151
|
Lindenstrøm T, Moguche A, Damborg M, Agger EM, Urdahl K, Andersen P. T Cells Primed by Live Mycobacteria Versus a Tuberculosis Subunit Vaccine Exhibit Distinct Functional Properties. EBioMedicine 2017; 27:27-39. [PMID: 29249639 PMCID: PMC5828549 DOI: 10.1016/j.ebiom.2017.12.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/22/2017] [Accepted: 12/05/2017] [Indexed: 01/10/2023] Open
Abstract
Despite inducing strong T cell responses, Mycobacterium tuberculosis (Mtb) infection fails to elicit protective immune memory. As such latently infected or successfully treated Tuberculosis (TB) patients are not protected against recurrent disease. Here, using a mouse model of aerosol Mtb infection, we show that memory immunity to H56/CAF01 subunit vaccination conferred sustained protection in contrast to the transient natural immunity conferred by Mtb infection. Loss of protection to re-infection in natural Mtb memory was temporally linked to an accelerated differentiation of ESAT-6- and to a lesser extent, Ag85B-specific CD4 T cells in both the lung parenchyma and vasculature. This phenotype was characterized by high KLRG1 expression and low, dual production of IFN-γ and TNF. In contrast, H56/CAF01 vaccination elicited cells that expressed low levels of KLRG1 with copious expression of IL-2 and IL-17A. Co-adoptive transfer studies revealed that H56/CAF01 induced memory CD4 T cells efficiently homed into the lung parenchyma of mice chronically infected with Mtb. In comparison, natural Mtb infection- and BCG vaccine-induced memory CD4 T cells exhibited a poor ability to home into the lung parenchyma. These studies suggest that impaired lung migratory capacity is an inherent trait of the terminally differentiated memory responses primed by mycobacteria/mycobacterial vectors. Differentiation state of M. tuberculosis (Mtb)-specific CD4 memory T cells differ depending on their initial priming Live mycobacteria prime fully differentiated CD4 memory T cells with lower lung homing capacity than subunit vaccination Lung parenchymal Mtb memory CD4 T cells produce fewer & less cytokines, express more KLRG1 and cannot sustain protection
People latently infected with M. tuberculosis or successfully treated for Tuberculosis are not protected against recurrent disease, even in the presence of strong T cell responses. Here, using a well-established mouse model, we show that in contrast to subunit vaccination, live mycobacteria prime CD4 T cells that are highly differentiated, have an inferior lung homing capacity and show impaired function once in the parenchyma leading to lack of sustained protection against challenge. This indicates a central shortcoming of natural immunity that needs to be addressed in order to develop improved vaccines against TB.
Collapse
Affiliation(s)
- Thomas Lindenstrøm
- Department of Infectious Disease Immunology, Statens Serum Institut, Denmark.
| | | | - Mie Damborg
- Department of Infectious Disease Immunology, Statens Serum Institut, Denmark
| | - Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, Denmark
| | - Kevin Urdahl
- Center for Infectious Disease Research, Seattle, USA
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Denmark
| |
Collapse
|
152
|
Tsuda B, Miyamoto A, Yokoyama K, Ogiya R, Oshitanai R, Terao M, Morioka T, Niikura N, Okamura T, Miyako H, Saito Y, Suzuki Y, Kametani Y, Tokuda Y. B-cell populations are expanded in breast cancer patients compared with healthy controls. Breast Cancer 2017; 25:284-291. [PMID: 29204848 PMCID: PMC5906508 DOI: 10.1007/s12282-017-0824-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/29/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND Historically, humoral immunity was considered unimportant in anti-tumor immunity, and the differentiation and anti-tumor activity of B cells in breast cancer are poorly understood. However, it was recently discovered that B cells participate in tumor immunity through both antibody production and immunosuppressive mechanisms. We analyzed the expression of B-cell differentiation markers in detail using fluorescence-activated cell sorting to investigate the relationship between B-cell subsets and breast cancer etiology. METHODS Blood samples were taken from breast cancer patients and healthy donors, and peripheral blood mononuclear cells were collected. B cells at various stages of differentiation were identified by the expression of combinations of the cell surface markers CD5, CD19, CD21, CD24, CD27, CD38, CD45, and IgD. Statistical analysis of the proportions of each B-cell subtype in the different patient groups was then performed. RESULTS Twenty-seven breast cancer patients and 12 controls were considered. The proportion of total B cells was significantly higher in cancer patients than in controls (11.51 ± 2.059 vs 8.905 ± 0.379%, respectively; p = 0.001). Breast cancer patients were then classified as High-B or Low-B for further analysis. A significantly higher proportion of memory B cells was found in the High-B group than in the Low-B or control groups (p = 0.003 and p = 0.043, respectively). CONCLUSIONS Breast cancer patients generally have a higher proportion of B cells than healthy controls, but this is highly variable. Analysis of the major B-cell surface markers indicates that memory B cells in particular are significantly expanded, or more robust, in breast cancer patients.
