1
|
Friis T, Pedersen KB, Hougaard D, Houen G. Cytochemical and Histochemical Staining with Peptide Antibodies. Methods Mol Biol 2024; 2821:249-263. [PMID: 38997495 DOI: 10.1007/978-1-0716-3914-6_20] [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: 07/14/2024]
Abstract
Peptide antibodies are particularly useful for immunocytochemistry (ICC) and immunohistochemistry (IHC), where antigens may denature due to fixation of tissues and cells. Peptide antibodies can be made to any defined sequence, including unknown putative proteins and posttranslationally modified sequences. Moreover, the availability of large amounts of the antigen (peptide) allows inhibition/absorption controls, which are important in ICC/IHC, due to the many possibilities for false-positive reactions caused by immunoglobulin Fc receptors, nonspecific reactions and cross-reactivity of primary and secondary antibodies with other antigens and endogenous immunoglobulins, respectively. Here, simple protocols for ICC and IHC are described together with recommendations for appropriate controls.
Collapse
Affiliation(s)
- Tina Friis
- Department of Congenital Diseases, Statens Serum Institut, Copenhagen S, Denmark
| | - Klaus Boberg Pedersen
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Copenhagen, Denmark
| | - David Hougaard
- Department of Congenital Diseases, Statens Serum Institut, Copenhagen, Denmark
| | - Gunnar Houen
- Department of Neurology and Translational Research Center, Rigshospitalet, Glostrup, Denmark.
| |
Collapse
|
2
|
Malati ZA, Pourfathollah AA, Dabbaghi R, Balagholi S, Javan MR. Evaluation of a New Method of Leukocyte Extractions from the Leukoreduction Filter. Indian J Hematol Blood Transfus 2023; 39:478-486. [PMID: 37304478 PMCID: PMC10247650 DOI: 10.1007/s12288-022-01618-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 12/19/2022] [Indexed: 01/15/2023] Open
Abstract
This study's purpose was to optimize the leukocyte extraction protocol and evaluate the efficacy of this new protocol. 12BioR blood filters were collected from Tehran Blood Transfusion Center. A twosyringe system and Multi-step rinsing were designed for cell extraction. The final purpose of this optimization was: (1) removed the residual RBCs, (2) reversed the leukocyte trapping process, and (3) remove the microparticles to obtain the high yield of target cells. Finally, Extracted cells were evaluated by Automated Cell count; Samples smear differential cell count, Trypan blue, and Annexin-PI staining. The results showed that on average 11.88 × 108 ± 3.32 leukocytes recovered after indirect washing and that the mean count of granulocytes, lymphocytes, and Monocyte in this sample was 5.24 ± 2.18 × 108, 5.57 ± 1.74 × 108, and 0.56 ± 0.38 × 108 respectively. Also, the mean percent of manual differential cell count after concentration was 42.81%, 41.80%, and 15.82% for granulocytes, lymphocytes, and monocytes respectively. Moreover, viability and apoptosis assay showed > 95% viability in mononuclear cells recovered from LRFs. It is concluded that the use of a double-syringe system and RBC and microparticles removal from leukoreduction filters lead to acceptable viable leukocyte count that can be used in in vitro and in vivo studies.
Collapse
Affiliation(s)
- Zahra Abbasi Malati
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Ali Akbar Pourfathollah
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
- Immunology Department Faculty Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Rasul Dabbaghi
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Balagholi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Javan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
3
|
Beignon AS, Galeotti C, Menager MM, Schvartz A. Trained immunity as a possible newcomer in autoinflammatory and autoimmune diseases pathophysiology. Front Med (Lausanne) 2023; 9:1085339. [PMID: 36743677 PMCID: PMC9896524 DOI: 10.3389/fmed.2022.1085339] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Autoimmune disorders have been well characterized over the years and many pathways-but not all of them-have been found to explain their pathophysiology. Autoinflammatory disorders, on the other hand, are still hiding most of their molecular and cellular mechanisms. During the past few years, a newcomer has challenged the idea that only adaptive immunity could display memory response. Trained immunity is defined by innate immune responses that are faster and stronger to a second stimulus than to the first one, being the same or not. In response to the trained immunity inducer, and through metabolic and epigenetic changes of hematopoietic stem and progenitor cells in the bone marrow that are transmitted to their cellular progeny (peripheral trained immunity), or directly of tissue-resident cells (local innate immunity), innate cells responsiveness and functions upon stimulation are improved in the long-term. Innate immunity can be beneficial, but it could also be detrimental when maladaptive. Here, we discuss how trained immunity could contribute to the physiopathology of autoimmune and autoinflammatory diseases.
Collapse
Affiliation(s)
- Anne-Sophie Beignon
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases/Infectious Diseases Models and Innovative Technologies (IMVA-HB/IDMIT), U1184, Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Caroline Galeotti
- Department of Pediatric Rheumatology, Reference Center for AutoInflammatory Diseases and Amyloidosis (CEREMAIA), Hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France
| | - Mickael M. Menager
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases/Infectious Diseases Models and Innovative Technologies (IMVA-HB/IDMIT), U1184, Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Adrien Schvartz
- Department of Pediatric Rheumatology, Reference Center for AutoInflammatory Diseases and Amyloidosis (CEREMAIA), Hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France,*Correspondence: Adrien Schvartz,
| |
Collapse
|
4
|
Cunningham S, Hackstein H. Rapid generation of monocyte-derived antigen-presenting cells with dendritic cell-like properties. Transfusion 2021; 61:1845-1855. [PMID: 33786883 DOI: 10.1111/trf.16385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND One of the major challenges in cellular therapy is the establishment and validation of simple and fast production protocols meeting good manufacturing practice (GMP) requirements. Dendritic cells (DCs) are of particular therapeutic interest, due to their critical role in T cell response initiation and regulation. Conventional wisdom states that DC generation from monocytes is a time-consuming protocol, taking up to 7-9 days. STUDY DESIGN AND METHODS This study systematically screened and validated numerous culture components and conditions to identify the minimal requirements, which can give rise to functional monocyte-derived antigen-presenting cells (MoAPCs) in less than 48 h (36 h MoAPC). A total of 36 h MoAPCs were evaluated in terms of surface marker expression, endocytic capability, and induction of antigen-specific T cell expansion via flow cytometry. RESULTS Screening of media compositions, glucose concentrations, and surface marker kinetics, particularly DC-SIGN as a DC-specific marker, allowed the generation of DC-like APCs in 36 h (36 h MoAPCs). A total of 36 h MoAPCs displayed a similar phenotype to 48 h MoAPC and standard 7 d MoDCs in terms of HLA-DP,DQ,DR, CD83, and DC-SIGN expression, while CD1a was preferentially expressed in standard MoDCs. Functional evaluation revealed that 36 h MoAPCs displayed reduced endocytosis capabilities and IL-12p70 production. However, 36 h MoAPCs were able to induce T cell expansion both in an allogenic and antigen-specific setting. CONCLUSION Our results indicate that mature 36 h MoAPCs possess DC-like capabilities by inducing antigen-specific T cell responses. This study has important implications for the generation of DC-based cellular therapies, allowing a more cost and time-efficient generation of APCs.
