1
|
Ma S, Yu J, Caligiuri MA. Natural killer cell-based immunotherapy for cancer. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf036. [PMID: 40246292 DOI: 10.1093/jimmun/vkaf036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/18/2025] [Indexed: 04/19/2025]
Abstract
Natural killer (NK) cells are emerging as a promising tool for cancer immunotherapy due to their innate ability to selectively recognize and eliminate cancer cells. Over the past 3 decades, strategies to harness NK cells have included cytokines, small molecules, antibodies, and the adoptive transfer of autologous or allogeneic NK cells, both unmodified and genetically engineered. Despite favorable safety profiles in clinical trials, challenges such as limited in vivo persistence, exhaustion, and the suppressive tumor microenvironment continue to hinder their efficacy and durability. This review categorizes NK cell-based therapies into 3 major approaches: (i) cellular therapies, including unmodified and chimeric antigen receptor-engineered NK cells; (ii) cytokine-based strategies such as interleukin-2 and interleukin-15 derivatives; and (iii) antibody-based therapies, including immune checkpoint inhibitors and NK cell engagers. We highlight these advancements, discuss current limitations, and propose strategies to optimize NK cell-based therapies for improved cancer treatment outcomes.
Collapse
Affiliation(s)
- Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, United States
| | - Jianhua Yu
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA, United States
- Institute for Precision Cancer Therapeutics and Immuno-Oncology, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, United States
- Clemons Family Center for Transformative Cancer Research, University of California, Irvine, Irvine, CA, United States
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, United States
| |
Collapse
|
2
|
Jost S, Reeves RK. Elephant in the room: natural killer cells don't forget HIV either. Curr Opin HIV AIDS 2025; 20:109-116. [PMID: 39773904 PMCID: PMC11802307 DOI: 10.1097/coh.0000000000000909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
PURPOSE OF REVIEW Like elephants (and T cells), accumulating evidence suggest natural killer (NK) cells never forget. The description of adaptive or memory NK cells, which can be induced by HIV/SIV infections and vaccines and associated with protective effects in persons with HIV (PWH), has dramatically increased the interest in leveraging NK cells to prevent HIV infection or suppress HIV reservoirs. However, harnessing their full antiviral potential has been hindered by an incomplete understanding of mechanisms underlying adaptive NK cell development and infected cell recognition. Herein, we outline the main discoveries around the adaptive functions of NK cells, with a focus on their involvement in HIV infection. RECENT FINDINGS NK cells with diverse adaptive capabilities, including antigen-specific memory, cytokine-induced and CMV-driven adaptive subsets, likely all play a role in HIV infection. Importantly, true antigen-specific memory NK cells have been identified that mediate recall responses against multiple infectious agents such as HIV, influenza, and SARS-CoV-2. The NKG2C receptor is pivotal for certain adaptive NK cell subsets, as it marks a population with enhanced antibody-dependent functions and has been described as the main receptor mediating antigen-specific responses via recognition of viral peptides presented by HLA-E. SUMMARY Antiviral functions of adaptive/memory NK cells have tremendous, but as of yet, untapped potential to be harnessed for vaccine design, curative, or other therapeutic interventions against HIV.
Collapse
Affiliation(s)
- Stephanie Jost
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | | |
Collapse
|
3
|
Kokiçi J, Preechanukul A, Arellano-Ballestero H, Gorou F, Peppa D. Emerging Insights into Memory Natural Killer Cells and Clinical Applications. Viruses 2024; 16:1746. [PMID: 39599860 PMCID: PMC11599065 DOI: 10.3390/v16111746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Natural killer (NK) cells are innate lymphocytes that can rapidly mount a response to their targets by employing diverse mechanisms. Due to their functional attributes, NK cells have been implicated in anti-viral and anti-tumour immune responses. Although traditionally known to mount non-specific, rapid immune responses, in recent years, the notion of memory NK cells with adaptive features has gained more recognition. Memory NK cells emerge in response to different stimuli, such as viral antigens and specific cytokine combinations. They form distinct populations, accompanied by transcriptional, epigenetic and metabolic reprogramming, resulting in unique phenotypic and functional attributes. Several clinical trials are testing the efficacy of memory NK cells due to their enhanced functionality, bioenergetic profile and persistence in vivo. The therapeutic potential of NK cells is being harnessed in viral infections, with wider applications in the cancer field. In this review, we summarise the current state of research on the generation of memory NK cells, along with their clinical applications in viral infection and cancer.
Collapse
Affiliation(s)
- Jonida Kokiçi
- Division of Infection and Immunity, University College London, London NW3 2PP, UK
| | - Anucha Preechanukul
- Division of Infection and Immunity, University College London, London NW3 2PP, UK
| | | | - Frances Gorou
- Division of Infection and Immunity, University College London, London NW3 2PP, UK
| | - Dimitra Peppa
- Division of Infection and Immunity, University College London, London NW3 2PP, UK
| |
Collapse
|
4
|
Ruppert Z, Neuperger P, Rákóczi B, Gémes N, Dukay B, Hajdu P, Péter M, Balogh G, Tiszlavicz L, Vígh L, Török Z, Puskás LG, Szebeni GJ, Tóth ME. Characterization of obesity-related diseases and inflammation using single cell immunophenotyping in two different diet-induced obesity models. Int J Obes (Lond) 2024; 48:1568-1576. [PMID: 39004641 PMCID: PMC11502477 DOI: 10.1038/s41366-024-01584-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Obesity is a growing problem worldwide and a major risk factor for many chronic diseases. The accumulation of adipose tissue leads to the release of significant amounts of pro-inflammatory cytokines and adipokines, resulting in a low-grade systemic inflammation. However, the mechanisms behind the development of obesity-related diseases are not fully understood. Therefore, our study aimed to investigate the pathological changes and inflammatory processes at systemic level and in individual organs in two different diet-induced mouse obesity models. METHODS Male C57BL6/J mice were fed by high-fat diet (HFD), high-fat/high-fructose diet (HFD + FR) or normal chow for 21 weeks starting at 3 months of age (n = 15 animals/group). Insulin resistance was tested by oral glucose tolerance test. Pathological changes were investigated on hematoxylin-eosin-stained liver and brown adipose tissue sections. The gene expression levels of adipokines and cytokines were analyzed by qPCR in adipose tissues, whereas serum protein concentrations were determined by multiplex immunoassays. Immunophenotyping of isolated blood, bone marrow and spleen cells was performed by single-cell mass cytometry. RESULTS Weight gain, glucose intolerance and hepatic steatosis were more severe in the HFD + FR group than in the control and HFD groups. This was accompanied by a higher level of systemic inflammation, as indicated by increased expression of pro-inflammatory genes in visceral white adipose tissue and by a higher serum TNFα level. In addition, immunophenotyping revealed the increase of the surface expressions of CD44 and CD69 on various cell types, such as CD8+ and CD4 + T-cells, B-cells and macrophages, in animals with obesity. CONCLUSIONS The combination of HFD with fructose supplementation promotes more properly the symptoms of metabolic syndrome. Therefore, the combined high-fat/high-fructose nutrition can be a more suitable model of the Western diet. However, despite these differences, both models showed immunophenotypic changes that may be associated with increased risk of obesity-related cancer.
Collapse
Affiliation(s)
- Zsófia Ruppert
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, Szeged, Hungary
- PhD School in Biology, University of Szeged, Szeged, Hungary
| | - Patrícia Neuperger
- PhD School in Biology, University of Szeged, Szeged, Hungary
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Bettina Rákóczi
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, Szeged, Hungary
- PhD School in Biology, University of Szeged, Szeged, Hungary
| | - Nikolett Gémes
- PhD School in Biology, University of Szeged, Szeged, Hungary
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Brigitta Dukay
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Petra Hajdu
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Mária Péter
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Gábor Balogh
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, Szeged, Hungary
| | - László Tiszlavicz
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - László Vígh
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Zsolt Török
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, Szeged, Hungary
| | - László G Puskás
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Gábor J Szebeni
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary.
