1
|
Evaluation of Immune Infiltration Based on Image Plus Helps Predict the Prognosis of Stage III Gastric Cancer Patients with Significantly Different Outcomes in Northeastern China. DISEASE MARKERS 2022; 2022:2893336. [PMID: 35371344 PMCID: PMC8975697 DOI: 10.1155/2022/2893336] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/18/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022]
Abstract
Gastric cancer (GC) might have significantly different outcomes within the same AJCC/UICC-TNM stage. The purpose of this study is to help predict the different prognosis through the pattern of immune cell infiltration. We retrospectively analyzed 2605 patients who underwent radical gastrectomy in the Harbin Medical University Cancer Hospital between 2002 and 2013. For stage III with significantly different survival probability, we analyzed the relationship between immune cell surface antigen and survival in TCGA dataset. Furthermore, 200 cases in stage III GC with different survival outcomes were randomly selected for immunohistochemical verification. Image Plus software was used to evaluate the area of immune cell infiltration. We found that patients in stage III had significantly different outcomes. Bioinformatics analysis showed that there was a significant negative correlation between the expression of immune cell surface antigen and prognosis. In order to investigate whether immune infiltration can distinguish GC patients in stage III with differences in prognosis, we verified by immunohistochemistry that CD4+ T cells, CD20+ B cells, and CD177+ neutrophils infiltrated more in group B with better prognosis; CD8+ T cells, CD68+ macrophages, and CD117+ mast cells infiltrated more in group A with poor prognosis. CD117+ mast cells have the same trend of predicting significance for prognosis in the RNA and protein levels. In conclusion, patients with GC in northeastern China have significant prognostic differences only in stage III. CD117+ mast cells may be important evaluation factors in further studies of Immunoscore.
Collapse
|
2
|
Multi-objective optimization reveals time- and dose-dependent inflammatory cytokine-mediated regulation of human stem cell derived T-cell development. NPJ Regen Med 2022; 7:11. [PMID: 35087040 PMCID: PMC8795204 DOI: 10.1038/s41536-022-00210-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 12/22/2021] [Indexed: 12/29/2022] Open
Abstract
The generation of T-cells from stem cells in vitro could provide an alternative source of cells for immunotherapies. T-cell development from hematopoietic stem and progenitor cells (HSPCs) is tightly regulated through Notch pathway activation by Delta-like (DL) ligands 1 and 4. Other molecules, such as stem cell factor (SCF) and interleukin (IL)-7, play a supportive role in regulating the survival, differentiation, and proliferation of developing T-cells. Numerous other signaling molecules influence T-lineage development in vivo, but little work has been done to understand and optimize their use for T-cell production. Using a defined engineered thymic niche system, we undertook a multi-stage statistical learning-based optimization campaign and identified IL-3 and tumor necrosis factor α (TNFα) as a stage- and dose-specific enhancers of cell proliferation and T-lineage differentiation. We used this information to construct an efficient three-stage process for generating conventional TCRαβ+CD8+ T-cells expressing a diverse TCR repertoire from blood stem cells. Our work provides new insight into T-cell development and a robust system for generating T-cells to enable clinical therapies for treating cancer and immune disorders.
Collapse
|
3
|
An in vitro platform supports generation of human innate lymphoid cells from CD34 + hematopoietic progenitors that recapitulate ex vivo identity. Immunity 2021; 54:2417-2432.e5. [PMID: 34453879 DOI: 10.1016/j.immuni.2021.07.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/12/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022]
Abstract
Innate lymphoid cells (ILCs) are critical effectors of innate immunity and inflammation, whose development and activation pathways make for attractive therapeutic targets. However, human ILC generation has not been systematically explored, and previous in vitro investigations relied on the analysis of few markers or cytokines, which are suboptimal to assign lineage identity. Here, we developed a platform that reliably generated human ILC lineages from CD34+ hematopoietic progenitors derived from cord blood and bone marrow. We showed that one culture condition is insufficient to generate all ILC subsets, and instead, distinct combination of cytokines and Notch signaling are essential. The identity of natural killer (NK)/ILC1s, ILC2s, and ILC3s generated in vitro was validated by protein expression, functional assays, and both global and single-cell transcriptome analysis, recapitulating the signatures and functions of their ex vivo ILC counterparts. These data represent a resource to aid in clarifying ILC biology and differentiation.
