51
|
Chuprin J, Buettner H, Seedhom MO, Greiner DL, Keck JG, Ishikawa F, Shultz LD, Brehm MA. Humanized mouse models for immuno-oncology research. Nat Rev Clin Oncol 2023; 20:192-206. [PMID: 36635480 PMCID: PMC10593256 DOI: 10.1038/s41571-022-00721-2] [Citation(s) in RCA: 162] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/14/2023]
Abstract
Immunotherapy has emerged as a promising treatment paradigm for many malignancies and is transforming the drug development landscape. Although immunotherapeutic agents have demonstrated clinical efficacy, they are associated with variable clinical responses, and substantial gaps remain in our understanding of their mechanisms of action and specific biomarkers of response. Currently, the number of preclinical models that faithfully recapitulate interactions between the human immune system and tumours and enable evaluation of human-specific immunotherapies in vivo is limited. Humanized mice, a term that refers to immunodeficient mice co-engrafted with human tumours and immune components, provide several advantages for immuno-oncology research. In this Review, we discuss the benefits and challenges of the currently available humanized mice, including specific interactions between engrafted human tumours and immune components, the development and survival of human innate immune populations in these mice, and approaches to study mice engrafted with matched patient tumours and immune cells. We highlight the latest advances in the generation of humanized mouse models, with the aim of providing a guide for their application to immuno-oncology studies with potential for clinical translation.
Collapse
Affiliation(s)
- Jane Chuprin
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Molecular, Cell and Cancer Biology, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Hannah Buettner
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Surgery, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Mina O Seedhom
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dale L Greiner
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | | | | | - Michael A Brehm
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
52
|
Mace EM. Human natural killer cells: Form, function, and development. J Allergy Clin Immunol 2023; 151:371-385. [PMID: 36195172 PMCID: PMC9905317 DOI: 10.1016/j.jaci.2022.09.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/22/2022] [Accepted: 09/02/2022] [Indexed: 02/07/2023]
Abstract
Human natural killer (NK) cells are innate lymphoid cells that mediate important effector functions in the control of viral infection and malignancy. Their ability to distinguish "self" from "nonself" and lyse virally infected and tumorigenic cells through germline-encoded receptors makes them important players in maintaining human health and a powerful tool for immunotherapeutic applications and fighting disease. This review introduces our current understanding of NK cell biology, including key facets of NK cell differentiation and the acquisition and execution of NK cell effector function. Further, it addresses the clinical relevance of NK cells in both primary immunodeficiency and immunotherapy. It is intended to provide an up-to-date and comprehensive overview of this important and interesting innate immune effector cell subset.
Collapse
Affiliation(s)
- Emily M Mace
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York.
| |
Collapse
|
53
|
Abeynaike SA, Huynh TR, Mehmood A, Kim T, Frank K, Gao K, Zalfa C, Gandarilla A, Shultz L, Paust S. Human Hematopoietic Stem Cell Engrafted IL-15 Transgenic NSG Mice Support Robust NK Cell Responses and Sustained HIV-1 Infection. Viruses 2023; 15:365. [PMID: 36851579 PMCID: PMC9960100 DOI: 10.3390/v15020365] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Mice reconstituted with human immune systems are instrumental in the investigation of HIV-1 pathogenesis and therapeutics. Natural killer (NK) cells have long been recognized as a key mediator of innate anti-HIV responses. However, established humanized mouse models do not support robust human NK cell development from engrafted human hematopoietic stem cells (HSCs). A major obstacle to human NK cell reconstitution is the lack of human interleukin-15 (IL-15) signaling, as murine IL-15 is a poor stimulator of the human IL-15 receptor. Here, we demonstrate that immunodeficient NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice expressing a transgene encoding human IL-15 (NSG-Tg(IL-15)) have physiological levels of human IL-15 and support long-term engraftment of human NK cells when transplanted with human umbilical-cord-blood-derived HSCs. These Hu-NSG-Tg(IL-15) mice demonstrate robust and long-term reconstitution with human immune cells, but do not develop graft-versus-host disease (GVHD), allowing for long-term studies of human NK cells. Finally, we show that these HSC engrafted mice can sustain HIV-1 infection, resulting in human NK cell responses in HIV-infected mice. We conclude that Hu-NSG-Tg(IL-15) mice are a robust novel model to study NK cell responses to HIV-1.
Collapse
Affiliation(s)
- Shawn A. Abeynaike
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tridu R. Huynh
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Scripps Research Translational Institute, La Jolla, CA 92037, USA
- Division of Internal Medicine, Scripps Clinic/Scripps Green Hospital, La Jolla, CA 92037, USA
| | - Abeera Mehmood
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Teha Kim
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kayla Frank
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kefei Gao
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Cristina Zalfa
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Angel Gandarilla
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Silke Paust
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
54
|
The Frequency and Function of NKG2C +CD57 + Adaptive NK Cells in Cytomagalovirus Co-Infected People Living with HIV Decline with Duration of Antiretroviral Therapy. Viruses 2023; 15:v15020323. [PMID: 36851537 PMCID: PMC9959045 DOI: 10.3390/v15020323] [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: 01/03/2023] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Human cytomegalovirus (CMV) infection drives the expansion and differentiation of natural killer (NK) cells with adaptive-like features. We investigated whether age and time on antiretroviral therapy (ART) influenced adaptive NK cell frequency and functionality. Flow cytometry was used to evaluate the frequency of adaptive and conventional NK cells in 229 CMV+ individuals of whom 170 were people living with HIV (PLWH). The frequency of these NK cell populations producing CD107a, CCL4, IFN-γ or TNF-α was determined following a 6-h antibody dependent (AD) stimulation. Though ART duration and age were correlated, longer time on ART was associated with a reduced frequency of adaptive NK cells. In general, the frequency and functionality of NK cells following AD stimulation did not differ significantly between treated CMV+PLWH and CMV+HIV- persons, suggesting that HIV infection, per se, did not compromise AD NK cell function. AD activation of adaptive NK cells from CMV+PLWH induced lower frequencies of IFN-γ or TNF-α secreting cells in older persons, when compared with younger persons.
Collapse
|
55
|
Smith MR, Satter LRF, Vargas-Hernández A. STAT5b: A master regulator of key biological pathways. Front Immunol 2023; 13:1025373. [PMID: 36755813 PMCID: PMC9899847 DOI: 10.3389/fimmu.2022.1025373] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/29/2022] [Indexed: 01/25/2023] Open
Abstract
The Signal Transducer and Activator of Transcription (STAT)-5 proteins are required in immune regulation and homeostasis and play a crucial role in the development and function of several hematopoietic cells. STAT5b activation is involved in the expression of genes that participate in cell development, proliferation, and survival. STAT5a and STAT5b are paralogs and only human mutations in STAT5B have been identified leading to immune dysregulation and hematopoietic malignant transformation. The inactivating STAT5B mutations cause impaired post-natal growth, recurrent infections and immune dysregulation, whereas gain of function somatic mutations cause dysregulated allergic inflammation. These mutations are rare, and they are associated with a wide spectrum of clinical manifestations which provide a disease model elucidating the biological mechanism of STAT5 by studying the consequences of perturbations in STAT5 activity. Further, the use of Jak inhibitors as therapy for a variety of autoimmune and malignant disorders has increased substantially heading relevant lessons for the consequences of Jak/STAT immunomodulation from the human model. This review summarizes the biology of the STAT5 proteins, human disease associate with molecular defects in STAT5b, and the connection between aberrant activation of STAT5b and the development of certain cancers.
Collapse
Affiliation(s)
- Madison R. Smith
- Department of Pediatrics, Division of Immunology, Allergy, and Retrovirology, Baylor College of Medicine, Houston, TX, United States,William T. Shearer Texas Children’s Hospital Center for Human Immunobiology, Houston, TX, United States
| | - Lisa R. Forbes Satter
- Department of Pediatrics, Division of Immunology, Allergy, and Retrovirology, Baylor College of Medicine, Houston, TX, United States,William T. Shearer Texas Children’s Hospital Center for Human Immunobiology, Houston, TX, United States
| | - Alexander Vargas-Hernández
- Department of Pediatrics, Division of Immunology, Allergy, and Retrovirology, Baylor College of Medicine, Houston, TX, United States,William T. Shearer Texas Children’s Hospital Center for Human Immunobiology, Houston, TX, United States,*Correspondence: Alexander Vargas-Hernández,
| |
Collapse
|
56
|
Kang G, Zhao X, Sun J, Cheng C, Wang C, Tao L, Zong L, Yin W, Cong J, Li J, Wang X. A2AR limits IL-15-induced generation of CD39 + NK cells with high cytotoxicity. Int Immunopharmacol 2023; 114:109567. [PMID: 36529024 DOI: 10.1016/j.intimp.2022.109567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
CD39-mediated inhibition of natural killer (NK) cell activity has been demonstrated, but the characteristics of CD39+ NK cells in humans are not known. We investigated the characteristics of human circulating CD39+ NK cells. In healthy donors, the proportion of circulating CD39+ NK cells in total NK cells was relatively low compared with that of CD39- NK cells. Nonetheless, a higher proportion of CD39+ NK cells expressed CD107a. Similarly, a higher proportion of CD39+ NK cells expressed CD107a in patients with hepatitis B virus or patients with hepatocellular carcinoma. Stimulation with NK-sensitive K562 cells or interleukin (IL)-12/IL-18 activated CD39+ NK cells to express higher levels of CD107a, IFN-γ and TNF-α, relative to CD39- NK cells. Importantly, IL-15 induced the generation of CD39+ NK cells. In contrast, A2A adenosine receptor (A2AR) ligation suppressed the generation of CD39+ NK cells by inhibiting IL-15 signaling. These data for the first time demonstrated that A2AR counteracts IL-15-induced generation of human CD39+ NK cells, which have a stronger cytotoxicity than CD39- NK cells. IL-15-induced human CD39+ NK cells might be better choice for immunotherapy based on adoptive transfer of NK cells.
Collapse
Affiliation(s)
- Guijie Kang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Xueqin Zhao
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Jiafeng Sun
- Dental Department, Health Service Center, Jianghai Community, Guangyi Street, Liangxi District, Wuxi 214000, Jiangsu, China
| | - Chen Cheng
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Cen Wang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Longxiang Tao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China
| | - Lu Zong
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China
| | - Wenwei Yin
- Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jingjing Cong
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China.
| | - Jing Li
- School of Life Sciences, Anhui Medical University, Hefei 230032, China.
| | - Xuefu Wang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
57
|
Sindaco P, Pandey H, Isabelle C, Chakravarti N, Brammer JE, Porcu P, Mishra A. The role of interleukin-15 in the development and treatment of hematological malignancies. Front Immunol 2023; 14:1141208. [PMID: 37153603 PMCID: PMC10157481 DOI: 10.3389/fimmu.2023.1141208] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/22/2023] [Indexed: 05/09/2023] Open
Abstract
Cytokines are a vital component of the immune system that controls the activation and growth of blood cells. However, chronic overexpression of cytokines can trigger cellular events leading to malignant transformation. The cytokine interleukin-15 (IL-15) is of particular interest, which has been shown to contribute to the development and progression of various hematological malignancies. This review will provide an overview of the impact of the immunopathogenic function of IL-15 by studying its role in cell survival, proliferation, inflammation, and treatment resistance. We will also review therapeutic approaches for inhibiting IL-15 in blood cancers.
Collapse
Affiliation(s)
- Paola Sindaco
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Hritisha Pandey
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Colleen Isabelle
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Nitin Chakravarti
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | | | - Pierluigi Porcu
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Anjali Mishra
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
- *Correspondence: Anjali Mishra,
| |
Collapse
|
58
|
Poon AYC, Sugimura R. The prospect of genetically engineering natural killer cells for cancer immunotherapy. Biol Open 2022; 11:bio059396. [PMID: 36445164 PMCID: PMC9729658 DOI: 10.1242/bio.059396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023] Open
Abstract
The use of natural killer (NK) cells in cancer immunotherapy demonstrates promising potential, yet its efficacy is often limited due to the loss of tumor-killing capacity and lack of specificity in vivo. Here, we review current approaches to confer enhanced tumor-killing capacity and specificity by genetic engineering. Increasing sensitivity to cytokines and protecting NK cells from the immune checkpoint endowed sustainability of NK cells in the tumor microenvironment. Transducing chimeric antigen receptor (CAR) in NK cells successfully targeted both hematologic and solid tumors in preclinical models. The use of human pluripotent stem cells as an expandable and genetically amenable platform offers a stable source of engineered NK cells for cancer immunotherapy. We highlight that CAR-NK cells from human pluripotent stem cells are a promising approach for cancer immunotherapy.