- Department of Internal Medicine, Hematology Centre, Faculty of Medicine, University of Szeged, H6725, Szeged, Hungary.
| | - Melinda E Tóth
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, HUN-REN Biological Research Centre, Szeged, Hungary.
| |
Collapse
|
5
|
Arellano-Ballestero H, Zubiak A, Dally C, Orchard K, Alrubayyi A, Charalambous X, Michael M, Torrance R, Eales T, Das K, Tran MGB, Sabry M, Peppa D, Lowdell MW. Proteomic and phenotypic characteristics of memory-like natural killer cells for cancer immunotherapy. J Immunother Cancer 2024; 12:e008717. [PMID: 39032940 PMCID: PMC11261707 DOI: 10.1136/jitc-2023-008717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Human and mouse natural killer (NK) cells have been shown to develop memory-like function after short-term exposure to the cocktail of IL-12/15/18 or to overnight co-culture with some tumor cell lines. The resulting cells retain enhanced lytic ability for up to 7 days as well as after cryopreservation, and memory-like NK cells (mlNK) have been shown to induce complete remissions in patients with hematological malignancies. No single phenotype has been described for mlNK and the physiological changes induced by the short-term cytokine or tumor-priming which are responsible for these enhanced functions have not been fully characterized. Here, we have generated mlNK by cytokine and tumor-priming to find commonalities to better define the nature of NK cell "memory" in vitro and, for the first time, in vivo. METHODS We initiated mlNK in vitro from healthy donors with cytokines (initiated cytokine-induced memory-like (iCIML)-NK) and by tumor priming (TpNK) overnight and compared them by high-dimensional flow cytometry, proteomic and metabolomic profiling. As a potential mechanism of enhanced cytolytic function, we analyzed the avidity of binding of the mlNK to NK-resistant tumors (z-Movi). We generated TpNK from healthy donors and from cancer patients to determine whether mlNK generated by interaction with a single tumor type could enhance lytic activity. Finally, we used a replication-incompetent tumor cell line (INKmune) to treat patients with myeloid leukaemias to potentiate NK cell function in vivo. RESULTS Tumor-primed mlNK from healthy donors and patients with cancer showed increased cytotoxicity against multiple tumor cell lines in vitro, analogous to iCIML-NK cells. Multidimensional cytometry identified distinct memory-like profiles of subsets of cells with memory-like characteristics; upregulation of CD57, CD69, CD25 and ICAM1. Proteomic profiling identified 41 proteins restricted to mlNK cells and we identified candidate molecules for the basis of NK memory which can explain how mlNK overcome inhibition by resistant tumors. Finally, of five patients with myelodysplastic syndrome or refractory acute myeloid leukemia treated with INKmune, three responded to treatment with measurable increases in NK lytic function and systemic cytokines. CONCLUSIONS NK cell "memory" is a physiological state associated with resistance to MHC-mediated inhibition, increased metabolic function, mitochondrial fitness and avidity to NK-resistant target cells.
Collapse
Affiliation(s)
| | - Agnieszka Zubiak
- Cancer Institute, University College London, London, UK
- INmuneBio Inc, Boca Raton, Florida, USA
| | - Chris Dally
- Department of Haematology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Kim Orchard
- Department of Haematology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | | | | | | | - Trinity Eales
- Cancer Institute, University College London, London, UK
| | | | - Maxine G. B. Tran
- Department of Biotechnology, Division of Surgery and Interventional Science, University College London, London, UK
- Specialist Centre for Kidney Cancer, Royal Free Hospital, London, UK
| | - May Sabry
- Cancer Institute, University College London, London, UK
- INmuneBio Inc, Boca Raton, Florida, USA
| | - Dimitra Peppa
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Mark W. Lowdell
- Cancer Institute, University College London, London, UK
- INmuneBio Inc, Boca Raton, Florida, USA
| |
Collapse
|
6
|
Koedijk JB, van der Werf I, Penter L, Vermeulen MA, Barneh F, Perzolli A, Meesters-Ensing JI, Metselaar DS, Margaritis T, Fiocco M, de Groot-Kruseman HA, Moeniralam R, Bang Christensen K, Porter B, Pfaff K, Garcia JS, Rodig SJ, Wu CJ, Hasle H, Nierkens S, Belderbos ME, Zwaan CM, Heidenreich O. A multidimensional analysis reveals distinct immune phenotypes and the composition of immune aggregates in pediatric acute myeloid leukemia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.03.03.23286485. [PMID: 37961528 PMCID: PMC10635226 DOI: 10.1101/2023.03.03.23286485] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Because of the low mutational burden and consequently, fewer potential neoantigens, children with acute myeloid leukemia (AML) are thought to have a T cell-depleted or 'cold' tumor microenvironment and may have a low likelihood of response to T cell-directed immunotherapies. Understanding the composition, phenotype, and spatial organization of T cells and other microenvironmental populations in the pediatric AML bone marrow (BM) is essential for informing future immunotherapeutic trials about targetable immune-evasion mechanisms specific to pediatric AML. Here, we conducted a multidimensional analysis of the tumor immune microenvironment in pediatric AML and non-leukemic controls. We demonstrated that nearly one-third of pediatric AML cases has an immune-infiltrated BM, which is characterized by a decreased ratio of M2-to M1-like macrophages. Furthermore, we detected the presence of large T cell networks, both with and without colocalizing B cells, in the BM and dissected the cellular composition of T- and B cell-rich aggregates using spatial transcriptomics. These analyses revealed that these aggregates are hotspots of CD8 + T cells, memory B cells, plasma cells and/or plasmablasts, and M1-like macrophages. Collectively, our study provides a multidimensional characterization of the BM immune microenvironment in pediatric AML and indicates starting points for further investigations into immunomodulatory mechanisms in this devastating disease.
Collapse
|
7
|
Bullock KK, Shattuck-Brandt R, Scalise C, Luo W, Chen SC, Saleh N, Gonzalez-Ericsson PI, Garcia G, Sanders ME, Ayers GD, Yan C, Richmond A. Endogenous pAKT activity is associated with response to AKT inhibition alone and in combination with immune checkpoint inhibition in murine models of TNBC. Cancer Lett 2024; 586:216681. [PMID: 38311054 PMCID: PMC11622984 DOI: 10.1016/j.canlet.2024.216681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/22/2023] [Accepted: 01/25/2024] [Indexed: 02/06/2024]
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous and challenging-to-treat breast cancer subtype. The clinical introduction of immune checkpoint inhibitors (ICI) for TNBC has had mixed results, and very few patients achieved a durable response. The PI3K/AKT pathway is frequently mutated in breast cancer. Given the important roles of the PI3K pathway in immune and tumor cell signaling, there is an interest in using inhibitors of this pathway to increase the response to ICI. This study sought to determine if AKT inhibition could enhance the response to ICI in murine TNBC models. We further sought to understand underlying mechanisms of response or non-response to AKT inhibition in combination with ICI. Using four murine TNBC-like cell lines and corresponding orthotopic mouse tumor models, we found that hyperactivity of the PI3K pathway, as evidenced by levels of phospho-AKT rather than PI3K pathway mutational status, was associated with response to AKT inhibition alone and in combination with ICI. Additional mutations in other growth regulatory pathways could override the response of PI3K pathway mutant tumors to AKT inhibition. Furthermore, we observed that AKT inhibition enhanced the response to ICI in an already sensitive model. However, AKT inhibition failed to convert ICI-resistant tumors, to responsive tumors. These findings suggest that analysis of both the mutational status and phospho-AKT protein levels may be beneficial in predicting which TNBC tumors will respond to AKT inhibition in combination with ICI.
Collapse
Affiliation(s)
- Kennady K Bullock
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Department of Pharmacology, Vanderbilt School of Medicine, Nashville, TN, USA
| | - Rebecca Shattuck-Brandt
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Department of Pharmacology, Vanderbilt School of Medicine, Nashville, TN, USA
| | - Carly Scalise
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Department of Pharmacology, Vanderbilt School of Medicine, Nashville, TN, USA
| | - Weifeng Luo
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Department of Pharmacology, Vanderbilt School of Medicine, Nashville, TN, USA
| | - Sheau-Chiann Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nabil Saleh
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Department of Pharmacology, Vanderbilt School of Medicine, Nashville, TN, USA
| | - Paula I Gonzalez-Ericsson
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Guadalupe Garcia
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Melinda E Sanders
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gregory D Ayers
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chi Yan
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Department of Pharmacology, Vanderbilt School of Medicine, Nashville, TN, USA.
| | - Ann Richmond
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Department of Pharmacology, Vanderbilt School of Medicine, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
8
|
Agrez M, Chandler C, Thurecht KJ, Fletcher NL, Liu F, Subramaniam G, Howard CB, Blyth B, Parker S, Turner D, Rzepecka J, Knox G, Nika A, Hall AM, Gooding H, Gallagher L. An immunomodulating peptide with potential to suppress tumour growth and autoimmunity. Sci Rep 2023; 13:19741. [PMID: 37957274 PMCID: PMC10643673 DOI: 10.1038/s41598-023-47229-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/10/2023] [Indexed: 11/15/2023] Open
Abstract
Cancers and autoimmune diseases commonly co-exist and immune checkpoint inhibitor therapy (ICI) exacerbates autoimmune pathologies. We recently described a lipidic peptide, designated IK14004, that promotes expansion of immunosuppressive T regulatory (Treg) cells and uncouples interleukin-2 from interferon-gamma production while activating CD8+ T cells. Herein, we report IK14004-mediated inhibition of Lewis lung cancer (LLC) growth and re-invigoration of splenocyte-derived exhausted CD4+ T cells. In human immune cells from healthy donors, IK14004 modulates expression of the T cell receptor α/β subunits, induces Type I IFN expression, stimulates natural killer (NK) cells to express NKG2D/NKp44 receptors and enhances K562 cytotoxicity. In both T and NK cells, IK14004 alters the IL-12 receptor β1/β2 chain ratio to favour IL-12p70 binding. Taken together, this novel peptide offers an opportunity to gain further insight into the complexity of ICI immunotherapy so that autoimmune responses may be minimised without promoting tumour evasion from the immune system.