Collapse
|
4
|
Silva CS, Reis RL, Martins A, Neves NM. Recapitulation of Thymic Function by Tissue Engineering Strategies. Adv Healthc Mater 2021; 10:e2100773. [PMID: 34197034 DOI: 10.1002/adhm.202100773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Indexed: 11/06/2022]
Abstract
The thymus is responsible for the development and selection of T lymphocytes, which in turn also participate in the maturation of thymic epithelial cells. These events occur through the close interactions between hematopoietic stem cells and developing thymocytes with the thymic stromal cells within an intricate 3D network. The complex thymic microenvironment and function, and the current therapies to induce thymic regeneration or to overcome the lack of a functional thymus are herein reviewed. The recapitulation of the thymic function using tissue engineering strategies has been explored as a way to control the body's tolerance to external grafts and to generate ex vivo T cells for transplantation. In this review, the main advances in the thymus tissue engineering field are disclosed, including both scaffold- and cell-based strategies. In light of the current gaps and limitations of the developed systems, the design of novel biomaterials for this purpose with unique features is also discussed.
Collapse
Affiliation(s)
- Catarina S. Silva
- 3B's Research Group I3Bs – Research Institute on Biomaterials Biodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine ICVS/3B's – PT Government Associate Laboratory AvePark, Parque da Ciência e Tecnologia, Zona Industrial da Gandra 4805‐017 Barco Guimarães Portugal
| | - Rui L. Reis
- 3B's Research Group I3Bs – Research Institute on Biomaterials Biodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine ICVS/3B's – PT Government Associate Laboratory AvePark, Parque da Ciência e Tecnologia, Zona Industrial da Gandra 4805‐017 Barco Guimarães Portugal
| | - Albino Martins
- 3B's Research Group I3Bs – Research Institute on Biomaterials Biodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine ICVS/3B's – PT Government Associate Laboratory AvePark, Parque da Ciência e Tecnologia, Zona Industrial da Gandra 4805‐017 Barco Guimarães Portugal
| | - Nuno M. Neves
- 3B's Research Group I3Bs – Research Institute on Biomaterials Biodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine ICVS/3B's – PT Government Associate Laboratory AvePark, Parque da Ciência e Tecnologia, Zona Industrial da Gandra 4805‐017 Barco Guimarães Portugal
| |
Collapse
|
5
|
DL4-μbeads induce T cell lineage differentiation from stem cells in a stromal cell-free system. Nat Commun 2021; 12:5023. [PMID: 34408144 PMCID: PMC8373879 DOI: 10.1038/s41467-021-25245-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/26/2021] [Indexed: 11/08/2022] Open
Abstract
T cells are pivotal effectors of the immune system and can be harnessed as therapeutics for regenerative medicine and cancer immunotherapy. An unmet challenge in the field is the development of a clinically relevant system that is readily scalable to generate large numbers of T-lineage cells from hematopoietic stem/progenitor cells (HSPCs). Here, we report a stromal cell-free, microbead-based approach that supports the efficient in vitro development of both human progenitor T (proT) cells and T-lineage cells from CD34+cells sourced from cord blood, GCSF-mobilized peripheral blood, and pluripotent stem cells (PSCs). DL4-μbeads, along with lymphopoietic cytokines, induce an ordered sequence of differentiation from CD34+ cells to CD34+CD7+CD5+ proT cells to CD3+αβ T cells. Single-cell RNA sequencing of human PSC-derived proT cells reveals a transcriptional profile similar to the earliest thymocytes found in the embryonic and fetal thymus. Furthermore, the adoptive transfer of CD34+CD7+ proT cells into immunodeficient mice demonstrates efficient thymic engraftment and functional maturation of peripheral T cells. DL4-μbeads provide a simple and robust platform to both study human T cell development and facilitate the development of engineered T cell therapies from renewable sources. T cells derived from stem cells can be harnessed for regenerative medicine and cancer immunotherapy, but current technologies limit production and translation. Here, the authors present a serum-free, stromal-cell free DLL4-coated microbead method for the scalable production of T-lineage cells from multiple sources of stem cells.
