated stimulation with peptide-loaded autologous DC. As ABCB5-reactive CD8 T cells can be found in the peripheral blood of melanoma patients and an ABCB5-specific response can be induced in vitro in naive donors, ABCB5 could be a new target for immunotherapies in melanoma.
Collapse
Affiliation(s)
- S Borchers
- RHEACELL GmbH & Co. KG, Heidelberg, Germany
| | - C Maβlo
- RHEACELL GmbH & Co. KG, Heidelberg, Germany
| | | | - A Tahedl
- TICEBA GmbH, Heidelberg, Germany
| | | | - Y Nowak
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - V Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | | | - M H Frank
- Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.,School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - C Ganss
- RHEACELL GmbH & Co. KG, Heidelberg, Germany.,TICEBA GmbH, Heidelberg, Germany
| | - M A Kluth
- RHEACELL GmbH & Co. KG, Heidelberg, Germany.,TICEBA GmbH, Heidelberg, Germany
| | - J Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| |
Collapse
|
16
|
Mehrotra S, Britten CD, Chin S, Garrett-Mayer E, Cloud CA, Li M, Scurti G, Salem ML, Nelson MH, Thomas MB, Paulos CM, Salazar AM, Nishimura MI, Rubinstein MP, Li Z, Cole DJ. Vaccination with poly(IC:LC) and peptide-pulsed autologous dendritic cells in patients with pancreatic cancer. J Hematol Oncol 2017; 10:82. [PMID: 28388966 PMCID: PMC5384142 DOI: 10.1186/s13045-017-0459-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 03/30/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Dendritic cells (DCs) enhance the quality of anti-tumor immune response in patients with cancer. Thus, we posit that DC-based immunotherapy, in conjunction with toll-like receptor (TLR)-3 agonist poly-ICLC, is a promising approach for harnessing immunity against metastatic or locally advanced unresectable pancreatic cancer (PC). METHODS We generated autologous DCs from the peripheral blood of HLA-A2+ patients with PC. DCs were pulsed with three distinct A2-restricted peptides: 1) human telomerase reverse transcriptase (hTERT, TERT572Y), 2) carcinoembryonic antigen (CEA; Cap1-6D), and 3) survivin (SRV.A2). Patients received four intradermal injections of 1 × 107 peptide-pulsed DC vaccines every 2 weeks (Day 0, 14, 28, and 42). Concurrently, patients received intramuscular administration of Poly-ICLC at 30 μg/Kg on vaccination days (i.e., day 0, 14, 28, and 42), as well as on days 3, 17, 21, 31, 37, and 45. Our key objective was to assess safety and feasibility. The effect of DC vaccination on immune response was measured at each DC injection time point by enumerating the phenotype and function of patient T cells. RESULTS Twelve patients underwent apheresis: nine patients with metastatic disease, and three patients with locally advanced unresectable disease. Vaccines were successfully manufactured from all individuals. We found that this treatment was well-tolerated, with the most common symptoms being fatigue and/or self-limiting flu-like symptoms. Among the eight patients who underwent imaging on day 56, four patients experienced stable disease while four patients had disease progression. The median overall survival was 7.7 months. One patient survived for 28 months post leukapheresis. MHC class I -tetramer analysis before and after vaccination revealed effective generation of antigen-specific T cells in three patients with stable disease. CONCLUSION Vaccination with peptide-pulsed DCs in combination with poly-ICLC is safe and induces a measurable tumor specific T cell population in patients with advanced PC. TRIAL REGISTRATION NCT01410968 ; Name of registry: clinicaltrials.gov; Date of registration: 08/04/2011).
Collapse
Affiliation(s)
- Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA.
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Present address: Gibbs Cancer Center and Research Institute, 380 Serpentine Drive, Spartanburg, SC, 29303, USA.
| | - Carolyn D Britten
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Steve Chin
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
- Present address: Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Elizabeth Garrett-Mayer
- Departmet of Population Sciences, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Colleen A Cloud
- Department of Surgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
| | - Mingli Li
- Department of Surgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
| | - Gina Scurti
- Department of Surgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
- Department of Surgery, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Mohamed L Salem
- Center of Excellence in Cancer Research and Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Michelle H Nelson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Melanie B Thomas
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
- Present address: Gibbs Cancer Center and Research Institute, 380 Serpentine Drive, Spartanburg, SC, 29303, USA
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Andres M Salazar
- Oncovir Inc., 3202 Cleaveland Avenue NW, Washington, DC, 20008, USA
| | - Michael I Nishimura
- Department of Surgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
- Department of Surgery, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Mark P Rubinstein
- Department of Surgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Zihai Li
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - David J Cole
- Department of Surgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA.
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
17
|
Advances in Monitoring Cell-Based Therapies with Magnetic Resonance Imaging: Future Perspectives. Int J Mol Sci 2017; 18:ijms18010198. [PMID: 28106829 PMCID: PMC5297829 DOI: 10.3390/ijms18010198] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/05/2017] [Accepted: 01/10/2017] [Indexed: 01/07/2023] Open
Abstract
Cell-based therapies are currently being developed for applications in both regenerative medicine and in oncology. Preclinical, translational, and clinical research on cell-based therapies will benefit tremendously from novel imaging approaches that enable the effective monitoring of the delivery, survival, migration, biodistribution, and integration of transplanted cells. Magnetic resonance imaging (MRI) offers several advantages over other imaging modalities for elucidating the fate of transplanted cells both preclinically and clinically. These advantages include the ability to image transplanted cells longitudinally at high spatial resolution without exposure to ionizing radiation, and the possibility to co-register anatomical structures with molecular processes and functional changes. However, since cellular MRI is still in its infancy, it currently faces a number of challenges, which provide avenues for future research and development. In this review, we describe the basic principle of cell-tracking with MRI; explain the different approaches currently used to monitor cell-based therapies; describe currently available MRI contrast generation mechanisms and strategies for monitoring transplanted cells; discuss some of the challenges in tracking transplanted cells; and suggest future research directions.
Collapse
|