Collapse
Affiliation(s)
- Sarah Cunningham
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
5
|
Cunningham S, Hackstein H. Recent Advances in Good Manufacturing Practice-Grade Generation of Dendritic Cells. Transfus Med Hemother 2020; 47:454-463. [PMID: 33442340 DOI: 10.1159/000512451] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/11/2020] [Indexed: 12/23/2022] Open
Abstract
Dendritic cells (DCs) are pivotal regulators of immune responses, specialized in antigen presentation and bridging the gap between the innate and adaptive immune system. Due to these key features, DCs have become a pillar of the continuously growing field of cellular therapies. Here we review recent advances in good manufacturing practice strategies and their individual specificities in relation to DC production for clinical applications. These take into account both small-scale experimental approaches as well as automated systems for patient care.
Collapse
Affiliation(s)
- Sarah Cunningham
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
6
|
Caffiyar MY, Lim KP, Basha IHK, Hamid NH, Cheong SC, Ho ETW. Label-Free, High-Throughput Assay of Human Dendritic Cells from Whole-Blood Samples with Microfluidic Inertial Separation Suitable for Resource-Limited Manufacturing. MICROMACHINES 2020; 11:mi11050514. [PMID: 32438709 PMCID: PMC7281724 DOI: 10.3390/mi11050514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/31/2020] [Accepted: 04/05/2020] [Indexed: 02/07/2023]
Abstract
Microfluidics technology has not impacted the delivery and accessibility of point-of-care health services, like diagnosing infectious disease, monitoring health or delivering interventions. Most microfluidics prototypes in academic research are not easy to scale-up with industrial-scale fabrication techniques and cannot be operated without complex manipulations of supporting equipment and additives, such as labels or reagents. We propose a label- and reagent-free inertial spiral microfluidic device to separate red blood, white blood and dendritic cells from blood fluid, for applications in health monitoring and immunotherapy. We demonstrate that using larger channel widths, in the range of 200 to 600 µm, allows separation of cells into multiple focused streams, according to different size ranges, and we utilize a novel technique to collect the closely separated focused cell streams, without constricting the channel. Our contribution is a method to adapt spiral inertial microfluidic designs to separate more than two cell types in the same device, which is robust against clogging, simple to operate and suitable for fabrication and deployment in resource-limited populations. When tested on actual human blood cells, 77% of dendritic cells were separated and 80% of cells remained viable after our assay.
Collapse
Affiliation(s)
- Mohamed Yousuff Caffiyar
- Department of Electrical & Electronics Engineering, Universiti Teknologi PETRONAS, Perak 32610, Malaysia; (M.Y.C.); (I.H.K.B.); (N.H.H.)
- Department of Electronics and Communication Engineering, C. Abdul Hakeem College of Engineering and Technology, Melvisharam, Tamil Nadu 632509, India
| | - Kue Peng Lim
- Head and Neck Cancer Research Group, Cancer Research Malaysia, Selangor 47500, Malaysia; (K.P.L.); (S.C.C.)
| | - Ismail Hussain Kamal Basha
- Department of Electrical & Electronics Engineering, Universiti Teknologi PETRONAS, Perak 32610, Malaysia; (M.Y.C.); (I.H.K.B.); (N.H.H.)
| | - Nor Hisham Hamid
- Department of Electrical & Electronics Engineering, Universiti Teknologi PETRONAS, Perak 32610, Malaysia; (M.Y.C.); (I.H.K.B.); (N.H.H.)
| | - Sok Ching Cheong
- Head and Neck Cancer Research Group, Cancer Research Malaysia, Selangor 47500, Malaysia; (K.P.L.); (S.C.C.)
- Department of Oral & Maxillofacial Clinical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Eric Tatt Wei Ho
- Department of Electrical & Electronics Engineering, Universiti Teknologi PETRONAS, Perak 32610, Malaysia; (M.Y.C.); (I.H.K.B.); (N.H.H.)
- Correspondence: ; Tel.: +60-5-368-7899
| |
Collapse
|
7
|
Sasani N, Roghanian R, Emtiazi G, Jalali SM, Nikougoftar Zarif M, Aghaie A. Design and introduction of a rational mechanical eluting system for leukocyte recovery from leukoreduction filters: A cell differential approach. Transfus Clin Biol 2020; 27:172-178. [PMID: 32340867 DOI: 10.1016/j.tracli.2020.03.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 11/25/2022]
Abstract
OBJECTIVES The use of leukoreduction filters has been highly increased in Iranian Blood Transfusion Centers within the last decade to provide sufficient leukoreduced blood products from healthy repeated donors for alloimmunized or sensitive recipients. Leucoflex LCR5, the dominant brand which procured by the Iranian Blood Transfusion Organization, is the most updated generation of the filters used around the world. MATERIAL AND METHODS In this study, we recovered trapped leukocytes from these filters using different buffer solutions and optimized elution method. The count of recovered cells assessed by cell counter, and cell viability was detected using trypan blue staining. The percent of leukocyte subpopulations was evaluated using a panel of monoclonal antibodies and flow cytometric analysis. RESULTS It illustrated that a buffer solution consistent with PBS in pH 7.2 containing 2mM EDTA and 4% (w/w) Dextran 40 was the best buffer for LCR5 filter backflushing. The white cell counted as 4.56×108 Granulocytes, 3.34×108 Lymphocytes, and 0.64×108 Monocytes according to analysis with auto hemoanalysis and flow cytometric methods. CONCLUSION The study guides and assists blood management systems in arranging a national blood profile database for future cell therapy strategies. Also, the recovered cells could be of significance in stem cell research, cellular interaction studies as well as novel molecular developments in drug discovery.