Collapse
Affiliation(s)
- Michael Agrez
- InterK Peptide Therapeutics Limited, New South Wales, Australia.
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia.
| | | | - Kristofer J Thurecht
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Feifei Liu
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Gayathri Subramaniam
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Christopher B Howard
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Benjamin Blyth
- Department of Oncology,, Peter MacCallum Cancer Centre and Sir Peter MacCallum, University of Melbourne, Melbourne, Australia
| | - Stephen Parker
- InterK Peptide Therapeutics Limited, New South Wales, Australia
| | | | | | - Gavin Knox
- Concept Life Sciences, Edinburgh, Scotland
| | | | | | | | | |
Collapse
|
9
|
Arellano-Ballestero H, Sabry M, Lowdell MW. A Killer Disarmed: Natural Killer Cell Impairment in Myelodysplastic Syndrome. Cells 2023; 12:633. [PMID: 36831300 PMCID: PMC9954109 DOI: 10.3390/cells12040633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Myelodysplastic syndrome (MDS) treatment remains a big challenge due to the heterogeneous nature of the disease and its ability to progress to acute myeloid leukemia (AML). The only curative option is allogeneic hematopoietic stem cell transplantation (HSCT), but most patients are unfit for this procedure and are left with only palliative treatment options, causing a big unmet need in the context of this disease. Natural killer (NK) cells are attractive candidates for MDS immunotherapy due to their ability to target myeloid leukemic cells without prior sensitization, and in recent years we have seen an arising number of clinical trials in AML and, recently, MDS. NK cells are reported to be highly dysfunctional in MDS patients, which can be overcome by adoptive NK cell immunotherapy or activation of endogenous NK cells. Here, we review the role of NK cells in MDS, the contribution of the tumor microenvironment (TME) to NK cell impairment, and the most recent data from NK cell-based clinical trials in MDS.
Collapse
Affiliation(s)
| | - May Sabry
- Department of Haematology, University College London, London NW3 5PF, UK
- InmuneBio Inc., Boca Raton, FL 33432, USA
- Novamune Ltd., London WC2R 1DJ, UK
| | - Mark W. Lowdell
- Department of Haematology, University College London, London NW3 5PF, UK
- InmuneBio Inc., Boca Raton, FL 33432, USA
| |
Collapse
|
10
|
Al-Kadhimi Z, Callahan M, Fehniger T, Cole KE, Vose J, Hinrichs S. Enrichment of innate immune cells from PBMC followed by triple cytokine activation for adoptive immunotherapy. Int Immunopharmacol 2022; 113:109387. [DOI: 10.1016/j.intimp.2022.109387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
|
11
|
Choi SH, Kim HJ, Park JD, Ko ES, Lee M, Lee DK, Choi JH, Jang HJ, Kim I, Jung HY, Park KH, Park KS. Chemical priming of natural killer cells with branched polyethylenimine for cancer immunotherapy. J Immunother Cancer 2022; 10:jitc-2022-004964. [PMID: 36028281 PMCID: PMC9422841 DOI: 10.1136/jitc-2022-004964] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2022] [Indexed: 11/08/2022] Open
Abstract
Background Due to their powerful immune surveillance activity and ability to kill and clear cancer cells, natural killer (NK) cells are an emerging anticancer immunotherapeutic agent. Therefore, there is much interest in developing efficient technologies that further enhance the therapeutic antitumor efficacy of NK cells. Methods To produce chemically primed NK cells, we screened polymers with various electric charges and examined their ability to enhance the cytotoxicity of NK cells. The effect of primary amine and electric charges of 25 kDa branched polyethylenimine (25KbPEI) was investigated by fluorination of the chemical. The role of 25KbPEI in determining the major priming mechanism was investigated in terms of calcium influx into NK cells. In vivo therapeutic efficacy of chemically primed NK cells was evaluated against solid tumor mouse model of triple negative breast and ovarian cancers. Results Chem_NK that was produced by the priming activity of 25KbPEI showed potent antitumor activity to various cancer cells. Chem_NK showed an activated phenotype, which manifests as increased expression of activating/adhesion/chemokine receptors and perforin accumulation, leading to enhanced migration ability and antitumor activity. Chem_NK display potent therapeutic efficacy against in vivo mouse model of triple negative breast and ovarian cancers. Fluorination of the primary amine group reduces the activity of 25KbPEI to prime NK cells, indicating that the cationic charge on the chemical plays a critical role in NK cell activation. A major priming mechanism was 25KbPEI-mediated calcium influx into NK cells, which occurred mainly via the Ca2+-permeable non-selective cation channel transient receptor potential melastatin 2. Conclusions NK cells can be chemically primed with 25KbPEI to express potent antitumor activity as well as enhanced migration ability. Because PEI is a biocompatible and Food and Drug Administration-approved chemical for biomedical use, these results suggest a cost-effective and simple method of producing therapeutic NK cells.
Collapse
Affiliation(s)
- Seung Hee Choi
- Department of Biomedical Science, CHA University, Seongnam-si, Korea (the Republic of)
| | - Hye Jin Kim
- Department of Biomedical Science, CHA University, Seongnam-si, Korea (the Republic of)
| | - Joo Dong Park
- Department of Biomedical Science, CHA University, Seongnam-si, Korea (the Republic of)
| | - Eun-Su Ko
- Department of Biomedical Science, CHA University, Seongnam-si, Korea (the Republic of)
| | - Minwook Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Korea (the Republic of)
| | - Dae-Keum Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Korea (the Republic of)
| | - Jin-Ho Choi
- Department of Biomedical Science, CHA University, Seongnam-si, Korea (the Republic of)
| | - Hye Jung Jang
- Department of Biomedical Science, CHA University, Seongnam-si, Korea (the Republic of)
| | - Isaac Kim
- Department of Surgery, Bundang CHA Medical Center, CHA University, Seongnam-si, Korea (the Republic of)
| | - Hae-Yun Jung
- Department of Biomedical Science, CHA University, Seongnam-si, Korea (the Republic of)
| | - Keun-Hong Park
- Department of Biomedical Science, CHA University, Seongnam-si, Korea (the Republic of)
| | - Kyung-Soon Park
- Department of Biomedical Science, CHA University, Seongnam-si, Korea (the Republic of)
| |
Collapse
|
12
|
Terrén I, Orrantia A, Astarloa-Pando G, Amarilla-Irusta A, Zenarruzabeitia O, Borrego F. Cytokine-Induced Memory-Like NK Cells: From the Basics to Clinical Applications. Front Immunol 2022; 13:884648. [PMID: 35603208 PMCID: PMC9114299 DOI: 10.3389/fimmu.2022.884648] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells are lymphocytes with a key role in the defense against viral infections and tumor cells. Although NK cells are classified as innate lymphoid cells (ILCs), under certain circumstances they exhibit adaptive and memory-like features. The latter may be achieved, among others, by a brief stimulation with interleukin (IL)-12, IL-15 and IL-18. These cytokine-induced memory-like (CIML) NK cells resemble the trained immunity observed in myeloid cells. CIML NK cells undergo transcriptional, epigenetic and metabolic reprogramming that, along with changes in the expression of cell surface receptors and components of cytotoxic granules, are responsible for their enhanced effector functions after a resting period. In addition, these memory-like NK cells persist for a long time, which make them a good candidate for cancer immunotherapy. Currently, several clinical trials are testing CIML NK cells infusions to treat tumors, mostly hematological malignancies. In relapse/refractory acute myeloid leukemia (AML), the adoptive transfer of CIML NK cells is safe and complete clinical remissions have been observed. In our review, we sought to summarize the current knowledge about the generation and molecular basis of NK cell memory-like responses and the up-to-date results from clinical trials with CIML NK cells.