Collapse
|
6
|
Pievani A, Savoldelli R, Poelchen J, Mattioli E, Anselmi G, Girardot A, Utikal J, Bourdely P, Serafini M, Guermonprez P. Harnessing Mesenchymal Stromal Cells for the Engineering of Human Hematopoietic Niches. Front Immunol 2021; 12:631279. [PMID: 33790904 PMCID: PMC8006008 DOI: 10.3389/fimmu.2021.631279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/10/2021] [Indexed: 01/02/2023] Open
Abstract
Tissue engineering opens multiple opportunities in regenerative medicine, drug testing, and modeling of the hematopoiesis in health and disease. Recapitulating the organization of physiological microenvironments supporting leukocyte development is essential to model faithfully the development of immune cells. Hematopoietic organs are shaped by spatially organized niches defined by multiple cellular contributions. A shared feature of immune niches is the presence of mesenchymal stromal cells endowed with unique roles in organizing niche development, maintenance, and function. Here, we review challenges and opportunities in harnessing stromal cells for the engineering of artificial immune niches and hematopoietic organoids recapitulating leukocyte ontogeny both in vitro and in vivo.
Collapse
Affiliation(s)
- Alice Pievani
- Department of Pediatrics, M. Tettamanti Research Center, University of Milano-Bicocca, Monza, Italy
| | - Roberto Savoldelli
- The Peter Gorer Department of Immunobiology, Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom.,Cancer Research UK King's Health Partner Cancer Centre, King's College London, London, United Kingdom
| | - Juliane Poelchen
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Elisa Mattioli
- The Peter Gorer Department of Immunobiology, Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom.,Cancer Research UK King's Health Partner Cancer Centre, King's College London, London, United Kingdom
| | - Giorgio Anselmi
- MRC Molecular Hematology Unit, Radcliffe Department of Medicine, Medical Research Council, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Alice Girardot
- Centre for Inflammation Research, CNRS ERL8252, INSERM1149, Hopital Bichat, Université de Paris, Paris, France
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Pierre Bourdely
- Centre for Inflammation Research, CNRS ERL8252, INSERM1149, Hopital Bichat, Université de Paris, Paris, France
| | - Marta Serafini
- Department of Pediatrics, M. Tettamanti Research Center, University of Milano-Bicocca, Monza, Italy
| | - Pierre Guermonprez
- Centre for Inflammation Research, CNRS ERL8252, INSERM1149, Hopital Bichat, Université de Paris, Paris, France
| |
Collapse
|
7
|
A clinically applicable and scalable method to regenerate T-cells from iPSCs for off-the-shelf T-cell immunotherapy. Nat Commun 2021; 12:430. [PMID: 33462228 PMCID: PMC7814014 DOI: 10.1038/s41467-020-20658-3] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Clinical successes demonstrated by chimeric antigen receptor T-cell immunotherapy have facilitated further development of T-cell immunotherapy against wide variety of diseases. One approach is the development of “off-the-shelf” T-cell sources. Technologies to generate T-cells from pluripotent stem cells (PSCs) may offer platforms to produce “off-the-shelf” and synthetic allogeneic T-cells. However, low differentiation efficiency and poor scalability of current methods may compromise their utilities. Here we show improved differentiation efficiency of T-cells from induced PSCs (iPSCs) derived from an antigen-specific cytotoxic T-cell clone, or from T-cell receptor (TCR)-transduced iPSCs, as starting materials. We additionally describe feeder-free differentiation culture systems that span from iPSC maintenance to T-cell proliferation phases, enabling large-scale regenerated T-cell production. Moreover, simultaneous addition of SDF1α and a p38 inhibitor during T-cell differentiation enhances T-cell commitment. The regenerated T-cells show TCR-dependent functions in vitro and are capable of in vivo anti-tumor activity. This system provides a platform to generate a large number of regenerated T-cells for clinical application and investigate human T-cell differentiation and biology. T-cell immunotherapies, such as CAR-T immunotherapy, are being developed against a wide variety of diseases. Here the authors report the feeder-free, scalable differentiation of human induced pluripotent cells (iPSCs) to T-cells with T-cell receptor dependent anti-tumour function in vitro and in vivo.
