1
|
Liu C, Cai Y, Mou S. Liquid biopsy in lung cancer: The role of circulating tumor cells in diagnosis, treatment, and prognosis. Biomed Pharmacother 2024; 181:117726. [PMID: 39612860 DOI: 10.1016/j.biopha.2024.117726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2024] Open
Abstract
Despite numerous therapeutic advancements, such as immune checkpoint inhibitors, lung cancer continues to be the leading cause of cancer-related mortality. Therefore, the identification of cancer at an early stage is becoming a significant subject in contemporary oncology. Despite significant advancements in early detection tactics in recent decades, they continue to provide challenges because of the inconspicuous symptoms observed during the early stages of the primary tumor. Presently, tumor biomarkers and imaging techniques are extensively employed across different forms of cancer. Nevertheless, every approach has its own set of constraints. In certain instances, the detriments outweigh the advantages. Hence, there is an urgent need to enhance early detection methods. Currently, liquid biopsy is considered more flexible and not intrusive method in comparison to conventional test for early detection. Circulating tumor cells (CTCs) are crucial components of liquid biopsy and have a pivotal function in the spread and formation of secondary tumors. These indicators show great promise in the early identification of cancer. This study presents a comprehensive examination of the methodologies employed for the isolation and enrichment of circulating tumor cells (CTCs) in lung cancer. Additionally, it explores the formation of clusters of CTCs, which have a pivotal function in facilitating the effective dissemination of cancer to distant organs. In addition, we discuss the importance of CTCs in the detection, treatment, and prognosis of lung cancer.
Collapse
Affiliation(s)
- Chibo Liu
- Department of Clinical Laboratory, Taizhou Municipal Hospital, Taizhou, Zhejiang, China.
| | - Yanqun Cai
- Department of Clinical Laboratory, Taizhou Municipal Hospital, Taizhou, Zhejiang, China
| | - Sihua Mou
- Department of Clinical Laboratory, Taizhou Municipal Hospital, Taizhou, Zhejiang, China.
| |
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
|
Understanding and improving cellular immunotherapies against cancer: From cell-manufacturing to tumor-immune models. Adv Drug Deliv Rev 2021; 179:114003. [PMID: 34653533 DOI: 10.1016/j.addr.2021.114003] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022]
Abstract
The tumor microenvironment (TME) is shaped by dynamic metabolic and immune interactions between precancerous and cancerous tumor cells and stromal cells like epithelial cells, fibroblasts, endothelial cells, and hematopoietically-derived immune cells. The metabolic states of the TME, including the hypoxic and acidic niches, influence the immunosuppressive phenotypes of the stromal and immune cells, which confers resistance to both host-mediated tumor killing and therapeutics. Numerous in vitro TME platforms for studying immunotherapies, including cell therapies, are being developed. However, we do not yet understand which immune and stromal components are most critical and how much model complexity is needed to answer specific questions. In addition, scalable sourcing and quality-control of appropriate TME cells for reproducibly manufacturing these platforms remain challenging. In this regard, lessons from the manufacturing of immunomodulatory cell therapies could provide helpful guidance. Although immune cell therapies have shown unprecedented results in hematological cancers and hold promise in solid tumors, their manufacture poses significant scale, cost, and quality control challenges. This review first provides an overview of the in vivo TME, discussing the most influential cell populations in the tumor-immune landscape. Next, we summarize current approaches for cell therapies against cancers and the relevant manufacturing platforms. We then evaluate current immune-tumor models of the TME and immunotherapies, highlighting the complexity, architecture, function, and cell sources. Finally, we present the technical and fundamental knowledge gaps in both cell manufacturing systems and immune-TME models that must be addressed to elucidate the interactions between endogenous tumor immunity and exogenous engineered immunity.
Collapse
|
4
|
Zhang H, Wang Y, Wang QT, Sun SN, Li SY, Shang H, He YW. Enhanced Human T Lymphocyte Antigen Priming by Cytokine-Matured Dendritic Cells Overexpressing Bcl-2 and IL-12. Front Cell Dev Biol 2020; 8:205. [PMID: 32292785 PMCID: PMC7118208 DOI: 10.3389/fcell.2020.00205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 03/10/2020] [Indexed: 01/08/2023] Open
Abstract
Dendritic cell (DC)-based vaccination is a promising immunotherapeutic strategy for cancer. However, clinical trials have shown only limited efficacy, suggesting the need to optimize protocols for human DC vaccine preparation. In this study, we systemically compared five different human DC vaccine maturation protocols used in clinical trials: (1) a four-cytokine cocktail (TNF-α, IL-6, IL-1β, and PGE2); (2) an α-DC-cytokine cocktail (TNF-α, IL-1β, IFN-α, IFN-γ, and poly I:C); (3) lipopolysaccharide (LPS)/IFN-γ; (4) TNF-α and PGE2; and (5) TriMix (mRNAs encoding CD40L, CD70, and constitutively active Toll-like receptor 4 electroporated into immature DCs). We found that the four-cytokine cocktail induced high levels of costimulatory and HLA molecules, as well as CCR7, in DCs. Mature DCs (mDCs) matured with the four-cytokine cocktail had higher viability than those obtained with the other protocols. Based on these features, we chose the four-cytokine cocktail protocol to further improve the immunizing capability of DCs by introducing exogenous genes. We showed that introducing exogenous Bcl-2 increased DC survival. Furthermore, introducing IL-12p70 rescued the inhibition of IL-12 secretion by PGE2 without impairing the DC phenotype. Introducing both Bcl-2 and IL-12p70 mRNAs into DCs induced enhanced cytomegalovirus pp65-specific CD8+ T cells secreting IFN-γ and TNF-α. Taken together, our data suggest that DC matured by the four-cytokine cocktail combined with exogenous Bcl-2 and IL-12p70 gene expression represents a promising approach for clinical applications in cancer immunotherapy.
Collapse
Affiliation(s)
- Hui Zhang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yu Wang
- Life Science Institute, Jinzhou Medical University, Jinzhou, China
| | | | - Sheng-Nan Sun
- Beijing Tricision Biotherapeutics Inc., Beijing, China
| | - Shi-You Li
- Beijing Tricision Biotherapeutics Inc., Beijing, China
| | - Hong Shang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - You-Wen He
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
5
|
Hopewell EL, Cox C. Manufacturing Dendritic Cells for Immunotherapy: Monocyte Enrichment. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 16:155-160. [PMID: 32055643 PMCID: PMC7005329 DOI: 10.1016/j.omtm.2019.12.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dendritic cells play a key role in activation of the immune system as potent antigen-presenting cells. This pivotal position, along with the ability to generate dendritic cells from monocytes and ready uptake of antigen, makes them an intriguing vehicle for immunotherapy for a variety of indications. Since the first reported trial using dendritic cells in 1995, they have been used in trials all over the world for a plethora of indications. Monocyte-derived dendritic cells are generated from whole blood or apheresis products by culturing enriched monocytes in the presence of interleukin (IL)-4 and granulocyte-macrophage colony-stimulating factor (GM-CSF). A variety of methods can be used for enrichment of monocytes for generation of clinical-grade dendritic cells and are summarized herein.
Collapse
Affiliation(s)
- Emily L Hopewell
- Cell and Gene Therapy Manufacturing, Indiana University, Indianapolis, IN, USA.,Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
| | - Cheryl Cox
- Cellular Therapy Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
6
|
Boudousquié C, Boand V, Lingre E, Dutoit L, Balint K, Danilo M, Harari A, Gannon PO, Kandalaft LE. Development and Optimization of a GMP-Compliant Manufacturing Process for a Personalized Tumor Lysate Dendritic Cell Vaccine. Vaccines (Basel) 2020; 8:vaccines8010025. [PMID: 31947581 PMCID: PMC7157441 DOI: 10.3390/vaccines8010025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 12/27/2019] [Accepted: 01/04/2020] [Indexed: 12/29/2022] Open
Abstract
With the emergence of immune checkpoint inhibitors and adoptive T-cell therapies, there is a considerable interest in using personalized autologous dendritic cell (DC) vaccines in combination with T cell-targeting immunotherapies to potentially maximize the therapeutic impact of DC vaccines. Here, we describe the development and optimization of a Good Manufacturing Practice (GMP)-compliant manufacturing process based on tumor lysate as a tumor antigen source for the production of an oxidized tumor cell lysate loaded DC (OC-DC) vaccine. The manufacturing process required one day for lysate preparation and six days for OC-DC vaccine production. Tumor lysate production was standardized based on an optimal tumor digestion protocol and the immunogenicity was improved through oxidation using hypochloric acid prior to freeze-thaw cycles resulting in the oxidized tumor cell lysate (OC-L). Next, monocytes were selected using the CliniMACS prodigy closed system and were placed in culture in cell factories in the presence of IL-4 and GM-CSF. Immature DCs were loaded with OC-L and matured using MPLA-IFNγ. After assessing the functionality of the OC-DC cells (IL12p70 secretion and COSTIM assay), the OC-DC vaccine was cryopreserved in multiple doses for single use. Finally, the stability of the formulated doses was tested and validated. We believe this GMP-compliant DC vaccine manufacturing process will facilitate access of patients to personalized DC vaccines, and allow for multi-center clinical trials.