Collapse
Affiliation(s)
- Iñigo Terrén
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Ane Orrantia
- Immunopathology Group, 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
|
13
|
Perna F, Espinoza-Gutarra MR, Bombaci G, Farag SS, Schwartz JE. Immune-Based Therapeutic Interventions for Acute Myeloid Leukemia. Cancer Treat Res 2022; 183:225-254. [PMID: 35551662 DOI: 10.1007/978-3-030-96376-7_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive, clonally heterogeneous, myeloid malignancy, with a 5-year overall survival of approximately 27%. It constitutes the most common acute leukemia in adults, with an incidence of 3-5 cases per 100,000 in the United States. Despite great advances in understanding the molecular mechanisms underpinning leukemogenesis, the past several decades had seen little change to the backbone of therapy, comprised of an anthracycline-based induction regimen for those who are fit enough to receive it, followed by risk-stratified post-remission therapy with consolidation cytarabine or allogeneic stem cell transplantation (allo-SCT). Allo-SCT is the most fundamental form of immunotherapy in which donor cytotoxic T and NK cells recognize and eradicate residual AML in the graft-versus-leukemia (GvL) effect. Building on that, several alternative or synergistic approaches to exploit both self and foreign immunity against AML have been developed. Checkpoint inhibitors, for example, CTLA-4 inhibitors, PD-1 inhibitors, and PD-L1 inhibitors block proteins found on T cells or cancer cells that stop the immune system from attacking the cancer cells. They have been used with limited success in both the AML relapsed/refractory (R/R) and post SCT settings. AML tumor mutational burden is low compared to solid tumors and thus, it is less likely to generate neoantigens and respond to antibody-mediated checkpoint blockade that has shown unprecedented results in solid tumors. Therefore, alternative therapeutic strategies that work independently of the T cell receptor (TCR) specificity have been developed. They include bispecific antibodies, which recruit T cells through CD3 engagement, and in AML have shown an overall response rate ranging between 14 and 30% in early phase trials. Chimeric Antigen Receptor (CAR) T cell therapy is a type of treatment in which T cells are genetically engineered to produce a recombinant receptor that redirects the specificity and function of T lymphocytes. However, lack of cell surface targets exclusively expressed on AML cells including Leukemic Stem Cells (LSCs) combined with clonal heterogeneity represents the biggest challenge in developing CAR therapy for AML. Antibody-Drug Conjugates (ADC) constitute the only FDA-approved immunotherapy to treat AML with Gemtuzumab Ozogamicin, a CD33-specific ADC used in CEBPα-mutated AML. The identification of additional cell surface targets is critical for the development of other ADC's potentially useful in the induction and maintenance regimens, given the ease at which these reagents can be generated and managed. Here, we will review those immune-based therapeutic interventions and highlight active areas of research investigations toward fulfillment of the great promise of immunotherapy to AML.
Collapse
Affiliation(s)
- Fabiana Perna
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, USA.
| | - Manuel R Espinoza-Gutarra
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, USA
| | - Giuseppe Bombaci
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, USA
| | - Sherif S Farag
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, USA
| | - Jennifer E Schwartz
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, USA
| |
Collapse
|
14
|
Tarannum M, Romee R, Shapiro RM. Innovative Strategies to Improve the Clinical Application of NK Cell-Based Immunotherapy. Front Immunol 2022; 13:859177. [PMID: 35401529 PMCID: PMC8990319 DOI: 10.3389/fimmu.2022.859177] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/28/2022] [Indexed: 12/31/2022] Open
Abstract
Natural killer cells constitute a part of the innate immune system that mediates an effective immune response towards virus-infected and malignant cells. In recent years, research has focused on exploring and advancing NK cells as an active immunotherapy platform. Despite major advances, there are several key challenges that need to be addressed for the effective translation of NK cell research to clinical applications. This review highlights some of these challenges and the innovative strategies being developed to overcome them, including in vitro expansion, in vivo persistence, infiltration to the tumor site, and prevention of exhaustion.
Collapse
Affiliation(s)
- Mubin Tarannum
- Division of Stem Cell Transplant and Cellular Therapy, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Rizwan Romee
- Division of Stem Cell Transplant and Cellular Therapy, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Roman M Shapiro
- Division of Stem Cell Transplant and Cellular Therapy, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
15
|
Novel insights in CAR-NK cells beyond CAR-T cell technology; promising advantages. Int Immunopharmacol 2022; 106:108587. [PMID: 35149294 DOI: 10.1016/j.intimp.2022.108587] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
Abstract
CAR-T (chimeric antigen receptor T cell) technology, which has recently showed successful results in the treatment of hematological tumors, has been the focus of attention as one of the most potent approaches in tumor immunotherapy. However, side effects and limitations of this application, such as the risk of graft versus host disease (GvHD), make it challenging to be as accessible as other treatments. Natural killer cells (NK) could be transplanted without alloreactivity, making them as an off-the-shelf product. CAR-NK (chimeric antigen receptor NK cell) therapy can circumvent some serious limitations of CAR-T cell therapy. Application of CAR-NK cells have some considerable advantages over CAR-T cells. These include lack of cytokine release syndrome (CRS), neurotoxicity, and GvHD when using allogenic CAR-T cell. These features lessen the risk of tumor antigen loss and disease relapse. Moreover, NK cells which were derived from different sources, can make the CAR therapy more feasible. In this narrative review, we outlined the key features of CAR-NK cells as an alternative to CAR-T cell therapy in cancer immunotherapy and highlighted the main advantages.
Collapse
|
16
|
Karmakar S, Pal P, Lal G. Key Activating and Inhibitory Ligands Involved in the Mobilization of Natural Killer Cells for Cancer Immunotherapies. Immunotargets Ther 2021; 10:387-407. [PMID: 34754837 PMCID: PMC8570289 DOI: 10.2147/itt.s306109] [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: 08/11/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are the most potent arm of the innate immune system and play an important role in immunity, alloimmunity, autoimmunity, and cancer. NK cells recognize “altered-self” cells due to oncogenic transformation or stress due to viral infection and target to kill them. The effector functions of NK cells depend on the interaction of the activating and inhibitory receptors on their surface with their cognate ligand expressed on the target cells. These activating and inhibitory receptors interact with major histocompatibility complex I (MHC I) expressed on the target cells and make decisions to mount an immune response. NK cell immune response includes cytolytic activity and secretion of cytokines to help with the ongoing immune response. The advancement of our knowledge on the expression of inhibitory and activating molecules led us to exploit these molecules in the treatment of cancer. This review discusses the importance of activating and inhibitory receptors on NK cells and their clinical importance in cancer immunotherapy.
Collapse
Affiliation(s)
- Surojit Karmakar
- National Centre for Cell Science (NCCS), Pune, MH, 411007, India
| | - Pradipta Pal
- National Centre for Cell Science (NCCS), Pune, MH, 411007, India
| | - Girdhari Lal
- National Centre for Cell Science (NCCS), Pune, MH, 411007, India
| |
Collapse
|
17
|
Isidori A, Cerchione C, Daver N, DiNardo C, Garcia-Manero G, Konopleva M, Jabbour E, Ravandi F, Kadia T, Burguera ADLF, Romano A, Loscocco F, Visani G, Martinelli G, Kantarjian H, Curti A. Immunotherapy in Acute Myeloid Leukemia: Where We Stand. Front Oncol 2021; 11:656218. [PMID: 34041025 PMCID: PMC8143531 DOI: 10.3389/fonc.2021.656218] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
In the past few years, our improved knowledge of acute myeloid leukemia (AML) pathogenesis has led to the accelerated discovery of new drugs and the development of innovative therapeutic approaches. The role of the immune system in AML development, growth and recurrence has gained increasing interest. A better understanding of immunological escape and systemic tolerance induced by AML blasts has been achieved. The extraordinary successes of immune therapies that harness the power of T cells in solid tumors and certain hematological malignancies have provided new stimuli in this area of research. Accordingly, major efforts have been made to develop immune therapies for the treatment of AML patients. The persistence of leukemia stem cells, representing the most relevant cause of relapse, even after allogeneic stem cell transplant (allo-SCT), remains a major hurdle in the path to cure for AML patients. Several clinical trials with immune-based therapies are currently ongoing in the frontline, relapsed/refractory, post-allo-SCT and minimal residual disease/maintenance setting, with the aim to improve survival of AML patients. This review summarizes the available data with immune-based therapeutic modalities such as monoclonal antibodies (naked and conjugated), T cell engagers, adoptive T-cell therapy, adoptive-NK therapy, checkpoint blockade via PD-1/PD-L1, CTLA4, TIM3 and macrophage checkpoint blockade via the CD47/SIRPa axis, and leukemia vaccines. Combining clinical results with biological immunological findings, possibly coupled with the discovery of biomarkers predictive for response, will hopefully allow us to determine the best approaches to immunotherapy in AML.