Collapse
Affiliation(s)
- Angie Yu Ching Poon
- School of Biomedical Science, University of Hong Kong, 21 Sassoon Rd, Hong Kong999077
| | - Ryohichi Sugimura
- School of Biomedical Science, University of Hong Kong, 21 Sassoon Rd, Hong Kong999077
| |
Collapse
|
59
|
Sungur CM, Wang Q, Ozantürk AN, Gao H, Schmitz AJ, Cella M, Yokoyama WM, Shan L. Human NK cells confer protection against HIV-1 infection in humanized mice. J Clin Invest 2022; 132:e162694. [PMID: 36282589 PMCID: PMC9753998 DOI: 10.1172/jci162694] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/04/2022] [Indexed: 12/24/2022] Open
Abstract
The role of NK cells against HIV-1 infections remains to be elucidated in vivo. While humanized mouse models potentially could be used to directly evaluate human NK cell responses during HIV-1 infection, improved functional development of human NK cells in these hosts is needed. Here, we report the humanized MISTRG-6-15 mouse model, in which NK cells were quick to expand and exhibit degranulation, cytotoxicity, and proinflammatory cytokine production in nonlymphoid organs upon HIV-1 infection but had reduced functionality in lymphoid organs. Although HIV-1 infection induced functional impairment of NK cells, antiretroviral therapy reinvigorated NK cells in response to HIV-1 rebound after analytic treatment interruption. Moreover, a broadly neutralizing antibody, PGT121, enhanced NK cell function in vivo, consistent with antibody-dependent cellular cytotoxicity. Monoclonal antibody depletion of NK cells resulted in higher viral loads in multiple nonlymphoid organs. Overall, our results in humanized MISTRG-6-15 mice demonstrated that NK cells provided direct anti-HIV-1 responses in vivo but were limited in their responses in lymphoid organs.
Collapse
Affiliation(s)
| | - Qiankun Wang
- Division of Infectious Diseases, Department of Medicine
| | | | - Hongbo Gao
- Division of Infectious Diseases, Department of Medicine
| | | | | | - Wayne M. Yokoyama
- Division of Rheumatology, Department of Medicine
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Liang Shan
- Division of Infectious Diseases, Department of Medicine
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
60
|
Bourayou E, Golub R. Inflammatory-driven NK cell maturation and its impact on pathology. Front Immunol 2022; 13:1061959. [PMID: 36569860 PMCID: PMC9780665 DOI: 10.3389/fimmu.2022.1061959] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
NK cells are innate lymphocytes involved in a large variety of contexts and are crucial in the immunity to intracellular pathogens as well as cancer due to their ability to kill infected or malignant cells. Thus, they harbor a strong potential for clinical and therapeutic use. NK cells do not require antigen exposure to get activated; their functional response is rather based on a balance between inhibitory/activating signals and on the diversity of germline-encoded receptors they express. In order to reach optimal functional status, NK cells go through a step-wise development in the bone marrow before their egress, and dissemination into peripheral organs via the circulation. In this review, we summarize bone marrow NK cell developmental stages and list key factors involved in their differentiation before presenting newly discovered and emerging factors that regulate NK cell central and peripheral maturation. Lastly, we focus on the impact inflammatory contexts themselves can have on NK cell development and functional maturation.
Collapse
Affiliation(s)
- Elsa Bourayou
- Institut Pasteur, Université Paris Cité, INSERM U1223, Lymphocyte and Immunity Unit, Paris, France
| | - Rachel Golub
- Institut Pasteur, Université Paris Cité, INSERM U1223, Lymphocyte and Immunity Unit, Paris, France
| |
Collapse
|
61
|
Salinas SA, Mace EM, Conte MI, Park CS, Li Y, Rosario-Sepulveda JI, Mahapatra S, Moore EK, Hernandez ER, Chinn IK, Reed AE, Lee BJ, Frumovitz A, Gibbs RA, Posey JE, Forbes Satter LR, Thatayatikom A, Allenspach EJ, Wensel TG, Lupski JR, Lacorazza HD, Orange JS. An ELF4 hypomorphic variant results in NK cell deficiency. JCI Insight 2022; 7:e155481. [PMID: 36477361 PMCID: PMC9746917 DOI: 10.1172/jci.insight.155481] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/13/2022] [Indexed: 12/12/2022] Open
Abstract
NK cell deficiencies (NKD) are a type of primary immune deficiency in which the major immunologic abnormality affects NK cell number, maturity, or function. Since NK cells contribute to immune defense against virally infected cells, patients with NKD experience higher susceptibility to chronic, recurrent, and fatal viral infections. An individual with recurrent viral infections and mild hypogammaglobulinemia was identified to have an X-linked damaging variant in the transcription factor gene ELF4. The variant does not decrease expression but disrupts ELF4 protein interactions and DNA binding, reducing transcriptional activation of target genes and selectively impairing ELF4 function. Corroborating previous murine models of ELF4 deficiency (Elf4-/-) and using a knockdown human NK cell line, we determined that ELF4 is necessary for normal NK cell development, terminal maturation, and function. Through characterization of the NK cells of the proband, expression of the proband's variant in Elf4-/- mouse hematopoietic precursor cells, and a human in vitro NK cell maturation model, we established this ELF4 variant as a potentially novel cause of NKD.
Collapse
Affiliation(s)
- Sandra Andrea Salinas
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Emily M. Mace
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Matilde I. Conte
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | | | - Yu Li
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | | | - Sanjana Mahapatra
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, Texas, USA
| | - Emily K. Moore
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Evelyn R. Hernandez
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Ivan K. Chinn
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, Texas, USA
| | - Abigail E. Reed
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Barclay J. Lee
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Alexander Frumovitz
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Richard A. Gibbs
- Department of Molecular and Human Genetics, and
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | | | - Lisa R. Forbes Satter
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, Texas, USA
| | - Akaluck Thatayatikom
- Division of Pediatric Allergy, Immunology, and Rheumatology, Department of Pediatrics, University of Florida, Shands Children’s Hospital, Gainesville, Florida, USA
| | - Eric J. Allenspach
- Division of Immunology, Seattle Children’s Hospital, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | | | - James R. Lupski
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, Texas, USA
- Department of Molecular and Human Genetics, and
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | | | - Jordan S. Orange
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
62
|
Podaza E, Kuo HH, Nguyen J, Elemento O, Martin ML. Next generation patient derived tumor organoids. Transl Res 2022; 250:84-97. [PMID: 35964899 DOI: 10.1016/j.trsl.2022.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/24/2022] [Accepted: 08/03/2022] [Indexed: 11/18/2022]
Abstract
Patient-derived tumor organoids (PDTOs) have emerged as exceptional pre-clinical models as they preserved, in most of the cases, the mutational landscape and tumor-clonal heterogeneity of the primary tumors. Despite being extensively used in disease modelling as well as in precision medicine for drug testing and discovery, they still have some limitations. The main limitation is that during their establishment they lose all components of the tumor microenvironment (TME) which are known modulators of tumor response to therapeutic treatment as well as disease progression. In this review we address the effects of different players of the TME such as immune cells, fibroblasts, endothelial cells and the extracellular matrix composition on tumor behavior and response to treatment as well as the different culture and co-culture strategies that could improve PDTOs value as pre-clinical models leading to the development of next generation PDTOs.
Collapse
Affiliation(s)
- Enrique Podaza
- Weill Cornell Medicine, Caryl and Israel Englander Institute for Precision Medicine, New York, New York
| | - Hui-Hsuan Kuo
- Weill Cornell Medicine, Caryl and Israel Englander Institute for Precision Medicine, New York, New York
| | - John Nguyen
- Weill Cornell Medicine, Caryl and Israel Englander Institute for Precision Medicine, New York, New York
| | - Olivier Elemento
- Weill Cornell Medicine, Caryl and Israel Englander Institute for Precision Medicine, New York, New York
| | - M Laura Martin
- Weill Cornell Medicine, Caryl and Israel Englander Institute for Precision Medicine, New York, New York.
| |
Collapse
|
63
|
Conte MI, Poli MC, Taglialatela A, Leuzzi G, Chinn IK, Salinas SA, Rey-Jurado E, Olivares N, Veramendi-Espinoza L, Ciccia A, Lupski JR, Aldave Becerra JC, Mace EM, Orange JS. Partial loss-of-function mutations in GINS4 lead to NK cell deficiency with neutropenia. JCI Insight 2022; 7:e154948. [PMID: 36345943 PMCID: PMC9675456 DOI: 10.1172/jci.insight.154948] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 09/14/2022] [Indexed: 11/09/2022] Open
Abstract
Human NK cell deficiency (NKD) is a primary immunodeficiency in which the main clinically relevant immunological defect involves missing or dysfunctional NK cells. Here, we describe a familial NKD case in which 2 siblings had a substantive NKD and neutropenia in the absence of other immune system abnormalities. Exome sequencing identified compound heterozygous variants in Go-Ichi-Ni-San (GINS) complex subunit 4 (GINS4, also known as SLD5), an essential component of the human replicative helicase, which we demonstrate to have a damaging impact upon the expression and assembly of the GINS complex. Cells derived from affected individuals and a GINS4-knockdown cell line demonstrate delayed cell cycle progression, without signs of improper DNA synthesis or increased replication stress. By modeling partial GINS4 depletion in differentiating NK cells in vitro, we demonstrate the causal relationship between the genotype and the NK cell phenotype, as well as a cell-intrinsic defect in NK cell development. Thus, biallelic partial loss-of-function mutations in GINS4 define a potentially novel disease-causing gene underlying NKD with neutropenia. Together with the previously described mutations in other helicase genes causing NKD, and with the mild defects observed in other human cells, these variants underscore the importance of this pathway in NK cell biology.
Collapse
Affiliation(s)
- Matilde I. Conte
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - M. Cecilia Poli
- Faculty of Medicine, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
- Immunology and Rheumatology Unit, Hospital Roberto del Rio, Santiago, Chile
| | - Angelo Taglialatela
- Department of Genetics and Development, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Giuseppe Leuzzi
- Department of Genetics and Development, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Ivan K. Chinn
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Division of Immunology, Allergy, and Retrovirology, Texas Children’s Hospital, Houston, Texas, USA
| | - Sandra A. Salinas
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Emma Rey-Jurado
- Faculty of Medicine, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Nixa Olivares
- Faculty of Medicine, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Liz Veramendi-Espinoza
- Allergy and Clinical Immunology, Hospital Nacional Edgardo Rebagliati Martins, Lima, Peru
| | - Alberto Ciccia
- Department of Genetics and Development, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
| | - James R. Lupski
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | | | - Emily M. Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Jordan S. Orange
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
64
|
Yu H, Jacquelot N, Belz GT. Metabolic features of innate lymphoid cells. J Exp Med 2022; 219:e20221140. [PMID: 36301303 PMCID: PMC9617479 DOI: 10.1084/jem.20221140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/31/2022] [Accepted: 10/04/2022] [Indexed: 12/02/2022] Open
Abstract
Innate and adaptive immune cells are found in distinct tissue niches where they orchestrate immune responses. This requires intrinsic and temporal metabolic adaptability to coordinately activate the immune response cascade. Dysregulation of this program is a key feature of immunosuppression. Direct or indirect metabolic immune cell reprogramming may offer new approaches to modulate immune cells behavior for therapy to overcome dysregulation. In this review, we explored how metabolism regulates lymphocytes beyond the classical T cell subsets. We focus on the innate lymphoid cell (ILC) family, highlighting the distinct metabolic characteristics of these cells, the impact of environmental factors, and the receptors that could alter immune cell functions through manipulation of metabolic pathways to potentially prevent or treat various diseases.