Collapse
Affiliation(s)
- N Sasani
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, 81746-79441 Isfahan, Iran
| | - R Roghanian
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, 81746-79441 Isfahan, Iran
| | - G Emtiazi
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, 81746-79441 Isfahan, Iran
| | - S M Jalali
- IBRF-1 plasma collection center, Iranian Blood Research and Fractionation Company, Fardis, Alborz, Iran
| | - M Nikougoftar Zarif
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, 14665-1157 Tehran, Iran
| | - A Aghaie
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, 14665-1157 Tehran, Iran.
| |
Collapse
|
8
|
Models for Monocytic Cells in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32036607 DOI: 10.1007/978-3-030-35723-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Monocytes (Mos) are immune cells that critically regulate cancer, enabling tumor growth and modulating metastasis. Mos can give rise to tumor-associated macrophages (TAMs) and Mo-derived dendritic cells (moDCs), all of which shape the tumor microenvironment (TME). Thus, understanding their roles in the TME is key for improved immunotherapy. Concurrently, various biological and mechanical factors including changes in local cytokines, extracellular matrix production, and metabolic changes in the TME affect the roles of monocytic cells. As such, relevant TME models are critical to achieve meaningful insight on the precise functions, mechanisms, and effects of monocytic cells. Notably, murine models have yielded significant insight into human Mo biology. However, many of these results have yet to be confirmed in humans, reinforcing the need for improved in vitro human TME models for the development of cancer interventions. Thus, this chapter (1) summarizes current insight on the tumor biology of Mos, TAMs, and moDCs, (2) highlights key therapeutic applications relevant to these cells, and (3) discusses various TME models to study their TME-related activity. We conclude with a perspective on the future research trajectory of this topic.
Collapse
|
9
|
Cid J, Carbassé G, Alba C, Perea D, Lozano M. Leukocytapheresis in nonmobilized donors for cellular therapy protocols: Evaluation of factors affecting collection efficiency of cells. J Clin Apher 2019; 34:672-679. [DOI: 10.1002/jca.21745] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/05/2019] [Accepted: 08/12/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Joan Cid
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and HemostasisICMHO, Hospital Clínic, University of Barcelona Barcelona Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
| | - Gloria Carbassé
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and HemostasisICMHO, Hospital Clínic, University of Barcelona Barcelona Spain
| | - Cristina Alba
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and HemostasisICMHO, Hospital Clínic, University of Barcelona Barcelona Spain
| | - Dolores Perea
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and HemostasisICMHO, Hospital Clínic, University of Barcelona Barcelona Spain
| | - Miquel Lozano
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and HemostasisICMHO, Hospital Clínic, University of Barcelona Barcelona Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
| |
Collapse
|
10
|
Green DS, Nunes AT, Tosh KW, David-Ocampo V, Fellowes VS, Ren J, Jin J, Frodigh SE, Pham C, Procter J, Tran C, Ekwede I, Khuu H, Stroncek DF, Highfill SL, Zoon KC, Annunziata CM. Production of a cellular product consisting of monocytes stimulated with Sylatron ® (Peginterferon alfa-2b) and Actimmune ® (Interferon gamma-1b) for human use. J Transl Med 2019; 17:82. [PMID: 30871636 PMCID: PMC6419352 DOI: 10.1186/s12967-019-1822-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/22/2019] [Indexed: 12/21/2022] Open
Abstract
Background Monocytes are myeloid cells that reside in the blood and bone marrow and respond to inflammation. At the site of inflammation, monocytes express cytokines and chemokines. Monocytes have been shown to be cytotoxic to tumor cells in the presence of pro-inflammatory cytokines such as Interferon Alpha, Interferon Gamma, and IL-6. We have previously shown that monocytes stimulated with both interferons (IFNs) results in synergistic killing of ovarian cancer cells. We translated these observations to an ongoing clinical trial using adoptive cell transfer of autologous monocytes stimulated ex vivo with IFNs and infused into the peritoneal cavity of patients with advanced, chemotherapy resistant, ovarian cancer. Here we describe the optimization of the monocyte elutriation protocol and a cryopreservation protocol of the monocytes isolated from peripheral blood. Methods Counter flow elutriation was performed on healthy donors or women with ovarian cancer. The monocyte-containing, RO-fraction was assessed for total monocyte number, purity, viability, and cytotoxicity with and without a cryopreservation step. All five fractions obtained from the elutriation procedure were also assessed by flow cytometry to measure the percent of immune cell subsets in each fraction. Results Both iterative monocyte isolation using counter flow elutriation or cryopreservation following counter flow elutriation can yield over 2 billion monocytes for each donor with high purity. We also show that the monocytes are stable, viable, and retain cytotoxic functions when cultured with IFNs. Conclusion Large scale isolation of monocytes from both healthy donors and patients with advanced, chemotherapy resistant ovarian cancer, can be achieved with high total number of monocytes. These monocytes can be cryopreserved and maintain viability and cytotoxic function. All of the elutriated cell fractions contain ample immune cells which could be used for other cell therapy-based applications. Electronic supplementary material The online version of this article (10.1186/s12967-019-1822-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniel S Green
- Women's Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive RM 3B43C, Bethesda, MD, 20892, USA
| | - Ana T Nunes
- Women's Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive RM 3B43C, Bethesda, MD, 20892, USA
| | - Kevin W Tosh
- Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Virginia David-Ocampo
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA.,Office of Tissues and Advanced Therapies, Center for Biologics and Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Vicki S Fellowes
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Jiaqiang Ren
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Jianjian Jin
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Sue-Ellen Frodigh
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Chauha Pham
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Jolynn Procter
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Celina Tran
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Irene Ekwede
- Women's Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive RM 3B43C, Bethesda, MD, 20892, USA
| | - Hanh Khuu
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA.,Office of Tissues and Advanced Therapies, Center for Biologics and Evaluation and Research, FDA, Silver Spring, MD, USA
| | - David F Stroncek
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Steven L Highfill
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Kathryn C Zoon
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christina M Annunziata
- Women's Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive RM 3B43C, Bethesda, MD, 20892, USA.
