1
|
Deshpande P, Dornbrand-Lo M, Phondge V, Kelly P, Wong AK. Tissue engineering approaches for lymphedema: biomaterial innovations and clinical potential. Front Cell Dev Biol 2025; 13:1537050. [PMID: 40302939 PMCID: PMC12037638 DOI: 10.3389/fcell.2025.1537050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
The lymphatic system plays a critical role in maintaining fluid balance and immune regulation. Lymphedema, and other lymphatic disorders, highlight the need for advanced therapeutic approaches, including tissue engineering. This review examines the latest developments in artificial lymphatic tissue engineering, focusing on scaffold materials, lymphangiogenic factors, and regenerative strategies to replicate the intricacy of lymphatic vessels and nodes. We conducted a thorough literature review of current practices and applications in lymphatic tissue engineering. Findings show that biomaterials such as hydrogels, decellularized matrices, and synthetic polymers provide effective scaffolds for lymphatic endothelial cell proliferation and lymphangiogenesis. Advances in growth factor delivery and stem-cell based therapies have further enhanced the viability of engineered lymphatic tissues. Despite promising progress, challenges in achieving functional replication of lymphatic structures and clinical translation of research remain. Ongoing research must address scaffold biocompatibility, optimized growth factor targeting, and scalable production to advance therapeutic options for lymphatic disorders. This review underscores the potential for transformative patient outcomes through innovative bioengineering solutions.
Collapse
Affiliation(s)
| | | | | | | | - Alex K. Wong
- Rutgers New Jersey Medical School, Division of Plastic and Reconstructive Surgery, Newark, NJ, United States
| |
Collapse
|
2
|
Kim HM, Bruno TC. An Introduction to Tertiary Lymphoid Structures in Cancer. Methods Mol Biol 2025; 2864:1-19. [PMID: 39527214 DOI: 10.1007/978-1-0716-4184-2_1] [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: 11/16/2024]
Abstract
Immunotherapy has revolutionized therapeutics for cancer patients, which signifies the importance of effective antitumor immunity in combatting cancer. However, the benefit of immunotherapies is limited to specific patient populations and tumor types, suggesting the overt need for new immunotherapeutic targets. Tertiary lymphoid structures (TLS) are ectopic lymph node-like structures that develop at the sites of chronic inflammation such as cancer. TLS are correlated with favorable clinical outcomes across multiple solid tumors and are associated with increased tumor-infiltrating lymphocytes (TILs), particularly effector memory CD8+ T cells. Despite strong clinical data in humans, there are still major knowledge gaps on the function of TLS in cancer. Herein, we highlight the known biology and clinical impact of TLS, which offer further evidence to harness TLS for improved immunotherapeutics.
Collapse
Affiliation(s)
- Hye Mi Kim
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Tumor Microenvironment Center (TMC), UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Tumor Microenvironment Center (TMC), UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Cancer Immunology and Immunotherapy Program (CIIP), UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Kolarzyk AM, Wong G, Lee E. Lymphatic Tissue and Organ Engineering for In Vitro Modeling and In Vivo Regeneration. Cold Spring Harb Perspect Med 2022; 12:a041169. [PMID: 35288402 PMCID: PMC9435571 DOI: 10.1101/cshperspect.a041169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The lymphatic system has an important role in maintaining fluid homeostasis and transporting immune cells and biomolecules, such as dietary fat, metabolic products, and antigens in different organs and tissues. Therefore, impaired lymphatic vessel function and/or lymphatic vessel deficiency can lead to numerous human diseases. The discovery of lymphatic endothelial markers and prolymphangiogenic growth factors, along with a growing number of in vitro and in vivo models and technologies has expedited research in lymphatic tissue and organ engineering, advancing therapeutic strategies. In this article, we describe lymphatic tissue and organ engineering in two- and three-dimensional culture systems and recently developed microfluidics and organ-on-a-chip systems in vitro. Next, we discuss advances in lymphatic tissue and organ engineering in vivo, focusing on biomaterial and scaffold engineering and their applications for lymphatic vessels and lymphoid organ regeneration. Last, we provide expert perspective and prospects in the field of lymphatic tissue engineering.