Collapse
Affiliation(s)
- Huiyang Yu
- The University of Queensland, Diamantina Institute, Brisbane, Queensland, Australia
| | - Nicolas Jacquelot
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Gabrielle T. Belz
- The University of Queensland, Diamantina Institute, Brisbane, Queensland, Australia
| |
Collapse
|
65
|
Wang X, Yang X, Yuan X, Wang W, Wang Y. Chimeric antigen receptor-engineered NK cells: new weapons of cancer immunotherapy with great potential. Exp Hematol Oncol 2022; 11:85. [PMID: 36324149 PMCID: PMC9628181 DOI: 10.1186/s40164-022-00341-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/29/2022] [Indexed: 11/18/2022] Open
Abstract
Chimeric antigen receptor (CAR)-engineered T (CAR-T) cells have obtained prominent achievement in the clinical immunotherapy of hematological malignant tumors, leading to a rapid development of cellular immunotherapy in cancer treatment. Scientists are also aware of the prospective advantages of CAR engineering in cellular immunotherapy. Due to various limitations such as the serious side effects of CAR-T therapy, researchers began to investigate other immune cells for CAR modification. Natural killer (NK) cells are critical innate immune cells with the characteristic of non-specifically recognizing target cells and with the potential to become "off-the-shelf" products. In recent years, many preclinical studies on CAR-engineered NK (CAR-NK) cells have shown their remarkable efficacy in cancer therapy and their superiority over autologous CAR-T cells. In this review, we summarize the generation, mechanisms of anti-tumor activity and unique advantages of CAR-NK cells, and then analyze some challenges and recent clinical trials about CAR-NK cells therapy. We believe that CAR-NK therapy is a promising prospect for cancer immunotherapy in the future.
Collapse
Affiliation(s)
- Xiao Wang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xuejiao Yang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xiang Yuan
- grid.13291.380000 0001 0807 1581Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Wenbo Wang
- grid.24516.340000000123704535Department of Oncology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Yueying Wang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
66
|
Ward Grados DF, Ahmadi H, Griffith TS, Warlick CA. Immunotherapy for Bladder Cancer: Latest Advances and Ongoing Clinical Trials. Immunol Invest 2022; 51:2226-2251. [PMID: 36083246 DOI: 10.1080/08820139.2022.2118606] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
For nearly 50 years, immunotherapy has been used in patients with bladder cancer in the form of Mycobacterium bovis Bacillus Calmette-Guerin (BCG), which is still the first-line therapy for non-muscle invasive disease. However, the remarkable results obtained with checkpoint inhibitor drugs, including Pembrolizumab and Atezolizumab, have fueled the quest to optimize these and other forms of immunotherapy for both non-muscle invasive as well as advanced bladder cancer. In this review we summarize the current state of the rapidly evolving field of immunotherapy in bladder cancer highlighting novel approaches and ongoing trials in this exciting area of research.
Collapse
Affiliation(s)
| | - Hamed Ahmadi
- Department of Urology, University of Minnesota, Minneapolis, MN, USA
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Christopher A Warlick
- Department of Urology, University of Minnesota, Minneapolis, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
67
|
SARS-CoV-2 Variant-Specific Infectivity and Immune Profiles Are Detectable in a Humanized Lung Mouse Model. Viruses 2022; 14:v14102272. [PMID: 36298826 PMCID: PMC9612296 DOI: 10.3390/v14102272] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/09/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022] Open
Abstract
Small animal models that accurately model pathogenesis of SARS-CoV-2 variants are required for ongoing research efforts. We modified our human immune system mouse model to support replication of SARS-CoV-2 by implantation of human lung tissue into the mice to create TKO-BLT-Lung (L) mice and compared infection with two different variants in a humanized lung model. Infection of TKO-BLT-L mice with SARS-CoV-2 recapitulated the higher infectivity of the B.1.1.7 variant with more animals becoming infected and higher sustained viral loads compared to mice challenged with an early B lineage (614D) virus. Viral lesions were observed in lung organoids but no differences were detected between the viral variants as expected. Partially overlapping but distinct immune profiles were also observed between the variants with a greater Th1 profile in VIDO-01 and greater Th2 profile in B.1.1.7 infection. Overall, the TKO-BLT-L mouse supported SARS-CoV-2 infection, recapitulated key known similarities and differences in infectivity and pathogenesis as well as revealing previously unreported differences in immune responses between the two viral variants. Thus, the TKO-BLT-L model may serve as a useful animal model to study the immunopathobiology of newly emerging variants in the context of genuine human lung tissue and immune cells.
Collapse
|
68
|
Waldmann TA, Waldmann R, Lin JX, Leonard WJ. The implications of IL-15 trans-presentation on the immune response. Adv Immunol 2022; 156:103-132. [PMID: 36410873 DOI: 10.1016/bs.ai.2022.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Interleukin-15 is a pleiotropic cytokine type I four alpha-helical bundle cytokine that along with IL-2, IL-4, IL-7, IL-9, and IL-21 shares the common cytokine receptor γ chain, γc. IL-15 is vital for the development, survival, and expansion of natural killer cells and for the development of CD8+ memory T cells. Whereas other family γc cytokines signal by directly binding to their target cells, IL-15 is distinctive in that it binds to IL-15Rα, a sushi domain containing binding protein that is expressed on a number of cell types, including monocytes and dendritic cells as well as T cells, and then is trans-presented to responding cells that express IL-2Rβ and γc. This distinctive mechanism for IL-15 relates to its role in signaling in the context of cell-cell interactions and signaling synapses. The actions of IL-15 and ways of manipulating its actions to potential therapeutic benefit are discussed.
Collapse
Affiliation(s)
- Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | | | - Jian-Xin Lin
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Warren J Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
69
|
Antosova Z, Podzimkova N, Tomala J, Augustynkova K, Sajnerova K, Nedvedova E, Sirova M, de Martynoff G, Bechard D, Moebius U, Kovar M, Spisek R, Adkins I. SOT101 induces NK cell cytotoxicity and potentiates antibody-dependent cell cytotoxicity and anti-tumor activity. Front Immunol 2022; 13:989895. [PMID: 36300122 PMCID: PMC9590108 DOI: 10.3389/fimmu.2022.989895] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
SOT101 is a superagonist fusion protein of interleukin (IL)-15 and the IL-15 receptor α (IL-15Rα) sushi+ domain, representing a promising clinical candidate for the treatment of cancer. SOT101 among other immune cells specifically stimulates natural killer (NK) cells and memory CD8+ T cells with no significant expansion or activation of the regulatory T cell compartment. In this study, we showed that SOT101 induced expression of cytotoxic receptors NKp30, DNAM-1 and NKG2D on human NK cells. SOT101 stimulated dose-dependent proliferation and the relative expansion of both major subsets of human NK cells, CD56brightCD16- and CD56dimCD16+, and these displayed an enhanced cytotoxicity in vitro. Using human PBMCs and isolated NK cells, we showed that SOT101 added concomitantly or used for immune cell pre-stimulation potentiated clinically approved monoclonal antibodies Cetuximab, Daratumumab and Obinutuzumab in killing of tumor cells in vitro. The anti-tumor efficacy of SOT101 in combination with Daratumumab was assessed in a solid multiple myeloma xenograft in CB17 SCID mouse model testing several combination schedules of administration in the early and late therapeutic setting of established tumors in vivo. SOT101 and Daratumumab monotherapies decreased with various efficacy tumor growth in vivo in dependence on the advancement of the tumor development. The combination of both drugs showed the strongest anti-tumor efficacy. Specifically, the sequencing of both drugs did not matter in the early therapeutic setting where a complete tumor regression was observed in all animals. In the late therapeutic treatment of established tumors Daratumumab followed by SOT101 administration or a concomitant administration of both drugs showed a significant anti-tumor efficacy over the respective monotherapies. These results suggest that SOT101 might significantly augment the anti-tumor activity of therapeutic antibodies by increasing NK cell-mediated activity in patients. These results support the evaluation of SOT101 in combination with Daratumumab in clinical studies and present a rationale for an optimal clinical dosing schedule selection.
Collapse
Affiliation(s)
| | - Nada Podzimkova
- Preclinical Department, SOTIO Biotech a.s, Prague, Czechia
- Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
| | - Jakub Tomala
- Laboratory of Tumor Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | | | | | - Eva Nedvedova
- Preclinical Department, SOTIO Biotech a.s, Prague, Czechia
| | - Milada Sirova
- Laboratory of Tumor Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | | | | | - Ulrich Moebius
- Preclinical Department, SOTIO Biotech a.s, Prague, Czechia
| | - Marek Kovar
- Laboratory of Tumor Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Radek Spisek
- Preclinical Department, SOTIO Biotech a.s, Prague, Czechia
- Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
| | - Irena Adkins
- Preclinical Department, SOTIO Biotech a.s, Prague, Czechia
- Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
- *Correspondence: Irena Adkins,
| |
Collapse
|
70
|
Bottino C, Della Chiesa M, Sorrentino S, Morini M, Vitale C, Dondero A, Tondo A, Conte M, Garaventa A, Castriconi R. Strategies for Potentiating NK-Mediated Neuroblastoma Surveillance in Autologous or HLA-Haploidentical Hematopoietic Stem Cell Transplants. Cancers (Basel) 2022; 14:cancers14194548. [PMID: 36230485 PMCID: PMC9559312 DOI: 10.3390/cancers14194548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary High-risk neuroblastomas (HR-NB) are malignant tumors of childhood that are treated with a very aggressive and life-threatening approach; this includes autologous hemopoietic stem cell transplantation (HSCT) and the infusion of a mAb targeting the GD2 tumor-associated antigen. Although the current treatment provided benefits, the 5-year overall survival remains below 50% due to relapses and refractoriness to therapy. Thus, there is an urgent need to ameliorate the standard therapeutic protocol, particularly improving the immune-mediated anti-tumor responses. Our review aims at summarizing and critically discussing novel immunotherapeutic strategies in HR-NB, including NK cell-based therapies and HLA-haploidentical HSCT from patients’ family. Abstract High-risk neuroblastomas (HR-NB) still have an unacceptable 5-year overall survival despite the aggressive therapy. This includes standardized immunotherapy combining autologous hemopoietic stem cell transplantation (HSCT) and the anti-GD2 mAb. The treatment did not significantly change for more than one decade, apart from the abandonment of IL-2, which demonstrated unacceptable toxicity. Of note, immunotherapy is a promising therapeutic option in cancer and could be optimized by several strategies. These include the HLA-haploidentical αβT/B-depleted HSCT, and the antibody targeting of novel NB-associated antigens such as B7-H3, and PD1. Other approaches could limit the immunoregulatory role of tumor-derived exosomes and potentiate the low antibody-dependent cell cytotoxicity of CD16 dim/neg NK cells, abundant in the early phase post-transplant. The latter effect could be obtained using multi-specific tools engaging activating NK receptors and tumor antigens, and possibly holding immunostimulatory cytokines in their construct. Finally, treatments also consider the infusion of novel engineered cytokines with scarce side effects, and cell effectors engineered with chimeric antigen receptors (CARs). Our review aims to discuss several promising strategies that could be successfully exploited to potentiate the NK-mediated surveillance of neuroblastoma, particularly in the HSCT setting. Many of these approaches are safe, feasible, and effective at pre-clinical and clinical levels.