Collapse
Affiliation(s)
| | - Claudio Cerchione
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Naval Daver
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, United States
| | - Courtney DiNardo
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, United States
| | | | - Marina Konopleva
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, United States
| | - Elias Jabbour
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, United States
| | - Farhad Ravandi
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, United States
| | - Tapan Kadia
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, United States
| | | | - Alessandra Romano
- Dipartimento di Chirurgia e Specialità Medico-Chirurgiche, Sezione di Ematologia, Università degli Studi di Catania, Catania, Italy
| | | | - Giuseppe Visani
- Haematology and Stem Cell Transplant Center, AORMN, Pesaro, Italy
| | - Giovanni Martinelli
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Hagop Kantarjian
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Antonio Curti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| |
Collapse
|
18
|
Sheppard S, Sun JC. Virus-specific NK cell memory. J Exp Med 2021; 218:211913. [PMID: 33755720 PMCID: PMC7992500 DOI: 10.1084/jem.20201731] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/25/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
NK cells express a limited number of germline-encoded receptors that identify infected or transformed cells, eliciting cytotoxicity, effector cytokine production, and in some circumstances clonal proliferation and memory. To maximize the functional diversity of NK cells, the array and expression level of surface receptors vary between individual NK cell “clones” in mice and humans. Cytomegalovirus infection in both species can expand a population of NK cells expressing receptors critical to the clearance of infected cells and generate a long-lived memory pool capable of targeting future infection with greater efficacy. Here, we discuss the pathways and factors that regulate the generation and maintenance of effector and memory NK cells and propose how this understanding may be harnessed therapeutically.
Collapse
Affiliation(s)
- Sam Sheppard
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Joseph C Sun
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY.,Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY
| |
Collapse
|
19
|
Lee H, Da Silva IP, Palendira U, Scolyer RA, Long GV, Wilmott JS. Targeting NK Cells to Enhance Melanoma Response to Immunotherapies. Cancers (Basel) 2021; 13:cancers13061363. [PMID: 33802954 PMCID: PMC8002669 DOI: 10.3390/cancers13061363] [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] [Received: 02/24/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/23/2022] Open
Abstract
Natural killer (NK) cells are a key component of an innate immune system. They are important not only in initiating, but also in augmenting adaptive immune responses. NK cell activation is mediated by a carefully orchestrated balance between the signals from inhibitory and activating NK cell receptors. NK cells are potent producers of proinflammatory cytokines and are also able to elicit strong antitumor responses through secretion of perforin and granzyme B. Tumors can develop many mechanisms to evade NK cell antitumor responses, such as upregulating ligands for inhibitory receptors, secreting anti-inflammatory cytokines and recruiting immunosuppressive cells. Enhancing NK cell responses will likely augment the effectiveness of immunotherapies, and strategies to accomplish this are currently being evaluated in clinical trials. A comprehensive understanding of NK cell biology will likely provide additional opportunities to further leverage the antitumor effects of NK cells. In this review, we therefore sought to highlight NK cell biology, tumor evasion of NK cells and clinical trials that target NK cells.
Collapse
Affiliation(s)
- Hansol Lee
- Melanoma Institute Australia, The University of Sydney, Sydney 2006, Australia; (H.L.); (I.P.D.S.); (U.P.); (R.A.S.); (J.S.W.)
- Faculty of Medicine and Health Sciences, The University of Sydney, Sydney 2006, Australia
| | - Inês Pires Da Silva
- Melanoma Institute Australia, The University of Sydney, Sydney 2006, Australia; (H.L.); (I.P.D.S.); (U.P.); (R.A.S.); (J.S.W.)
| | - Umaimainthan Palendira
- Melanoma Institute Australia, The University of Sydney, Sydney 2006, Australia; (H.L.); (I.P.D.S.); (U.P.); (R.A.S.); (J.S.W.)
- Department of Infectious Diseases and Immunology, The Charles Perkins Centre, School of Medical Sciences, The University of Sydney, Sydney 2006, Australia
| | - Richard A. Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney 2006, Australia; (H.L.); (I.P.D.S.); (U.P.); (R.A.S.); (J.S.W.)
- Faculty of Medicine and Health Sciences, The University of Sydney, Sydney 2006, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney 2006, Australia
| | - Georgina V. Long
- Melanoma Institute Australia, The University of Sydney, Sydney 2006, Australia; (H.L.); (I.P.D.S.); (U.P.); (R.A.S.); (J.S.W.)
- Department of Medical Oncology, Royal North Shore Hospital and Mater Hospital, Sydney 2065, Australia
- Sydney Medical School, The University of Sydney, Sydney 2006, Australia
- Correspondence: ; Tel.: +61-2-9911-7336
| | - James S. Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney 2006, Australia; (H.L.); (I.P.D.S.); (U.P.); (R.A.S.); (J.S.W.)
- Faculty of Medicine and Health Sciences, The University of Sydney, Sydney 2006, Australia
| |
Collapse
|
20
|
Esteves AM, Papaevangelou E, Dasgupta P, Galustian C. Combination of Interleukin-15 With a STING Agonist, ADU-S100 Analog: A Potential Immunotherapy for Prostate Cancer. Front Oncol 2021; 11:621550. [PMID: 33777767 PMCID: PMC7988118 DOI: 10.3389/fonc.2021.621550] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/12/2021] [Indexed: 01/08/2023] Open
Abstract
Prostate cancer is the second most commonly diagnosed cancer in men with mortality rates, overtaking those for breast cancer in the last 2 years in the UK. Despite advances in prostate cancer treatments, over 25% of men do not survive over 5 years with advanced disease. Due to the success of immunotherapies in treating other cancers, this treatment modality has been investigated for Prostate cancer, however, the sole FDA approved immunotherapy so far (Provenge™) only extends life by a few months. Therefore, finding immunotherapeutic agents to treat prostate cancer is of major interest. Our group has previously shown that Interleukin-15 (IL-15), unlike other therapeutic cytokines such as IL-2 and IL-12, can stimulate expansion and activity of CD8 T cells and NK cells in vitro when they are exposed to prostate cancer cells, while studies in mice have shown a 50% reduction in tumor size with no apparent toxicity. In this study, we aim to examine potencies of IL-15 in combination with a cyclic dinucleotide (CDN) that activates the Stimulator of Interferon-Gene (STING) receptor. Selected CDNs (also known as STING agonists) have previously been shown to activate both T cells and dendritic cells through STING. We hypothesize that the combination of STING agonists and IL-15 can additively increase NK and T cell activity as they act to increase type I interferons (IFNs) through STING activation and IFN-γ through IL-15. In prostate cancer-lymphocyte co-cultures we now show that combination of IL-15 and the STING agonist ADU-S100 analog induces a marked killing of cancer cells above that seen with IL-15 or ADU-S100 alone. We show that this is related to a potent activation of NK cells resulting in increased perforin and CD69 expression, and up to a 13-fold increase in IFNγ secretion in the co-cultures. NK cells are responsible for killing of the cancer cells, as shown by a lack of cytotoxicity in NK depleted lymphocyte-tumor cell co-cultures, or in co-cultures of B and T cells with tumor cells. In summary, we propose that the combination of IL-15 and the sting agonist ADU-S100 analog may be potently effective in treatment of prostate cancer.
Collapse
Affiliation(s)
- Ana M Esteves
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Efthymia Papaevangelou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Prokar Dasgupta
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom.,Urology Centre, Guy's Hospital, London, United Kingdom
| | - Christine Galustian
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom
| |
Collapse
|
21
|
Liu S, Galat V, Galat Y, Lee YKA, Wainwright D, Wu J. NK cell-based cancer immunotherapy: from basic biology to clinical development. J Hematol Oncol 2021; 14:7. [PMID: 33407739 PMCID: PMC7788999 DOI: 10.1186/s13045-020-01014-w] [Citation(s) in RCA: 396] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cell is a specialized immune effector cell type that plays a critical role in immune activation against abnormal cells. Different from events required for T cell activation, NK cell activation is governed by the interaction of NK receptors with target cells, independent of antigen processing and presentation. Due to relatively unsophisticated cues for activation, NK cell has gained significant attention in the field of cancer immunotherapy. Many efforts are emerging for developing and engineering NK cell-based cancer immunotherapy. In this review, we provide our current understandings of NK cell biology, ongoing pre-clinical and clinical development of NK cell-based therapies and discuss the progress, challenges, and future perspectives.