Collapse
Affiliation(s)
- Caroline Boudousquié
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; (V.B.); (E.L.); (L.D.); (K.B.); (A.H.); (P.O.G.)
- Correspondence: (C.B.); (L.E.K.)
| | - Valérie Boand
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; (V.B.); (E.L.); (L.D.); (K.B.); (A.H.); (P.O.G.)
| | - Emilie Lingre
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; (V.B.); (E.L.); (L.D.); (K.B.); (A.H.); (P.O.G.)
| | - Laeticia Dutoit
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; (V.B.); (E.L.); (L.D.); (K.B.); (A.H.); (P.O.G.)
| | - Klara Balint
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; (V.B.); (E.L.); (L.D.); (K.B.); (A.H.); (P.O.G.)
| | - Maxime Danilo
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland;
| | - Alexandre Harari
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; (V.B.); (E.L.); (L.D.); (K.B.); (A.H.); (P.O.G.)
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland;
| | - Philippe O. Gannon
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; (V.B.); (E.L.); (L.D.); (K.B.); (A.H.); (P.O.G.)
| | - Lana E. Kandalaft
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; (V.B.); (E.L.); (L.D.); (K.B.); (A.H.); (P.O.G.)
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland;
- Correspondence: (C.B.); (L.E.K.)
| |
Collapse
|
7
|
Hlavackova E, Pilatova K, Cerna D, Selingerova I, Mudry P, Mazanek P, Fedorova L, Merhautova J, Jureckova L, Semerad L, Pacasova R, Flajsarova L, Souckova L, Demlova R, Sterba J, Valik D, Zdrazilova-Dubska L. Dendritic Cell-Based Immunotherapy in Advanced Sarcoma and Neuroblastoma Pediatric Patients: Anti-cancer Treatment Preceding Monocyte Harvest Impairs the Immunostimulatory and Antigen-Presenting Behavior of DCs and Manufacturing Process Outcome. Front Oncol 2019; 9:1034. [PMID: 31709173 PMCID: PMC6823179 DOI: 10.3389/fonc.2019.01034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/24/2019] [Indexed: 01/27/2023] Open
Abstract
Despite efforts to develop novel treatment strategies, refractory and relapsing sarcoma, and high-risk neuroblastoma continue to have poor prognoses and limited overall survival. Monocyte-derived dendritic cell (DC)-based anti-cancer immunotherapy represents a promising treatment modality in these neoplasias. A DC-based anti-cancer vaccine was evaluated for safety in an academic phase-I/II clinical trial for children, adolescents, and young adults with progressive, recurrent, or primarily metastatic high-risk tumors, mainly sarcomas and neuroblastomas. The DC vaccine was loaded with self-tumor antigens obtained from patient tumor tissue. DC vaccine quality was assessed in terms of DC yield, viability, immunophenotype, production of IL-12 and IL-10, and stimulation of allogenic donor T-cells and autologous T-cells in allo-MLR and auto-MLR, respectively. Here, we show that the outcome of the manufacture of DC-based vaccine is highly variable in terms of both DC yield and DC immunostimulatory properties. In 30% of cases, manufacturing resulted in a product that failed to meet medicinal product specifications and therefore was not released for administration to a patient. Focusing on the isolation of monocytes and the pharmacotherapy preceding monocyte harvest, we show that isolation of monocytes by elutriation is not superior to adherence on plastic in terms of DC yield, viability, or immunostimulatory capacity. Trial patients having undergone monocyte-interfering pharmacotherapy prior to monocyte harvest was associated with an impaired DC-based immunotherapy product outcome. Certain combinations of anti-cancer treatment resulted in a similar pattern of inadequate DC parameters, namely, a combination of temozolomide with irinotecan was associated with DCs showing poor maturation and decreased immunostimulatory features, and a combination of pazopanib, topotecan, and MTD-based cyclophosphamide was associated with poor monocyte differentiation and decreased DC immunostimulatory parameters. Searching for a surrogate marker predicting an adverse outcome of DC manufacture in the peripheral blood complete blood count prior to monocyte harvest, we observed an association between an increased number of immature granulocytes in peripheral blood and decreased potency of the DC-based product as quantified by allo-MLR. We conclude that the DC-manufacturing yield and the immunostimulatory quality of anti-cancer DC-based vaccines generated from the monocytes of patients were not influenced by the monocyte isolation modality but were detrimentally affected by the specific combination of anti-cancer agents used prior to monocyte harvest.
Collapse
Affiliation(s)
- Eva Hlavackova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia.,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.,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Dasa Cerna
- Department of Pediatric Oncology, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Iveta Selingerova
- 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
| | - Pavel Mazanek
- Department of Pediatric Oncology, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Lenka Fedorova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Jana Merhautova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Lucie Jureckova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Lukas Semerad
- Department of Internal Medicine-Hematology and Oncology, University Hospital and Medical Faculty, Masaryk University, Brno, Czechia
| | - Rita Pacasova
- Transfusion and Tissue Department, University Hospital Brno, Brno, Czechia
| | - Lucie Flajsarova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Lenka Souckova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia.,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
| | - 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
| | - Dalibor Valik
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Lenka Zdrazilova-Dubska
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| |
Collapse
|
8
|
Abstract
PURPOSES Bone tumours have been a tragedy for the patient in any time period. In the majority of the cases it occurs in children or young adults. In the past the affected limbs could not be spared and the overall prognosis was poor. METHODS Chemotherapy successfully introduced for the purpose of overcoming the poor overall prognosis (Rosen and Jaffe) and tumour prostheses were invented for the purpose of salvaging the affected limbs (Marcove, Scales, Campanacci, Sivas, Salzer). According to the Vienna Tumour Registry in 1968, the first custom-made Vitallium prosthesis for the proximal femur was implanted in a parosteal osteosarcoma. RESULTS In Vienna, as a result of the successful chemotherapy the surgical methods for bone tumours changed to limb sparing methods also. A modular ceramic prosthesis for the proximal humerus was introduced by Salzer. From 1975 -1982 16 custom-made endoprosthesis (1) for the knee region were implanted which were replaced by the KMFTR in 1982 (2, Kotz modular femur tibia reconstruction system) which was introduced at the "2nd ISOLS" to an international group of experts. The successful system was followed by the HMRS (Howmedica modular resection system) in 1988. At that time, especially in children, the rotation-plasty of Borgreve was adopted for tumours of the knee region (2). A scientific survey of 70 patients with rotation-plasty until 1991 showed excellent clinical and oncologic results. Later a similar approach was used in upper extremity tumours as "resection replantation" with surprisingly good results. Sophisticated technologies with growing mechanisms allowed the use of endoprostheses even in children (3) for the purpose of substitution since the mutilating rotation-plasty in 1996. CONCLUSION For almost 100 years efforts have been undertaken to improve the treatment of bone tumours. Surgery was aiming to keep the function of the limbs by tumour resection instead of amputation. Together with successful chemotherapy, which saves lives, an adequate surgery could stepwise salvage the function of the limb. Body integrity was the final aim for the diseased. Finally, by the effort of the International Societies like ISOLS and EMSOS the survival of malignant bone tumour patients improved from 20% to 80 % with good function quality by sophisticated operative techniques and improved tumour prostheses.
Collapse
|
9
|
Abstract
With the spotlight on cancer immunotherapy and the expanding use of immune checkpoint inhibitors, strategies to improve the response rate and duration of current cancer immunotherapeutics are highly sought. In that sense, investigators around the globe have been putting spurs on the development of effective cancer vaccines in humans after decades of efforts that led to limited clinical success. In more than three decades of research in pursuit of targeted and personalized immunotherapy, several platforms have been incorporated into the list of cancer vaccines from live viral or bacterial agents harboring antigens to synthetic peptides with the hope of stronger and durable immune responses that will tackle cancers better. Unlike adoptive cell therapy, cancer vaccines can take advantage of using a patient's entire immune system that can include more than engineered receptors or ligands in developing antigen-specific responses. Advances in molecular technology also secured the use of genetically modified genes or proteins of interest to enhance the chance of stronger immune responses. The formulation of vaccines to increase chances of immune recognition such as nanoparticles for peptide delivery is another area of great interest. Studies indicate that cancer vaccines alone may elicit tumor-specific cellular or humoral responses in immunologic assays and even regression or shrinkage of the cancer in select trials, but novel strategies, especially in combination with other cancer therapies, are under study and are likely to be critical to achieve and optimize reliable objective responses and survival benefit. In this review, cancer vaccine platforms with different approaches to deliver tumor antigens and boost immunity are discussed with the intention of summarizing what we know and what we need to improve in the clinical trial setting.