Collapse
Affiliation(s)
- Cristina Bottino
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
- Laboratory of Clinical and Experimental Immunology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
- Correspondence: ; Tel.: +39-01056363855
| | - Mariella Della Chiesa
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| | | | - Martina Morini
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Chiara Vitale
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| | - Alessandra Dondero
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| | - Annalisa Tondo
- Department of Pediatric Hematology/Oncology and HSCT, Meyer Children’s University Hospital, 50139 Florence, Italy
| | - Massimo Conte
- Pediatric Oncology Unit-IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Alberto Garaventa
- Pediatric Oncology Unit-IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Roberta Castriconi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| |
Collapse
|
71
|
Aryee K, Burzenski LM, Yao L, Keck JG, Greiner D, Shultz LD, Brehm MA. Enhanced development of functional human NK cells in NOD-scid-IL2rg null mice expressing human IL15. FASEB J 2022; 36:e22476. [PMID: 35959876 PMCID: PMC9383543 DOI: 10.1096/fj.202200045r] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 07/08/2022] [Accepted: 07/19/2022] [Indexed: 01/09/2023]
Abstract
Human innate immunity plays a critical role in tumor surveillance and in immunoregulation within the tumor microenvironment. Natural killer (NK) cells are innate lymphoid cells that have opposing roles in the tumor microenvironment, including NK cell subsets that mediate tumor cell cytotoxicity and subsets with regulatory function that contribute to the tumor immune suppressive environment. The balance between effector and regulatory NK cell subsets has been studied extensively in murine models of cancer, but there is a paucity of models to study human NK cell function in tumorigenesis. Humanized mice are a powerful alternative to syngeneic mouse tumor models for the study of human immuno-oncology and have proven effective tools to test immunotherapies targeting T cells. However, human NK cell development and survival in humanized NOD-scid-IL2rgnull (NSG) mice are severely limited. To enhance NK cell development, we have developed NSG mice that constitutively expresses human Interleukin 15 (IL15), NSG-Tg(Hu-IL15). Following hematopoietic stem cell engraftment of NSG-Tg(Hu-IL15) mice, significantly higher levels of functional human CD56+ NK cells are detectable in blood and spleen, as compared to NSG mice. Hematopoietic stem cell (HSC)-engrafted NSG-Tg(Hu-IL15) mice also supported the development of human CD3+ T cells, CD20+ B cells, and CD33+ myeloid cells. Moreover, the growth kinetics of a patient-derived xenograft (PDX) melanoma were significantly delayed in HSC-engrafted NSG-Tg(Hu-IL15) mice as compared to HSC-engrafted NSG mice demonstrating that human NK cells have a key role in limiting the tumor growth. Together, these data demonstrate that HSC-engrafted NSG-Tg(Hu-IL15) mice support enhanced development of functional human NK cells, which limit the growth of PDX tumors.
Collapse
Affiliation(s)
- Ken‐Edwin Aryee
- Program in Molecular MedicineDiabetes Center of Excellence, University of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | | | - Li‐Chin Yao
- The Jackson LaboratorySacramentoCaliforniaUSA
| | | | - Dale L. Greiner
- Program in Molecular MedicineDiabetes Center of Excellence, University of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | | | - Michael A. Brehm
- Program in Molecular MedicineDiabetes Center of Excellence, University of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| |
Collapse
|
72
|
Stem Cell Therapy and Innate Lymphoid Cells. Stem Cells Int 2022; 2022:3530520. [PMID: 35958032 PMCID: PMC9363162 DOI: 10.1155/2022/3530520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 11/17/2022] Open
Abstract
Innate lymphoid cells have the capability to communicate with other immune cell types to coordinate the immune system functioning during homeostasis and inflammation. However, these cells behave differently at the functional level, unlike T cells, these cells do not need antigen receptors for activation because they are activated by the interaction of their receptor ligation. In hematopoietic stem cell transplantation (HSCT), T cells and NK cells have been extensively studied but very few studies are available on ILCs. In this review, an attempt has been made to provide current information related to NK and ILCs cell-based stem cell therapies and role of the stem cells in the regulation of ILCs as well. Also, the latest information on the differentiation of NK cells and ILCs from CD34+ hematopoietic stem cells is covered in the article.
Collapse
|
73
|
Interleukin 15 in Cell-Based Cancer Immunotherapy. Int J Mol Sci 2022; 23:ijms23137311. [PMID: 35806311 PMCID: PMC9266896 DOI: 10.3390/ijms23137311] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 02/01/2023] Open
Abstract
Cell-based cancer immunotherapy, such as chimeric antigen receptor (CAR) engineered T and natural killer (NK) cell therapies, has become a revolutionary new pillar in cancer treatment. Interleukin 15 (IL-15), a potent immunostimulatory cytokine that potentiates T and NK cell immune responses, has demonstrated the reliability and potency to potentially improve the therapeutic efficacy of current cell therapy. Structurally similar to interleukin 2 (IL-2), IL-15 supports the persistence of CD8+ memory T cells while inhibiting IL-2-induced T cell death that better maintains long-term anti-tumor immunity. In this review, we describe the biology of IL-15, studies on administrating IL-15 and/or its derivatives as immunotherapeutic agents, and IL-15-armored immune cells in adoptive cell therapy. We also discuss the advantages and challenges of incorporating IL-15 in cell-based immunotherapy and provide directions for future investigation.
Collapse
|
74
|
Yang S, Liu Q, Yang J, Wu J, Wang S. Increased Levels of Serum IL-15 and TNF-β Indicate the Progression of Human Intracranial Aneurysm. Front Aging Neurosci 2022; 14:903619. [PMID: 35783134 PMCID: PMC9247574 DOI: 10.3389/fnagi.2022.903619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/31/2022] [Indexed: 01/07/2023] Open
Abstract
Objective Existing evidence suggests that chronic inflammation promotes the progression of human intracranial aneurysm (IA) and many cytokines have been detected to participate in the process of inflammation. However, rare cytokines in plasma have been used as proxies for progression of IA. This study aimed to identify novel cytokines as biomarkers to predict the development of IA. Methods Patients with unruptured intracranial aneurysms (UIAs) undergoing microsurgical clipping were prospectively recruited from January 2017 to June 2020 and were separated into two groups based on their ELAPSS score (low risk group < 10, intermediate-high risk group ≥ 10). Propensity score matching (PSM) was used to reduce imbalances in the baseline characteristics between groups. All blood samples were collected before surgery. A human serum 48-cytokines examination was performed to analyze the concentrations of serological cytokines. Clinical data and cytokines were compared between groups. Results A total of 184 patients were enrolled in this study. The low risk group contained 77 patients and 107 patients were included in the intermediate-high risk group. Finally, there were 69 patients in each group after PSM with a matching rate of 1:1. The concentrations of 3 serum cytokines were significantly increased in intermediate-high risk patients, namely, interleukin-15 (IL-15), monocyte chemoattractant protein-1 (MCP-1), and tumor necrosis factor-β (TNF-β) (P < 0.05, |log2 fold change| > 2). The result of receiver operator characteristic (ROC)curve revealed that TNF-β had the highest predictive accuracy, with an area under the curve (AUC) value of 0.725 [95% confidence interval (CI) 0.639–0.811, P < 0.001] followed by IL-15 (AUC = 0.691, 95% CI 0.602–0.781, P < 0.001) and MCP-1 (AUC = 0.661, 95% CI 0.569–0.753, P = 0.001). Multivariate logistic analysis demonstrated high IL-15 [odds ratio (OR), 3.23; 95% CI, 1.47–7.12; P = 0.004] and high TNF-β (OR, 8.30; 95% CI, 3.25–21.25; P < 0.001) as the risk factors that correlated with intermediate-high risk of IA progression. Conclusion UIA patients with intermediate-high growth risk exhibited increased serum levels of IL-15, MCP-1, and TNF-β. Serum IL-15, and TNF-β could serve as biomarkers to predict the progression of UIAs.
Collapse
Affiliation(s)
- Shuzhe Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Qingyuan Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Junhua Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Jun Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Shuo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
- *Correspondence: Shuo Wang,
| |
Collapse
|
75
|
Tan S, Fang M, Fan W, Wang Z, Lv Y, Zou J, Wang X, Liu B, Yang YG, Hu Z. Improvement of human myeloid and natural killer cell development in humanized mice via hydrodynamic injection of transposon plasmids containing multiple human cytokine genes. Immunol Cell Biol 2022; 100:624-635. [PMID: 35662247 DOI: 10.1111/imcb.12564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/12/2021] [Accepted: 06/03/2022] [Indexed: 11/29/2022]
Abstract
Humanized mice reconstituted with a functional human immune system (HIS) are instrumental in studying human immunity and immune disorders in vivo. The poor or lack of cross-reactivity between mouse cytokines and human cells limits the development and/or function of human immune cell subsets including human myeloid, natural killer and B cells. Here we explored the potential to achieve long-term production of human cytokines in immunodeficient mice using a transposon-plasmid-based hydrodynamic injection approach. We constructed a transposon-plasmid carrying five human cytokine coding sequences (named PB-5F), and observed that four of the cytokines (granulocyte-macrophage colony-stimulating factor, interleukin (IL)-15, IL-6 and IL-3) were detectable in sera and three (granulocyte-macrophage colony-stimulating factor, IL-15 and IL-6) showed long-term production in immunodeficient mice that received a single hydrodynamic injection of PB-5F plus the transposase plasmid (Super PB). Furthermore, a single injection of PB-5F/Super PB markedly enhanced the reconstitution of human myeloid cells and natural killer cells, and promoted human B-cell maturation in HIS mice. Taken together, our data revealed that hydrodynamic injection of the PB-5F/Super PB vectors may serve as a convenient and efficacious means to promote human immune function in HIS mice.
Collapse
Affiliation(s)
- Shulian Tan
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
| | - Minghui Fang
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
| | - Wei Fan
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Zhaowei Wang
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Yanan Lv
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Jun Zou
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Xue Wang
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
| | - Bin Liu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China.,International Center of Future Science, Jilin University, Changchun, China
| | - Zheng Hu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
76
|
Alexandrova M, Manchorova D, Dimova T. Immunity at maternal-fetal interface: KIR/HLA (Allo)recognition. Immunol Rev 2022; 308:55-76. [PMID: 35610960 DOI: 10.1111/imr.13087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/28/2022] [Accepted: 05/09/2022] [Indexed: 12/15/2022]
Abstract
Both KIR and HLA are the most variable gene families in the human genome. The recognition of the semi-allogeneic embryo-derived trophoblasts by maternal decidual NK (dNK) cells is essential for the establishment of the functional placenta. This recognition is based on the KIR-HLA interactions and trophoblast expresses a specific HLA profile that constitutes classical polymorphic HLA-C and non-classical oligomorphic HLA-E, HLA-F, and HLA-G molecules. This review highlights some features of the KIR/HLA-C (allo)recognition by decidual NK (dNK) cells as a main immune cell population specifically enriched at maternal-fetal interface during human early pregnancy. How KIR/HLA-C axis operates in pregnancy disorders and in the context of transplacental infections is discussed as well. We summarized old and new data on dNK-cell functional plasticity, their selective expression of KIR and fetal maternal/paternal HLA-C haplotypes present. Results showed that KIR-HLA-C combinations and the corresponding axis operate differently in each pregnancy, determined by the variability of both maternal KIR haplotypes and fetus' maternal/paternal HLA-C allotype combinations. Moreover, the maturation of NK cells strongly depends on if or not HLA allotypes for certain KIR are present. We suggest that the unique KIR/HLA combinations reached in each pregnancy (normal and pathological) should be studied according to well-defined guidelines and unified methodologies to have comparable results ease to interpret and use in clinics.
Collapse
Affiliation(s)
- Marina Alexandrova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Diana Manchorova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Tanya Dimova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
77
|
Caveolin-1 temporal modulation enhances antibody drug efficacy in heterogeneous gastric cancer. Nat Commun 2022; 13:2526. [PMID: 35534471 PMCID: PMC9085816 DOI: 10.1038/s41467-022-30142-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 04/19/2022] [Indexed: 11/11/2022] Open
Abstract
Resistance mechanisms and heterogeneity in HER2-positive gastric cancers (GC) limit Trastuzumab benefit in 32% of patients, and other targeted therapies have failed in clinical trials. Using patient samples, patient-derived xenografts (PDXs), partially humanized biological models, and HER2-targeted imaging technologies we demonstrate the role of caveolin-1 (CAV1) as a complementary biomarker in GC selection for Trastuzumab therapy. In retrospective analyses of samples from patients enrolled on Trastuzumab trials, the CAV1-high profile associates with low membrane HER2 density and low patient survival. We show a negative correlation between CAV1 tumoral protein levels – a major protein of cholesterol-rich membrane domains – and Trastuzumab-drug conjugate TDM1 tumor uptake. Finally, CAV1 depletion using knockdown or pharmacologic approaches (statins) increases antibody drug efficacy in tumors with incomplete HER2 membranous reactivity. In support of these findings, background statin use in patients associates with enhanced antibody efficacy. Together, this work provides preclinical justification and clinical evidence that require prospective investigation of antibody drugs combined with statins to delay drug resistance in tumors. Clinical evidences have demonstrated limited efficacy of HER2-targeted therapies in patients with gastric cancer (GC). Here the authors show that survival benefit to anti-HER2 antibody Trastuzumab is reduced in GC patients with high levels of the caveolin-1 and that, in preclinical cancer models, antibody drug efficacy can be improved by modulating caveolin-1 levels with cholesterol-depleting drugs, statins.