Collapse
Affiliation(s)
- Sizhe Liu
- Department of Urology, Feinberg School of Medicine, Northwestern University, 303 E. Superior St., Lurie Research Building 6-117, Chicago, IL, 60611, USA
| | - Vasiliy Galat
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pediatrics, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yekaterina Galat
- Department of Pediatrics, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | | | - Derek Wainwright
- Departments of Neurological Surgery, Medicine-Hematology and Oncology, Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jennifer Wu
- Department of Urology, Feinberg School of Medicine, Northwestern University, 303 E. Superior St., Lurie Research Building 6-117, Chicago, IL, 60611, USA.
| |
Collapse
|
22
|
Song X, Xu C, Wu X, Zhao X, Fan J, Meng S. The potential markers of NK-92 associated to cytotoxicity against K562 cells. Biologicals 2020; 68:46-53. [PMID: 32933840 DOI: 10.1016/j.biologicals.2020.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 01/21/2020] [Accepted: 08/19/2020] [Indexed: 12/28/2022] Open
Abstract
Markers associated to NK cytolytic activity are in a great need to regulate NK cell immunotherapy products. We assume that biomarkers which response to cytolysis will change their transcription, expression or secretion. To find NK-92 indicator to cytolytic activity, we have evaluated the potential markers by quantifying the expression of well-known cytotoxicity functional molecules (cytokine IFN-γ, Granzyme B, perforin, CD69 and CD107a), and explored candidate markers by a sweeping transcription picture of NK-92 using a direct cytolysis model (incubation with K562). We found that IFN-γ secretion was highly correlated to cytotoxicity of NK-92, neither Granzyme B, perforin secretion, nor CD69, CD107a positive population were upregulated by K562 stimulation. RNAseq revealed 432 genes expression changed during cytolysis, several genes (BIRC3, CSF2, VCAM1 and TNFRSF9) mRNA expression were validated by real time RT-PCR under K562 being killed or protected from being killed conditions. Results suggested IFN-γ secretion, BIRC3 and TNFRSF9 transcription in NK-92 were responsive to K562 cytolysis. In a word, our results confirmed one marker and reveal an array of novel candidate markers associated with NK-92 cytotoxicity. Further studies are greatly needed to determine the roles these new makers play in NK-92 cytolysis process.
Collapse
Affiliation(s)
- Xue Song
- National Cell Collection and Research Center, Institute for Biological Product Control, National Institutes for Food and Drug Control, Tiantan Xili, Dongcheng District, Beijing, 100050, China
| | - Chongfeng Xu
- National Cell Collection and Research Center, Institute for Biological Product Control, National Institutes for Food and Drug Control, Tiantan Xili, Dongcheng District, Beijing, 100050, China
| | - Xueling Wu
- National Cell Collection and Research Center, Institute for Biological Product Control, National Institutes for Food and Drug Control, Tiantan Xili, Dongcheng District, Beijing, 100050, China
| | - Xiang Zhao
- National Cell Collection and Research Center, Institute for Biological Product Control, National Institutes for Food and Drug Control, Tiantan Xili, Dongcheng District, Beijing, 100050, China
| | - Jinping Fan
- National Cell Collection and Research Center, Institute for Biological Product Control, National Institutes for Food and Drug Control, Tiantan Xili, Dongcheng District, Beijing, 100050, China
| | - Shufang Meng
- National Cell Collection and Research Center, Institute for Biological Product Control, National Institutes for Food and Drug Control, Tiantan Xili, Dongcheng District, Beijing, 100050, China.
| |
Collapse
|
23
|
Natural killer and NKT cells in the male reproductive tract. J Reprod Immunol 2020; 142:103178. [PMID: 32739646 DOI: 10.1016/j.jri.2020.103178] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/01/2020] [Accepted: 07/09/2020] [Indexed: 01/01/2023]
Abstract
Natural killer (NK) cells are important effector lymphocytes that play a pivotal role in the innate and adaptive immune responses to tumors and viral infection. NKT cells are a heterogeneous group of T cells that share properties with both T cells and NK cells. They display immunoregulatory properties as they facilitate the cell-mediated immune response to tumors and infectious diseases, and inhibit cell-mediated immunity associated with autoimmune diseases and allograft rejection. However, the roles of NK and NKT cells in the male reproductive tract remain largely unexplored, in particular, NKT cells, tissue distribution, and state of health or disease. Infection and inflammation of the male genital tract are thought to be the primary etiological factors of male infertility. In this review, we considered this complex and rapidly growing field. We summarize the recent findings and the characterization and roles of NK and NKT cells in the male reproductive tract, including the testis, epididymis, prostate, seminal vesicle, and semen, to enhance our understanding of the immunological mechanisms of male infertility and for the design effective vaccines for male reproductive health in the future.
Collapse
|
24
|
Villalba M, Alexia C, Bellin-Robert A, Fayd'herbe de Maudave A, Gitenay D. Non-Genetically Improving the Natural Cytotoxicity of Natural Killer (NK) Cells. Front Immunol 2020; 10:3026. [PMID: 31998309 PMCID: PMC6970430 DOI: 10.3389/fimmu.2019.03026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022] Open
Abstract
The innate lymphocyte lineage natural killer (NK) is now the target of multiple clinical applications, although none has received an agreement from any regulatory agency yet. Transplant of naïve NK cells has not proven efficient enough in the vast majority of clinical trials. Hence, new protocols wish to improve their medical use by producing them from stem cells and/or modifying them by genetic engineering. These techniques have given interesting results but these improvements often hide that natural killers are mainly that: natural. We discuss here different ways to take advantage of NK physiology to improve their clinical activity without the need of additional modifications except for in vitro activation and expansion and allograft in patients. Some of these tactics include combination with monoclonal antibodies (mAb), drugs that change metabolism and engraftment of specific NK subsets with particular activity. Finally, we propose to use specific NK cell subsets found in certain patients that show increase activity against a specific disease, including the use of NK cells derived from patients.
Collapse
Affiliation(s)
- Martin Villalba
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France.,IRMB, CHU Montpellier, Montpellier, France
| | - Catherine Alexia
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | | | | | - Delphine Gitenay
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
| |
Collapse
|
25
|
Sakellariou C, Elhage O, Papaevangelou E, Giustarini G, Esteves AM, Smolarek D, Smith RA, Dasgupta P, Galustian C. Prostate cancer cells enhance interleukin-15-mediated expansion of NK cells. BJU Int 2019; 125:89-102. [PMID: 31392791 DOI: 10.1111/bju.14893] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To identify cytokines that can activate and expand NK cells in the presence of prostate cancer cells in order to determine whether these agents may be useful in future intra-tumoural administration in pre-clinical and clinical prostate cancer trials. MATERIALS AND METHODS Lymphocytes isolated from normal donor blood were set up in co-cultures with either cancer or non-cancerous prostate cell lines, together with each of the cytokines interleukin (IL)-2, IL-12, IL-15, interferon (IFN)-γ or IL-21 for a period of 7 days. Then, expansion of NK cells, NKT cells and CD8 T cells was measured by flow cytometry and compared with the expansion of the same cells in the absence of prostate cells. The cytotoxic activity of NK cells, as measured by perforin and tumour cell killing, was also assessed. NK cell receptors and their corresponding ligands on prostate tumour cells were analysed to determine whether any of these were modulated by co-culture. The role of the tumour-secreted heat shock proteins HSP90 and HSP70 in the expansion of NK cells in the co-cultures was also investigated because of their effects on NK and CD8 T-cell activation. RESULTS We showed that, among a panel of cytokines known to cause NK cell activation and expansion, only IL-15 could actively induce expansion of NK, NKT and CD8 T cells in the presence of prostate cancer cell lines. Furthermore, the expansion of NK cells was far greater (up to 50% greater) in the presence of the cancer cells (LNCaP, PC3) than when lymphocytes were incubated alone. In contrast, non-cancerous cell lines (PNT2 and WPMY-1) did not exert any expansion of NK cells. The cytolytic activity of the NK cells, as measured by perforin, CD107a and killing of tumour cells, was also greatest in co-cultures with IL-15. Examination of NK cell receptors shows that NKG2D is upregulated to a greater degree in the presence of prostate cancer cells, compared with the upregulation with IL-15 in lymphocytes alone. However, blocking of NKG2D does not inhibit the enhanced expansion of NK cells in the presence of tumour cells. CONCLUSIONS Among a panel of NK cell-activating cytokines, IL-15 was the only cytokine that could stimulate expansion of NK cells in the presence of prostate cancer cells; therefore IL-15 may be a good candidate for novel future intra-tumoural therapy of the disease.