Collapse
Affiliation(s)
- Hoyoung M. Maeng
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jay A. Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
10
|
|
11
|
Kikete S, Luo L, Jia B, Wang L, Ondieki G, Bian Y. Plant-derived polysaccharides activate dendritic cell-based anti-cancer immunity. Cytotechnology 2018; 70:1097-1110. [PMID: 29556897 PMCID: PMC6081929 DOI: 10.1007/s10616-018-0202-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 02/05/2018] [Indexed: 02/07/2023] Open
Abstract
Today, cancers pose a major public health burden. Although a myriad of cancer treatments are available, only a few have achieved clinical efficacy. This is partly attributed to cancers capability to evade host immunity by converting dendritic cells (DCs) from potent stimulators to negative modulators of immunity. Dendritic cell-based immunotherapy attempts to resolve this problem by manipulating the functional characteristics of DCs. Plant-derived polysaccharides (PDPs) can stimulate the maturation of DCs conferring on them the capacity to present internalised tumorigenic antigens to naïve T cells and subsequently priming T cells to eliminate tumours. PDPs have been used as immune modulators and later as anti-cancer agents by Traditional Chinese Medicine practitioners for centuries. They are abundant in nature and form a large group of heterogeneous though structurally related macromolecules that exhibit diverse immunological properties. They can induce antigen pulsed DCs to acquire functional characteristics in vitro which can subsequently be re-introduced into cancer patients. They can also be used as adjuvants in DC-based vaccines or independently for their intrinsic anti-tumour activities. Clinically, some in vitro generated DCs have been shown to be both safe and immunogenic although their clinical application is limited in part by unsatisfactory functional maturation as well as impaired migration to draining lymph nodes where T cells reside. We review the relative potencies of individual PDPs to induce both phenotypic and functional maturation in DCs, their relative abilities to activate anti-cancer immunity, the possible mechanisms by which they act and also the challenges surrounding their clinical application.
Collapse
Affiliation(s)
- Siambi Kikete
- Tianjin University of Traditional Chinese Medicine, No. 88, Yuquan Road, Nan Kai District, Tianjin, 300193, People's Republic of China
| | - Li Luo
- Department of Rheumatology and Immunology, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830000, People's Republic of China
| | - Beitian Jia
- Tianjin University of Traditional Chinese Medicine, No. 88, Yuquan Road, Nan Kai District, Tianjin, 300193, People's Republic of China
| | - Li Wang
- Tianjin Second People's Hospital, Nan Kai District, Tianjin, 300192, People's Republic of China
| | - Gregory Ondieki
- Tianjin University of Traditional Chinese Medicine, No. 88, Yuquan Road, Nan Kai District, Tianjin, 300193, People's Republic of China
| | - Yuhong Bian
- Tianjin University of Traditional Chinese Medicine, No. 88, Yuquan Road, Nan Kai District, Tianjin, 300193, People's Republic of China.
| |
Collapse
|
12
|
Dobrovolskienė N, Pašukonienė V, Darinskas A, Kraśko JA, Žilionytė K, Mlynska A, Gudlevičienė Ž, Mišeikytė-Kaubrienė E, Schijns V, Lubitz W, Kudela P, Strioga M. Tumor lysate-loaded Bacterial Ghosts as a tool for optimized production of therapeutic dendritic cell-based cancer vaccines. Vaccine 2018; 36:4171-4180. [PMID: 29895501 DOI: 10.1016/j.vaccine.2018.06.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/14/2018] [Accepted: 06/05/2018] [Indexed: 12/18/2022]
Abstract
Cancer immunotherapy with dendritic cell (DC)-based vaccines has been used to treat various malignancies for more than two decades, however generally showed a limited clinical success. Among various factors responsible for their modest clinical activity is the lack of universally applied, standardized protocols for the generation of clinical-grade DC vaccines, capable of inducing effective anti-tumor immune responses. We investigated Bacterial Ghosts (BGs) - empty envelopes of Gram-negative bacteria - as a tool for optimized production of DC vaccines. BGs possess various intact cell surface structures, exhibiting strong adjuvant properties required for the induction of DC maturation, whereas their empty internal space can be easily filled with a source tumor antigens, e.g. tumor lysate. Hence BGs emerge as an excellent platform for both the induction of immunogenic DC maturation and loading with tumor antigens in a single-step procedure. We compared the phenotype, cytokine secretion profile, functional activity and ability to induce immunogenic T-cell responses in vitro of human monocyte-derived DCs generated using BG platform and DCs matured with widely used lipopolysaccharide (LPS) plus interferon-γ cocktail and loaded with tumor lysate. Both approaches induced DC maturation, however BG-based protocol was superior to LPS-based protocol in terms of the ability to induce DCs with a lower tolerogenic potential, resulting in a more robust CD8+ T cell activation and their functional activity as well as significantly lower induction of regulatory T cells. These superior parameters are attributed, at least in part, to the ability of BG-matured DCs to resist potential immunosuppressive and pro-tolerogenic activity of various tumor cell lysates, including melanoma, renal carcinoma and glioblastoma.
Collapse
Affiliation(s)
- N Dobrovolskienė
- National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania.
| | - V Pašukonienė
- National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania.
| | - A Darinskas
- National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania; JSC "Froceth", Linkmenų g. 28, LT-08217 Vilnius, Lithuania
| | - J A Kraśko
- National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania; JSC "Froceth", Linkmenų g. 28, LT-08217 Vilnius, Lithuania.
| | - K Žilionytė
- National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania.
| | - A Mlynska
- National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania.
| | - Ž Gudlevičienė
- National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania.
| | - E Mišeikytė-Kaubrienė
- National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania; Faculty of Medicine, Vilnius University, M.K. Čiurlionio g. 21, LT-03101 Vilnius, Lithuania
| | - V Schijns
- Cell Biology and Immunology, Wageningen University, P.O. Box 338, 6700 AH Wageningen, The Netherlands; Epitopoietic Research Corporation (ERC), ERC-The Netherlands, 5374 RE Schaijk, The Netherlands.
| | - W Lubitz
- BIRD-C GmbH & Co KG, Dr. Bohrgasse 2-8/14/1, A-1030 Vienna, Austria.
| | - P Kudela
- BIRD-C GmbH & Co KG, Dr. Bohrgasse 2-8/14/1, A-1030 Vienna, Austria
| | - M Strioga
- National Cancer Institute, Santariškių g. 1, LT-08660 Vilnius, Lithuania; Faculty of Medicine, Vilnius University, M.K. Čiurlionio g. 21, LT-03101 Vilnius, Lithuania.
| |
Collapse
|
13
|
Abstract
Metastasis contributes to poor prognosis in many types of cancer and is the leading cause of cancer-related deaths. Tumor cells metastasize to distant sites via the circulatory and lymphatic systems. In this review, we discuss the potential of circulating tumor cells for diagnosis and describe the experimental therapeutics that aim to target these disseminating cancer cells. We discuss the advantages and limitations of such strategies and how they may lead to the development of the next generation of antimetastasis treatments.
Collapse
Affiliation(s)
- Eric Lin
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Thong Cao
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| |
Collapse
|
14
|
Bigenzahn S, Juergens B, Mahr B, Pratschke J, Koenigsrainer A, Becker T, Fuchs D, Brandacher G, Kainz A, Muehlbacher F, Wekerle T. No augmentation of indoleamine 2,3-dioxygenase (IDO) activity through belatacept treatment in liver transplant recipients. Clin Exp Immunol 2018; 192:233-241. [PMID: 29271486 DOI: 10.1111/cei.13093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/30/2017] [Accepted: 11/20/2017] [Indexed: 01/18/2023] Open
Abstract
Belatacept is a second-generation cytotoxic T lymphocyte antigen (CTLA)-4 immunoglobulin (Ig) fusion protein approved for immunosuppression in renal transplant recipients. It was designed intentionally to interrupt co-stimulation via CD28 by binding to its ligands B7·1 and B7·2. Experimental evidence suggests a potential additional mechanism for CTLA-4 Ig compounds through binding to B7 molecules expressed on antigen-presenting cells (APCs) and up-regulation of indoleamine 2,3-dioxygenase (IDO), an immunomodulating enzyme that catalyzes the degradation of tryptophan to kynurenine and that down-regulates T cell immunity. So far it remains unknown whether belatacept up-regulates IDO in transplant recipients. We therefore investigated whether belatacept therapy enhances IDO activity in liver transplant recipients enrolled in a multi-centre, investigator-initiated substudy of the Phase II trial of belatacept in liver transplantation (IM103-045). Tryptophan and kynurenine serum levels were measured during the first 6 weeks post-transplant in liver transplant patients randomized to receive either belatacept or tacrolimus-based immunosuppression. There was no significant difference in IDO activity, as indicated by the kynurenine/tryptophan ratio, between belatacept and tacrolimus-treated patients in per-protocol and in intent-to-treat analyses. Moreover, no evidence was found that belatacept affects IDO in human dendritic cells (DC) in vitro. These data provide evidence that belatacept is not associated with detectable IDO induction in the clinical transplant setting compared to tacrolimus-treated patients.