Collapse
Affiliation(s)
- Banri Tsuda
- Department of Breast and Endocrine Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan.
| | - Asuka Miyamoto
- Department of Breast and Endocrine Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Kozue Yokoyama
- Department of Breast and Endocrine Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Rin Ogiya
- Department of Breast and Endocrine Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Risa Oshitanai
- Department of Breast and Endocrine Surgery, Tokai University Hachioji Hospital, Hachioji, Japan
| | - Mayako Terao
- Department of Breast and Endocrine Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Toru Morioka
- Department of Breast and Endocrine Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Naoki Niikura
- Department of Breast and Endocrine Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Takuho Okamura
- Department of Breast and Endocrine Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | | | - Yuki Saito
- Department of Breast and Endocrine Surgery, Tokai University Hachioji Hospital, Hachioji, Japan
| | - Yasuhiro Suzuki
- Department of Breast and Endocrine Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Yoshie Kametani
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Japan
| | - Yutaka Tokuda
- Department of Breast and Endocrine Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| |
Collapse
|
153
|
Sadelain M, Rivière I, Riddell S. Therapeutic T cell engineering. Nature 2017; 545:423-431. [PMID: 28541315 DOI: 10.1038/nature22395] [Citation(s) in RCA: 602] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/26/2017] [Indexed: 12/13/2022]
Abstract
Genetically engineered T cells are powerful new medicines, offering hope for curative responses in patients with cancer. Chimaeric antigen receptors (CARs) are a class of synthetic receptors that reprogram lymphocyte specificity and function. CARs targeting CD19 have demonstrated remarkable potency in B cell malignancies. Engineered T cells are applicable in principle to many cancers, pending further progress to identify suitable target antigens, overcome immunosuppressive tumour microenvironments, reduce toxicities, and prevent antigen escape. Advances in the selection of optimal T cells, genetic engineering, and cell manufacturing are poised to broaden T-cell-based therapies and foster new applications in infectious diseases and autoimmunity.
Collapse
Affiliation(s)
- Michel Sadelain
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Isabelle Rivière
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Stanley Riddell
- Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| |
Collapse
|
154
|
Abstract
INTRODUCTION Recent breakthrough advances in Multiple Myeloma (MM) immunotherapy have been achieved with the approval of the first two monoclonal antibodies, elotuzumab and daratumumab. Adoptive cell therapy (ACT) represents yet another, maybe the most powerful modality of immunotherapy, in which allogeneic or autologous effector cells are expanded and activated ex vivo followed by their re-infusion back into patients. Infused effector cells belong to two categories: naturally occurring, non-engineered cells (donor lymphocyte infusion, myeloma infiltrating lymphocytes, deltagamma T cells) or genetically- engineered antigen-specific cells (chimeric antigen receptor T or NK cells, TCR-engineered cells). Areas covered: This review article summarizes our up-to-date knowledge on ACT in MM, its promises, and upcoming strategies to both overcome its toxicity and to integrate it into future treatment paradigms. Expert opinion: Early results of clinical studies using CAR T cells or TCR- engineered T cells in relapsed and refractory MM are particularly exciting, indicating the potential of long-term disease control or even cure. Despite several caveats including toxicity, costs and restricted availability in particular, these forms of immunotherapy are likely to once more revolutionize MM therapy.
Collapse
Affiliation(s)
- Sonia Vallet
- a Department of Internal Medicine , Karl Landsteiner University of Health Sciences, University Hospital , Krems an der Donau , Austria
| | - Martin Pecherstorfer
- a Department of Internal Medicine , Karl Landsteiner University of Health Sciences, University Hospital , Krems an der Donau , Austria
| | - Klaus Podar
- a Department of Internal Medicine , Karl Landsteiner University of Health Sciences, University Hospital , Krems an der Donau , Austria
| |
Collapse
|
155
|
Schober K, Busch DH. TIL 2.0: More effective and predictive T-cell products by enrichment for defined antigen specificities. Eur J Immunol 2017; 46:1335-9. [PMID: 27280482 DOI: 10.1002/eji.201646436] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 04/17/2016] [Accepted: 04/21/2016] [Indexed: 12/21/2022]
Abstract
Adoptive transfer of in vitro-expanded T cells derived from tumor-infiltrating lymphocytes (TILs) in melanoma patients started the era of tumor immunotherapy three decades ago. The approach has demonstrated remarkable clinical responses in several studies since. Reinfusion of TIL-derived T cells represents a highly personalized form of immunotherapy, taking into account the enormous interindividual tumor heterogeneity. However, despite its successes, TIL therapy does not lead to objective clinical responses in all cases. It is thus crucial to find out which tumor antigens are particularly valuable targets and to develop strategies to enhance the reactivity of T-cell products toward them. In this issue of the European Journal of Immunology, Kelderman et al. [Eur. J. Immunol. 2016. 46: 1351-1360] present a platform for the generation of antigen-specific TIL therapy. Combining recently developed technologies for clinical identification and enrichment of antigen-specific CD8(+) T cells, such as MHC Streptamers and UV-mediated peptide exchange, the authors could enrich T-cell populations with defined antigen specificities from melanoma-derived TILs. This T-cell product showed higher reactivity against autologous tumor cell lines than bulk TIL-derived T cells. The novel platform might enable the generation of more effective and predictable TIL-derived T-cell products for future clinical applications.