| |
Collapse
|
11
|
Roddie C, O'Reilly M, Dias Alves Pinto J, Vispute K, Lowdell M. Manufacturing chimeric antigen receptor T cells: issues and challenges. Cytotherapy 2019; 21:327-340. [PMID: 30685216 DOI: 10.1016/j.jcyt.2018.11.009] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/25/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Clinical trials of adoptively transferred CD19 chimeric antigen receptor (CAR) T cells have delivered unprecedented responses in patients with relapsed refractory B-cell malignancy. These results have prompted Food and Drug Administration (FDA) approval of two CAR T-cell products in this high-risk patient population. The widening range of indications for CAR T-cell therapy and increasing patient numbers present a significant logistical challenge to manufacturers aiming for reproducible delivery systems for high-quality clinical CAR T-cell products. This review discusses current and novel CAR T-cell processing methodologies and the quality control systems needed to meet the increasing clinical demand for these exciting new therapies.
Collapse
Affiliation(s)
- Claire Roddie
- Research Department of Haematology, University College London, London, UK; Department of Haematology, University College London Hospitals National Health Service (NHS) Foundation Trust, London.
| | - Maeve O'Reilly
- Research Department of Haematology, University College London, London, UK; Department of Haematology, University College London Hospitals National Health Service (NHS) Foundation Trust, London
| | | | - Ketki Vispute
- Research Department of Haematology, University College London, London, UK
| | - Mark Lowdell
- Research Department of Haematology, University College London, London, UK
| |
Collapse
|
12
|
CECHIM GIOVANA, CHIES JOSÉA. In vitro generation of human monocyte-derived dendritic cells methodological aspects in a comprehensive review. ACTA ACUST UNITED AC 2019. [DOI: 10.1590/0001-3765201920190310] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
13
|
da Silva LT, Santillo BT, de Almeida A, Duarte AJDS, Oshiro TM. Using Dendritic Cell-Based Immunotherapy to Treat HIV: How Can This Strategy be Improved? Front Immunol 2018; 9:2993. [PMID: 30619346 PMCID: PMC6305438 DOI: 10.3389/fimmu.2018.02993] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/04/2018] [Indexed: 11/13/2022] Open
Abstract
Harnessing dendritic cells (DC) to treat HIV infection is considered a key strategy to improve anti-HIV treatment and promote the discovery of functional or sterilizing cures. Although this strategy represents a promising approach, the results of currently published trials suggest that opportunities to optimize its performance still exist. In addition to the genetic and clinical characteristics of patients, the efficacy of DC-based immunotherapy depends on the quality of the vaccine product, which is composed of precursor-derived DC and an antigen for pulsing. Here, we focus on some factors that can interfere with vaccine production and should thus be considered to improve DC-based immunotherapy for HIV infection.
Collapse
Affiliation(s)
- Laís Teodoro da Silva
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Bruna Tereso Santillo
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alexandre de Almeida
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alberto Jose da Silva Duarte
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Telma Miyuki Oshiro
- Laboratorio de Investigacao em Dermatologia e Imunodeficiencias, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
14
|
Pfeiffer H, Völkl S, Gary R, Mackensen A, Achenbach S, Strasser E, Aigner M. Impact of collection programs for the generation of monocyte apheresis products on product quality and composition as starting material for the generation of cellular therapeutics. Transfusion 2018; 58:2175-2183. [DOI: 10.1111/trf.14817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Hella Pfeiffer
- Department of Transfusion Medicine and Haemostaseology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| | - Simon Völkl
- Department of Medicine 5, Haematology and Oncology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| | - Regina Gary
- Department of Medicine 5, Haematology and Oncology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| | - Andreas Mackensen
- Department of Medicine 5, Haematology and Oncology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| | - Susanne Achenbach
- Department of Transfusion Medicine and Haemostaseology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| | - Erwin Strasser
- Department of Transfusion Medicine and Haemostaseology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| | - Michael Aigner
- Department of Medicine 5, Haematology and Oncology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| |
Collapse
|
15
|
Anyanwu A, Sitzmann N, Hetjens S, Klüter H, Wuchter P. Low-Volume Leukapheresis in Non-Cytokine-Stimulated Donors for the Collection of Mononuclear Cells. Transfus Med Hemother 2018; 45:323-330. [PMID: 30498410 DOI: 10.1159/000490859] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/11/2018] [Indexed: 11/19/2022] Open
Abstract
Background There is an increasing demand for products containing mononuclear cells (MNCs) for cellular immune therapy. Hence, leukapheresis is increasingly performed in healthy volunteer donors. Methods We evaluated 147 low-volume leukapheresis procedures from 77 healthy non-cytokine-stimulated donors. Complete blood counts (CBCs) of the donors were measured before and directly after the procedures as well as from the MNC products. Follow-up CBCs were collected from donors within 21 days. Results The product hematocrit within a range from 1.2 to 6.0% did not correlate with the collection efficiency of any cell population or the granulocyte and platelet yield. There was a strong correlation between the CBC values before leukapheresis and the cell yield of lymphocytes and monocytes as well as a perfect negative correlation between cell recruitment and cell loss in all cell populations. Furthermore, we observed a significant decrease in the CBC values in all cell populations directly after leukapheresis, which recovered within a mean of 16.1 days (SD ± 2.1 days) and even showed a significant increase in granulocytes and platelets. Conclusion Low-volume leukapheresis is feasible for the collection of MNCs in which the product hematocrit is negligible for the collection efficiency, cell yield, or contamination of residual cells under operational settings recommended by the manufacturer. Our data suggests that cell recruitment is regulated by the number of cells removed, which may also be the stimulus to induce granulo- and thrombopoiesis within the first days after leukapheresis.