Collapse
Affiliation(s)
- S Bigenzahn
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Austria
| | - B Juergens
- Division of Transplantation Immunology, Children's Cancer Research Institute, St Anna Children's Hospital, Vienna, Austria
| | - B Mahr
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Austria
| | - J Pratschke
- Department of General, Visceral, and Transplantation Surgery, Charité, Berlin, Germany
| | - A Koenigsrainer
- Department of General, Visceral and Transplant Surgery, University Hospital Tuebingen, Tuebingen, Germany
| | - T Becker
- Department of General, Visceral and Transplant Surgery, Hanover Medical School, Hanover, Germany
| | - D Fuchs
- Division of Biological Chemistry, Biocentre, Innsbruck Medical University, Innsbruck, Austria
| | - G Brandacher
- Department of General and Transplant Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - A Kainz
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - F Muehlbacher
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Austria
| | - T Wekerle
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Austria
| |
Collapse
|
15
|
Aichhorn S, Linhardt A, Halfmann A, Nadlinger M, Kirchberger S, Stadler M, Dillinger B, Distel M, Dohnal A, Teasdale I, Schöfberger W. A pH-sensitive Macromolecular Prodrug as TLR7/8 Targeting Immune Response Modifier. Chemistry 2017; 23:17721-17726. [PMID: 28758266 PMCID: PMC5763314 DOI: 10.1002/chem.201702942] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Indexed: 11/09/2022]
Abstract
The chemical synthesis and biological activity of novel functionalized imidazoquinoline derivatives (ImQ) to generate Toll-like receptor (TLR) 7/8 specific prodrugs are presented. In vivo activity of ImQs to induce inflammation was confirmed in zebrafish larvae. After covalent ligation to fully biodegradable polyphosphazenes (ImQ-polymer), the macromolecular prodrugs were designed to undergo intracellular pH-sensitive release of ImQs to induce inflammation through binding to endosomal TLR7/8 (danger signal). We showed ImQ dissociation from prodrugs at a pH 5 pointing towards endosomal prodrug degradability. ImQ-polymers strongly activated ovalbumin-specific T cells in murine splenocytes as shown by increased proliferation and expression of the IL-2 receptor (CD25) on CD8+ T cells accompanied by strong IFN-γ release. ImQ prodrugs presented here are suggested to form the basis of novel nanovaccines, for example, for intravenous or intratumoral cancer immunotherapeutic applications to trigger physiological antitumor immune responses.
Collapse
Affiliation(s)
- Stefan Aichhorn
- Institute of Organic ChemistryJohannes Kepler UniversityAltenberger Straße 694040LinzAustria
| | - Anne Linhardt
- Institute of Polymer ChemistryJohannes Kepler UniversityAltenberger Straße 694040LinzAustria
| | - Angela Halfmann
- Tumorimmunology and Innovative Cancer Models, St. Anna Kinderkrebsforschung e.V. CCRI-Children's Cancer Research InstituteZimmermannplatz 101090ViennaAustria
| | - Markus Nadlinger
- Institute of Organic ChemistryJohannes Kepler UniversityAltenberger Straße 694040LinzAustria
| | - Stefanie Kirchberger
- Tumorimmunology and Innovative Cancer Models, St. Anna Kinderkrebsforschung e.V. CCRI-Children's Cancer Research InstituteZimmermannplatz 101090ViennaAustria
| | - Manuela Stadler
- Tumorimmunology and Innovative Cancer Models, St. Anna Kinderkrebsforschung e.V. CCRI-Children's Cancer Research InstituteZimmermannplatz 101090ViennaAustria
| | - Barbara Dillinger
- Tumorimmunology and Innovative Cancer Models, St. Anna Kinderkrebsforschung e.V. CCRI-Children's Cancer Research InstituteZimmermannplatz 101090ViennaAustria
| | - Martin Distel
- Tumorimmunology and Innovative Cancer Models, St. Anna Kinderkrebsforschung e.V. CCRI-Children's Cancer Research InstituteZimmermannplatz 101090ViennaAustria
| | - Alexander Dohnal
- Tumorimmunology and Innovative Cancer Models, St. Anna Kinderkrebsforschung e.V. CCRI-Children's Cancer Research InstituteZimmermannplatz 101090ViennaAustria
| | - Ian Teasdale
- Institute of Polymer ChemistryJohannes Kepler UniversityAltenberger Straße 694040LinzAustria
| | - Wolfgang Schöfberger
- Institute of Organic ChemistryJohannes Kepler UniversityAltenberger Straße 694040LinzAustria
| |
Collapse
|
16
|
Dwarshuis NJ, Parratt K, Santiago-Miranda A, Roy K. Cells as advanced therapeutics: State-of-the-art, challenges, and opportunities in large scale biomanufacturing of high-quality cells for adoptive immunotherapies. Adv Drug Deliv Rev 2017. [PMID: 28625827 DOI: 10.1016/j.addr.2017.06.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Therapeutic cells hold tremendous promise in treating currently incurable, chronic diseases since they perform multiple, integrated, complex functions in vivo compared to traditional small-molecule drugs or biologics. However, they also pose significant challenges as therapeutic products because (a) their complex mechanisms of actions are difficult to understand and (b) low-cost bioprocesses for large-scale, reproducible manufacturing of cells have yet to be developed. Immunotherapies using T cells and dendritic cells (DCs) have already shown great promise in treating several types of cancers, and human mesenchymal stromal cells (hMSCs) are now extensively being evaluated in clinical trials as immune-modulatory cells. Despite these exciting developments, the full potential of cell-based therapeutics cannot be realized unless new engineering technologies enable cost-effective, consistent manufacturing of high-quality therapeutic cells at large-scale. Here we review cell-based immunotherapy concepts focused on the state-of-the-art in manufacturing processes including cell sourcing, isolation, expansion, modification, quality control (QC), and culture media requirements. We also offer insights into how current technologies could be significantly improved and augmented by new technologies, and how disciplines must converge to meet the long-term needs for large-scale production of cell-based immunotherapies.
Collapse
Affiliation(s)
- Nate J Dwarshuis
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Atlanta, GA 30332-0313, United States; The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States.
| | - Kirsten Parratt
- The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States; Department of Material Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30332, United States.
| | - Adriana Santiago-Miranda
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Atlanta, GA 30332-0313, United States; The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States.
| | - Krishnendu Roy
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Atlanta, GA 30332-0313, United States; The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States.
| |
Collapse
|
17
|
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
|
18
|
Pathogen-Associated Molecular Patterns Induced Crosstalk between Dendritic Cells, T Helper Cells, and Natural Killer Helper Cells Can Improve Dendritic Cell Vaccination. Mediators Inflamm 2016; 2016:5740373. [PMID: 26980946 PMCID: PMC4766350 DOI: 10.1155/2016/5740373] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/30/2015] [Indexed: 12/29/2022] Open
Abstract
A coordinated cellular interplay is of crucial importance in both host defense against pathogens and malignantly transformed cells. The various interactions of Dendritic Cells (DC), Natural Killer (NK) cells, and T helper (Th) cells can be influenced by a variety of pathogen-associated molecular patterns (PAMPs) and will lead to enhanced CD8+ effector T cell responses. Specific Pattern Recognition Receptor (PRR) triggering during maturation enables DC to enhance Th1 as well as NK helper cell responses. This effect is correlated with the amount of IL-12p70 released by DC. Activated NK cells are able to amplify the proinflammatory cytokine profile of DC via the release of IFN-γ. The knowledge on how PAMP recognition can modulate the DC is of importance for the design and definition of appropriate therapeutic cancer vaccines. In this review we will discuss the potential role of specific PAMP-matured DC in optimizing therapeutic DC-based vaccines, as some of these DC are efficiently activating Th1, NK cells, and cytotoxic T cells. Moreover, to optimize these vaccines, also the inhibitory effects of tumor-derived suppressive factors, for example, on the NK-DC crosstalk, should be taken into account. Finally, the suppressive role of the tumor microenvironment in vaccination efficacy and some proposals to overcome this by using combination therapies will be described.
Collapse
|
19
|
Nava S, Lisini D, Pogliani S, Dossena M, Bersano A, Pellegatta S, Parati E, Finocchiaro G, Frigerio S. Safe and Reproducible Preparation of Functional Dendritic Cells for Immunotherapy in Glioblastoma Patients. Stem Cells Transl Med 2015; 4:1164-72. [PMID: 26273063 DOI: 10.5966/sctm.2015-0091] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/06/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Cell therapy based on dendritic cells (DCs) pulsed with tumor lysate is a promising approach in addition to conventional therapy for the treatment of patients with glioblastoma (GB). The success of this approach strongly depends on the ability to generate high-quality, functionally mature DCs (mDCs), with a high level of standardization and in compliance with Good Manufacturing Practices. In the cell factory of the Carlo Besta Foundation, two phase I clinical trials on immunotherapy with tumor lysate-loaded DCs as treatment for GB are ongoing. From 2010 to 2014, 54 patients were enrolled in the studies and 54 batches of DCs were prepared. We retrospectively analyzed the results of the quality control tests carried out on each produced batch, evaluating yield of mDCs and their quality in terms of microbiological safety and immunological efficacy. The number of mDCs obtained allowed the treatment of all the enrolled patients. All 54 batches were sterile, conformed to acceptable endotoxin levels, and were free of Mycoplasma species and adventitious viruses. During culture, cells maintained a high percentage of viability (87%-98%), and all batches showed high viability after thawing (mean±SD: 94.6%±2.9%). Phenotype evaluation of mDCs showed an evident upregulation of markers typical of DC maturation; mixed lymphocyte reaction tests for the functional evaluation of DCs demonstrated that all batches were able to induce lymphocyte responses. These results demonstrated that our protocol for DC preparation is highly reproducible and permits generation of large numbers of safe and functional DCs for in vivo use in immunotherapy approaches. SIGNIFICANCE Cell therapy based on antigen-pulsed dendritic cells (DCs) is a promising approach for the treatment of glioblastoma patients. The success of this approach strongly depends on the ability to generate high-quality, functional DCs with a high level of standardization, ensuring reproducibility, efficacy, and safety of the final product. This article summarizes the results of the quality controls on 54 batches, to demonstrate the feasibility of producing a therapeutic cell-based vaccine via a well-controlled Good Manufacturing Practices (GMP)-compliant production process. The findings may be of scientific interest to those working in the field of preparation of GMP-compliant products for cell-therapy applications.