Collapse
Affiliation(s)
- Kilian Schober
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany.,DZIF - National Centre for Infection Research, Munich, Germany.,Focus Group "Clinical Cell Processing and Purification,", Institute for Advanced Study, Technische Universität München, Munich, Germany
| |
Collapse
|
156
|
Gattinoni L, Speiser DE, Lichterfeld M, Bonini C. T memory stem cells in health and disease. Nat Med 2017; 23:18-27. [PMID: 28060797 DOI: 10.1038/nm.4241] [Citation(s) in RCA: 439] [Impact Index Per Article: 54.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 10/28/2016] [Indexed: 12/11/2022]
Abstract
T memory stem (TSCM) cells are a rare subset of memory lymphocytes endowed with the stem cell-like ability to self-renew and the multipotent capacity to reconstitute the entire spectrum of memory and effector T cell subsets. Cumulative evidence in mice, nonhuman primates and humans indicates that TSCM cells are minimally differentiated cells at the apex of the hierarchical system of memory T lymphocytes. Here we describe emerging findings demonstrating that TSCM cells, owing to their extreme longevity and robust potential for immune reconstitution, are central players in many physiological and pathological human processes. We also discuss how TSCM cell stemness could be leveraged therapeutically to enhance the efficacy of vaccines and adoptive T cell therapies for cancer and infectious diseases or, conversely, how it could be disrupted to treat TSCM cell driven and sustained diseases, such as autoimmunity, adult T cell leukemia and HIV-1.
Collapse
Affiliation(s)
- Luca Gattinoni
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel E Speiser
- Department of Oncology, Ludwig Cancer Research, Lausanne University Hospital, Lausanne, Switzerland
| | - Mathias Lichterfeld
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Chiara Bonini
- Experimental Hematology Unit, Division of Immunology Transplantation and Infectious Diseases, Leukemia Unit, San Raffaele Scientific Institute, Milan, Italy.,Hematology Department, Vita Salute San Raffaele University, Milan, Italy
| |
Collapse
|
157
|
Francian A, Mann K, Kullberg M. Complement C3-dependent uptake of targeted liposomes into human macrophages, B cells, dendritic cells, neutrophils, and MDSCs. Int J Nanomedicine 2017; 12:5149-5161. [PMID: 28790822 PMCID: PMC5529385 DOI: 10.2147/ijn.s138787] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Antitumor immunity in cancer patients is heavily modulated by cells of the innate immune system. Antigen-presenting cells, including dendritic cells, macrophages, and B cells, initiate immune recognition of tumor antigen by displaying antigen to effector cells. Countering this immune stimulation are immunosuppressive cells which include M2 macrophages, N2 neutrophils, and myeloid-derived suppressor cells (MDSCs). To create effective cancer immunotherapies, it is critical that we can target these important cell types of the immune system with immunostimulatory compounds. A commonality of these cell types is the complement receptor, which recognizes pathogens that are bound to activated complement C3 in human blood. To target the complement receptor, we have created a liposome that has a small molecule, orthopyridyl disulfide (OPSS), conjugated to its surface. OPSS forms a disulfide bond with activated complement C3, which then targets liposomes for uptake by dendritic cells, macrophages, B cells, MDSCs, and neutrophils in human blood. Internalization is efficient and specific to cells that display the complement receptor. Liposomes are a versatile drug delivery device. Possible applications for this system include delivery of toll-receptor agonists or tumor antigen to antigen-presenting cells and delivery of immunostimulatory drugs to M2, N2, and MDSC immunosuppressive cells.