Collapse
Affiliation(s)
- Adamma Anyanwu
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | - Nicole Sitzmann
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | - Svetlana Hetjens
- Institute of Medical Statistics, Biomathematics and Informatics, Heidelberg University, Medical Faculty Mannheim, Mannheim, Germany
| | - Harald Klüter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| |
Collapse
|
16
|
Marques GS, Silva Z, Videira PA. Antitumor Efficacy of Human Monocyte-Derived Dendritic Cells: Comparing Effects of two Monocyte Isolation Methods. Biol Proced Online 2018; 20:4. [PMID: 29434528 PMCID: PMC5796591 DOI: 10.1186/s12575-018-0069-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/17/2018] [Indexed: 02/08/2023] Open
Abstract
Background Dendritic cells (DCs), which can be used as anti-cancer vaccines, are generally obtained in vitro from isolated CD14+ monocytes (MoDCs). This generates high cell numbers and allows instructing DCs to guarantee effective antitumor responses. However, the impact of the monocyte isolation step in the antitumor effectiveness of the generated MoDCs is still unknown. Here, we compared the most used immunomagnetic technologies for monocyte isolation: magnetic activated cell sorting (MACS) from Miltenyi Biotec and EasySep from STEM CELL. Results MACS technology allowed a higher monocyte yield and purity and, by flow cytometry, monocytes displayed higher size and lower granularity. In the resting state, EasySep_MoDCs showed a higher basal expression of HLA-DR, and no significant response to stimulation by LPS and TNF-α. When stimulated with whole tumor cells lysates, both MoDCs expressed similar levels of maturation and co-stimulatory markers. However, when cultured with autologous T cells, MACS_MoDCs induced significantly higher IFN-γ secretion than EasySep_MoDCs, indicating a stronger induction of Th1 cell response profile. Concordantly, T cells induced by MACS_MoDCs also showed a higher release of cytotoxic granules when in contact with tumor cells. Conclusions Overall, both the MACS and the EasySep isolation immunomagnetic technologies provide monocytes that differentiate into viable and functional MoDCs. In our experimental settings, resting EasySep_MoDCs showed a higher basal level of maturation but show less responsivity to stimuli. On the other hand, MACS_MoDCs, when stimulated with tumor antigens, showed better ability to stimulate Th1 responses and to induce T cell cytotoxicity against tumor cells. Thus, monocyte isolation techniques crucially affect MoDCs’ function and, therefore, should be carefully selected to obtain the desired functionality.
Collapse
Affiliation(s)
- Graça S Marques
- 1CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Zélia Silva
- 1CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal.,2UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Paula A Videira
- 1CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal.,2UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Lisbon, Portugal.,3CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
17
|
Mfarrej B, Tresoldi E, Stabilini A, Paganelli A, Caldara R, Secchi A, Battaglia M. Generation of donor-specific Tr1 cells to be used after kidney transplantation and definition of the timing of their in vivo infusion in the presence of immunosuppression. J Transl Med 2017; 15:40. [PMID: 28222739 PMCID: PMC5319067 DOI: 10.1186/s12967-017-1133-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/03/2017] [Indexed: 02/20/2023] Open
Abstract
Background Operational tolerance is an alternative to lifelong immunosuppression after transplantation. One strategy to achieve tolerance is by T regulatory cells. Safety and feasibility of a T regulatory type 1 (Tr1)-cell—based therapy to prevent graft versus host disease in patients with hematological malignancies has been already proven. We are now planning to perform a Tr1-cell—based therapy after kidney transplantation. Methods Upon tailoring the lab-grade protocol to patients on dialysis, aims of the current work were to develop a clinical-grade compatible protocol to generate a donor-specific Tr1-cell—enriched medicinal product (named T10 cells) and to test the Tr1-cell sensitivity to standard immunosuppression in vivo to define the best timing of cell infusion. Results We developed a medicinal product that was enriched in Tr1 cells, anergic to donor-cell stimulation, able to suppress proliferation upon donor- but not third-party stimulation in vitro, and stable upon cryopreservation. The protocol was reproducible upon up scaling to leukapheresis from patients on dialysis and was effective in yielding the expected number of T10 cells necessary for the planned infusions. The tolerogenic gene signature of circulating Tr1 cells was minimally compromised in kidney transplant recipients under standard immunosuppression and it eventually started to recover 36 weeks post-transplantation, providing rationale for selecting the timings of the cell infusions. Conclusions These data provide solid ground for proceeding with the trial and establish robust rationale for defining the correct timing of cell infusion during concomitant immunosuppressive treatment. Electronic supplementary material The online version of this article (doi:10.1186/s12967-017-1133-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bechara Mfarrej
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy
| | - Eleonora Tresoldi
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy.,Human T Cell Laboratory, Saint Vincent's Institute of Medical Research, Melbourne, Australia
| | - Angela Stabilini
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy
| | - Alessia Paganelli
- Department of Internal Medicine, Transplantation Medicine, San Raffaele Hospital, Milan, Italy
| | - Rossana Caldara
- Department of Internal Medicine, Transplantation Medicine, San Raffaele Hospital, Milan, Italy
| | - Antonio Secchi
- Department of Internal Medicine, Transplantation Medicine, San Raffaele Hospital, Milan, Italy
| | - Manuela Battaglia
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy.