Collapse
Affiliation(s)
- Sara Nava
- Cell Therapy Production Unit, Laboratory of Cellular Neurobiology, Cerebrovascular Unit, and Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniela Lisini
- Cell Therapy Production Unit, Laboratory of Cellular Neurobiology, Cerebrovascular Unit, and Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Simona Pogliani
- Cell Therapy Production Unit, Laboratory of Cellular Neurobiology, Cerebrovascular Unit, and Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Marta Dossena
- Cell Therapy Production Unit, Laboratory of Cellular Neurobiology, Cerebrovascular Unit, and Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Anna Bersano
- Cell Therapy Production Unit, Laboratory of Cellular Neurobiology, Cerebrovascular Unit, and Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Serena Pellegatta
- Cell Therapy Production Unit, Laboratory of Cellular Neurobiology, Cerebrovascular Unit, and Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Eugenio Parati
- Cell Therapy Production Unit, Laboratory of Cellular Neurobiology, Cerebrovascular Unit, and Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Gaetano Finocchiaro
- Cell Therapy Production Unit, Laboratory of Cellular Neurobiology, Cerebrovascular Unit, and Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Simona Frigerio
- Cell Therapy Production Unit, Laboratory of Cellular Neurobiology, Cerebrovascular Unit, and Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
20
|
Van Gool SW. Brain Tumor Immunotherapy: What have We Learned so Far? Front Oncol 2015; 5:98. [PMID: 26137448 PMCID: PMC4470276 DOI: 10.3389/fonc.2015.00098] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 04/13/2015] [Indexed: 12/17/2022] Open
Abstract
High grade glioma is a rare brain cancer, incurable in spite of modern neurosurgery, radiotherapy, and chemotherapy. Novel approaches are in research, and immunotherapy emerges as a promising strategy. Clinical experiences with active specific immunotherapy demonstrate feasibility, safety and most importantly, but incompletely understood, prolonged long-term survival in a fraction of the patients. In relapsed patients, we developed an immunotherapy schedule and we categorized patients into clinically defined risk profiles. We learned how to combine immunotherapy with standard multimodal treatment strategies for newly diagnosed glioblastoma multiforme patients. The developmental program allows further improvements related to newest scientific insights. Finally, we developed a mode of care within academic centers to organize cell-based therapies for experimental clinical trials in a large number of patients.
Collapse
|
21
|
Datta J, Berk E, Cintolo JA, Xu S, Roses RE, Czerniecki BJ. Rationale for a Multimodality Strategy to Enhance the Efficacy of Dendritic Cell-Based Cancer Immunotherapy. Front Immunol 2015; 6:271. [PMID: 26082780 PMCID: PMC4451636 DOI: 10.3389/fimmu.2015.00271] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/15/2015] [Indexed: 02/03/2023] Open
Abstract
Dendritic cells (DC), master antigen-presenting cells that orchestrate interactions between the adaptive and innate immune arms, are increasingly utilized in cancer immunotherapy. Despite remarkable progress in our understanding of DC immunobiology, as well as several encouraging clinical applications – such as DC-based sipuleucel-T for metastatic castration-resistant prostate cancer – clinically effective DC-based immunotherapy as monotherapy for a majority of tumors remains a distant goal. The complex interplay between diverse molecular and immune processes that govern resistance to DC-based vaccination compels a multimodality approach, encompassing a growing arsenal of antitumor agents which target these distinct processes and synergistically enhance DC function. These include antibody-based targeted molecular therapies, immune checkpoint inhibitors, therapies that inhibit immunosuppressive cellular elements, conventional cytotoxic modalities, and immune potentiating adjuvants. It is likely that in the emerging era of “precision” cancer therapeutics, tangible clinical benefits will only be realized with a multifaceted – and personalized – approach combining DC-based vaccination with adjunctive strategies.
Collapse
Affiliation(s)
- Jashodeep Datta
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Erik Berk
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Jessica A Cintolo
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Shuwen Xu
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Robert E Roses
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Brian J Czerniecki
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA ; Rena Rowen Breast Center, Hospital of the University of Pennsylvania , Philadelphia, PA , USA
| |
Collapse
|
22
|
Nazarkina Z, Laktionov P. Preparation of dendritic cells for cancer immunotherapy. ACTA ACUST UNITED AC 2015; 61:30-40. [DOI: 10.18097/pbmc20156101030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Development of new effective method for cancer therapy is one of the most important trends in the modern medicine. Along with surgery, chemotherapy and radiotherapy, induction of an immune response against the tumor cells is a promising approach for therapy of cancer, particularly metastatic, slowly dividing tumors and cancer stem cells. Induction of the antitumor T-cell immune response involves activation of antigen-presenting cells, which can efficiently present the cancer antigens and activate T-lymphocytes. The immune response may be activated by dendritic cells (DC) loaded with tumor antigens, such as tumor-specific proteins, tumor cell lysates, apoptotic or necrotic tumor cells, as well as nucleic acids encoding tumor antigens. Regardless of the selected source of the tumor antigen, preparation of mature DC is a principal step in the development of anticancer vaccines aimed at the induction of the cytotoxic T-cell immune response. Recently, various research groups have proposed several strategies for producing mature DC, differed by the set of agents used. It has been shown that the maturation strategy influences both their phenotype and the ability to induce the immune response. In this review we have analyzed the results of studies on the various strategies of preparation of mature DCs.
Collapse
Affiliation(s)
- Zh.K. Nazarkina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - P.P. Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
23
|
Datta J, Terhune JH, Lowenfeld L, Cintolo JA, Xu S, Roses RE, Czerniecki BJ. Optimizing dendritic cell-based approaches for cancer immunotherapy. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2014; 87:491-518. [PMID: 25506283 PMCID: PMC4257036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Dendritic cells (DC) are professional antigen-presenting cells uniquely suited for cancer immunotherapy. They induce primary immune responses, potentiate the effector functions of previously primed T-lymphocytes, and orchestrate communication between innate and adaptive immunity. The remarkable diversity of cytokine activation regimens, DC maturation states, and antigen-loading strategies employed in current DC-based vaccine design reflect an evolving, but incomplete, understanding of optimal DC immunobiology. In the clinical realm, existing DC-based cancer immunotherapy efforts have yielded encouraging but inconsistent results. Despite recent U.S. Federal and Drug Administration (FDA) approval of DC-based sipuleucel-T for metastatic castration-resistant prostate cancer, clinically effective DC immunotherapy as monotherapy for a majority of tumors remains a distant goal. Recent work has identified strategies that may allow for more potent "next-generation" DC vaccines. Additionally, multimodality approaches incorporating DC-based immunotherapy may improve clinical outcomes.
Collapse
Affiliation(s)
- Jashodeep Datta
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Julia H. Terhune
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania,Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Lea Lowenfeld
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jessica A. Cintolo
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Shuwen Xu
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Robert E. Roses
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Brian J. Czerniecki
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania,To whom all correspondence should be addressed: Brian J. Czerniecki, MD PhD, Department of Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Drive, Philadelphia, PA 19104; Tele: 215-615-1696; Fax: 215-615-0555;
| |
Collapse
|
24
|
Eyrich M, Schreiber SC, Rachor J, Krauss J, Pauwels F, Hain J, Wölfl M, Lutz MB, de Vleeschouwer S, Schlegel PG, Van Gool SW. Development and validation of a fully GMP-compliant production process of autologous, tumor-lysate-pulsed dendritic cells. Cytotherapy 2014; 16:946-64. [DOI: 10.1016/j.jcyt.2014.02.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 02/04/2014] [Accepted: 02/27/2014] [Indexed: 01/01/2023]
|
25
|
Abstract
Since 1922 surgical approaches toward limb salvage in bone and soft tissue tumours have been documented. There is the famous "Umkippplastik" of Sauerbruch, the "Tikhoff-Linberg" inter-scapulo-thoracic resection or in 1943 a metallic tumour prosthesis for the hip joint in the United States (Moore, Bohlman). Since 1960 acrylic prostheses and metallic prosthesis with bone cement have been in use. Cement-free implants and the first modular ceramic prostheses were implanted in the 1970s in Vienna. At the same time successful chemotherapy in bone sarcomas was introduced by Gerald Rosen and Norman Jaffe. This was mainly the decade of custom-made prostheses. In the 1980s modular tumour prostheses with cone connection to be adopted to the needs of the patient were built intra-operatively. Since 1981 biannual international meetings (ISOLS) have pushed forward the field of bone tumour treatment to allow also tumour resection in wide borders for spine and pelvic tumours. New hope for resistant tumours could be monoclonal antibodies or even dendritic cell therapy.