Collapse
|
8
|
Singh J, Mohtashami M, Anderson G, Zúñiga-Pflücker JC. Thymic Engraftment by in vitro-Derived Progenitor T Cells in Young and Aged Mice. Front Immunol 2020; 11:1850. [PMID: 32973763 PMCID: PMC7462002 DOI: 10.3389/fimmu.2020.01850] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
T cells play a critical role in mediating antigen-specific and long-term immunity against viral and bacterial pathogens, and their development relies on the highly specialized thymic microenvironment. T cell immunodeficiency can be acquired in the form of inborn errors, or can result from perturbations to the thymus due to aging or irradiation/chemotherapy required for cancer treatment. Hematopoietic stem cell transplant (HSCT) from compatible donors is a cornerstone for the treatment of hematological malignancies and immunodeficiency. Although it can restore a functional immune system, profound impairments exist in recovery of the T cell compartment. T cells remain absent or low in number for many months after HSCT, depending on a variety of factors including the age of the recipient. While younger patients have a shorter refractory period, the prolonged T cell recovery observed in older patients can lead to a higher risk of opportunistic infections and increased predisposition to relapse. Thus, strategies for enhancing T cell recovery in aged individuals are needed to counter thymic damage induced by radiation and chemotherapy toxicities, in addition to naturally occurring age-related thymic involution. Preclinical results have shown that robust and rapid long-term thymic reconstitution can be achieved when progenitor T cells, generated in vitro from HSCs, are co-administered during HSCT. Progenitor T cells appear to rely on lymphostromal crosstalk via receptor activator of NF-κB (RANK) and RANK-ligand (RANKL) interactions, creating chemokine-rich niches within the cortex and medulla that likely favor the recruitment of bone marrow-derived thymus seeding progenitors. Here, we employed preclinical mouse models to demonstrate that in vitro-generated progenitor T cells can effectively engraft involuted aged thymuses, which could potentially improve T cell recovery. The utility of progenitor T cells for aged recipients positions them as a promising cellular therapy for immune recovery and intrathymic repair following irradiation and chemotherapy, even in a post-involution thymus.
Collapse
Affiliation(s)
| | | | - Graham Anderson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Juan Carlos Zúñiga-Pflücker
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
9
|
Generation and function of progenitor T cells from StemRegenin-1-expanded CD34+ human hematopoietic progenitor cells. Blood Adv 2020; 3:2934-2948. [PMID: 31648315 DOI: 10.1182/bloodadvances.2018026575] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 09/08/2019] [Indexed: 12/19/2022] Open
Abstract
Broader clinical application of umbilical cord blood (UCB), as a source of hematopoietic stem/progenitor cells (HSPCs), is limited by low CD34+ and T-cell numbers, contributing to slow lymphohematopoietic recovery, infection, and relapse. Studies have evaluated the safety, feasibility, and expedited neutrophil recovery associated with the transplantation of CD34+ HSPCs from ex vivo expansion cultures using the aryl hydrocarbon receptor antagonist StemRegenin-1 (SR1). In a phase 1/2 study of 17 patients who received combined SR1-expanded and unexpanded UCB units, a considerable advantage for enhancing T-cell chimerism was not observed. We previously showed that progenitor T (proT) cells generated in vitro from HSPCs accelerated T-cell reconstitution and restored immunity after hematopoietic stem cell transplantation (HSCT). To expedite immune recovery, we hypothesized that SR1-expanded HSPCs together with proT cells could overcome the known T-cell immune deficiency that occurs post-HSCT. Here, we show that SR1-expanded UCB can induce >250-fold expansion of CD34+ HSPCs, which can generate large numbers of proT cells upon in vitro differentiation. When compared with nonexpanded naive proT cells, SR1 proT cells also showed effective thymus-seeding and peripheral T-cell functional capabilities in vivo despite having an altered phenotype. In a competitive transfer approach, both naive and SR1 proT cells showed comparable thymus-engrafting capacities. Single-cell RNA sequencing of peripheral CD3+ T cells from mice injected with either naive or SR1 proT cells revealed functional subsets of T cells with polyclonal T-cell receptor repertoires. Our findings support the use of SR1-expanded UCB grafts combined with proT-cell generation for decreasing T-cell immunodeficiency post-HSCT.
Collapse
|
10
|
Melatonin and Mesenchymal Stem Cells as a Key for Functional Integrity for Liver Cancer Treatment. Int J Mol Sci 2020; 21:ijms21124521. [PMID: 32630505 PMCID: PMC7350224 DOI: 10.3390/ijms21124521] [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: 05/25/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common hepatobiliary malignancy with limited therapeutic options. On the other hand, melatonin is an indoleamine that modulates a variety of potential therapeutic effects. In addition to its important role in the regulation of sleep–wake rhythms, several previous studies linked the biologic effects of melatonin to various substantial endocrine, neural, immune and antioxidant functions, among others. Furthermore, the effects of melatonin could be influenced through receptor dependent and receptor independent manner. Among the other numerous physiological and therapeutic effects of melatonin, controlling the survival and differentiation of mesenchymal stem cells (MSCs) has been recently discussed. Given its controversial interaction, several previous reports revealed the therapeutic potential of MSCs in controlling the hepatocellular carcinoma (HCC). Taken together, the intention of the present review is to highlight the effects of melatonin and mesenchymal stem cells as a key for functional integrity for liver cancer treatment. We hope to provide solid piece of information that may be helpful in designing novel drug targets to control HCC.