Collapse
Affiliation(s)
- Anna M Kolarzyk
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| | - Gigi Wong
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biological Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| |
Collapse
|
4
|
Kang W, Feng Z, Luo J, He Z, Liu J, Wu J, Rong P. Tertiary Lymphoid Structures in Cancer: The Double-Edged Sword Role in Antitumor Immunity and Potential Therapeutic Induction Strategies. Front Immunol 2021; 12:689270. [PMID: 34394083 PMCID: PMC8358404 DOI: 10.3389/fimmu.2021.689270] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/05/2021] [Indexed: 12/14/2022] Open
Abstract
The complex tumor microenvironment (TME) plays a vital role in cancer development and dramatically determines the efficacy of immunotherapy. Tertiary lymphoid structures (TLSs) within the TME are well recognized and consist of T cell-rich areas containing dendritic cells (DCs) and B cell-rich areas containing germinal centers (GCs). Accumulating research has indicated that there is a close association between tumor-associated TLSs and favorable clinical outcomes in most types of cancers, though a minority of studies have reported an association between TLSs and a poor prognosis. Overall, the double-edged sword role of TLSs in the TME and potential mechanisms need to be further investigated, which will provide novel therapeutic perspectives for antitumor immunoregulation. In this review, we focus on discussing the main functions of TLSs in the TME and recent advances in the therapeutic manipulation of TLSs through multiple strategies to enhance local antitumor immunity.
Collapse
Affiliation(s)
- Wendi Kang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhichao Feng
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Molecular Imaging Research Center, Central South University, Changsha, China
| | - Jianwei Luo
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhenhu He
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jun Liu
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jianzhen Wu
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Molecular Imaging Research Center, Central South University, Changsha, China
| |
Collapse
|
5
|
Nosenko MA, Moysenovich AM, Arkhipova AY, Atretkhany KSN, Nedospasov SA, Drutskaya MS, Moisenovich MM. Fibroblasts upregulate expression of adhesion molecules and promote lymphocyte retention in 3D fibroin/gelatin scaffolds. Bioact Mater 2021; 6:3449-3460. [PMID: 33817420 PMCID: PMC7988350 DOI: 10.1016/j.bioactmat.2021.03.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/05/2021] [Accepted: 03/04/2021] [Indexed: 01/16/2023] Open
Abstract
Bioengineered scaffolds are crucial components in artificial tissue construction. In general, these scaffolds provide inert three-dimensional (3D) surfaces supporting cell growth. However, some scaffolds can affect the phenotype of cultured cells, especially, adherent stromal cells, such as fibroblasts. Here we report on unique properties of 3D fibroin/gelatin materials, which may rapidly induce expression of adhesion molecules, such as ICAM-1 and VCAM-1, in cultured primary murine embryonic fibroblasts (MEFs). In contrast, two-dimensional (2D) fibroin/gelatin films did not show significant effects on gene expression profiles in fibroblasts as compared to 3D culture conditions. Interestingly, TNF expression was induced in MEFs cultured in 3D fibroin/gelatin scaffolds, while genetic or pharmacological TNF ablation resulted in diminished ICAM-1 and VCAM-1 expression by these cells. Using selective MAPK inhibitors, we uncovered critical contribution of JNK to 3D-induced upregulation of these adhesion molecules. Moreover, we observed ICAM-1/VCAM-1-dependent adhesion of lymphocytes to fibroblasts cultured in 3D fibroin/gelatin scaffolds, but not on 2D fibroin/gelatin films, suggesting functional reprogramming in stromal cells, when exposed to 3D environment. Finally, we observed significant infiltration of lymphocytes into 3D fibroin/gelatin, but not into collagen scaffolds in vivo upon subcapsular kidney implantation in mice. Together our data highlight the important features of fibroin/gelatin scaffolds, when they are produced as 3D sponges rather than 2D films, which should be considered when using these materials for tissue engineering. 3D, but not 2D fibroin-based scaffolds promote expression of adhesion molecules in murine fibroblasts. Overexpression of adhesion molecules in 3D fibroin/gelatin-cultured fibroblasts is TNF- and JNK-dependent. Culturing of fibroblasts in 3D fibroin/gelatin scaffolds promotes adhesion of T-lymphocytes. Implantation of 3D fibroin/gelatin scaffolds in vivo induces infiltration and clustering of T- and B-lymphocytes.