Collapse
Affiliation(s)
| | - Kristine Mann
- WWAMI Medical Education Program.,Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK, USA
| | | |
Collapse
|
158
|
Neal LR, Bailey SR, Wyatt MM, Bowers JS, Majchrzak K, Nelson MH, Haupt C, Paulos CM, Varela JC. The Basics of Artificial Antigen Presenting Cells in T Cell-Based Cancer Immunotherapies. JOURNAL OF IMMUNOLOGY RESEARCH AND THERAPY 2017; 2:68-79. [PMID: 28825053 PMCID: PMC5560309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Adoptive T cell transfer (ACT) can mediate objective responses in patients with advanced malignancies. There have been major advances in this field, including the optimization of the ex vivo generation of tumor-reactive lymphocytes to ample numbers for effective ACT therapy via the use of natural and artificial antigen presenting cells (APCs). Herein we review the basic properties of APCs and how they have been manufactured through the years to augment vaccine and T cell-based cancer therapies. We then discuss how these novel APCs impact the function and memory properties of T cells. Finally, we propose new ways to synthesize aAPCs to augment the therapeutic effectiveness of antitumor T cells for ACT therapy.
Collapse
Affiliation(s)
- Lillian R. Neal
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
- Department of Hematology and Oncology, Medical University of South Carolina, Charleston, 29425
| | - Stefanie R. Bailey
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Megan M. Wyatt
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Jacob S. Bowers
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Kinga Majchrzak
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Michelle H. Nelson
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Carl Haupt
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Hematology and Oncology, Medical University of South Carolina, Charleston, 29425
| | - Chrystal M. Paulos
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Juan C. Varela
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Hematology and Oncology, Medical University of South Carolina, Charleston, 29425
| |
Collapse
|
159
|
彭 耀, 吴 其, 刘 鸿, 赵 健, 危 华. [Construction of specific artificial antigen-presenting cells for in vitro activation of CD19 chimeric antigen receptor T cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:581-587. [PMID: 28539278 PMCID: PMC6780484 DOI: 10.3969/j.issn.1673-4254.2017.05.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To construct CD19-specific artificial antigen-presenting cells (aAPCs) for in vitro activation and expansion of CD19 chimeric antigen receptor (CAR)-modified T cells (CD19-CAR-T) and investigate their cytotoxic effect. METHODS CD19-specific aAPCs (NIH3T3-CD19/86, NIH3T3-CD19/86/137L) expressing costimulatory molecules CD86 and/or CD137L were prepared on the basis of NIH3T3 backbone cells by lentivirus-mediated gene transfer. Irradiated CD19-specific aAPCs were co-cultured with CD19-CAR-T cells to activate and amplify CD19-CAR-T cells. The growth curve of CD19-CAR-T cells was determined by trypan blue exclusion assay, and CD19CAR expression and phenotype on CD19-CAR-T cells were detected by flow cytometry. The in vitro cytotoxicity of CD19-CAR-T cells against the target cells was evaluated by bioluminescence-based cytotoxicity assay. RESULTS Flow cytometry showed that NIH3T3-CD19/86 and NIH3T3-CD19/86/137L expressed high levels of CD19, CD86 and/or CD137L. Both NIH3T3-CD19/86 and NIH3T3-CD19/86/137L cells could amplify CD19-CAR-T cells efficiently, but NIH3T3-CD19/86/137L cells had better amplification effect. After 14 days of co-culture with NIH3T3-CD19/86/137L cells, the number of CD19-CAR-T cells was significantly greater than that of NIH3T3-CD19/86 cells (P<0.05), and the proportion of CD19-CAR-T cells in the total T cells increased significantly (P<0.05). CD19-CAR-T cells amplified by CD19-specific aAPCs produced target-specific cytotoxicity and were able to specifically kill CD19-positive target cells. About 20% central memory T cells were present in the final products expanded by NIH3T3-CD19/86/137L. CONCLUSION We successfully prepared CD19-specific aAPCs that can specifically amplify functional CD19-CART cells in vitro, which facilitates the acquisition of clinical-scale high-quality CD19-CART cells.
Collapse
Affiliation(s)
- 耀军 彭
- 解放军总医院肿瘤中心实验室,北京 100853Cancer Center Key Laboratory, General Hospital of PLA, Beijing 100853, China
| | - 其艳 吴
- 解放军总医院肿瘤中心实验室,北京 100853Cancer Center Key Laboratory, General Hospital of PLA, Beijing 100853, China
| | - 鸿宇 刘
- 南开大学医学院,天津 300071College of Medicine, Nankai University, Tianjin 300071, China
| | - 健 赵
- 解放军总医院肿瘤中心实验室,北京 100853Cancer Center Key Laboratory, General Hospital of PLA, Beijing 100853, China
- 第二军医大学肿瘤研究所,上海 200433International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, China
| | - 华锋 危
- 解放军总医院肿瘤中心实验室,北京 100853Cancer Center Key Laboratory, General Hospital of PLA, Beijing 100853, China
| |
Collapse
|
160
|
Affiliation(s)
- Thomas S Watkins
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia.,School of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - John J Miles
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia.,School of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
161
|
Chimeric Antigen Receptors: A Cell and Gene Therapy Perspective. Mol Ther 2017; 25:1117-1124. [PMID: 28456379 DOI: 10.1016/j.ymthe.2017.03.034] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 03/28/2017] [Accepted: 03/28/2017] [Indexed: 02/08/2023] Open
Abstract
Chimeric antigen receptors (CARs) are synthetic receptors that reprogram T lymphocytes to target chosen antigens. The targeting of CD19, a cell surface molecule expressed in the vast majority of leukemias and lymphomas, has been successfully translated in the clinic, earning CAR therapy a special distinction in the selection of "cancer immunotherapy" by Science as the breakthrough of the year in 2013. CD19 CAR therapy is predicated on advances in genetic engineering, T cell biology, tumor immunology, synthetic biology, target identification, cell manufacturing sciences, and regulatory compliance-the central tenets of CAR therapy. Here, we review two of these foundations: the genetic engineering approaches and cell types to engineer.