Collapse
|
11
|
Suraiya AB, Hun ML, Truong VX, Forsythe JS, Chidgey AP. Gelatin-Based 3D Microgels for In Vitro T Lineage Cell Generation. ACS Biomater Sci Eng 2020; 6:2198-2208. [PMID: 33455336 DOI: 10.1021/acsbiomaterials.9b01610] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
T cells are predominantly produced by the thymus and play a significant role in maintaining our adaptive immune system. Physiological involution of the thymus occurs gradually with age, compromising naive T cell output, which can have severe clinical complications. Also, T cells are utilized as therapeutic agents in cancer immunotherapies. Therefore, there is an increasing need for strategies aimed at generating naive T cells. The majority of in vitro T cell generation studies are performed in two-dimensional (2D) cultures, which ignore the physiological thymic microenvironment and are not scalable; therefore, we applied a new three-dimensional (3D) approach. Here, we use a gelatin-based 3D microgel system for T lineage induction by co-culturing OP9-DL4 cells and mouse fetal-liver-derived hematopoietic stem cells (HSCs). Flow cytometric analysis revealed that microgel co-cultures supported T lineage induction similar to 2D cultures while providing a 3D environment. We also encapsulated mouse embryonic thymic epithelial cells (TECs) within the microgels to provide a defined 3D culture platform. The microgel system supported TEC maintenance and retained their phenotype. Together, these data show that our microgel system has the capacity for TEC maintenance and induction of in vitro T lineage differentiation with potential for scalability.
Collapse
Affiliation(s)
- Anisha B Suraiya
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia.,Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | - Michael L Hun
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | - Vinh X Truong
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | - Ann P Chidgey
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| |
Collapse
|
12
|
Trotman-Grant A, Zúñiga-Pflücker JC. Close Quarters Can Be a Good Fit for Stem Cells to Become T Cells. Cell Stem Cell 2019; 24:345-347. [PMID: 30849360 DOI: 10.1016/j.stem.2019.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Human pluripotent stem cells (PSCs) could potentially provide a potentially unlimited supply of T cells. In this issue of Cell Stem Cell, Montel-Hagen et al. (2019) take advantage of a three-dimensional mouse stromal cell-based system that combines mesoderm specification with organoid-induced hematopoietic differentiation to enable commitment, selection, and maturation of conventional human T cells.
Collapse
Affiliation(s)
- Ashton Trotman-Grant
- Department of Immunology, University of Toronto and Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Juan Carlos Zúñiga-Pflücker
- Department of Immunology, University of Toronto and Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada.
| |
Collapse
|
13
|
Pires CF, Rosa FF, Kurochkin I, Pereira CF. Understanding and Modulating Immunity With Cell Reprogramming. Front Immunol 2019; 10:2809. [PMID: 31921109 PMCID: PMC6917620 DOI: 10.3389/fimmu.2019.02809] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/15/2019] [Indexed: 12/30/2022] Open
Abstract
Cell reprogramming concepts have been classically developed in the fields of developmental and stem cell biology and are currently being explored for regenerative medicine, given its potential to generate desired cell types for replacement therapy. Cell fate can be experimentally reversed or modified by enforced expression of lineage specific transcription factors leading to pluripotency or attainment of another somatic cell type identity. The possibility to reprogram fibroblasts into induced dendritic cells (DC) competent for antigen presentation creates a paradigm shift for understanding and modulating the immune system with direct cell reprogramming. PU.1, IRF8, and BATF3 were identified as sufficient and necessary to impose DC fate in unrelated cell types, taking advantage of Clec9a, a C-type lectin receptor with restricted expression in conventional DC type 1. The identification of such minimal gene regulatory networks helps to elucidate the molecular mechanisms governing development and lineage heterogeneity along the hematopoietic hierarchy. Furthermore, the generation of patient-tailored reprogrammed immune cells provides new and exciting tools for the expanding field of cancer immunotherapy. Here, we summarize cell reprogramming concepts and experimental approaches, review current knowledge at the intersection of cell reprogramming with hematopoiesis, and propose how cell fate engineering can be merged to immunology, opening new opportunities to understand the immune system in health and disease.