Collapse
Affiliation(s)
- Maxim A Nosenko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | | | - Anastasia Y Arkhipova
- Biological Faculty, Lomonosov Moscow State University, Moscow, 119991, Russia.,Biological Faculty, Shenzhen MSU-BIT University, Shenzhen, 518172, China
| | - Kamar-Sulu N Atretkhany
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Sergei A Nedospasov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.,Biological Faculty, Lomonosov Moscow State University, Moscow, 119991, Russia.,Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, 354340, Russia
| | - Marina S Drutskaya
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | | |
Collapse
|
6
|
Magrone T, Jirillo E. Development and Organization of the Secondary and Tertiary Lymphoid Organs: Influence of Microbial and Food Antigens. Endocr Metab Immune Disord Drug Targets 2019; 19:128-135. [DOI: 10.2174/1871530319666181128160411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 12/11/2022]
Abstract
Background:Secondary lymphoid organs (SLO) are distributed in many districts of the body and, especially, lymph nodes, spleen and gut-associated lymphoid tissue are the main cellular sites. On the other hand, tertiary lymphoid organs (TLO) are formed in response to inflammatory, infectious, autoimmune and neoplastic events. </P><P> Developmental Studies: In the present review, emphasis will be placed on the developmental differences of SLO and TLO between small intestine and colon and on the role played by various chemokines and cell receptors. Undoubtedly, microbiota is indispensable for the formation of SLO and its absence leads to their poor formation, thus indicating its strict interaction with immune and non immune host cells. Furthermore, food antigens (for example, tryptophan derivatives, flavonoids and byphenils) bind the aryl hydrocarbon receptor on innate lymphoid cells (ILCs), thus promoting the development of postnatal lymphoid tissues. Also retinoic acid, a metabolite of vitamin A, contributes to SLO development during embryogenesis. Vitamin A deficiency seems to account for reduction of ILCs and scarce formation of solitary lymphoid tissue. </P><P> Translational Studies: The role of lymphoid organs with special reference to intestinal TLO in the course of experimental and human disease will also be discussed. </P><P> Future Perspectives: Finally, a new methodology, the so-called “gut-in-a dish”, which has facilitated the in vitro interaction study between microbe and intestinal immune cells, will be described.
Collapse
Affiliation(s)
- Thea Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, School of Medicine, Bari, Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, School of Medicine, Bari, Italy
| |
Collapse
|
7
|
Kaiser P, Werner M, Jérôme V, Freitag R. Scale-up of the ex vivo expansion of encapsulated primary human T lymphocytes. Biotechnol Bioeng 2018; 115:2632-2642. [PMID: 29959863 DOI: 10.1002/bit.26786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/23/2018] [Accepted: 06/26/2018] [Indexed: 02/02/2023]
Abstract
A number of evolving medical therapies call for the controlled expansion of primary human T lymphocytes. After encapsulation in sodium cellulose sulfate-poly(diallyldimethyl) ammonium chloride polyelectrolyte capsules, T lymphocytes can be expanded without persisting activation. Here, the challenge of scaling up this process is addressed. Encapsulated T lymphocytes were cultured in spinner flasks as well as in several types of the bioreactor, including fixed and fluidized beds, a waved cell bag, and a standard stirred tank reactor (STR; 1-L scale). Two proprietary T lymphocyte culture media as well as a standard RPMI-based medium were used. As before, encapsulation coincided with the presence of only a low fraction of activated T lymphocytes (peripheral blood T cells) in the total population. Unexpectedly, growth rates were lower in well-mixed reactors than those in cultivations under static conditions, that is, in T-flasks. Switching the STR to low oxygen conditions (40% air saturation) improved the growth rates to the level of the static cultures and thus forms the potential basis for efficient scale-up of T lymphocyte expansion.