Collapse
Affiliation(s)
- Christina Sakellariou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | | | - Efthymia Papaevangelou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Giulio Giustarini
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Ana M Esteves
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Dorota Smolarek
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Richard A Smith
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Prokar Dasgupta
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK.,Urology Centre, Guys Hospital, London, UK
| | - Christine Galustian
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| |
Collapse
|
26
|
Sabry M, Zubiak A, Hood SP, Simmonds P, Arellano-Ballestero H, Cournoyer E, Mashar M, Pockley AG, Lowdell MW. Tumor- and cytokine-primed human natural killer cells exhibit distinct phenotypic and transcriptional signatures. PLoS One 2019; 14:e0218674. [PMID: 31242243 PMCID: PMC6594622 DOI: 10.1371/journal.pone.0218674] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/06/2019] [Indexed: 11/19/2022] Open
Abstract
An emerging cellular immunotherapy for cancer is based on the cytolytic activity of natural killer (NK) cells against a wide range of tumors. Although in vitro activation, or “priming,” of NK cells by exposure to pro-inflammatory cytokines, such as interleukin (IL)-2, has been extensively studied, the biological consequences of NK cell activation in response to target cell interactions have not been thoroughly characterized. We investigated the consequences of co-incubation with K562, CTV-1, Daudi RPMI-8226, and MCF-7 tumor cell lines on the phenotype, cytokine expression profile, and transcriptome of human NK cells. We observe the downregulation of several activation receptors including CD16, CD62L, C-X-C chemokine receptor (CXCR)-4, natural killer group 2 member D (NKG2D), DNAX accessory molecule (DNAM)-1, and NKp46 following tumor-priming. Although this NK cell phenotype is typically associated with NK cell dysfunction in cancer, we reveal the upregulation of NK cell activation markers, such as CD69 and CD25; secretion of pro-inflammatory cytokines, including macrophage inflammatory proteins (MIP-1) α /β and IL-1β/6/8; and overexpression of numerous genes associated with enhanced NK cell cytotoxicity and immunomodulatory functions, such as FAS, TNFSF10, MAPK11, TNF, and IFNG. Thus, it appears that tumor-mediated ligation of receptors on NK cells may induce a primed state which may or may not lead to full triggering of the lytic or cytokine secreting machinery. Key signaling molecules exclusively affected by tumor-priming include MAP2K3, MARCKSL1, STAT5A, and TNFAIP3, which are specifically associated with NK cell cytotoxicity against tumor targets. Collectively, these findings help define the phenotypic and transcriptional signature of NK cells following their encounters with tumor cells, independent of cytokine stimulation, and provide insight into tumor-specific NK cell responses to inform the transition toward harnessing the therapeutic potential of NK cells in cancer.
Collapse
Affiliation(s)
- May Sabry
- Department of Haematology, University College London, London, United Kingdom
| | - Agnieszka Zubiak
- Department of Haematology, University College London, London, United Kingdom
| | - Simon P. Hood
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| | - Poppy Simmonds
- Department of Haematology, University College London, London, United Kingdom
| | | | - Eily Cournoyer
- Department of Haematology, University College London, London, United Kingdom
| | - Meghavi Mashar
- Department of Haematology, University College London, London, United Kingdom
| | - A. Graham Pockley
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| | - Mark W. Lowdell
- Department of Haematology, University College London, London, United Kingdom
- InmuneBio Inc., La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
27
|
Wagner AK, Alici E, Lowdell MW. Characterization of human natural killer cells for therapeutic use. Cytotherapy 2019; 21:315-326. [PMID: 30910383 DOI: 10.1016/j.jcyt.2018.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/04/2018] [Indexed: 11/25/2022]
Abstract
As a part of the innate immune system, natural killer (NK) cells are cytotoxic lymphocytes that can exert cytotoxic activity against infected or transformed cells. Furthermore, due to their expression of a functional Fc receptor, they have also been eluded as a major effector fraction in antibody-dependent cellular cytotoxicity. These characteristics have led to multiple efforts to use them for adoptive immunotherapy against various malignancies. There are now at least 70 clinical trials testing the safety and efficacy of NK cell products around the world in early-phase clinical trials. NK cells are also being tested in the context of tumor retargeting via chimeric antigen receptors, other genetic modification strategies, as well as tumor-specific activation strategies such as bispecific engagers with or without cytokine stimulations. One advantage of the use of NK cells for adoptive immunotherapy is their potential to overcome HLA barriers. This has led to a plethora of sources, such as cord blood hematopoietic stem cells and induced pluripotent stem cells, which can generate comparatively high cytotoxic NK cells to peripheral blood counterparts. However, the variety of the sources has led to a heterogeneity in the characterization of the final infusion product. Therefore, in this review, we will discuss a comparative assessment strategy, from characterization of NK cells at collection to final product release by various phenotypic and functional assays, in an effort to predict potency of the cellular product.
Collapse
Affiliation(s)
- Arnika K Wagner
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Evren Alici
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Mark W Lowdell
- Department of Haematology, Cancer Institute, University College London, London, UK.
| |
Collapse
|
28
|
Isvoranu G, Surcel M, Huică RI, Munteanu AN, Pîrvu IR, Ciotaru D, Constantin C, Bratu O, Neagu M, Ursaciuc C. Natural killer cell monitoring in cutaneous melanoma - new dynamic biomarker. Oncol Lett 2019; 17:4197-4206. [PMID: 30944615 PMCID: PMC6444282 DOI: 10.3892/ol.2019.10069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/20/2018] [Indexed: 12/12/2022] Open
Abstract
Melanoma is responsible for most skin cancer deaths in humans. The immune system plays a major role in regulating tumor cell proliferation by initiating defence responses against tumor aggression. Research on murine cancer models allow for a better understanding of immune response in malignancies, revealing specific changes of the immune status in the presence of tumors. Melanoma resistance to conventional therapies and its high immunogenicity justify the development of new therapies. These features reinforce melanoma as a suitable model for studying antitumor immunity. Recent findings on NK cell activation in cancer patients indicate that several important parameters, such as tumor capacity to modulate the function and phenotype of NK cells, require consideration for the choice of an NK-based therapy. In this study, we investigated T-CD4+ and T-CD8+ lymphocytes, B lymphocytes and NK cells in peripheral blood and spleen cells suspension from melanoma-bearing mice compared to healthy controls in order to assess the potential for tumor growth-promoting immunosuppression. Our results indicate that in a melanoma-bearing mouse model the percentage of NK cells in spleen is reduced and that their phenotype is different compared to control mouse NK cells.