Collapse
|
162
|
Karuturi BVK, Tallapaka SB, Yeapuri P, Curran SM, Sanderson SD, Vetro JA. Encapsulation of an EP67-Conjugated CTL Peptide Vaccine in Nanoscale Biodegradable Particles Increases the Efficacy of Respiratory Immunization and Affects the Magnitude and Memory Subsets of Vaccine-Generated Mucosal and Systemic CD8 + T Cells in a Diameter-Dependent Manner. Mol Pharm 2017; 14:1469-1481. [PMID: 28319404 DOI: 10.1021/acs.molpharmaceut.6b01088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The diameter of biodegradable particles used to coencapsulate immunostimulants and subunit vaccines affects the magnitude of memory CD8+ T cells generated by systemic immunization. Possible effects on the magnitude of CD8+ T cells generated by mucosal immunization or memory subsets that potentially correlate more strongly with protection against certain pathogens, however, are unknown. In this study, we conjugated our novel host-derived mucosal immunostimulant, EP67, to the protective MCMV CTL epitope, pp89, through a lysosomal protease-labile double arginine linker (pp89-RR-EP67) and encapsulated in PLGA 50:50 micro- or nanoparticles. We then compared total magnitude, effector/central memory (CD127/KRLG1/CD62L), and IFN-γ/TNF-α/IL-2 secreting subsets of pp89-specific CD8+ T cells as well as protection of naive female BALB/c mice against primary respiratory infection with MCMV 21 days after respiratory immunization. We found that decreasing the diameter of encapsulating particle from ∼5.4 μm to ∼350 nm (i) increased the magnitude of pp89-specific CD8+ T cells in the lungs and spleen; (ii) partially changed CD127/KLRG1 effector memory subsets in the lungs but not the spleen; (iii) changed CD127/KRLG1/CD62L effector/central memory subsets in the spleen; (iv) changed pp89-responsive IFN-γ/TNF-α/IL-2 secreting subsets in the lungs and spleen; (v) did not affect the extent to which encapsulation increased efficacy against primary MCMV respiratory infection over unencapsulated pp89-RR-EP67. Thus, although not observed under our current experimental conditions with MCMV, varying the diameter of nanoscale biodegradable particles may increase the efficacy of mucosal immunization with coencapsulated immunostimulant/subunit vaccines against certain pathogens by selectively increasing memory subset(s) of CD8+ T cells that correlate the strongest with protection.