Collapse
Affiliation(s)
- Patrick Kaiser
- Department of Process Biotechnology, University of Bayreuth, Bayreuth, Germany
| | - Melanie Werner
- Department of Process Biotechnology, University of Bayreuth, Bayreuth, Germany
| | - Valérie Jérôme
- Department of Process Biotechnology, University of Bayreuth, Bayreuth, Germany
| | - Ruth Freitag
- Department of Process Biotechnology, University of Bayreuth, Bayreuth, Germany
| |
Collapse
|
8
|
Zhu G, Nemoto S, Mailloux AW, Perez-Villarroel P, Nakagawa R, Falahat R, Berglund AE, Mulé JJ. Induction of Tertiary Lymphoid Structures With Antitumor Function by a Lymph Node-Derived Stromal Cell Line. Front Immunol 2018; 9:1609. [PMID: 30061886 PMCID: PMC6054958 DOI: 10.3389/fimmu.2018.01609] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/28/2018] [Indexed: 02/03/2023] Open
Abstract
Tertiary lymphoid structures (TLSs) associate with better prognosis in certain cancer types, but their underlying formation and immunological benefit remain to be determined. We established a mouse model of TLSs to study their contribution to antitumor immunity. Because the stroma in lymph nodes (sLN) participates in architectural support, lymphogenesis, and lymphocyte recruitment, we hypothesized that TLSs can be created by sLN. We selected a sLN line with fibroblast morphology that expressed sLN surface markers and lymphoid chemokines. The subcutaneous injection of the sLN line successfully induced TLSs that attracted infiltration of host immune cell subsets. Injection of MC38 tumor lysate-pulsed dendritic cells activated TLS-residing lymphocytes to demonstrate specific cytotoxicity. The presence of TLSs suppressed MC38 tumor growth in vivo by improving antitumor activity of tumor-infiltrating lymphocytes with downregulated immune checkpoint proteins (PD-1 and Tim-3). Future engineering of sLN lines may allow for further enhancements of TLS functions and immune cell compositions.
Collapse
Affiliation(s)
- Genyuan Zhu
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Satoshi Nemoto
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Adam W Mailloux
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | | | - Ryosuke Nakagawa
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Rana Falahat
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Anders E Berglund
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, United States
| | - James J Mulé
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States.,Cutaneous Oncology Program, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
9
|
Hadamitzky C, Perić H, Theobald SJ, Gratz KF, Spohr H, Pabst R, Vogt PM. Effect of cryopreservation on lymph node fragment regeneration after autologous transplantation in the minipig model. Innov Surg Sci 2018; 3:139-146. [PMID: 31579777 PMCID: PMC6604575 DOI: 10.1515/iss-2018-0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/10/2018] [Indexed: 02/02/2023] Open
Abstract
Introduction Lymphoedema is a worldwide pandemic causing swelling of tissues due to dysfunctional transport of lymph fluid. Present management concepts are based in conservative palliation of symptoms through manual lymphatic drainage, use of compression garments, manual lymph drainage, exercise, and skin care. Nevertheless, some curative options as autologous lymph node transplantation were shown to reduce lymphoedema in selected cases. Lately, some concern has arisen due to reports of donor site morbidity. A possible solution could be the development of artificial lymph node scaffolds as niches of lymphatic regeneration. Engineering these scaffolds has included cryopreservation of lymph node stroma. However, the effects of cryopreservation on the regeneration capacities of these organs were unknown. Materials and methods Here, we used the minipig animal model to assess lymphatic regeneration processes after cryopreservation of autologous lymph nodes. Superficial inguinal lymph nodes were excised and conserved at -80°C for 1 month. Thereafter, lymph node fragments were transplanted in the subcutaneous tissue. Results Regeneration of the lymph nodes was assessed five months after transplantation. We show that lymph node fragment regeneration takes place in spite of former cryopreservation. Transplanted fragments presented typical histological appearance. Their draining capacity was documented by macroscopic transport of Berlin Blue dye as well as through SPECT-CT hybrid imaging. Discussion In conclusion, our results suggest that processes of cryopreservation can be used in the creation of artificial lymph node scaffolds without major impairment of lymph node fragments regeneration.
Collapse
Affiliation(s)
- Catarina Hadamitzky
- Practice for Lympho-Vascular Diseases Bahnhofstrasse 12, Hannover, Germany.,Catarina Hadamitzky and Hanes Perić contributed equally to the manuscript
| | - Hanes Perić
- myDent GmbH Tiergartenstrasse 114, 30559 Hannover, Germany.,Catarina Hadamitzky and Hanes Perić contributed equally to the manuscript
| | - Sebastian J Theobald
- Clinic of Haematology, Haemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | | | - Hendrik Spohr
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Reinhard Pabst
- Institute of Immunomorphology, Hannover Medical School, Hannover, Germany
| | - Peter M Vogt
- Clinic of Plastic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
10
|
Zhu G, Falahat R, Wang K, Mailloux A, Artzi N, Mulé JJ. Tumor-Associated Tertiary Lymphoid Structures: Gene-Expression Profiling and Their Bioengineering. Front Immunol 2017; 8:767. [PMID: 28713385 PMCID: PMC5491937 DOI: 10.3389/fimmu.2017.00767] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/16/2017] [Indexed: 11/15/2022] Open
Abstract
Tertiary lymphoid structures (TLSs) have been identified in the parenchyma and/or in the peripheral margins of human solid tumors. Uncovering the functional nature of these structures is the subject of much intensive investigation. Studies have shown a direct correlation of the presence of human tumor-localized TLS and better patient outcome (e.g., increase in overall survival) in certain solid tumor histologies, but not all. We had identified a tumor-derived immune gene-expression signature, encoding 12 distinct chemokines, which could reliably identify the presence of TLSs, of different degrees, in various human solid tumors. We are focused on understanding the influence of TLSs on the tumor microenvironment and leveraging this understanding to both manipulate the antitumor immune response and potentially enhance immunotherapy applications. Moreover, as not all human solid tumors show the presence of these lymphoid structures, we are embarking on bioengineering approaches to design and build “designer” TLSs to address, and potentially overcome, an unmet medical need in cancer patients whose tumors lack such lymphoid structures.