Collapse
|
78
|
Elmas E, Saljoughian N, de Souza Fernandes Pereira M, Tullius BP, Sorathia K, Nakkula RJ, Lee DA, Naeimi Kararoudi M. CRISPR Gene Editing of Human Primary NK and T Cells for Cancer Immunotherapy. Front Oncol 2022; 12:834002. [PMID: 35449580 PMCID: PMC9016158 DOI: 10.3389/fonc.2022.834002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Antitumor activity of immune cells such as T cells and NK cells has made them auspicious therapeutic regimens for adaptive cancer immunotherapy. Enhancing their cytotoxic effects against malignancies and overcoming their suppression in tumor microenvironment (TME) may improve their efficacy to treat cancers. Clustered, regularly interspaced short palindromic repeats (CRISPR) genome editing has become one of the most popular tools to enhance immune cell antitumor activity. In this review we highlight applications and practicability of CRISPR/Cas9 gene editing and engineering strategies for cancer immunotherapy. In addition, we have reviewed several approaches to study CRISPR off-target effects.
Collapse
Affiliation(s)
- Ezgi Elmas
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, United States
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Noushin Saljoughian
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- CRISPR/Gene Editing Core, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Marcelo de Souza Fernandes Pereira
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Brian P. Tullius
- Pediatric Cellular Therapy, AdventHealth for Children, Orlando, FL, United States
| | - Kinnari Sorathia
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Robin J. Nakkula
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Dean A. Lee
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| | - Meisam Naeimi Kararoudi
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- CRISPR/Gene Editing Core, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
79
|
Experimental models for ovarian cancer research. Exp Cell Res 2022; 416:113150. [DOI: 10.1016/j.yexcr.2022.113150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/30/2022] [Accepted: 04/05/2022] [Indexed: 11/23/2022]
|
80
|
Goldenson BH, Hor P, Kaufman DS. iPSC-Derived Natural Killer Cell Therapies - Expansion and Targeting. Front Immunol 2022; 13:841107. [PMID: 35185932 PMCID: PMC8851389 DOI: 10.3389/fimmu.2022.841107] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/13/2022] [Indexed: 12/15/2022] Open
Abstract
Treatment of cancer with allogeneic natural killer (NK) cell therapies has seen rapid development, especially use against hematologic malignancies. Clinical trials of NK cell-based adoptive transfer to treat relapsed or refractory malignancies have used peripheral blood, umbilical cord blood and pluripotent stem cell-derived NK cells, with each approach undergoing continued clinical development. Improving the potency of these therapies relies on genetic modifications to improve tumor targeting and to enhance expansion and persistence of the NK cells. Induced pluripotent stem cell (iPSC)-derived NK cells allow for routine targeted introduction of genetic modifications and expansion of the resulting NK cells derived from a clonal starting cell population. In this review, we discuss and summarize recent important advances in the development of new iPSC-derived NK cell therapies, with a focus on improved targeting of cancer. We then discuss improvements in methods to expand iPSC-derived NK cells and how persistence of iPSC-NK cells can be enhanced. Finally, we describe how these advances may combine in future NK cell-based therapy products for the treatment of both hematologic malignancies and solid tumors.
Collapse
Affiliation(s)
- Benjamin H Goldenson
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Pooja Hor
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Dan S Kaufman
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
81
|
Preservation of lymphocyte functional fitness in perinatally-infected and treated HIV+ pediatric patients displaying sub-optimal viral control. COMMUNICATIONS MEDICINE 2022; 2. [PMID: 35434722 PMCID: PMC9012494 DOI: 10.1038/s43856-022-00085-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Abstract
Background
Host–pathogen dynamics associated with HIV infection are quite distinct in children versus adults. We interrogated the functional fitness of the lymphocyte responses in two cohorts of perinatally infected HIV+ pediatric subjects with early anti-retroviral therapy (ART) initiation but divergent patterns of virologic control. We hypothesized that sub-optimal viral control would compromise immune functional fitness.
Methods
The immune responses in the two HIV+ cohorts (n = 6 in each cohort) were benchmarked against the responses measured in age-range matched, uninfected healthy control subjects (n = 11) by utilizing tests for normality, and comparison [the Kruskal–Wallis test, and the two-tailed Mann–Whitney U test (where appropriate)]. Lymphocyte responses were examined by intra-cellular cytokine secretion, degranulation assays as well as phosflow. A subset of these data were further queried by an automated clustering algorithm. Finally, we evaluated the humoral immune responses to four childhood vaccines in all three cohorts.
Results
We demonstrate that contrary to expectations pediatric HIV+ patients with sub-optimal viral control display no significant deficits in immune functional fitness. In fact, the patients that display better virologic control lack functional Gag-specific T cell responses and compared to healthy controls they display signaling deficits and an enrichment of mitogen-stimulated CD3 negative and positive lymphocyte clusters with suppressed cytokine production.
Conclusions
These results highlight the immune resilience in HIV+ children on ART with sub-optimal viral control. With respect to HIV+ children on ART with better viral control, our data suggest that this cohort might potentially benefit from targeted interventions that might mitigate cell-mediated immune functional quiescence.
Collapse
|
82
|
Mortlock RD, Wu C, Potter EL, Abraham DM, Allan DSJ, Hong SG, Roederer M, Dunbar CE. Tissue Trafficking Kinetics of Rhesus Macaque Natural Killer Cells Measured by Serial Intravascular Staining. Front Immunol 2022; 12:772332. [PMID: 35095846 PMCID: PMC8790741 DOI: 10.3389/fimmu.2021.772332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/16/2021] [Indexed: 11/13/2022] Open
Abstract
The in vivo tissue distribution and trafficking patterns of natural killer (NK) cells remain understudied. Animal models can help bridge the gap, and rhesus macaque (RM) primates faithfully recapitulate key elements of human NK cell biology. Here, we profiled the tissue distribution and localization patterns of three NK cell subsets across various RM tissues. We utilized serial intravascular staining (SIVS) to investigate the tissue trafficking kinetics at steady state and during recovery from CD16 depletion. We found that at steady state, CD16+ NK cells were selectively retained in the vasculature while CD56+ NK cells had a shorter residence time in peripheral blood. We also found that different subsets of NK cells had distinct trafficking kinetics to and from the lymph node as well as other lymphoid and non-lymphoid tissues. Lastly, we found that following administration of CD16-depleting antibody, CD16+ NK cells and their putative precursors retained a high proportion of continuously circulating cells, suggesting that regeneration of the CD16 NK compartment may take place in peripheral blood or the perivascular compartments of tissues.
Collapse
Affiliation(s)
- Ryland D Mortlock
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Chuanfeng Wu
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - E Lake Potter
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Diana M Abraham
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - David S J Allan
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - So Gun Hong
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Cynthia E Dunbar
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
83
|
Ju C, Liang J, Zhang M, Zhao J, Li L, Chen S, Zhao J, Gao X. The mouse resource at National Resource Center for Mutant Mice. Mamm Genome 2022; 33:143-156. [PMID: 35138443 DOI: 10.1007/s00335-021-09940-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 12/10/2021] [Indexed: 10/19/2022]
Abstract
Mouse models are essential for dissecting disease mechanisms and defining potential drug targets. There are more than 18,500 mouse strains available for research communities in National Resource Center for Mutant Mice (NRCMM) of China, affiliated with Model Animal Research Center of Nanjing University and Gempharmatech Company. In 2019, Gempharmatech launched the Knockout All Project (KOAP) aiming to generate null mutants and gene floxed strains for all protein-coding genes in mouse genome within 5 years. So far, KOAP has generated 8,004 floxed strains and 9,769 KO (knockout) strains (updated to Oct, 2021). NRCMM also created hundreds of Cre transgenic lines, mutant knock-in models, immuno-deficient models, and humanized mouse models. As a member of the international mouse phenotyping consortium (IMPC), NRCMM provides comprehensive phenotyping services for mouse models. In summary, NRCMM will continue to support biomedical community with new mouse models as well as related services.
Collapse
Affiliation(s)
| | | | | | | | | | - Shuai Chen
- Model Animal Research Center of Nanjing University, Nanjing, China.,Nanjing Biomedical Research Institute of Nanjing University, Nanjing, China
| | - Jing Zhao
- GemPharmatech Co., Ltd, Nanjing, China.
| | - Xiang Gao
- National Resource Center for Mutant Mice, Nanjing, China. .,GemPharmatech Co., Ltd, Nanjing, China. .,Model Animal Research Center of Nanjing University, Nanjing, China.
| |
Collapse
|
84
|
The use of supercytokines, immunocytokines, engager cytokines, and other synthetic cytokines in immunotherapy. Cell Mol Immunol 2022; 19:192-209. [PMID: 35043005 PMCID: PMC8803834 DOI: 10.1038/s41423-021-00786-6] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/25/2021] [Indexed: 02/08/2023] Open
Abstract
Cytokines exert powerful immunomodulatory effects that are critical to physiology and pathology in humans. The application of natural cytokines in clinical studies has not been clearly established, and there are often problems associated with toxicity or lack of efficacy. The key reasons can be attributed to the pleiotropy of cytokine receptors and undesired activation of off-target cells. With a deeper understanding of the structural principles and functional signals of cytokine-receptor interactions, artificial modification of cytokine signaling through protein engineering and synthetic immunology has become an increasingly feasible and powerful approach. Engineered cytokines are designed to selectively target cells. Herein, the theoretical and experimental evidence of cytokine engineering is reviewed, and the "supercytokines" resulting from structural enhancement and the "immunocytokines" generated by antibody fusion are described. Finally, the "engager cytokines" formed by the crosslinking of cytokines and bispecific immune engagers and other synthetic cytokines formed by nonnatural analogs are also discussed.
Collapse
|
85
|
Wang R, Bao W, Pal M, Liu Y, Yazdanbakhsh K, Zhong H. Intermediate monocytes induced by IFN-γ inhibit cancer metastasis by promoting NK cell activation through FOXO1 and interleukin-27. J Immunother Cancer 2022; 10:jitc-2021-003539. [PMID: 35091454 PMCID: PMC8804695 DOI: 10.1136/jitc-2021-003539] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2021] [Indexed: 11/05/2022] Open
Abstract
Background Circulating monocytes are functionally heterogeneous and can be divided into classical (CMo), intermediate (IMo), and non-CMo/patrolling monocyte (PMo) subsets. CMo can differentiate into PMo through IMo. PMos have been shown to inhibit cancer metastasis but the role of IMo is unclear. To date, no strategy has been developed to inhibit cancer metastasis through enhancing PMo/IMo differentiation. Methods We screened multiple inflammatory cytokines/chemokines activity of modulating PMo/IMo associated cell markers expression using human monocyte in vitro culture system. We tested our candidate cytokine activity in vivo using multiple mice models. We identified critical key factors and cytokines for our candidate cytokine activity by using gene-knockout mice and neutralization antibodies. Results We identified IFN-γ as a candidate inflammatory cytokine in the regulation of human IMo/PMo marker expression. Our in vivo data demonstrated that IMo expansion was induced by short-term (3 days) IFN-γ treatment through increasing CMo-IMo differentiation and blocking IMo-PMo differentiation. The IMo induced by IFN-γ (IFN-IMo), but not IFN-γ activated CMo (IFN-CMo), inhibited cancer metastasis by 90%. Surprizing, the effect of IFN-γ is greater in PMo deficiency mice, indicating the effect of IFN-IMo is not mediated through further differentiation into PMo. We also found that IFN-IMos induced by short-term IFN-γ treatment robustly boosted NK cell expansion for threefold and promoted NK differentiation and function through IL-27 and CXCL9. Furthermore, we identified that FOXO1, a key molecule controlling cellular energy metabolism, mediated the effect of IFN-γ induced IL-27 expression, and that NR4A1, a key molecule controlling PMo differentiation and inhibiting cancer metastasis, inhibited the pro-NK cell and anti-metastasis activity of IFN-IMo by suppressing CXCL9 expression. Conclusions We have discovered the antimetastasis and pro-NK cell activity of IFN-IMo, identified FOXO1 as a key molecule for IFN-γ driven monocyte differentiation and function, and found NR4A1 as an inhibitory molecule for IFN-IMo activity. Our study has not only shown novel mechanisms for a classical antitumor cytokine but also provided potential target for developing superior monocytic cell therapy against cancer metastasis.