| |
Collapse
|
18
|
Scheid E, Major P, Bergeron A, Finn OJ, Salter RD, Eady R, Yassine-Diab B, Favre D, Peretz Y, Landry C, Hotte S, Mukherjee SD, Dekaban GA, Fink C, Foster PJ, Gaudet J, Gariepy J, Sekaly RP, Lacombe L, Fradet Y, Foley R. Tn-MUC1 DC Vaccination of Rhesus Macaques and a Phase I/II Trial in Patients with Nonmetastatic Castrate-Resistant Prostate Cancer. Cancer Immunol Res 2016; 4:881-892. [PMID: 27604597 DOI: 10.1158/2326-6066.cir-15-0189] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 08/08/2016] [Indexed: 11/16/2022]
Abstract
MUC1 is a glycoprotein expressed on the apical surface of ductal epithelial cells. Malignant transformation results in loss of polarization and overexpression of hypoglycosylated MUC1 carrying truncated carbohydrates known as T or Tn tumor antigens. Tumor MUC1 bearing Tn carbohydrates (Tn-MUC1) represent a potential target for immunotherapy. We evaluated the Tn-MUC1 glycopeptide in a human phase I/II clinical trial for safety that followed a preclinical study of different glycosylation forms of MUC1 in rhesus macaques, whose MUC1 is highly homologous to human MUC1. Either unglycosylated rhesus macaque MUC1 peptide (rmMUC1) or Tn-rmMUC1 glycopeptide was mixed with an adjuvant or loaded on autologous dendritic cells (DC), and responses were compared. Unglycosylated rmMUC1 peptide induced negligible humoral or cellular responses compared with the Tn-rmMUC1 glycopeptide. Tn-rmMUC1 loaded on DCs induced the highest anti-rmMUC1 T-cell responses and no clinical toxicity. In the phase I/II clinical study, 17 patients with nonmetastatic castrate-resistant prostate cancer (nmCRPC) were tested with a Tn-MUC1 glycopeptide-DC vaccine. Patients were treated with multiple intradermal and intranodal doses of autologous DCs, which were loaded with the Tn-MUC1 glycopeptide (and KLH as a positive control for immune reactivity). PSA doubling time (PSADT) improved significantly in 11 of 16 evaluable patients (P = 0.037). Immune response analyses detected significant Tn-MUC1-specific CD4+ and/or CD8+ T-cell intracellular cytokine responses in 5 out of 7 patients evaluated. In conclusion, vaccination with Tn-MUC1-loaded DCs in nmCRPC patients appears to be safe, able to induce significant T-cell responses, and have biological activity as measured by the increase in PSADT following vaccination. Cancer Immunol Res; 4(10); 881-92. ©2016 AACR.
Collapse
Affiliation(s)
| | - Pierre Major
- McMaster University, Hamilton, Ontario, Canada. Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Alain Bergeron
- Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada. Centre de Recherche sur le Cancer de l'Université Laval, Québec, Canada
| | - Olivera J Finn
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Russell D Salter
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Robin Eady
- Hamilton Health Sciences, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | - Corby Fink
- Robarts Research Institute, London, Ontario, Canada
| | | | | | - Jean Gariepy
- Sunnybrook Research Institute, Toronto, Ontario, Canada
| | | | - Louis Lacombe
- Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada. Centre de Recherche sur le Cancer de l'Université Laval, Québec, Canada
| | - Yves Fradet
- Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada. Centre de Recherche sur le Cancer de l'Université Laval, Québec, Canada
| | - Ronan Foley
- McMaster University, Hamilton, Ontario, Canada. Hamilton Health Sciences, Hamilton, Ontario, Canada.
| |
Collapse
|
19
|
Jin X, Kruth HS. Culture of Macrophage Colony-stimulating Factor Differentiated Human Monocyte-derived Macrophages. J Vis Exp 2016. [PMID: 27404952 DOI: 10.3791/54244] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A protocol is presented for cell culture of macrophage colony-stimulating factor (M-CSF) differentiated human monocyte-derived macrophages. For initiation of experiments, fresh or frozen monocytes are cultured in flasks for 1 week with M-CSF to induce their differentiation into macrophages. Then, the macrophages can be harvested and seeded into culture wells at required cell densities for carrying out experiments. The use of defined numbers of macrophages rather than defined numbers of monocytes to initiate macrophage cultures for experiments yields macrophage cultures in which the desired cell density can be more consistently attained. Use of cryopreserved monocytes reduces dependency on donor availability and produces more homogeneous macrophage cultures.
Collapse
Affiliation(s)
- Xueting Jin
- Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute, National Institutes of Health
| | - Howard S Kruth
- Experimental Atherosclerosis Section, National Heart, Lung, and Blood Institute, National Institutes of Health;
| |
Collapse
|
20
|
Fesnak A, Lin C, Siegel DL, Maus MV. CAR-T Cell Therapies From the Transfusion Medicine Perspective. Transfus Med Rev 2016; 30:139-45. [PMID: 27067907 DOI: 10.1016/j.tmrv.2016.03.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/21/2016] [Indexed: 02/06/2023]
Abstract
The use of chimeric antigen receptor (CAR)-T cell therapy for the treatment of hematologic malignancies has generated significant excitement over the last several years. From a transfusion medicine perspective, the implementation of CAR-T therapy as a potential mainstay treatment for not only hematologic but also solid-organ malignancies represents a significant opportunity for growth and expansion. In this review, we will describe the rationale for the development of genetically redirected T cells as a cancer therapeutic, the different elements that are required to engineer these cells, as well as an overview of the process by which patient cells are harvested and processed to create and subsequently validate CAR-T cells. Finally, we will briefly describe some of the toxicities and clinical efficacy of CAR-T cells in the setting of patients with advanced malignancy.
Collapse
Affiliation(s)
- Andrew Fesnak
- Division of Transfusion Medicine & Therapeutic Pathology, Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - ChieYu Lin
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Don L Siegel
- Division of Transfusion Medicine & Therapeutic Pathology, Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Marcela V Maus
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA.
| |
Collapse
|
21
|
Monocyte Activation in Immunopathology: Cellular Test for Development of Diagnostics and Therapy. J Immunol Res 2016; 2016:4789279. [PMID: 26885534 PMCID: PMC4739459 DOI: 10.1155/2016/4789279] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/20/2015] [Accepted: 12/21/2015] [Indexed: 12/14/2022] Open
Abstract
Several highly prevalent human diseases are associated with immunopathology. Alterations in the immune system are found in such life-threatening disorders as cancer and atherosclerosis. Monocyte activation followed by macrophage polarization is an important step in normal immune response to pathogens and other relevant stimuli. Depending on the nature of the activation signal, macrophages can acquire pro- or anti-inflammatory phenotypes that are characterized by the expression of distinct patterns of secreted cytokines and surface antigens. This process is disturbed in immunopathologies resulting in abnormal monocyte activation and/or bias of macrophage polarization towards one or the other phenotype. Such alterations could be used as important diagnostic markers and also as possible targets for the development of immunomodulating therapy. Recently developed cellular tests are designed to analyze the phenotype and activity of living cells circulating in patient's bloodstream. Monocyte/macrophage activation test is a successful example of cellular test relevant for atherosclerosis and oncopathology. This test demonstrated changes in macrophage activation in subclinical atherosclerosis and breast cancer and could also be used for screening a panel of natural agents with immunomodulatory activity. Further development of cellular tests will allow broadening the scope of their clinical implication. Such tests may become useful tools for drug research and therapy optimization.