Collapse
Affiliation(s)
- Rainer I Kotz
- Wiener Privatklinik, Pelikangasse 15, 1090, Vienna, Austria,
| |
Collapse
|
26
|
Geyeregger R, Freimüller C, Stemberger J, Fischer G, Witt V, Fritsch G. Human AdV-specific T cells: persisting in vitro functionality despite lethal irradiation. Bone Marrow Transplant 2014; 49:934-41. [DOI: 10.1038/bmt.2014.86] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 01/15/2014] [Accepted: 03/05/2014] [Indexed: 11/09/2022]
|
27
|
MicroRNA profiling of activated and tolerogenic human dendritic cells. Mediators Inflamm 2014; 2014:259689. [PMID: 24799764 PMCID: PMC3995309 DOI: 10.1155/2014/259689] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/19/2014] [Accepted: 02/21/2014] [Indexed: 01/11/2023] Open
Abstract
Dendritic cells (DCs) belong to the immune system and are particularly studied for their potential to direct either an activated or tolerogenic immune response. The roles of microRNAs (miRNAs) in posttranscriptional gene expression regulation are being increasingly investigated. This study's aim is to evaluate the miRNAs' expression changes in prepared human immature (iDCs), activated (aDCs), and tolerogenic dendritic cells (tDCs). The dendritic cells were prepared using GM-CSF and IL-4 (iDC) and subsequently maturated by adding LPS and IFN-γ (aDC) or IL-10 and TGF-β (tDC). Surface markers, cytokine profiles, and miRNA profiles were evaluated in iDC, tDC, and aDC at 6 h and 24 h of maturation. We identified 4 miRNAs (miR-7, miR-9, miR-155 and miR-182), which were consistently overexpressed in aDC after 6 h and 24 h of maturation and 3 miRNAs (miR-17, miR-133b, and miR-203) and miR-23b cluster solely expressed in tDC. We found 5 miRNAs (miR-10a, miR-203, miR-210, miR-30a, and miR-449b) upregulated and 3 miRNAs downregulated (miR-134, miR-145, and miR-149) in both tDC and aDC. These results indicate that miRNAs are specifically modulated in human DC types. This work may contribute to identifying specific modulating miRNAs for aDC and tDC, which could in the future serve as therapeutic targets in the treatment of cancer and autoimmune diseases.
Collapse
|
28
|
Nazarkina ZK, Laktionov PP. Preparation of dendritic cells for cancer immunotherapy. BIOCHEMISTRY (MOSCOW) SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2014. [DOI: 10.1134/s1990750814020085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Short-term in-vitro expansion improves monitoring and allows affordable generation of virus-specific T-cells against several viruses for a broad clinical application. PLoS One 2013; 8:e59592. [PMID: 23630567 PMCID: PMC3632539 DOI: 10.1371/journal.pone.0059592] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 02/15/2013] [Indexed: 12/04/2022] Open
Abstract
Adenoviral infections are a major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT) in pediatric patients. Adoptive transfer of donor-derived human adenovirus (HAdV)-specific T-cells represents a promising treatment option. However, the difficulty in identifying and selecting rare HAdV-specific T-cells, and the short time span between patients at high risk for invasive infection and viremia are major limitations. We therefore developed an IL-15-driven 6 to 12 day short-term protocol for in vitro detection of HAdV-specific T cells, as revealed by known MHC class I multimers and a newly identified adenoviral CD8 T-cell epitope derived from the E1A protein for the frequent HLA-type A*02∶01 and IFN-γ. Using this novel and improved diagnostic approach we observed a correlation between adenoviral load and reconstitution of CD8+ and CD4+ HAdV-specific T-cells including central memory cells in HSCT-patients. Adaption of the 12-day protocol to good manufacturing practice conditions resulted in a 2.6-log (mean) expansion of HAdV-specific T-cells displaying high cytolytic activity (4-fold) compared to controls and low or absent alloreactivity. Similar protocols successfully identified and rapidly expanded CMV-, EBV-, and BKV-specific T-cells. Our approach provides a powerful clinical-grade convertible tool for rapid and cost-effective detection and enrichment of multiple virus-specific T-cells that may facilitate broad clinical application.
Collapse
|
30
|
Luger R, Valookaran S, Knapp N, Vizzardelli C, Dohnal AM, Felzmann T. Toll-like receptor 4 engagement drives differentiation of human and murine dendritic cells from a pro- into an anti-inflammatory mode. PLoS One 2013; 8:e54879. [PMID: 23408948 PMCID: PMC3569454 DOI: 10.1371/journal.pone.0054879] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 12/17/2012] [Indexed: 01/21/2023] Open
Abstract
The dendritic cell (DC) coordinates innate and adaptive immunity to fight infections and cancer. Our observations reveal that DCs exposed to the microbial danger signal lipopolysaccharide (LPS) in the presence of interferon-γ (IFN-γ) acquire a continuously changing activation/maturation phenotype. The DCs’ initial mode of action is pro-inflammatory via up-regulation among others of the signaling molecule interleukin (IL) 12, which polarizes IFN-γ secreting type 1 helper T-cells (Th1). Within 24 hours the same DC switches from the pro- into an anti-inflammatory phenotype. This is mediated by autocrine IL-10 release and secretion of soluble IL-2 receptor alpha (sIL-2RA) molecules. T-cells, when contacted with DCs during their anti-inflammatory phase loose their proliferative capacity and develop regulatory T-cell (Treg) -like anti-inflammatory functions indicated by IL-10 secretion and elevated FoxP3 levels. Studying the kinetics of IL-12 and IL-10 expression from LPS/IFN-γ activated myeloid DCs on a single cell level confirmed these observations. When T-cells are separated from DCs within 24 hours, they are spared from the anti-inflammatory DC activity. We conclude that, in addition to differentiation of DCs into distinct subsets, the observed sequential functional phases of DC differentiation permit the fine-tuning of an immune response. A better understanding of time-kinetic DC features is required for optimally exploiting the therapeutic capacity of DCs in cancer immune therapy.
Collapse
Affiliation(s)
- Romana Luger
- St. Anna Children’s Cancer Research Institute, Laboratory of Tumor Immunology, Department of Pediatrics, Medical University Vienna, Austria
| | - Sneha Valookaran
- St. Anna Children’s Cancer Research Institute, Laboratory of Tumor Immunology, Department of Pediatrics, Medical University Vienna, Austria
| | - Natalie Knapp
- St. Anna Children’s Cancer Research Institute, Laboratory of Tumor Immunology, Department of Pediatrics, Medical University Vienna, Austria
| | - Caterina Vizzardelli
- St. Anna Children’s Cancer Research Institute, Laboratory of Tumor Immunology, Department of Pediatrics, Medical University Vienna, Austria
| | - Alexander M. Dohnal
- St. Anna Children’s Cancer Research Institute, Laboratory of Tumor Immunology, Department of Pediatrics, Medical University Vienna, Austria
| | - Thomas Felzmann
- St. Anna Children’s Cancer Research Institute, Laboratory of Tumor Immunology, Department of Pediatrics, Medical University Vienna, Austria
- Activartis Biotech GmbH, Vienna, Austria
- * E-mail:
| |
Collapse
|
31
|
Neurokinin-1 receptor agonists bias therapeutic dendritic cells to induce type 1 immunity by licensing host dendritic cells to produce IL-12. Blood 2013; 121:2923-33. [PMID: 23365459 DOI: 10.1182/blood-2012-07-446054] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Substance-P and hemokinin-1 are proinflammatory neuropeptides with potential to promote type 1 immunity through agonistic binding to neurokinin-1 receptor (NK1R). Dendritic cells (DCs) are professional antigen-presenting cells that initiate and regulate the outcome of innate and adaptive immune responses. Immunostimulatory DCs are highly desired for the development of positive immunization techniques. DCs express functional NK1R; however, regardless of their potential DC-stimulatory function, the ability of NK1R agonists to promote immunostimulatory DCs remains unexplored. Here, we demonstrate that NK1R signaling activates therapeutic DCs capable of biasing type 1 immunity by inhibition of interleukin-10 (IL-10) synthesis and secretion, without affecting their low levels of IL-12 production. The potent type 1 effector immune response observed following cutaneous administration of NK1R-signaled DCs required their homing in skin-draining lymph nodes (sDLNs) where they induced inflammation and licensed endogenous-conventional sDLN-resident and -recruited inflammatory DCs to secrete IL-12. Our data demonstrate that NK1R signaling promotes immunostimulatory DCs, and provide relevant insight into the mechanisms used by neuromediators to regulate innate and adaptive immune responses.