Collapse
Affiliation(s)
- Rikang Wang
- Laboratory of Immune Regulation, New York Blood Center, New York, New York, USA
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Weili Bao
- Laboratory of Complement Biology, New York Blood Center, New York, New York, USA
| | - Mouli Pal
- Laboratory of Complement Biology, New York Blood Center, New York, New York, USA
| | - Yunfeng Liu
- Laboratory of Complement Biology, New York Blood Center, New York, New York, USA
| | - Karina Yazdanbakhsh
- Laboratory of Complement Biology, New York Blood Center, New York, New York, USA
| | - Hui Zhong
- Laboratory of Immune Regulation, New York Blood Center, New York, New York, USA
| |
Collapse
|
86
|
Moquin-Beaudry G, Benabdallah B, Maggiorani D, Le O, Li Y, Colas C, Raggi C, Ellezam B, M'Callum MA, Dal Soglio D, Guimond JV, Paganelli M, Haddad E, Beauséjour C. Autologous humanized mouse models of iPSC-derived tumors enable characterization and modulation of cancer-immune cell interactions. CELL REPORTS METHODS 2022; 2:100153. [PMID: 35474871 PMCID: PMC9017190 DOI: 10.1016/j.crmeth.2021.100153] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/03/2021] [Accepted: 12/21/2021] [Indexed: 01/21/2023]
Abstract
Modeling the tumor-immune cell interactions in humanized mice is complex and limits drug development. Here, we generated easily accessible tumor models by transforming either primary skin fibroblasts or induced pluripotent stem cell-derived cell lines injected in immune-deficient mice reconstituted with human autologous immune cells. Our results showed that fibroblastic, hepatic, or neural tumors were all efficiently infiltrated and partially or totally rejected by autologous immune cells in humanized mice. Characterization of tumor-immune infiltrates revealed high expression levels of the dysfunction markers Tim3 and PD-1 in T cells and an enrichment in regulatory T cells, suggesting rapid establishment of immunomodulatory phenotypes. Inhibition of PD-1 by Nivolumab in humanized mice resulted in increased immune cell infiltration and a slight decrease in tumor growth. We expect that these versatile and accessible cancer models will facilitate preclinical studies and the evaluation of autologous cancer immunotherapies across a range of different tumor cell types.
Collapse
Affiliation(s)
- Gaël Moquin-Beaudry
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Pharmacologie et Physiologie, Montréal, QC, Canada
| | - Basma Benabdallah
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Damien Maggiorani
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Oanh Le
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Yuanyi Li
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Chloé Colas
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Microbiologie, Immunologie et Infectiologie, Montréal, QC, Canada
| | - Claudia Raggi
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Benjamin Ellezam
- Département de Neurosciences, Montréal, QC, Canada
- Département de Pathologie, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Marie-Agnès M'Callum
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Biologie Moléculaire, Montréal, QC, Canada
| | - Dorothée Dal Soglio
- Département de Pathologie et Biologie Moléculaire, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
- Département de Pathologie, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Jean V. Guimond
- CIUSSS du Centre-Sud-de-l’Ile-de-Montréal, Montréal, QC, Canada
| | - Massimiliano Paganelli
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Biologie Moléculaire, Montréal, QC, Canada
- Division of Gastroenterology, Hepatology and Nutrition and Pediatric Liver Transplantation Program at CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
- Département de Pédiatrie, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Elie Haddad
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Microbiologie, Immunologie et Infectiologie, Montréal, QC, Canada
- Département de Pédiatrie, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Christian Beauséjour
- Centre de Recherche du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montréal, QC H3T 1C5, Canada
- Département de Pharmacologie et Physiologie, Montréal, QC, Canada
| |
Collapse
|
87
|
Kim JT, Zhang TH, Carmona C, Lee B, Seet CS, Kostelny M, Shah N, Chen H, Farrell K, Soliman MSA, Dimapasoc M, Sinani M, Blanco KYR, Bojorquez D, Jiang H, Shi Y, Du Y, Komarova NL, Wodarz D, Wender PA, Marsden MD, Sun R, Zack JA. Latency reversal plus natural killer cells diminish HIV reservoir in vivo. Nat Commun 2022; 13:121. [PMID: 35013215 PMCID: PMC8748509 DOI: 10.1038/s41467-021-27647-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/03/2021] [Indexed: 02/06/2023] Open
Abstract
HIV is difficult to eradicate due to the persistence of a long-lived reservoir of latently infected cells. Previous studies have shown that natural killer cells are important to inhibiting HIV infection, but it is unclear whether the administration of natural killer cells can reduce rebound viremia when anti-retroviral therapy is discontinued. Here we show the administration of allogeneic human peripheral blood natural killer cells delays viral rebound following interruption of anti-retroviral therapy in humanized mice infected with HIV-1. Utilizing genetically barcoded virus technology, we show these natural killer cells efficiently reduced viral clones rebounding from latency. Moreover, a kick and kill strategy comprised of the protein kinase C modulator and latency reversing agent SUW133 and allogeneic human peripheral blood natural killer cells during anti-retroviral therapy eliminated the viral reservoir in a subset of mice. Therefore, combinations utilizing latency reversal agents with targeted cellular killing agents may be an effective approach to eradicating the viral reservoir.
Collapse
Affiliation(s)
- Jocelyn T Kim
- Department of Medicine, Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Tian-Hao Zhang
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Camille Carmona
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Bryanna Lee
- Department of Medicine, Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Christopher S Seet
- Department of Medicine, Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Matthew Kostelny
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Nisarg Shah
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Hongying Chen
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Kylie Farrell
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Mohamed S A Soliman
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Melanie Dimapasoc
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Michelle Sinani
- Department of Medicine, Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Kenia Yazmin Reyna Blanco
- Department of Medicine, Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - David Bojorquez
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Hong Jiang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Yuan Shi
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Yushen Du
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Natalia L Komarova
- Department of Mathematics, University of California, Irvine, Irvine, CA, 92697, USA
| | - Dominik Wodarz
- Department of Population Health and Disease Prevention, Program in Public Health Susan and Henry Samueli College of Health Sciences, University of California, Irvine, Irvine, CA, 92697, USA
| | - Paul A Wender
- Department of Chemistry and Department of Chemical and Systems Biology, Stanford University, Stanford, CA, 94305, USA
| | - Matthew D Marsden
- Department of Microbiology and Molecular Genetics and Department of Medicine, Division of Infectious Diseases, School of Medicine, University of California, Irvine, Irvine, CA, 92697, USA
| | - Ren Sun
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA.,School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jerome A Zack
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, 90095, USA.,Department of Medicine, Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
88
|
Current Status, Barriers, and Future Directions for Humanized Mouse Models to Evaluate Stem Cell–Based Islet Cell Transplant. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1387:89-106. [DOI: 10.1007/5584_2022_711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
89
|
Kenesei Á, Volkó J, Szalóki N, Mocsár G, Jambrovics K, Balajthy Z, Bodnár A, Tóth K, Waldmann TA, Vámosi G. IL-15 Trans-Presentation Is an Autonomous, Antigen-Independent Process. THE JOURNAL OF IMMUNOLOGY 2021; 207:2489-2500. [PMID: 34654688 DOI: 10.4049/jimmunol.2100277] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 09/10/2021] [Indexed: 11/19/2022]
Abstract
IL-15 plays a pivotal role in the long-term survival of T cells and immunological memory. Its receptor consists of three subunits (IL-15Rα, IL-2/15Rβ, and γc). IL-15 functions mainly via trans-presentation (TP), during which an APC expressing IL-15 bound to IL-15Rα presents the ligand to the βγc receptor-heterodimer on a neighboring T/NK cell. To date, no direct biophysical evidence for the intercellular assembly of the IL-15R heterotrimer exists. Ag presentation (AP), the initial step of T cell activation, is also based on APC-T cell interaction. We were compelled to ask whether AP has any effect on IL-15 TP or whether they are independent processes. In our human Raji B cell-Jurkat T cell model system, we monitored inter-/intracellular protein interactions upon formation of IL-15 TP and AP receptor complexes by Förster resonance energy transfer measurements. We detected enrichment of IL-15Rα and IL-2/15Rβ at the synapse and positive Förster resonance energy transfer efficiency if Raji cells were pretreated with IL-15, giving direct biophysical evidence for IL-15 TP. IL-15Rα and MHC class II interacted and translocated jointly to the immunological synapse when either ligand was present, whereas IL-2/15Rβ and CD3 moved independently of each other. IL-15 TP initiated STAT5 phosphorylation in Jurkat cells, which was not further enhanced by AP. Conversely, IL-15 treatment slightly attenuated Ag-induced phosphorylation of the CD3ζ chain. Our studies prove that in our model system, IL-15 TP and AP can occur independently, and although AP enhances IL-15R assembly, it has no significant effect on IL-15 signaling during TP. Thus, IL-15 TP can be considered an autonomous, Ag-independent process.
Collapse
Affiliation(s)
- Ádám Kenesei
- Department of Biophysics and Cell Biology, Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Julianna Volkó
- Department of Biophysics and Cell Biology, Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nikoletta Szalóki
- Department of Biophysics and Cell Biology, Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Mocsár
- Department of Biophysics and Cell Biology, Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Károly Jambrovics
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Balajthy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Andrea Bodnár
- Department of Biophysics and Cell Biology, Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Katalin Tóth
- Division of Biophysics of Macromolecules, German Cancer Research Center, Heidelberg, Germany; and
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - György Vámosi
- Department of Biophysics and Cell Biology, Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary;
| |
Collapse
|
90
|
Orrantia A, Terrén I, Astarloa-Pando G, González C, Uranga A, Mateos-Mazón JJ, García-Ruiz JC, Riñón M, Rey M, Pérez-Fernandez S, Zenarruzabeitia O, Borrego F. NK Cell Reconstitution After Autologous Hematopoietic Stem Cell Transplantation: Association Between NK Cell Maturation Stage and Outcome in Multiple Myeloma. Front Immunol 2021; 12:748207. [PMID: 34675932 PMCID: PMC8524090 DOI: 10.3389/fimmu.2021.748207] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/16/2021] [Indexed: 02/02/2023] Open
Abstract
Autologous hematopoietic stem cell transplantation (autoHSCT) is a standard of care for transplant-eligible patients with multiple myeloma (MM). Among factors that influence outcome after autoHSCT, it has been suggested that the number of natural killer (NK) cells plays an important role. However, the impact that different NK cell subsets and their phenotype could have in disease progression after autoHSCT are less clear. For this reason, we have phenotypically and functionally characterized NK cells during immune system reconstitution after autoHSCT in 54 MM patients. Shortly after leukocyte recovery, an extensive redistribution of NK cell subsets occurs in these patients. In addition, NK cells undergo a profound phenotypic change characterized, among others, by their increased proliferative capacity and immature phenotype. Importantly, MM patients who showed lower frequencies of the mature highly differentiated NKG2A-CD57+ NK cell subset at +30 and +100 days after autoHSCT experienced superior progression-free survival and had a longer time to the next treatment than those with higher frequencies. Our results provide significant insights into NK cell reconstitution after autoHSCT and suggest that the degree of NK cell maturation after autoHSCT affects the clinical outcome of MM patients treated with this therapeutic strategy.
Collapse
Affiliation(s)
- Ane Orrantia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Iñigo Terrén
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | | | - Carmen González
- Biodonostia Health Research Institute, Hematology and Hemotherapy Service, Donostia University Hospital, Donostia-San Sebastián, Spain
| | - Alasne Uranga
- Biodonostia Health Research Institute, Hematology and Hemotherapy Service, Donostia University Hospital, Donostia-San Sebastián, Spain
| | - Juan J Mateos-Mazón
- Hematological Cancer Group, Biocruces Bizkaia Health Research Institute, Hematology and Hemotherapy Service, Cruces University Hospital, Barakaldo, Spain
| | - Juan C García-Ruiz
- Hematological Cancer Group, Biocruces Bizkaia Health Research Institute, Hematology and Hemotherapy Service, Cruces University Hospital, Barakaldo, Spain
| | - Marta Riñón
- Regulation of the Immune System Group, Biocruces Bizkaia Health Research Institute, Immunology Service, Cruces University Hospital, Barakaldo, Spain
| | - Mercedes Rey
- Biodonostia Health Research Institute, Immunology Service, Donostia University Hospital, Donostia-San Sebastián, Spain
| | - Silvia Pérez-Fernandez
- Scientific Coordination Facility, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Francisco Borrego
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
91
|
Gordon SM. Interleukin-15 in Outcomes of Pregnancy. Int J Mol Sci 2021; 22:11094. [PMID: 34681751 PMCID: PMC8541205 DOI: 10.3390/ijms222011094] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/11/2021] [Accepted: 10/11/2021] [Indexed: 01/15/2023] Open
Abstract
Interleukin-15 (IL-15) is a pleiotropic cytokine that classically acts to support the development, maintenance, and function of killer lymphocytes. IL-15 is abundant in the uterus prior to and during pregnancy, but it is subject to tight spatial and temporal regulation. Both mouse models and human studies suggest that homeostasis of IL-15 is essential for healthy pregnancy. Dysregulation of IL-15 is associated with adverse outcomes of pregnancy. Herein, we review producers of IL-15 and responders to IL-15, including non-traditional responders in the maternal uterus and fetal placenta. We also review regulation of IL-15 at the maternal-fetal interface and propose mechanisms of action of IL-15 to facilitate additional study of this critical cytokine in the context of pregnancy.