Collapse
Affiliation(s)
- Bala V K Karuturi
- Center for Drug Delivery and Nanomedicine and §Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center , 986025 Nebraska Medical Center, Omaha, Nebraska 68198-6025, United States
| | - Shailendra B Tallapaka
- Center for Drug Delivery and Nanomedicine and §Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center , 986025 Nebraska Medical Center, Omaha, Nebraska 68198-6025, United States
| | - Pravin Yeapuri
- Center for Drug Delivery and Nanomedicine and §Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center , 986025 Nebraska Medical Center, Omaha, Nebraska 68198-6025, United States
| | - Stephen M Curran
- Center for Drug Delivery and Nanomedicine and §Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center , 986025 Nebraska Medical Center, Omaha, Nebraska 68198-6025, United States
| | - Sam D Sanderson
- Center for Drug Delivery and Nanomedicine and §Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center , 986025 Nebraska Medical Center, Omaha, Nebraska 68198-6025, United States
| | - Joseph A Vetro
- Center for Drug Delivery and Nanomedicine and §Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center , 986025 Nebraska Medical Center, Omaha, Nebraska 68198-6025, United States
| |
Collapse
|
163
|
Sadelain M. Chimeric Antigen Receptors: A Paradigm Shift in Immunotherapy. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2017. [DOI: 10.1146/annurev-cancerbio-050216-034351] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
164
|
Seay HR, Putnam AL, Cserny J, Posgai AL, Rosenau EH, Wingard JR, Girard KF, Kraus M, Lares AP, Brown HL, Brown KS, Balavage KT, Peters LD, Bushdorf AN, Atkinson MA, Bluestone JA, Haller MJ, Brusko TM. Expansion of Human Tregs from Cryopreserved Umbilical Cord Blood for GMP-Compliant Autologous Adoptive Cell Transfer Therapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 4:178-191. [PMID: 28345003 PMCID: PMC5363324 DOI: 10.1016/j.omtm.2016.12.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/16/2016] [Indexed: 12/29/2022]
Abstract
Umbilical cord blood is a traditional and convenient source of cells for hematopoietic stem cell transplantation. Thymic regulatory T cells (Tregs) are also present in cord blood, and there is growing interest in the use of autologous Tregs to provide a low-risk, fully human leukocyte antigen (HLA)-matched cell product for treating autoimmune diseases, such as type 1 diabetes. Here, we describe a good manufacturing practice (GMP)-compatible Treg expansion protocol using fluorescence-activated cell sorting, resulting in a mean 2,092-fold expansion of Tregs over a 16-day culture for a median yield of 1.26 × 109 Tregs from single-donor cryopreserved units. The resulting Tregs passed prior clinical trial release criteria for Treg purity and sterility, including additional rigorous assessments of FOXP3 and Helios expression and epigenetic analysis of the FOXP3 Treg-specific demethylated region (TSDR). Compared with expanded adult peripheral blood Tregs, expanded cord blood Tregs remained more naive, as assessed by continued expression of CD45RA, produced reduced IFN-γ following activation, and effectively inhibited responder T cell proliferation. Immunosequencing of the T cell receptor revealed a remarkably diverse receptor repertoire within cord blood Tregs that was maintained following in vitro expansion. These data support the feasibility of generating GMP-compliant Tregs from cord blood for adoptive cell transfer therapies and highlight potential advantages in terms of safety, phenotypic stability, autoantigen specificity, and tissue distribution.
Collapse
Affiliation(s)
- Howard R Seay
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Amy L Putnam
- Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Judit Cserny
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Emma H Rosenau
- Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - John R Wingard
- Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | | | - Angela P Lares
- Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | - Kristi T Balavage
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Leeana D Peters
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Ashley N Bushdorf
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA; Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Jeffrey A Bluestone
- Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael J Haller
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| |
Collapse
|
165
|
Human parainfluenza virus-3 can be targeted by rapidly ex vivo expanded T lymphocytes. Cytotherapy 2016; 18:1515-1524. [PMID: 27692559 DOI: 10.1016/j.jcyt.2016.08.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/05/2016] [Accepted: 08/30/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND AIMS Human parainfluenza virus-3 (HPIV) is a common cause of respiratory infection in immunocompromised patients and currently has no effective therapies. Virus-specific T-cell therapy has been successful for the treatment or prevention of viral infections in immunocompromised patients but requires determination of T-cell antigens on targeted viruses. METHODS HPIV3-specific T cells were expanded from peripheral blood of healthy donors using a rapid generation protocol targeting four HPIV3 proteins. Immunophenotyping was performed by flow cytometry. Viral specificity was determined by interferon (IFN)-γ ELISpot, intracellular cytokine staining and cytokine measurements from culture supernatants by Luminex assay. Cytotoxic activity was tested by 51Cr release and CD107a mobilization assays. Virus-specific T cells targeting six viruses were then produced by rapid protocol, and the phenotype of HPIV3-specific T cells was determined by immunomagnetic sorting for IFN-γ-producing cells. RESULTS HPIV3-specific T cells were expanded from 13 healthy donors. HPIV3-specific T cells showed a CD4+ predominance (mean CD4:CD8 ratio 2.89) and demonstrated specificity for multiple HPIV3 antigens. The expanded T cells were polyfunctional based on cytokine production but only had a minor cytotoxic component. T cells targeting six viruses in a single product similarly showed HPIV3 specificity, with a predominant effector memory phenotype (CD3+/CD45RA-/CCR7-) in responder cells. DISCUSSION HPIV3-specific T cells can be produced using a rapid ex vivo protocol from healthy donors and are predominantly CD4+ T cells with Th1 activity. HPIV3 epitopes can also be successfully targeted alongside multiple other viral epitopes in production of six-virus T cells, without loss of HPIV3 specificity. These products may be clinically beneficial to combat HPIV3 infections by adoptive T-cell therapy in immune-compromised patients.