Collapse
Affiliation(s)
- Genyuan Zhu
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
| | - Rana Falahat
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
| | - Kui Wang
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Adam Mailloux
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
| | - Natalie Artzi
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - James J Mulé
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States.,Cutaneous Oncology Department, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
11
|
Wegner A. Chimeric antigen receptor T cells for the treatment of cancer and the future of preclinical models for predicting their toxicities. Immunotherapy 2017; 9:669-680. [DOI: 10.2217/imt-2017-0028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Chimeric antigen receptor T-cell therapy has achieved highly promising results in clinical trials, particularly in B-cell malignancies. However, reports of serious adverse events including a number of patient deaths have raised concerns about safety of this treatment. Presently available preclinical models are not designed for predicting toxicities seen in human patients. Besides choosing the right animal model, careful considerations must be taken in chimeric antigen receptor T-cell design and the amount of T cells infused. The development of more sophisticated in vitro models and humanized mouse models for preclinical modeling and toxicity tests will help us to improve the design of clinical trials in cancer immunotherapy.
Collapse
Affiliation(s)
- Anja Wegner
- Department of Research Oncology, King's College London, Guy's Hospital Campus, Great Maze Pond, London, SE1 9RT, UK
- Institute of Immunity & Transplantation, University College London, Royal Free Hospital, Roland Hill Street, London, NW3 2PF, UK
| |
Collapse
|
12
|
Jing F, Choi EY. Potential of Cells and Cytokines/Chemokines to Regulate Tertiary Lymphoid Structures in Human Diseases. Immune Netw 2016; 16:271-280. [PMID: 27799872 PMCID: PMC5086451 DOI: 10.4110/in.2016.16.5.271] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/22/2016] [Accepted: 08/27/2016] [Indexed: 02/06/2023] Open
Abstract
Tertiary lymphoid structures (TLS) are ectopic lymphoid tissues involved in chronic inflammation, autoimmune diseases, transplant rejection and cancer. They exhibit almost all the characteristics of secondary lymphoid organs (SLO), which are associated with adaptive immune responses; as such, they contain organized B-cell follicles with germinal centers, distinct areas containing T cells and dendritic cells, high endothelial venules, and lymphatics. In this review, we briefly describe the formation of SLO, and describe the cellular subsets and molecular cues involved in the formation and maintenance of TLS. Finally, we discuss the associations of TLS with human diseases, especially autoimmune diseases, and the potential for therapeutic targeting.
Collapse
Affiliation(s)
- Feifeng Jing
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
13
|
Kobayashi Y, Watanabe T. Gel-Trapped Lymphorganogenic Chemokines Trigger Artificial Tertiary Lymphoid Organs and Mount Adaptive Immune Responses In Vivo. Front Immunol 2016; 7:316. [PMID: 27597851 PMCID: PMC4992816 DOI: 10.3389/fimmu.2016.00316] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/05/2016] [Indexed: 12/22/2022] Open
Abstract
We previously generated artificial lymph node-like tertiary lymphoid organs (artTLOs) in mice using lymphotoxin α-expressing stromal cells. Here, we show the construction of transplantable and functional artTLOs by applying soluble factors trapped in slow-releasing gels in the absence of lymphoid tissue organizer stromal cells. The resultant artTLOs were easily removable, transplantable, and were capable of attracting memory B and T cells. Importantly, artTLOs induced a powerful antigen-specific secondary immune response, which was particularly pronounced in immune-compromised hosts. Synthesis of functionally stable immune tissues/organs like those described here may be a first step to eventually develop immune system-based therapeutics. Although much needs to be learned from the precise mechanisms of action, they may offer ways in the future to reestablish immune functions to overcome hitherto untreatable diseases, including severe infection, cancer, autoimmune diseases, and various forms of immune deficiencies, including immune-senescence during aging.