Collapse
Affiliation(s)
- Scott M. Gordon
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
92
|
Tourret M, Talvard-Balland N, Lambert M, Ben Youssef G, Chevalier MF, Bohineust A, Yvorra T, Morin F, Azarnoush S, Lantz O, Dalle JH, Caillat-Zucman S. Human MAIT cells are devoid of alloreactive potential: prompting their use as universal cells for adoptive immune therapy. J Immunother Cancer 2021; 9:jitc-2021-003123. [PMID: 34615705 PMCID: PMC8496386 DOI: 10.1136/jitc-2021-003123] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 12/20/2022] Open
Abstract
Background Mucosal-associated invariant T (MAIT) cells are semi-invariant T cells that recognize microbial antigens presented by the highly conserved MR1 molecule. MAIT cells are predominantly localized in the liver and barrier tissues and are potent effectors of antimicrobial defense. MAIT cells are very few at birth and accumulate gradually over a period of about 6 years during the infancy. The cytotoxic potential of MAIT cells, as well as their newly described regulatory and tissue repair functions, open the possibility of exploiting their properties in adoptive therapy. A prerequisite for their use as ‘universal’ cells would be a lack of alloreactive potential, which remains to be demonstrated. Methods We used ex vivo, in vitro and in vivo models to determine if human MAIT cells contribute to allogeneic responses. Results We show that recovery of MAIT cells after allogeneic hematopoietic stem cell transplantation recapitulates their slow physiological expansion in early childhood, independent of recovery of non-MAIT T cells. In vitro, signals provided by allogeneic cells and cytokines do not induce sustained MAIT cell proliferation. In vivo, human MAIT cells do not expand nor accumulate in tissues in a model of T-cell-mediated xenogeneic graft-versus-host disease in immunodeficient mice. Conclusions Altogether, these results provide evidence that MAIT cells are devoid of alloreactive potential and pave the way for harnessing their translational potential in universal adoptive therapy overcoming barriers of HLA disparity. Trial registration number ClinicalTrials.gov number NCT02403089.
Collapse
Affiliation(s)
- Marie Tourret
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Nana Talvard-Balland
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Marion Lambert
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Ghada Ben Youssef
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Mathieu F Chevalier
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Armelle Bohineust
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Thomas Yvorra
- INSERM UMR3666/U1143, Université PSL, Institut Curie, Paris, France
| | - Florence Morin
- Laboratoire d'Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Paris, France
| | - Saba Azarnoush
- Département d'Immuno-Hématologie, Hôpital Robert Debré, AP-HP, Université de Paris, Paris, France
| | - Olivier Lantz
- INSERM U932, Université PSL, Institut Curie, Paris, France.,Laboratoire d'immunologie clinique & Centre d'investigation Clinique en Biothérapie (CIC-BT1428), Institut Curie, Paris, France
| | - Jean-Hugues Dalle
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France.,Département d'Immuno-Hématologie, Hôpital Robert Debré, AP-HP, Université de Paris, Paris, France
| | - Sophie Caillat-Zucman
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France .,Laboratoire d'Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Paris, France
| |
Collapse
|
93
|
Evaluation of Comprehensive Gene Expression and NK Cell-Mediated Killing in Glioblastoma Cell Line-Derived Spheroids. Cancers (Basel) 2021; 13:cancers13194896. [PMID: 34638384 PMCID: PMC8508082 DOI: 10.3390/cancers13194896] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/16/2021] [Accepted: 09/27/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Glioblastoma (GBM) is the most aggressive primary malignant brain tumor in adults. Despite standard treatment, including surgery, chemotherapy, and radiotherapy, it is associated with poor survival. Immunotherapy is a promising alternative for patients with GBM. Natural killer (NK) cells are possible promising targets in GBM treatment because of their potent cytotoxic effect. We previously reported that highly activated and ex vivo-expanded NK cells, or genuine induced NK cells (GiNK), exert a greatly cytotoxic effect on GBM cells. In this study, we investigated the potential of NK cell-based immunotherapy for GBM, which we evaluated using an ex vivo three-dimensional GBM cell-derived spheroid model. Our results indicated that the NK cells had an anti-tumor effect on the spheroid models. Our findings could lead to the development of future NK cell-based immunotherapies for GBM. Abstract Glioblastoma (GBM) is the most common and aggressive primary brain tumor, with a dismal prognosis. Natural killer (NK) cells are large granular lymphocytes with natural cytotoxicity against tumor cells, and they should be established for the novel treatment of patients with GBM. We previously reported highly activated, and ex vivo-expanded NK cells derived from human peripheral blood, designated genuine induced NK cells (GiNK), which were induced by specific culture conditions and which exerted a cytotoxic effect on GBM cells via apoptosis. Here, we comprehensively summarize the molecular characteristics, especially focusing on the expression of stem cell markers, extracellular matrix markers, chemokines, chemokine receptors, and NK receptor ligands of spheroids derived from GBM cell lines as compared with that of two-dimensional (2D) adherent GBM cells via microarray. The spheroid had upregulated gene expression of stem cell markers, extracellular matrix markers, chemokines, chemokine receptors, and NK cell inhibitory receptor ligands compared with the 2D adherent GBM cells. Preclinical evaluation of the NK cells was performed via an ex vivo 3D spheroid model derived from GBM cell lines. In the model, the NK cells accumulated and infiltrated around the spheroids and induced GBM cell death. Flow cytometry-based apoptosis detection clearly showed that the NK cells induced GBM cell death via apoptosis. Our findings could provide pivotal information for NK cell-based immunotherapy for patients with GBM.
Collapse
|
94
|
Natural Killer Cells and Type 1 Innate Lymphoid Cells in Hepatocellular Carcinoma: Current Knowledge and Future Perspectives. Int J Mol Sci 2021; 22:ijms22169044. [PMID: 34445750 PMCID: PMC8396475 DOI: 10.3390/ijms22169044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 02/07/2023] Open
Abstract
Natural killer (NK) cells and type 1 innate lymphoid cells (ILC1) are specific innate lymphoid cell subsets that are key for the detection and elimination of pathogens and cancer cells. In liver, while they share a number of characteristics, they differ in many features. These include their developmental pathways, tissue distribution, phenotype and functions. NK cells and ILC1 contribute to organ homeostasis through the production of key cytokines and chemokines and the elimination of potential harmful bacteria and viruses. In addition, they are equipped with a wide range of receptors, allowing them to detect “stressed cells’ such as cancer cells. Our understanding of the role of innate lymphoid cells in hepatocellular carcinoma (HCC) is growing owing to the development of mouse models, the progress in immunotherapeutic treatment and the recent use of scRNA sequencing analyses. In this review, we summarize the current understanding of NK cells and ILC1 in hepatocellular carcinoma and discuss future strategies to take advantage of these innate immune cells in anti-tumor immunity. Immunotherapies hold great promise in HCC, and a better understanding of the role and function of NK cells and ILC1 in liver cancer could pave the way for new NK cell and/or ILC1-targeted treatment.
Collapse
|
95
|
Atkinson L, Martin F, Sturmey RG. Intraovarian injection of platelet-rich plasma in assisted reproduction: too much too soon? Hum Reprod 2021; 36:1737-1750. [PMID: 33963408 PMCID: PMC8366566 DOI: 10.1093/humrep/deab106] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/09/2021] [Indexed: 12/13/2022] Open
Abstract
The prospect of ovarian rejuvenation offers the tantalising prospect of treating age-related declines in fertility or in pathological conditions such as premature ovarian failure. The concept of ovarian rejuvenation was invigorated by the indication of the existence of oogonial stem cells (OSCs), which have been shown experimentally to have the ability to differentiate into functional follicles and generate oocytes; however, their clinical potential remains unknown. Furthermore, there is now growing interest in performing ovarian rejuvenation in situ. One proposed approach involves injecting the ovary with platelet rich plasma (PRP). PRP is a component of blood that remains after the in vitro removal of red and white blood cells. It contains blood platelets, tiny anucleate cells of the blood, which are responsible for forming athrombus to prevent bleeding. In addition, PRP contains an array of cytokines and growth factors, as well as a number of small molecules.The utility ofPRP has been investigatedin a range of regenerative medicine approaches and has been shown to induce differentiation of a range of cell types, presumably through the action of cytokines. A handful ofcasereports have described the use of PRP injections into the ovaryin the human, and while these clinical data report promising results, knowledge on the mechanisms and safety of PRP injections into the ovary remain limited.In this article, we summarise some of the physiological detail of platelets and PRP, before reviewing the existing emerging literature in this area. We then propose potential mechanisms by which PRP may be eliciting any effects before reflecting on some considerations for future studies in the area. Importantly, on the basis of our existing knowledge, we suggest that immediate use of PRP in clinical applications is perhaps premature and further fundamental and clinical research on the nature of ovarian insufficiency, as well as the mechanism by which PRP may act on the ovary, is needed to fully understand this promising development.
Collapse
Affiliation(s)
- Lloyd Atkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Francesca Martin
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Roger G Sturmey
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK.,Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, St Mary's Hospital, Manchester, UK
| |
Collapse
|
96
|
Ma R, Lu T, Li Z, Teng KY, Mansour AG, Yu M, Tian L, Xu B, Ma S, Zhang J, Barr T, Peng Y, Caligiuri MA, Yu J. An Oncolytic Virus Expressing IL15/IL15Rα Combined with Off-the-Shelf EGFR-CAR NK Cells Targets Glioblastoma. Cancer Res 2021; 81:3635-3648. [PMID: 34006525 PMCID: PMC8562586 DOI: 10.1158/0008-5472.can-21-0035] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/31/2021] [Accepted: 05/06/2021] [Indexed: 02/05/2023]
Abstract
IL15 is a pleiotropic cytokine with multiple roles that improve immune responses to tumor cells. Oncolytic viruses (OV) specifically lyse tumors and activate immune responses. Systemic administration of IL15 or its complex with the IL15Rα and chimeric antigen receptor (CAR) natural killer (NK) cells are currently being tested in the clinic. Here, we generated a herpes simplex 1-based OV-expressing human IL15/IL15Rα sushi domain fusion protein (named OV-IL15C), as well as off-the-shelf EGFR-CAR NK cells, and studied their monotherapy and combination efficacy in vitro and in multiple glioblastoma (GBM) mouse models. In vitro, soluble IL15/IL15Rα complex was secreted from OV-IL15C-infected GBM cells, which promoted GBM cytotoxicity and improved survival of NK and CD8+ T cells. Frozen, readily available off-the-shelf EGFR-CAR NK cells showed enhanced killing of tumor cells compared with empty vector-transduced NK cells. In vivo, OV-IL15C significantly inhibited tumor growth and prolonged survival of GBM-bearing mice in the presence of CD8+ T cells compared with parental OV. OV-IL15C plus EGFR-CAR NK cells synergistically suppressed tumor growth and significantly improved survival compared with either monotherapy, correlating with increased intracranial infiltration and activation of NK and CD8+ T cells and elevated persistence of CAR NK cells in an immunocompetent model. Collectively, OV-IL15C and off-the-shelf EGFR-CAR NK cells represent promising therapeutic strategies for GBM treatment to improve the clinical management of this devastating disease. SIGNIFICANCE: The combination of an oncolytic virus expressing the IL15/IL15Rα complex and frozen, ready-to-use EGFR-CAR NK cells elicits strong antitumor responses in glioblastoma.