Collapse
|
166
|
Technical Considerations for the Generation of Adoptively Transferred T Cells in Cancer Immunotherapy. Cancers (Basel) 2016; 8:cancers8090086. [PMID: 27657129 PMCID: PMC5040988 DOI: 10.3390/cancers8090086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/04/2016] [Accepted: 09/12/2016] [Indexed: 12/28/2022] Open
Abstract
A significant function of the immune system is the surveillance and elimination of aberrant cells that give rise to cancer. Even when tumors are well established and metastatic, immune-mediated spontaneous regressions have been documented. While there are have been various forms of immunotherapy, one of the most widely studied for almost 40 years is adoptive cellular immunotherapy, but its success has yet to be fully realized. Adoptive cell transfer (ACT) is a therapeutic modality that has intrigued physicians and researchers for its many theoretical benefits. Preclinical investigations and human trials have utilized natural killer (NK) cells, dendritic cells (DC), macrophages, T-cells or B-cells for ACT with the most intense research focused on T-cell ACT. T-cells are exquisitely specific to the target of its T-cell receptor (TCR), thus potentially reducing the amount of collateral damage and off-target effects from treatment. T-cells also possess a memory subset that may reduce the risk of recurrence of a cancer after the successful treatment of the primary disease. There are several options for the source of T-cells used in the generation of cells for ACT. Perhaps the most widely known source is T-cells generated from tumor-infiltrating lymphocytes (TILs). However, studies have also employed peripheral blood mononuclear cells (PBMCs), lymph nodes, and even induced pluripotent stem cells (IPSCs) as a source of T-cells. Several important technical considerations exist regarding benefits and limitations of each source of T-cells. Unique aspects of T-cells factor into their ability to be efficacious in ACT including the total number of cells available for ACT, the anti-tumor efficacy on a per cell basis, the repertoire of TCRs specific to tumor cells, and their ability to traffic to various organs that harbor tumor. Current research is attempting to unlock the full potential of these cells to effectively and safely treat cancer.
Collapse
|
167
|
Refining human T-cell immunotherapy of cytomegalovirus disease: a mouse model with 'humanized' antigen presentation as a new preclinical study tool. Med Microbiol Immunol 2016; 205:549-561. [PMID: 27539576 DOI: 10.1007/s00430-016-0471-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 08/03/2016] [Indexed: 12/12/2022]
Abstract
With the cover headline 'T cells on the attack,' the journal Science celebrated individualized cancer immunotherapy by adoptive transfer of T cells as the 'Breakthrough of the Year' 2013 (J. Couzin-Frankel in Science 342:1432-1433, 2013). It is less well recognized and appreciated that individualized T cell immunotherapy of cytomegalovirus (CMV) infection is approaching clinical application for preventing CMV organ manifestations, interstitial CMV pneumonia in particular. This coincident medical development is particularly interesting as reactivated CMV infection is a major viral complication in the state of transient immunodeficiency after the therapy of hematopoietic malignancies by hematopoietic cell transplantation (HCT). It may thus be attractive to combine T cell immunotherapy of 'minimal residual disease/leukemia (MRD)' and CMV-specific T cell immunotherapy to combat both risks in HCT recipients simultaneously, and ideally with T cells derived from the respective HLA-matched HCT donor. Although clinical trials of human CMV-specific T cell immunotherapy were promising in that the incidence of virus reactivation and disease was found to be reduced with statistical significance, animal models are still instrumental for providing 'proof of concept' by directly documenting the prevention of viral multiple-organ histopathology and organ failure under controlled conditions of the absence versus presence of the therapy, which obviously is not feasible in an individual human patient. Further, animal models can make predictions regarding parameters that determine the efficacy of T cell immunotherapy for improved study design in clinical investigations, and they allow for manipulating host and virus genetics. The latter is of particular value as it opens the possibility for epitope specificity controls that are inherently missing in clinical trials. Here, we review a recently developed new mouse model that is more approximated to human CMV-specific T cell immunotherapy by 'humanizing' antigen presentation using antigenically chimeric CMV and HLA-transgenic mice to allow for an in vivo testing of the antiviral function of human CMV-specific T cells. As an important new message, this model predicts that T cell immunotherapy is most efficient if CD4 T cells are equipped with a transduced TCR directed against an epitope presented by MHC/HLA class-I for local delivery of 'cognate' help to CD8 effector T cells at infected MHC/HLA class-II-negative host tissue cells.