Collapse
|
32
|
Grosse J, Meier K, Bauer TJ, Eilles C, Grimm D. Cell separation by countercurrent centrifugal elutriation: recent developments. Prep Biochem Biotechnol 2012; 42:217-33. [PMID: 22509848 DOI: 10.1080/10826068.2011.602799] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Countercurrent centrifugal elutriation (CCE) is a cell separation technique that separates particles predominantly according to their size, and to some degree according to their specific density, without a need for antibodies or ligands tagging cell surfaces. The principles of this technique have been known for half a century. Still, numerous recent publications confirmed that CCE is a valuable supplement to current cell separation technology. It is mainly applied when homogeneous populations of cells, which mirror an in vivo situation, are required for answering scientific questions or for clinical transplantation, while antibodies or ligands suitable for cell isolation are not available. Currently, new technical developments are expanding its application toward fractionation of healthy and malignant tissue cells and the preparation of dendritic cells for immunotherapy.
Collapse
Affiliation(s)
- Jirka Grosse
- Department of Nuclear Medicine, University of Regensburg, Regensburg, Germany
| | | | | | | | | |
Collapse
|
33
|
Kitawaki T, Kadowaki N, Fukunaga K, Kasai Y, Maekawa T, Ohmori K, Itoh T, Shimizu A, Kuzushima K, Kondo T, Ishikawa T, Uchiyama T. Cross-priming of CD8+ T cells in vivo by dendritic cells pulsed with autologous apoptotic leukemic cells in immunotherapy for elderly patients with acute myeloid leukemia. Exp Hematol 2011; 39:424-433.e2. [DOI: 10.1016/j.exphem.2011.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 12/28/2010] [Accepted: 01/01/2011] [Indexed: 10/18/2022]
|
34
|
Castiello L, Sabatino M, Jin P, Clayberger C, Marincola FM, Krensky AM, Stroncek DF. Monocyte-derived DC maturation strategies and related pathways: a transcriptional view. Cancer Immunol Immunother 2011; 60:457-66. [PMID: 21258790 DOI: 10.1007/s00262-010-0954-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 11/30/2010] [Indexed: 12/17/2022]
Abstract
Ex vivo production of highly stimulator mature dendritic cells (DCs) for cellular therapy has been used to treat different pathological conditions with the aim of inducing a specific immune response. In the last decade, several protocols have been developed to mature monocyte-derived DCs: each one has led to the generation of DCs showing different phenotypes and stimulatory abilities, but it is not yet known which one is the best for inducing effective immune responses. We grouped several different maturation protocols according to the downstream pathways they activated and reviewed the shared features at a transcriptomic level to reveal the potential of DCs matured by each protocol to develop Th-polarized immune responses.
Collapse
Affiliation(s)
- Luciano Castiello
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Vilekar P, Awasthi V, Lagisetty P, King C, Shankar N, Awasthi S. In vivo trafficking and immunostimulatory potential of an intranasally-administered primary dendritic cell-based vaccine. BMC Immunol 2010; 11:60. [PMID: 21143974 PMCID: PMC3018378 DOI: 10.1186/1471-2172-11-60] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 12/10/2010] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Coccidioidomycosis or Valley fever is caused by a highly virulent fungal pathogen: Coccidioides posadasii or immitis. Vaccine development against Coccidioides is of contemporary interest because a large number of relapses and clinical failures are reported with antifungal agents. An efficient Th1 response engenders protection. Thus, we have focused on developing a dendritic cell (DC)-based vaccine for coccidioidomycosis. In this study, we investigated the immunostimulatory characteristics of an intranasal primary DC-vaccine in BALB/c mouse strain that is most susceptible to coccidioidomycosis. The DCs were transfected nonvirally with Coccidioides-Ag2/PRA-cDNA. Expression of DC-markers, Ag2/PRA and cytokines were studied by flow cytometry, dot-immunoblotting and cytometric bead array methods, respectively. The T cell activation was studied by assessing the upregulation of activation markers in a DC-T cell co-culture assay. For trafficking, the DCs were co-transfected with a plasmid DNA encoding HSV1 thymidine kinase (TK) and administered intranasally into syngeneic mice. The trafficking and homing of TK-expressing DCs were monitored with positron emission tomography (PET) using 18F-FIAU probe. Based on the PET-probe accumulation in vaccinated mice, selected tissues were studied for antigen-specific response and T cell phenotypes using ELISPOT and flow cytometry, respectively. RESULTS We found that the primary DCs transfected with Coccidioides-Ag2/PRA-cDNA were of immature immunophenotype, expressed Ag2/PRA and activated naïve T cells. In PET images and subsequent biodistribution, intranasally-administered DCs were found to migrate in blood, lung and thymus; lymphocytes showed generation of T effector memory cell population (T(EM)) and IFN-γ release. CONCLUSIONS In conclusion, our results demonstrate that the intranasally-administered primary DC vaccine is capable of inducing Ag2/PRA-specific T cell response. Unique approaches utilized in our study represent an attractive and novel means of producing and evaluating an autologous DC-based vaccine.
Collapse
Affiliation(s)
- Prachi Vilekar
- Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Oklahoma City, OK 73117, USA
| | | | | | | | | | | |
Collapse
|
36
|
Eifler RL, Lind J, Falkenhagen D, Weber V, Fischer MB, Zeillinger R. Enrichment of circulating tumor cells from a large blood volume using leukapheresis and elutriation: proof of concept. CYTOMETRY PART B-CLINICAL CYTOMETRY 2010; 80:100-11. [PMID: 20954267 DOI: 10.1002/cyto.b.20560] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 07/07/2010] [Accepted: 07/21/2010] [Indexed: 12/25/2022]
Abstract
BACKGROUND The aim of this study was to determine the applicability of a sequential process using leukapheresis, elutriation, and fluorescence-activated cell sorting (FACS) to enrich and isolate circulating tumor cells from a large blood volume to allow further molecular analysis. METHODS Mononuclear cells were collected from 10 L of blood by leukapheresis, to which carboxyfluorescein succinimidyl ester prelabeled CaOV-3 tumor cells were spiked at a ratio of 26 to 10⁶ leukocytes. Elutriation separated the spiked leukapheresates primarily by cell size into distinct fractions, and leukocytes and tumor cells, characterized as carboxyfluorescein succinimidyl ester positive, EpCAM positive and CD45 negative events, were quantified by flow cytometry. Tumor cells were isolated from the last fraction using FACS or anti-EpCAM coupled immunomagnetic beads, and their recovery and purity determined by fluorescent microscopy and real-time PCR. RESULTS Leukapheresis collected 13.5 x 10⁹ mononuclear cells with 87% efficiency. In total, 53 to 78% of spiked tumor cells were pre-enriched in the last elutriation fraction among 1.6 x 10⁹ monocytes. Flow cytometry predicted a circulating tumor cell purity of ~90% giving an enrichment of 100,000-fold following leukapheresis, elutriation, and FACS, where CaOV-3 cells were identified as EpCAM positive and CD45 negative events. FACS confirmed this purity. Alternatively, immunomagnetic bead adsorption recovered 10% of tumor cells with a median purity of 3.5%. CONCLUSIONS This proof of concept study demonstrated that elutriation and FACS following leukapheresis are able to enrich and isolate tumor cells from a large blood volume for molecular characterization.
Collapse
Affiliation(s)
- Robert L Eifler
- Department of Clinical Medicine and Biotechnology, Danube University, Krems, Austria; Department of Blood Transfusion, Medical University of Vienna, Austria
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
Dendritic cells form the connection between innate and adoptive mechanisms of the immune system. As antigen-presenting cells, dendritic cells are capable of presenting tumour antigen and effectively stimulating immune response targeted against a tumour. A number of preclinical and clinical studies document dendritic cells' potential in anti-cancer treatment. Increasing knowledge of dendritic cell biology is leading to improved methods for their preparation for clinical application. Unfortunately, there is to date no consensus specifying optimal conditions for dendritic cell preparation in vitro. This review summarizes the methods used for preparing myeloid dendritic cells derived from monocytic precursors while focusing on cytokine cocktails used for their growth, maturation, and functional adjustment.