Collapse
Affiliation(s)
- Rui Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Chengdu, P.R. China
| | - Ting Lu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Zhenlong Li
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Kun-Yu Teng
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Anthony G Mansour
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Melissa Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Lei Tian
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Bo Xu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Los Angeles, California
| | - Tasha Barr
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Chengdu, P.R. China.
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California.
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Centre, Los Angeles, California
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, California
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California.
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Centre, Los Angeles, California
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, California
| |
Collapse
|
97
|
Desbois M, Béal C, Charrier M, Besse B, Meurice G, Cagnard N, Jacques Y, Béchard D, Cassard L, Chaput N. IL-15 superagonist RLI has potent immunostimulatory properties on NK cells: implications for antimetastatic treatment. J Immunother Cancer 2021; 8:jitc-2020-000632. [PMID: 32532840 PMCID: PMC7295443 DOI: 10.1136/jitc-2020-000632] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
Background As the immune system is compromised in patients with cancer, therapeutic strategies to stimulate immunity appear promising, to avoid relapse and increase long-term overall survival. Interleukin-15 (IL-15) has similar properties to IL-2, but does not cause activation-induced cell death nor activation and proliferation of regulatory T cells (Treg), which makes it a serious candidate for anticancer immunotherapy. However, IL-15 has a short half-life and high doses are needed to achieve responses. Designed to enhance its activity, receptor-linker-IL-15 (RLI) (SO-C101) is a fusion molecule of human IL-15 covalently linked to the human IL-15Rα sushi+ domain currently assessed in a phase I/Ib clinical trial on patients with advanced/metastatic solid cancer. Methods We investigated the antimetastatic activity of RLI in a 4T1 mouse mammary carcinoma that spontaneously metastasizes and evaluated its immunomodulatory role in the metastatic lung microenvironment. We further characterized the proliferation, maturation and cytotoxic functions of natural killer (NK) cells in tumor-free mice treated with RLI. Finally, we explored the effect of RLI on human NK cells from healthy donors and patients with non-small cell lung cancer (NSCLC). Results RLI treatment displayed antimetastatic properties in the 4T1 mouse model. By characterizing the lung microenvironment, we observed that RLI restored the balance between NK cells and neutrophils (CD11b+ Ly6Ghigh Ly6Clow) that massively infiltrate lungs of 4T1-tumor bearing mice. In addition, the ratio between NK cells and Treg was strongly increased by RLI treatment. Further pharmacodynamic studies in tumor-free mice revealed superior proliferative and cytotoxic functions on NK cells after RLI treatment compared with IL-15 alone. Characterization of the maturation stage of NK cells demonstrated that RLI favored accumulation of CD11b+ CD27high KLRG1+ mature NK cells. Finally, RLI demonstrated potent immunostimulatory properties on human NK cells by inducing proliferation and activation of NK cells from healthy donors and enhancing cytotoxic responses to NKp30 crosslinking in NK cells from patients with NSCLC. Conclusions Collectively, our work demonstrates superior activity of RLI compared with rhIL-15 in modulating and activating NK cells and provides additional evidences for a therapeutic strategy using RLI as antimetastatic molecule.
Collapse
Affiliation(s)
- Mélanie Desbois
- Analyse Moléculaire, Modélisation et Imagerie de la Maladie Cancéreuse, Laboratoire d'Immunomonitoring en Oncologie, Gustave Roussy Institute, INSERM, CNRS, Paris-Saclay University, Villejuif, Île-de-France, France.,Cytune Pharma, Nantes, France.,Faculté de Médecine, Paris-Saclay University, Le Kremlin-Bicêtre, Île-de-France, France
| | - Coralie Béal
- Centre d'investigation Clinique Biothérapie 1428, Gustave Roussy Institute, Villejuif, Île-de-France, France
| | - Mélinda Charrier
- Analyse Moléculaire, Modélisation et Imagerie de la Maladie Cancéreuse, Laboratoire d'Immunomonitoring en Oncologie, Gustave Roussy Institute, INSERM, CNRS, Paris-Saclay University, Villejuif, Île-de-France, France.,Faculté de Médecine, Paris-Saclay University, Le Kremlin-Bicêtre, Île-de-France, France.,Centre d'investigation Clinique Biothérapie 1428, Gustave Roussy Institute, Villejuif, Île-de-France, France
| | - Benjamin Besse
- Faculté de Médecine, Paris-Saclay University, Le Kremlin-Bicêtre, Île-de-France, France.,Comité de Pathologie Thoracique, Gustave Roussy Institute, Villejuif, Île-de-France, France
| | - Guillaume Meurice
- Plateforme de Bioinformatique, Gustave Roussy Institute, Villejuif, Île-de-France, France
| | - Nicolas Cagnard
- Plateforme de Bioinformatique, Université Paris Descartes, Paris, Île-de-France, France
| | | | | | - Lydie Cassard
- Analyse Moléculaire, Modélisation et Imagerie de la Maladie Cancéreuse, Laboratoire d'Immunomonitoring en Oncologie, Gustave Roussy Institute, INSERM, CNRS, Paris-Saclay University, Villejuif, Île-de-France, France
| | - Nathalie Chaput
- Analyse Moléculaire, Modélisation et Imagerie de la Maladie Cancéreuse, Laboratoire d'Immunomonitoring en Oncologie, Gustave Roussy Institute, INSERM, CNRS, Paris-Saclay University, Villejuif, Île-de-France, France .,Faculty of Pharmacy, University Paris-Saclay, Chatenay-Malabry, France
| |
Collapse
|
98
|
Rosser CJ, Tikhonenkov S, Nix JW, Chan OTM, Ianculescu I, Reddy S, Soon-Shiong P. Safety, Tolerability, and Long-Term Clinical Outcomes of an IL-15 analogue (N-803) Admixed with Bacillus Calmette-Guérin (BCG) for the Treatment of Bladder Cancer. Oncoimmunology 2021; 10:1912885. [PMID: 33996264 PMCID: PMC8096327 DOI: 10.1080/2162402x.2021.1912885] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Intravesical BCG is active against non-muscle invasive bladder cancer (NMIBC), but bladder cancer will recur and even progress in a significant number of patients. To improve the response rate, N-803, an IL-15 superagonist was administered in combination with BCG. To evaluate the safety and efficacy associated with the use of intravesical N-803 and BCG in patients with BCG-naïve NMIBC. This phase 1b clinical trial used a 3 + 3 dose-escalation design. Participants were enrolled from July 2014 and July 2015, with follow-up and analyses through January 15, 2021. Eligibility criteria included histologically confirmed non-muscle invasive urothelial carcinoma of intermediate or high risk who had not received prior treatment with intravesical BCG (ie, BCG-naïve). All 9 participants met the eligibility criteria, received treatment according to the protocol, and were included in all analyses. Treatment was done once weekly for 6 consecutive weeks with bladder infusion of the standard dose of BCG, 50 mg/instillation, in combination with increasing doses of N-803 (100, 200, or 400 µg N-803 per instillation). No DLTs were noted in any of the dose cohorts. All adverse events (AEs) were manageable and less than grade 3. During the 2-year follow-up, all 9 participants were disease free. Furthermore, 6 y after treatment, all 9 participants (100%) were disease free with no evidence of disease progression and an intact bladder. This phase 1b trial found the combination of intravesical N-803 and BCG to be associated with modest toxic effects, low immunogenicity, and substantial prolonged antitumoral activity; phase 2 trials are in progress.
Collapse
Affiliation(s)
- Charles J Rosser
- Clinical & Translational Research Program, University of Hawaii Cancer Center,Honolulu, Hawaii
| | - Sergei Tikhonenkov
- Clinical & Translational Research Program, University of Hawaii Cancer Center,Honolulu, Hawaii
| | - Jeffrey W Nix
- Department of Urology, University of Alabama, Birmingham, Alabama
| | - Owen T M Chan
- Clinical & Translational Research Program, University of Hawaii Cancer Center,Honolulu, Hawaii
| | | | - Sandeep Reddy
- NantHealth Inc, Culver City, California.,ImmunityBio, Inc., Culver City, California
| | | |
Collapse
|
99
|
Crinier A, Dumas PY, Escalière B, Piperoglou C, Gil L, Villacreces A, Vély F, Ivanovic Z, Milpied P, Narni-Mancinelli É, Vivier É. Single-cell profiling reveals the trajectories of natural killer cell differentiation in bone marrow and a stress signature induced by acute myeloid leukemia. Cell Mol Immunol 2021; 18:1290-1304. [PMID: 33239726 PMCID: PMC8093261 DOI: 10.1038/s41423-020-00574-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells are innate cytotoxic lymphoid cells (ILCs) involved in the killing of infected and tumor cells. Among human and mouse NK cells from the spleen and blood, we previously identified by single-cell RNA sequencing (scRNAseq) two similar major subsets, NK1 and NK2. Using the same technology, we report here the identification, by single-cell RNA sequencing (scRNAseq), of three NK cell subpopulations in human bone marrow. Pseudotime analysis identified a subset of resident CD56bright NK cells, NK0 cells, as the precursor of both circulating CD56dim NK1-like NK cells and CD56bright NK2-like NK cells in human bone marrow and spleen under physiological conditions. Transcriptomic profiles of bone marrow NK cells from patients with acute myeloid leukemia (AML) exhibited stress-induced repression of NK cell effector functions, highlighting the profound impact of this disease on NK cell heterogeneity. Bone marrow NK cells from AML patients exhibited reduced levels of CD160, but the CD160high group had a significantly higher survival rate.
Collapse
Affiliation(s)
- Adeline Crinier
- Aix Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Pierre-Yves Dumas
- CHU Bordeaux, Service d'Hématologie Clinique et de Thérapie Cellulaire, Bordeaux, France
- Bordeaux University, Bordeaux, France
- Institut National de la Santé et de la Recherche Médicale, U1035, Bordeaux, France
| | - Bertrand Escalière
- Aix Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | | | - Laurine Gil
- Aix Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Arnaud Villacreces
- Bordeaux University, Bordeaux, France
- Institut National de la Santé et de la Recherche Médicale, U1035, Bordeaux, France
| | - Frédéric Vély
- Aix Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- APHM, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France
| | - Zoran Ivanovic
- Institut National de la Santé et de la Recherche Médicale, U1035, Bordeaux, France
- Établissement Français du Sang Nouvelle Aquitaine, Bordeaux, France
| | - Pierre Milpied
- Aix Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Émilie Narni-Mancinelli
- Aix Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
| | - Éric Vivier
- Aix Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
- APHM, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France.
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France.
| |
Collapse
|
100
|
Horowitz NB, Mohammad I, Moreno-Nieves UY, Koliesnik I, Tran Q, Sunwoo JB. Humanized Mouse Models for the Advancement of Innate Lymphoid Cell-Based Cancer Immunotherapies. Front Immunol 2021; 12:648580. [PMID: 33968039 PMCID: PMC8100438 DOI: 10.3389/fimmu.2021.648580] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a branch of the immune system that consists of diverse circulating and tissue-resident cells, which carry out functions including homeostasis and antitumor immunity. The development and behavior of human natural killer (NK) cells and other ILCs in the context of cancer is still incompletely understood. Since NK cells and Group 1 and 2 ILCs are known to be important for mediating antitumor immune responses, a clearer understanding of these processes is critical for improving cancer treatments and understanding tumor immunology as a whole. Unfortunately, there are some major differences in ILC differentiation and effector function pathways between humans and mice. To this end, mice bearing patient-derived xenografts or human cell line-derived tumors alongside human genes or human immune cells represent an excellent tool for studying these pathways in vivo. Recent advancements in humanized mice enable unparalleled insights into complex tumor-ILC interactions. In this review, we discuss ILC behavior in the context of cancer, the humanized mouse models that are most commonly employed in cancer research and their optimization for studying ILCs, current approaches to manipulating human ILCs for antitumor activity, and the relative utility of various mouse models for the development and assessment of these ILC-related immunotherapies.
Collapse
Affiliation(s)
- Nina B Horowitz
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Department of Bioengineering, Stanford University School of Medicine and School of Engineering, Stanford, CA, United States
| | - Imran Mohammad
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Uriel Y Moreno-Nieves
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Ievgen Koliesnik
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Quan Tran
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - John B Sunwoo
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|