Collapse
|
22
|
Gelao L, Criscitiello C, Esposito A, De Laurentiis M, Fumagalli L, Locatelli MA, Minchella I, Santangelo M, De Placido S, Goldhirsch A, Curigliano G. Dendritic cell-based vaccines: clinical applications in breast cancer. Immunotherapy 2015; 6:349-60. [PMID: 24762078 DOI: 10.2217/imt.13.169] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Recent evidence suggests that the immune system is involved in the carcinogenesis process and the antitumor immune responses impact the clinical outcome, thus emphasizing the concept of cancer immune surveillance. In this context, dendritic cells (DCs) seem to play a crucial role, as they are the most potent antigen-presenting cells (APCs) and are able to stimulate naive T lymphocytes and to generate memory T lymphocytes. Immunotherapy with DC-based vaccines is a very attractive approach to treat cancer, offering the potential for high tumor-specific cytotoxicity. Although breast cancer (BC) is traditionally considered a poorly immunogenic tumor, increasing numbers of both preclinical and clinical studies demonstrate that vaccination with DCs is capable of inducing an antitumor-specific response, while being well tolerated and safe. However, clinical objective responses are still disappointing and many reasons may explain the difficulty of developing effective DC-based therapies for BC. In this review, we discuss the characteristics of DCs, and the major clinical indications for DC-based immunotherapy in BC with related drawbacks.
Collapse
Affiliation(s)
- Lucia Gelao
- Division of Early Drug Development for Innovative Therapies, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Orekhov AN, Sobenin IA, Gavrilin MA, Gratchev A, Kotyashova SY, Nikiforov NG, Kzhyshkowska J. Macrophages in immunopathology of atherosclerosis: a target for diagnostics and therapy. Curr Pharm Des 2015; 21:1172-9. [PMID: 25312739 PMCID: PMC4428062 DOI: 10.2174/1381612820666141013120459] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 10/01/2014] [Indexed: 01/22/2023]
Abstract
Immunopathology plays important roles in the development of different life-threatening diseases, such as atherosclerosis and its consequences (acute myocardial infarction and stroke), cancer, chronic inflammatory diseases. Effective modulation of the immune system may significantly increase the efficacy of prevention and therapy efforts. Currently there are no marketed drugs capable of normalizing immune system function in an intrinsic and comprehensive way. Here, we describe a test system designed for complex analysis of monocyte activity in individuals to diagnose immunopathology and monitor treatment efficacy. This cell-based test system may also be useful for screening compounds with an immune-correcting effects. Both diagnostic and screening systems are based on primary culture of human monocytes and/or monocyte-derived macrophages. This is the first step in creating a method for assessment of macrophage activity, which is required for further development of immune-correcting drugs. The existing preliminary data provide the basis for realization of this idea.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Julia Kzhyshkowska
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiyskaya Street, 125315 Moscow, Russia.
| |
Collapse
|
24
|
Friis T, Pedersen KB, Hougaard D, Houen G. Immunocytochemical and Immunohistochemical Staining with Peptide Antibodies. Methods Mol Biol 2015; 1348:311-325. [PMID: 26424283 DOI: 10.1007/978-1-4939-2999-3_27] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Peptide antibodies are particularly useful for immunocytochemistry (ICC) and immunohistochemistry (IHC), where antigens may denature due to fixation of tissues and cells. Peptide antibodies can be made to any defined sequence, including unknown putative proteins and posttranslationally modified sequences. Moreover, the availability of large amounts of the antigen (peptide) allows inhibition/adsorption controls, which are important in ICC/IHC, due to the many possibilities for false-positive reactions caused by immunoglobulin Fc receptors, nonspecific reactions, and cross-reactivity of primary and secondary antibodies with other antigens and endogenous immunoglobulins, respectively. Here, simple protocols for ICC and IHC are described together with recommendations for appropriate controls.
Collapse
Affiliation(s)
- Tina Friis
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Klaus Boberg Pedersen
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - David Hougaard
- Department of Congenital Diseases, Statens Serum Institut, Copenhagen, Denmark
| | - Gunnar Houen
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark.
| |
Collapse
|
25
|
Brauns T, Leblanc P, Gelfand JA, Poznanski M. Could mycobacterial Hsp70-containing fusion protein lead the way to an affordable therapeutic cancer vaccine? Expert Rev Vaccines 2014; 14:435-46. [PMID: 25496347 DOI: 10.1586/14760584.2015.979797] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cancer vaccine development efforts have recently gained momentum, but most vaccines showing clinical impact in human trials tend to be based on technology approaches that are very costly and difficult to produce at scale. With the projected doubling of the incidence of cancer and its related cost of care in the U.S. over the next two decades, the widespread clinical use of such vaccines will prove difficult to justify. Heat shock protein-based vaccines have shown the potential to elicit clinically meaningful immunologic responses in cancer, but the predominant development approach - heat shock protein-peptide complexes derived from a patient's own tumor - face similar challenges of cost and scalability. New innovative modalities for deploying heat shock proteins in cancer vaccines may open the door to vaccines that can generate potent cytotoxic responses against multiple tumor targets and can be made in a cost-effective and scalable manner.
Collapse
Affiliation(s)
- Timothy Brauns
- Vaccine and Immunotherapy Center, Massachusetts General Hospital - Medicine/Infectious Diseases, 149 13th Street, Mailstop 149-5-5246, Charlestown, MA 02129, USA
| | | | | | | |
Collapse
|
26
|
Mackern-Oberti JP, Vega F, Llanos C, Bueno SM, Kalergis AM. Targeting dendritic cell function during systemic autoimmunity to restore tolerance. Int J Mol Sci 2014; 15:16381-417. [PMID: 25229821 PMCID: PMC4200801 DOI: 10.3390/ijms150916381] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/29/2014] [Accepted: 09/05/2014] [Indexed: 12/11/2022] Open
Abstract
Systemic autoimmune diseases can damage nearly every tissue or cell type of the body. Although a great deal of progress has been made in understanding the pathogenesis of autoimmune diseases, current therapies have not been improved, remain unspecific and are associated with significant side effects. Because dendritic cells (DCs) play a major role in promoting immune tolerance against self-antigens (self-Ags), current efforts are focusing at generating new therapies based on the transfer of tolerogenic DCs (tolDCs) during autoimmunity. However, the feasibility of this approach during systemic autoimmunity has yet to be evaluated. TolDCs may ameliorate autoimmunity mainly by restoring T cell tolerance and, thus, indirectly modulating autoantibody development. In vitro induction of tolDCs loaded with immunodominant self-Ags and subsequent cell transfer to patients would be a specific new therapy that will avoid systemic immunosuppression. Herein, we review recent approaches evaluating the potential of tolDCs for the treatment of systemic autoimmune disorders.