Collapse
Affiliation(s)
- Yuka Kobayashi
- The Tazuke-Kofukai Medical Research Institute, Kitano Hospital, Kita-ku , Osaka , Japan
| | - Takeshi Watanabe
- The Tazuke-Kofukai Medical Research Institute, Kitano Hospital, Kita-ku , Osaka , Japan
| |
Collapse
|
14
|
Colombo BM, Scalvenzi T, Benlamara S, Pollet N. Microbiota and mucosal immunity in amphibians. Front Immunol 2015; 6:111. [PMID: 25821449 PMCID: PMC4358222 DOI: 10.3389/fimmu.2015.00111] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/26/2015] [Indexed: 12/11/2022] Open
Abstract
We know that animals live in a world dominated by bacteria. In the last 20 years, we have learned that microbes are essential regulators of mucosal immunity. Bacteria, archeas, and viruses influence different aspects of mucosal development and function. Yet, the literature mainly covers findings obtained in mammals. In this review, we focus on two major themes that emerge from the comparative analysis of mammals and amphibians. These themes concern: (i) the structure and functions of lymphoid organs and immune cells in amphibians, with a focus on the gut mucosal immune system; and (ii) the characteristics of the amphibian microbiota and its influence on mucosal immunity. Lastly, we propose to use Xenopus tadpoles as an alternative small-animal model to improve the fundamental knowledge on immunological functions of gut microbiota.
Collapse
Affiliation(s)
- Bruno M Colombo
- Institute of Systems and Synthetic Biology, Université d'Evry Val d'Essonne , Evry , France
| | - Thibault Scalvenzi
- Institute of Systems and Synthetic Biology, Université d'Evry Val d'Essonne , Evry , France
| | - Sarah Benlamara
- Institute of Systems and Synthetic Biology, Université d'Evry Val d'Essonne , Evry , France
| | - Nicolas Pollet
- Institute of Systems and Synthetic Biology, CNRS , Evry , France ; Evolution, Genome, Comportement et Ecologie, CNRS, Université Paris-Sud, IRD , Gif-sur-Yvette , France
| |
Collapse
|
15
|
Werner M, Schmoldt D, Hilbrig F, Jérôme V, Raup A, Zambrano K, Hübner H, Buchholz R, Freitag R. High cell density cultivation of human leukemia T cells (Jurkat cells) in semipermeable polyelectrolyte microcapsules. Eng Life Sci 2015. [DOI: 10.1002/elsc.201400186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- Melanie Werner
- Chair for Process BiotechnologyUniversität Bayreuth, Bayreuth Germany
| | - Daria Schmoldt
- Chair for Process BiotechnologyUniversität Bayreuth, Bayreuth Germany
| | - Frank Hilbrig
- Chair for Process BiotechnologyUniversität Bayreuth, Bayreuth Germany
| | - Valérie Jérôme
- Chair for Process BiotechnologyUniversität Bayreuth, Bayreuth Germany
| | - Alexander Raup
- Institute of Bioprocess EngineeringFriedrich‐Alexander‐Universität Erlangen‐Nürnberg, Erlangen Germany
| | - Kenny Zambrano
- Institute of Bioprocess EngineeringFriedrich‐Alexander‐Universität Erlangen‐Nürnberg, Erlangen Germany
| | - Holger Hübner
- Institute of Bioprocess EngineeringFriedrich‐Alexander‐Universität Erlangen‐Nürnberg, Erlangen Germany
| | - Rainer Buchholz
- Institute of Bioprocess EngineeringFriedrich‐Alexander‐Universität Erlangen‐Nürnberg, Erlangen Germany
| | - Ruth Freitag
- Chair for Process BiotechnologyUniversität Bayreuth, Bayreuth Germany
| |
Collapse
|
16
|
Giese C, Marx U. Human immunity in vitro - solving immunogenicity and more. Adv Drug Deliv Rev 2014; 69-70:103-22. [PMID: 24447895 DOI: 10.1016/j.addr.2013.12.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 12/19/2013] [Accepted: 12/28/2013] [Indexed: 12/24/2022]
Abstract
It has been widely recognised that the phylogenetic distance between laboratory animals and humans limits the former's predictive value for immunogenicity testing of biopharmaceuticals and nanostructure-based drug delivery and adjuvant systems. 2D in vitro assays have been established in conventional culture plates with little success so far. Here, we detail the status of various 3D approaches to emulate innate immunity in non-lymphoid organs and adaptive immune response in human professional lymphoid immune organs in vitro. We stress the tight relationship between the necessarily changing architecture of professional lymphoid organs at rest and when activated by pathogens, and match it with the immunity identified in vitro. Recommendations for further improvements of lymphoid tissue architecture relevant to the development of a sustainable adaptive immune response in vitro are summarized. In the end, we sketch a forecast of translational innovations in the field to model systemic innate and adaptive immunity in vitro.