Collapse
|
38
|
Michael Dohnal A, Luger R, Paul P, Fuchs D, Felzmann T. CD40 ligation restores type 1 polarizing capacity in TLR4-activated dendritic cells that have ceased interleukin-12 expression. J Cell Mol Med 2010; 13:1741-1750. [PMID: 20187300 DOI: 10.1111/j.1582-4934.2008.00584.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Inflammation triggered by microbial lipopolysaccharide (LPS) through Toll-like receptor (TLR) 4 in the presence of interferon (IFN)-gamma induces cytokine secretion in dendritic cells (DCs) tightly regulated by a defined differentiation program. This DC differentiation is characterized not only by a dynamic immune activating but also by tolerance-inducing phenotype associated with down-modulation of cytokines previously considered to be irreversible. CD40L on activated T cells further modifies DC differentiation. Using DNA micro-arrays, we showed down-regulated mRNA levels of TLR signalling molecules, whereas CD40/CD40L signalling molecules were up-regulated at a time when LPS/IFN-gamma-activated DCs had ceased cytokine expression. Accordingly, we demonstrated that CD40/CD40L but not TLR4 or TLR3 signalling mediated by LPS or poly (cytidylic-inosinic) acid (poly I:C) and dsRNA re-established the capacity for secreting interleukin (IL)-12 in primarily LPS/IFN-gamma-activated DCs, which have exhausted their potential for cytokine secretion. The resulting TH1 polarizing DC phenotype - which lacked accompanying secretion of the crucial immune suppressive factor IL-10 - maintained the potential for activation of cytotoxic T lymphocytes (CTLs). We therefore conclude that immune modulation is restricted to a secondary T-cell-mediated stimulus at an exhausted DC state, which prevents an immune tolerant DC phenotype. These findings impact on the rational design of TLR-activated DC-based cancer vaccines for the induction of anti-tumoural CTL responses.
Collapse
Affiliation(s)
- Alexander Michael Dohnal
- Laboratory of Tumor Immunology, St. Anna Children's Cancer Research Institute, Vienna, Austria.,Trimed Biotech, Vienna, Austria
| | - Romana Luger
- Laboratory of Tumor Immunology, St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Petra Paul
- Laboratory of Tumor Immunology, St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Thomas Felzmann
- Laboratory of Tumor Immunology, St. Anna Children's Cancer Research Institute, Vienna, Austria.,Trimed Biotech, Vienna, Austria
| |
Collapse
|
39
|
Labbe A, Nelles M, Walia J, Jia L, Furlonger C, Nonaka T, Medin JA, Paige CJ. IL-12 immunotherapy of murine leukaemia: comparison of systemic versus gene modified cell therapy. J Cell Mol Med 2010; 13:1962-1976. [PMID: 18624776 DOI: 10.1111/j.1582-4934.2008.00412.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The ability of IL-12 to initiate anti-leukaemia immune responses has been well established; however clinical outcomes fail to recapitulate the therapeutic benefits observed in the laboratory. To address this, we compared two systems of IL-12 therapy that elicit protective immune responses against the murine acute lymphoblastic leukaemia (ALL) cell line, 70Z/3. These systems differ in the method of IL-12 administration and ultimately result in leukaemia clearance by distinct mechanisms, emphasizing the importance of treatment vehicle. Injecting low-dose IL-12 was sufficient to elicit long-term protective immunity against an established leukaemia burden, mediated by both CD4(+) and CD8(+) T cells. These findings agree with the standard model of IL-12 activity. We compared this protocol to a cell-based approach in which a novel lentiviral vector (LV) expressing murine IL-12 was created, 70Z/3 cells transduced, and clones selected that stably secrete different amounts of IL-12. We found that only a small proportion (1%) of IL-12 secreting cells were required for rejection but that the amount of IL-12 produced per cell was critical for successful therapy. Importantly, the levels of IL-12 required were found to be higher than the levels reported to date in the human clinical trial literature. We found that the cell-based approach led to protective immunity that was both long-term and specific but dependent primarily on a CD4(+) cellular subset alone. Our results highlight that the mode of IL-12 delivery has a distinct impact on the immune response initiated, leading to leukaemia clearance by disparate mechanisms. We also establish a new and critical parameter, IL-12 production/cell, which may have significant implications for future therapeutic design.
Collapse
Affiliation(s)
- Alain Labbe
- Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada.,Departments of Immunology
| | - Megan Nelles
- Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada.,Medical Biophysics
| | - Jagdeep Walia
- Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | - Lintao Jia
- Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | - Caren Furlonger
- Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | - Takahiro Nonaka
- Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | - Jeffrey A Medin
- Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada.,Medical Biophysics.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Christopher J Paige
- Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada.,Departments of Immunology.,Medical Biophysics
| |
Collapse
|
40
|
Li Y, Xu J, Zou H, Wang C. 1-MT enhances potency of tumor cell lysate-pulsed dendritic cells against pancreatic adenocarcinoma by downregulating the percentage of Tregs. ACTA ACUST UNITED AC 2010; 30:344-8. [PMID: 20556579 DOI: 10.1007/s11596-010-0354-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Indexed: 12/23/2022]
Abstract
This study examined whether 1-methyl-tryptophan [1-MT, an indoleamine 2, 3-dioxygenase (IDO) inhibitor] could reduce CD4+CD25+ regulatory T cells (Tregs) proliferation and improve the anti-tumor efficacy of dendritic cells (DCs) pulsed with tumor cell lysate in the mice bearing pancreatic adenocarcinoma. The models of pancreatic adenocarcinoma were established in C57BL/6 mice by subcutaneous injection of Pan02 cells. Eight mice which were subcutaneously injected with PBS served as control. The expression of IDO was determined in tumor draining lymph nodes (TDLNs) and spleens of the murine pancreatic adenocarcinoma models. The prevalence of Tregs was measured in the TDLNs and spleens before and after 1-MT administration. The dendritic cells were pulsed with tumor cell lysate for preparing DC vaccine. The DC vaccine, as a single agent or in combination with 1-MT, was administered to pancreatic adenocarcinoma mice. The anti-tumor efficacy was determined after different treatments by regular observation of tumor size. The results showed that the levels of IDO mRNA and protein in tumor-bearing mice were significantly higher than those in the normal control mice. The percentage of Tregs in the spleen and TDLNs was also higer in tumor-bearing mice than in normal control mice (P<0.05). Foxp3 expression was significantly lower in the TDLNs and spleens of tumor-bearing mice administrated with 1-MT than that in normal control mice. Furthemore, in the mice that were administered 1-MT plus DC vaccine, the tumor was increased more slowly than in mice treated with DC vaccine or 1-MT alone, or PBS on day 36 (P<0.01). Our results indicated that 1-MT may enhance anti-tumor efficacy of dendritic cells pulsed with tumor cell lysate by downregulating the percentage of Tregs.
Collapse
Affiliation(s)
- Yuandong Li
- Pancreatic Surgery Center of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | | | | | | |
Collapse
|
41
|
Dendritic Cells Generated in Clinical Grade Bags Strongly Differ in Immune Functionality When Compared With Classical DCs Generated in Plates. J Immunother 2010; 33:352-63. [DOI: 10.1097/cji.0b013e3181cc266b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
42
|
Wolf B, Posnick D, Fisher JL, Lewis LD, Ernstoff MS. Indoleamine-2,3-dioxygenase enzyme expression and activity in polarized dendritic cells. Cytotherapy 2010; 11:1084-9. [PMID: 19929471 DOI: 10.3109/14653240903271230] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Polarized mature dendritic cells (DC) can activate cytolytic T-lymphocyte (CTL) responses and may be a more effective clinical strategy in DC-based cancer vaccines. A subset of mature DC can down-regulate the T-cell immune response through expression of indoleamine-2,3-dioxygenase (IDO). We determined whether polarizing DC ex vivo increased IDO expression and activity. METHODS Peripheral blood monocytes from healthy volunteers were cultured ex vivo in polarizing and non-polarizing culture conditions. DC IDO expression was detected by Western blot. IDO enzyme activity was determined by high-performance liquid chromatography (HPLC) measurement of kynurenine (K) and tryptophan (T) concentrations in culture supernatants. RESULTS IDO protein was markedly increased in DC after polarization (median 1222.4%, range 331.5-2113.3%) versus non-polarized DC (median 28.3%, range 3.7-119.8%; P=0.04). The median K/T ratio was significantly higher in polarized DC versus non-polarized DC (6.34, range 6.02-6.65, versus 0.047, range 0.004-0.541; P=0.04). IDO protein expression correlated with enzyme activity (r=0.80, P=0.002). CONCLUSIONS DC polarizing culture conditions increased expression of IDO protein and IDO enzyme activity. DC culture and maturation methodologies may impact the effectiveness of adoptive DC therapy.
Collapse
Affiliation(s)
- Benita Wolf
- Medical Oncology Immunotherapy Group, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire 03756, USA
| | | | | | | | | |
Collapse
|
43
|
Janssen WE, Ribickas A, Meyer LV, Smilee RC. Large-scale Ficoll gradient separations using a commercially available, effectively closed, system. Cytotherapy 2010; 12:418-24. [DOI: 10.3109/14653240903479663] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
44
|
Immune Suppression by γδ T-cells as a Potential Regulatory Mechanism After Cancer Vaccination With IL-12 Secreting Dendritic Cells. J Immunother 2010; 33:40-52. [DOI: 10.1097/cji.0b013e3181b51447] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
Gulen D, Abe F, Maas S, Reed E, Cowan K, Pirruccello S, Wisecarver J, Warkentin P, Northam M, Turken O, Coskun U, Senesac J, Talmadge JE. Closing the manufacturing process of dendritic cell vaccines transduced with adenovirus vectors. Int Immunopharmacol 2008; 8:1728-36. [DOI: 10.1016/j.intimp.2008.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Accepted: 08/12/2008] [Indexed: 10/21/2022]
|