Collapse
Affiliation(s)
- Juan P Mackern-Oberti
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Portugal 49, Santiago 8330025, Chile.
| | - Fabián Vega
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 350, Santiago 8330033, Chile.
| | - Carolina Llanos
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 350, Santiago 8330033, Chile.
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Portugal 49, Santiago 8330025, Chile.
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Portugal 49, Santiago 8330025, Chile.
| |
Collapse
|
27
|
The Functional Difference of Dendritic Cells in HBeAg Negative Chronic Hepatitis B Patients with Three Different Spleen Deficiency Syndromes and the Therapeutic Evaluation of Chinese Medicine Intervention Based on Syndrome Differentiation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:802402. [PMID: 25093028 PMCID: PMC4100377 DOI: 10.1155/2014/802402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/08/2014] [Accepted: 05/28/2014] [Indexed: 01/17/2023]
Abstract
Objective. To investigate the dendritic cells (DCs) maturity differences of HBeAg negative chronic hepatitis B (CHB) patients with different spleen deficiency (SD) syndromes and explore the role of syndrome differentiation in the therapeutic evaluation of Chinese medicine. Methods. 120 participants were recruited including three treatment groups in different SD syndrome categories as spleen deficiency with liver depression (SDLD), spleen deficiency with damp heat (SDDH), and spleen deficiency with kidney deficiency (SDKD) and one healthy control group; each group had 30 participants. Corresponding drugs were applied. The outcome measures included DC phenotype, liver function, IL-10, IL-12, and HBV-DNA levels. Results. The surface markers of mature DCs and cytokines levels were different in each group; the positive rate of CD80, CD1a, HLA-DR, and CD1a was the lowest in SDKD group. After 3-month intervention, the expression of CD80, CD86, CD1a, HLA-DR, and IL-12 significantly increased, while ALT, AST, and IL-10 significantly decreased (P < 0.05) in treatment groups. HBV-DNA level also significantly reduced in both SDKD and SDLD groups (P < 0.05). Conclusions. HBeAg negative patients had DCs dysmaturity, and there were differences between different SD syndromes. Chinese medicine intervention according to syndrome differentiation could advance the maturity and function of DCs and improve the therapeutic effect.
Collapse
|
28
|
Nucleic acid recognition in dendritic cells. Methods Mol Biol 2014; 1169:55-65. [PMID: 24957229 DOI: 10.1007/978-1-4939-0882-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
The immune system consists of specialized cell types with distinct functions in order to provide an effective innate and adaptive immune defense against harmful invading pathogens like bacteria, viruses, fungi, parasites, or other substances threatening the integrity of the organism. Once the immune system recognizes such pathogens via pattern recognition receptors (PRRs), they are taken up, processed, and presented as antigens on MHC class I and II to T lymphocytes by specialized cells called dendritic cells (DCs). At the same time pathogen components which bind to PRRs in DCs trigger potent cytokine and chemokine responses. Although other cell types like macrophages can also take up, process, and present antigens to naïve T lymphocytes, DCs are the cells with the greatest capacity to do so. Thus, DCs are also called professional antigen presenting cells (APCs), which induce a strong adaptive immune response and thereby act as a bridge between the innate and adaptive immune system. This chapter provides detailed instructions on how to generate various types of DCs from human peripheral blood mononuclear cells (PBMCs) and murine bone marrow, as well as stimulation conditions for activation of these cells by PRR ligands in vitro.
Collapse
|
29
|
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
|
30
|
Strasser EF, Weidinger T, Weiss DR, Strobel J, Zimmermann R, Eckstein R. Storage induced apoptosis of peripheral blood mononuclear cells obtained from leucoreduction system chambers. Vox Sang 2011; 101:106-11. [PMID: 21492183 DOI: 10.1111/j.1423-0410.2011.01473.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVES Recently, it was reported that leucocytes obtained from leucoreduction system chambers (LRSCs) after plateletapheresis show excellent quality due to culture of dendritic cells. This study analysed apoptosis of mononuclear cells derived from LRSCs of single platelet units (SPUs) and double platelet units (DPUs) during storage. MATERIALS AND METHODS This randomized prospective study compared eighteen single and double platelet units produced with the Trima Accel cell separator. Buffy coat was drained from the LRSCs and analysed after 1, 6, 24, 48 and 72 h. CD45+ lymphocytes and CD14+ monocytes cells as well as Annexin-V+ and 7-AAD+ mononuclear cells were measured by flow cytometry. RESULTS The WBC concentration of LRSCs obtained from SPUs and DPUs differed significantly (SPUs: 0·93 ± 0·32 ×10(5) per μl WBCs; DPUs: 1·71 ± 0·55 ×10(5) per μl WBCs; P<0·001). Processed blood volume (PBV) correlated significantly with WBC concentration (r(2)=0·75, P<0·001). Fifty percent of monocytes were Annexin-V-positive 1 h after production decreasing to 30% during 24 h of storage. Compared to that, the part of late apoptotic or necrotic PBMCs increased later on, after 24 h. After 24 h, Annexin-V- and 7-AAD-positive, late apoptotic and necrotic lymphocytes and monocytes doubled. CONCLUSION PBMCs stored in autologous plasma in PVC-bags at room temperature did not show an increase of 7-AAD-positive PBMCs during 24 h prior to cell processing but increased significantly thereafter.
Collapse
Affiliation(s)
- E F Strasser
- Department of Transfusion Medicine and Haemostaseology, University Hospital Erlangen, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
31
|
|