Collapse
|
168
|
Realizing the Potential for Recipient Immune Cells in Adoptive Immune Therapy. EBioMedicine 2016; 10:11-2. [PMID: 27397512 PMCID: PMC5006657 DOI: 10.1016/j.ebiom.2016.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 07/04/2016] [Indexed: 11/23/2022] Open
|
169
|
Sadelain M. Chimeric antigen receptors: driving immunology towards synthetic biology. Curr Opin Immunol 2016; 41:68-76. [PMID: 27372731 DOI: 10.1016/j.coi.2016.06.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/06/2016] [Accepted: 06/08/2016] [Indexed: 12/15/2022]
Abstract
The advent of second generation chimeric antigen receptors and the CD19 paradigm have ushered a new therapeutic modality in oncology. In contrast to earlier forms of adoptive cell therapy, which were based on the isolation and expansion of naturally occurring T cells, CAR therapy is based on the design and manufacture of engineered T cells with optimized properties. A new armamentarium, comprising not only CARs but also chimeric costimulatory receptors, chimeric cytokine receptors, inhibitory receptors and synthetic Notch receptors, expressed in naïve, central memory or stem cell-like memory T cells, is being developed for clinical use in a wide range of cancers. Immunological principles are thus finding a new purpose thanks to advances in genetic engineering, synthetic biology and cell manufacturing sciences.
Collapse
Affiliation(s)
- Michel Sadelain
- Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
170
|
Nguyen HH, Kim T, Song SY, Park S, Cho HH, Jung SH, Ahn JS, Kim HJ, Lee JJ, Kim HO, Cho JH, Yang DH. Naïve CD8(+) T cell derived tumor-specific cytotoxic effectors as a potential remedy for overcoming TGF-β immunosuppression in the tumor microenvironment. Sci Rep 2016; 6:28208. [PMID: 27306834 PMCID: PMC4910083 DOI: 10.1038/srep28208] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/23/2016] [Indexed: 01/04/2023] Open
Abstract
Despite of the potential implications for cancer immunotherapy, conventional approaches using in vitro expanded CD8+ T cells have suboptimal outcomes, mostly due to loss of functionality from cellular exhaustion. We therefore investigated the phenotypic and functional differences among in vitro activated CD8+ T cells of three different sources, namely naïve (NTeff), memory (MTeff) and tumor-infiltrating lymphocytes (TILeff) from human and mice, to better understand mechanisms behind potent effector functions and potential for overcoming current limitations. In line with the greater proliferation activity and longer telomere lengths of NTeff populations, cells of naïve origin exhibited significantly less amounts of T cell exhaustion markers than those of MTeff and TILeff, and moreover, acquired distinct expression patterns of memory-promoting transcription factors, T-bet and Eomes, induced in a rapid and sustainable manner. NTeff cells appeared to have lower expression of Foxp1 and were refractory to apoptosis upon TGF-β conditioning, implying better survival potential and resistance to tumor-induced immune suppression. Of CD8+ T cell pools activated to tumor-specific CTLs, naïve cell generated effectors possessed the most potent cytotoxic activity, validating implications for use in rational design of adoptive immunotherapy.
Collapse
Affiliation(s)
- Hong Hanh Nguyen
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Therasa Kim
- Department of Medical Sciences, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Sang Yun Song
- Department of Thoracic and Cardiovascular Surgery, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Somang Park
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Hyang Hee Cho
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Sung-Hoon Jung
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Jae-Sook Ahn
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Hee-Ok Kim
- Academy of Immunology &Microbiology (AIM), Institute for Basic Science (IBS), Pohang, Republic of Korea
| | - Jae-Ho Cho
- Academy of Immunology &Microbiology (AIM), Institute for Basic Science (IBS), Pohang, Republic of Korea
| | - Deok-Hwan Yang
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| |
Collapse
|
171
|
Höfer T, Barile M, Flossdorf M. Stem-cell dynamics and lineage topology from in vivo fate mapping in the hematopoietic system. Curr Opin Biotechnol 2016; 39:150-156. [PMID: 27107166 DOI: 10.1016/j.copbio.2016.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/29/2016] [Accepted: 04/01/2016] [Indexed: 12/21/2022]
Abstract
In recent years, sophisticated fate-mapping tools have been developed to study the behavior of stem cells in the intact organism. These experimental approaches are beginning to yield a quantitative picture of how cell numbers are regulated during steady state and in response to challenges. Focusing on hematopoiesis and immune responses, we discuss how novel mathematical approaches driven by these fate-mapping data have provided insights into the dynamics and topology of cellular differentiation pathways in vivo. The combination of experiment and theory has allowed to quantify the degree of self-renewal in stem and progenitor cells, shown how native hematopoiesis differs fundamentally from post-transplantation hematopoiesis, and uncovered that the diversification of T lymphocytes during immune responses resembles tissue renewal driven by stem cells.
Collapse
Affiliation(s)
- Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Bioquant Center, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.
| | - Melania Barile
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Bioquant Center, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Michael Flossdorf
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Bioquant Center, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| |
Collapse
|
172
|
Affiliation(s)
- Luca Gattinoni
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, 10 Center Drive, Bethesda, MD, USA.
| |
Collapse
|