Collapse
Affiliation(s)
| | - Uwe Marx
- Technische Universität Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| |
Collapse
|
17
|
Kukita A, Kukita T. Multifunctional properties of RANKL/RANK in cell differentiation, proliferation and metastasis. Future Oncol 2013; 9:1609-22. [DOI: 10.2217/fon.13.115] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
It is known that there are close relationships between bone destruction and tumor growth in bone metastasis. RANKL is a central factor in bone metastasis, inducing osteoclastogenesis mediated by its receptor RANK. Recent reports demonstrate that RANKL has important roles in organogenesis stimulating proliferation and differentiation of epithelial and stroma cells. RANKL is induced not only by cytokines and hormones but also by UV-irradiation, inflammation and carcinogens. Expression of RANK and RANKL is found in several human cancer cell lines, and RANK signaling stimulates proliferation, migration and epithelial–mesenchymal transition of cancer cells, which may be involved in metastasis via an autocrine/paracrine mechanism. RANKL regulates the number of Tregs that produce RANKL, which may affect cancer metastasis. In this review we discuss the multifunctional roles of RANKL/RANK in osteoclastogenesis, organogenesis, and the metastasis and tumorigenesis of cancer cells.
Collapse
Affiliation(s)
- Akiko Kukita
- Department of Microbiology, Medicine, Saga University, 5-1-1, Nabeshima, Saga, 849-8501, Japan
| | - Toshio Kukita
- Molecular Cell Biology & Oral Anatomy, Kyushu University, Maidashi, Fukuoka, Japan
| |
Collapse
|
18
|
Corsiero E, Bombardieri M, Manzo A, Bugatti S, Uguccioni M, Pitzalis C. Role of lymphoid chemokines in the development of functional ectopic lymphoid structures in rheumatic autoimmune diseases. Immunol Lett 2012; 145:62-7. [PMID: 22698185 DOI: 10.1016/j.imlet.2012.04.013] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 04/13/2012] [Indexed: 01/11/2023]
Abstract
A sizeable subset of patients with the two most common organ-specific rheumatic autoimmune diseases, rheumatoid arthritis (RA) and Sjögren's syndrome (SS) develop ectopic lymphoid structures (ELS) in the synovial tissue and salivary glands, respectively. These structures are characterized by perivascular (RA) and periductal (SS) clusters of T and B lymphocytes, differentiation of high endothelial venules and networks of stromal follicular dendritic cells (FDC). Accumulated evidence from other and our group demonstrated that the formation and maintenance of ELS in these chronic inflammatory conditions is critically dependent on the ectopic expression of lymphotoxins (LT) and lymphoid chemokines CXCL13, CCL19, CCL21 and CXCL12. In this review we discuss recent advances highlighting the cellular and molecular mechanisms, which regulate the formation of ELS in RA and SS, with particular emphasis on the role of lymphoid chemokines. In particular, we shall focus on the evidence that in the inflammatory microenvironment of the RA synovium and SS salivary glands, several cell types, including resident epithelial, stromal and endothelial cells as well as different subsets of infiltrating immune cells, have been shown to be capable of producing lymphoid chemokines. Finally, we summarize accumulating data supporting the conclusion that ELS in RA and SS represent functional niches for B cells to undergo affinity maturation, clonal selection and differentiation into plasma cells autoreactive against disease-specific antigens, thus contributing to humoral autoimmunity over and above that of secondary lymphoid organs.
Collapse
Affiliation(s)
- Elisa Corsiero
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | | | | | | | | | | |
Collapse
|