Collapse
Affiliation(s)
- Cristiana F. Pires
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Fábio F. Rosa
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ilia Kurochkin
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Carlos-Filipe Pereira
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
14
|
Singh J, Zúñiga-Pflücker JC. Producing proT cells to promote immunotherapies. Int Immunol 2019; 30:541-550. [PMID: 30102361 DOI: 10.1093/intimm/dxy051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 08/08/2018] [Indexed: 12/17/2022] Open
Abstract
T lymphocytes are critical mediators of the adaptive immune system and they can be harnessed as therapeutic agents against pathogens and in cancer immunotherapy. T cells can be isolated and expanded from patients and potentially generated in vitro using clinically relevant systems. An ultimate goal for T-cell immunotherapy is to establish a safe, universal effector cell type capable of transcending allogeneic and histocompatibility barriers. To this end, human pluripotent stem cells offer an advantage in generating a boundless supply of T cells that can be readily genetically engineered. Here, we review emerging T-cell therapeutics, including tumor-infiltrating lymphocytes, chimeric antigen receptors and progenitor T cells (proT cells) as well as feeder cell-free in vitro systems for their generation. Furthermore, we explore their potential for adoption in the clinic and highlight the challenges that must be addressed to increase the therapeutic success of a universal immunotherapy.
Collapse
Affiliation(s)
- Jastaranpreet Singh
- Department of Immunology, University of Toronto, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | | |
Collapse
|
15
|
Abstract
Cell lines and animal models have provided the foundation of cancer research for many years. However, human pluripotent stem cells (hPSCs) and organoids are increasingly enabling insights into tumor development, progression, and treatment. Here, we review recent studies using hPSCs to elucidate the reciprocal roles played by genetic alterations and cell identity in cancer formation. We also review studies using human organoids as models that recapitulate both intra- and inter-tumoral heterogeneity to gain new insights into tumorigenesis and treatment responses. Finally, we highlight potential opportunities for cancer research using hPSC-derived organoids and genome editing in the future.
Collapse
Affiliation(s)
- Ryan C Smith
- Department of Neurosurgery, Brain Tumor Center, and Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner, Jr., Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Viviane Tabar
- Department of Neurosurgery, Brain Tumor Center, and Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner, Jr., Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
16
|
Iriguchi S, Kaneko S. Toward the development of true "off-the-shelf" synthetic T-cell immunotherapy. Cancer Sci 2019; 110:16-22. [PMID: 30485606 PMCID: PMC6317915 DOI: 10.1111/cas.13892] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 11/19/2018] [Accepted: 11/22/2018] [Indexed: 12/24/2022] Open
Abstract
Recent outstanding clinical results produced by engineered T cells, including chimeric antigen receptors, have already facilitated further research that broadens their applicability. One such direction is to explore new T cell sources for allogeneic “off‐the‐shelf” adoptive immunotherapy. Human pluripotent stem cells could serve as an alternative cell source for this purpose due to their unique features of infinite propagation ability and pluripotency. Here, we describe the current state of engineered T cell transfer with the focus on cell manufacturing processes and the potentials and challenges of induced pluripotent stem cell‐derived T cells as a starting material to construct off‐the‐shelf T‐cell banks.
Collapse
Affiliation(s)
- Shoichi Iriguchi
- Center for iPS Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Shin Kaneko
- Center for iPS Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
17
|
Iriguchi S, Kaneko S. In Vitro Differentiation of T Cells: From Human Embryonic Stem Cells and Induced Pluripotent Stem Cells. Methods Mol Biol 2019; 2048:59-70. [PMID: 31396929 DOI: 10.1007/978-1-4939-9728-2_6] [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] [Indexed: 01/06/2023]
Abstract
In this chapter, we describe a protocol for hematopoietic differentiation of human pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) derived from non-T cells, followed by the differentiation of the T-cell lineage. Derivation of T cells from PSCs involves three steps: induction of PSCs to hematopoietic progenitor cells (HPCs), differentiation of HPCs into progenitor T cells, and maturation of progenitor T cells into mature T cells (CD8 single-positive (SP) or CD4 SP).
Collapse
Affiliation(s)
- Shoichi Iriguchi
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Research and Application, Kyoto University, Kyoto, Japan
| | - Shin Kaneko
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.
| |
Collapse
|
18
|
Human T cell development notched up a level. Nat Methods 2017; 14:477-478. [PMID: 28448067 DOI: 10.1038/nmeth.4277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|