Collapse
Affiliation(s)
- Gheorghița Isvoranu
- Animal Husbandry, 'Victor Babeș' National Institute of Pathology, 050096 Bucharest, Romania
| | - Mihaela Surcel
- Immunobiology Laboratory, 'Victor Babeș' National Institute of Pathology, 050096 Bucharest, Romania.,Doctoral School of Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Radu-Ionuț Huică
- Immunobiology Laboratory, 'Victor Babeș' National Institute of Pathology, 050096 Bucharest, Romania.,Division of Cellular and Molecular Biology and Histology, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Adriana Narcisa Munteanu
- Immunobiology Laboratory, 'Victor Babeș' National Institute of Pathology, 050096 Bucharest, Romania.,Doctoral School of Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Ioana Ruxandra Pîrvu
- Immunobiology Laboratory, 'Victor Babeș' National Institute of Pathology, 050096 Bucharest, Romania
| | - Dan Ciotaru
- Immunobiology Laboratory, 'Victor Babeș' National Institute of Pathology, 050096 Bucharest, Romania
| | - Carolina Constantin
- Immunobiology Laboratory, 'Victor Babeș' National Institute of Pathology, 050096 Bucharest, Romania.,Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | - Ovidiu Bratu
- Clinical Department 3, Central Military Hospital, 010825 Bucharest, Romania.,Academy of Romanian Scientists, 050085 Bucharest, Romania
| | - Monica Neagu
- Immunobiology Laboratory, 'Victor Babeș' National Institute of Pathology, 050096 Bucharest, Romania.,Doctoral School of Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania.,Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | - Cornel Ursaciuc
- Immunobiology Laboratory, 'Victor Babeș' National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
29
|
Hood SP, Foulds GA, Imrie H, Reeder S, McArdle SEB, Khan M, Pockley AG. Phenotype and Function of Activated Natural Killer Cells From Patients With Prostate Cancer: Patient-Dependent Responses to Priming and IL-2 Activation. Front Immunol 2019; 9:3169. [PMID: 30761160 PMCID: PMC6362408 DOI: 10.3389/fimmu.2018.03169] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/24/2018] [Indexed: 12/27/2022] Open
Abstract
Background: Although immunotherapy has emerged as the “next generation” of cancer treatments, it has not yet been shown to be successful in the treatment of patients with prostate cancer, for whom therapeutic options remain limited to radiotherapy and androgen (hormone) deprivation therapy. Previous studies have shown that priming natural killer (NK) cells isolated from healthy individuals via co-incubation with CTV-1 cells derived from an acute lymphoblastic leukemia (ALL) enhances their cytotoxicity against human DU145 (metastatic) prostate cancer cells, but it remains unknown to what extent NK cells from patients with prostate cancer can be triggered to kill. Herein, we explore the phenotype of peripheral blood NK cells in patients with prostate cancer and compare the capacity of CTV-1 cell-mediated priming and IL-2 stimulation to trigger NK cell-mediated killing of the human PC3 (metastatic) prostate cancer cell line. Methods: The phenotype of resting, primed (co-incubation with CTV-1 cells for 17 h) and IL-2 activated (100 IU/ml IL-2 for 17 h) NK cells isolated from frozen-thawed peripheral blood mononuclear cell (PBMC) preparations from patients with benign disease (n = 6) and prostate cancer (n = 18) and their cytotoxicity against PC3 and K562 cells was determined by flow cytometry. Relationship(s) between NK cell phenotypic features and cytotoxic potential were interrogated using Spearman Rank correlation matrices. Results and Conclusions: NK cell priming and IL-2 activation of patient-derived NK cells resulted in similar levels of cytotoxicity, but distinct NK cell phenotypes. Importantly, the capacity of priming and IL-2 stimulation to trigger cytotoxicity was patient-dependent and mutually exclusive, in that NK cells from ~50% of patients preferentially responded to priming whereas NK cells from the remaining patients preferentially responded to cytokine stimulation. In addition to providing more insight into the biology of primed and cytokine-stimulated NK cells, this study supports the use of autologous NK cell-based immunotherapies for the treatment of prostate cancer. However, our findings also indicate that patients will need to be stratified according to their potential responsiveness to individual therapeutic approaches.
Collapse
Affiliation(s)
- Simon P Hood
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Gemma A Foulds
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Heather Imrie
- School of Animal Rural and Environmental Sciences, Nottingham Trent University, Nottingham, United Kingdom
| | - Stephen Reeder
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Stéphanie E B McArdle
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Masood Khan
- Department of Urology, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Alan Graham Pockley
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
30
|
Pahl JHW, Cerwenka A, Ni J. Memory-Like NK Cells: Remembering a Previous Activation by Cytokines and NK Cell Receptors. Front Immunol 2018; 9:2796. [PMID: 30546366 PMCID: PMC6279934 DOI: 10.3389/fimmu.2018.02796] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/13/2018] [Indexed: 12/24/2022] Open
Abstract
Natural Killer (NK) cells are cytotoxic innate lymphoid cells serving at the front line against infection and cancer. In inflammatory microenvironments, multiple soluble and contact-dependent signals modulate NK cell responsiveness. Besides their innate cytotoxic and immunostimulatory activity, it has been uncovered in recent years that NK cells constitute a heterogeneous and versatile cell subset. Persistent memory-like NK populations that mount a robust recall response were reported during viral infection, contact hypersensitivity reactions, and after stimulation by pro-inflammatory cytokines or activating receptor pathways. In this review, we highlight recent findings on the generation, functionality, and clinical applicability of memory-like NK cells and describe common features in comparison to other recent concepts of memory NK cells. Understanding of these features will facilitate the conception and design of novel NK cell-based immunotherapies.
Collapse
Affiliation(s)
- Jens H W Pahl
- Department for Immunobiochemistry, Universitätsmedizin Mannheim, Medizinische Fakultät Mannheim, Universität Heidelberg, Mannheim, Germany
| | - Adelheid Cerwenka
- Department for Immunobiochemistry, Universitätsmedizin Mannheim, Medizinische Fakultät Mannheim, Universität Heidelberg, Mannheim, Germany
| | - Jing Ni
- Department for Immunobiochemistry, Universitätsmedizin Mannheim, Medizinische Fakultät Mannheim, Universität Heidelberg, Mannheim, Germany.,Innate Immunity, German Cancer Consortium, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
31
|
Ye W, Chew M, Hou J, Lai F, Leopold SJ, Loo HL, Ghose A, Dutta AK, Chen Q, Ooi EE, White NJ, Dondorp AM, Preiser P, Chen J. Microvesicles from malaria-infected red blood cells activate natural killer cells via MDA5 pathway. PLoS Pathog 2018; 14:e1007298. [PMID: 30286211 PMCID: PMC6171940 DOI: 10.1371/journal.ppat.1007298] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/26/2018] [Indexed: 11/23/2022] Open
Abstract
Natural killer (NK) cells provide the first line of defense against malaria parasite infection. However, the molecular mechanisms through which NK cells are activated by parasites are largely unknown, so is the molecular basis underlying the variation in NK cell responses to malaria infection in the human population. Here, we compared transcriptional profiles of responding and non-responding NK cells following exposure to Plasmodium-infected red blood cells (iRBCs) and identified MDA5, a RIG-I-like receptor involved in sensing cytosolic RNAs, to be differentially expressed. Knockout of MDA5 in responding human NK cells by CRISPR/cas9 abolished NK cell activation, IFN-γ secretion, lysis of iRBCs. Similarly, inhibition of TBK1/IKKε, an effector molecule downstream of MDA5, also inhibited activation of responding NK cells. Conversely, activation of MDA5 by liposome-packaged poly I:C restored non-responding NK cells to lyse iRBCs. We further show that microvesicles containing large parasite RNAs from iRBCs activated NK cells by fusing with NK cells. These findings suggest that NK cells are activated through the MDA5 pathway by parasite RNAs that are delivered to the cytoplasm of NK cells by microvesicles from iRBCs. The difference in MDA5 expression between responding and non-responding NK cells following exposure to iRBCs likely contributes to the variation in NK cell responses to malaria infection in the human population. Malaria is an important parasitic disease with a major public health concern. Malaria pathogenesis involves a complex interplay between parasitic and host factors. A better understanding of early host response and the determinants of immunity are essential to developing innovative therapeutic approaches. Natural killer (NK) cells are important immune cells in protection against malaria infection but show significant differences in their responses in the human population. Here we analyze the differences between human NK cells that respond to and don’t respond to malaria infection. We found that human NK cells that respond to malaria-infected red blood cells (iRBC) have higher levels of a pathogen recognition receptor, MDA5. This receptor is activated by small vesicles released from iRBC. By activating MDA5 with a small molecule agonist, we can improve non-responder NK cells to clear iRBC. Our study provides new insights into the mechanism by which NK cells control malaria infection and possible NK cell-based intervention of malaria infection in human.
Collapse
Affiliation(s)
- Weijian Ye
- School of Biological Sciences, Nanyang Technological University, Singapore.,Singapore-MIT Alliance for Research and Technology, Infectious Disease Interdisciplinary Research Group, Singapore
| | - Marvin Chew
- School of Biological Sciences, Nanyang Technological University, Singapore.,Singapore-MIT Alliance for Research and Technology, Infectious Disease Interdisciplinary Research Group, Singapore
| | - Jue Hou
- Singapore-MIT Alliance for Research and Technology, Infectious Disease Interdisciplinary Research Group, Singapore
| | - Fritz Lai
- Humanized Mouse Unit, Institute of Molecular and Cell Biology, Agency of Science, Technology and Research, Singapore
| | - Stije J Leopold
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Hooi Linn Loo
- Singapore-MIT Alliance for Research and Technology, Infectious Disease Interdisciplinary Research Group, Singapore
| | - Aniruddha Ghose
- Department of Internal Medicine, Chittagong Medical College Hospital, Chittagong, Bangladesh
| | - Ashok K Dutta
- Department of Internal Medicine, Chittagong Medical College Hospital, Chittagong, Bangladesh
| | - Qingfeng Chen
- Humanized Mouse Unit, Institute of Molecular and Cell Biology, Agency of Science, Technology and Research, Singapore
| | - Eng Eong Ooi
- Singapore-MIT Alliance for Research and Technology, Infectious Disease Interdisciplinary Research Group, Singapore.,Duke-National University of Singapore Medical School, Singapore
| | - Nicholas J White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Arjen M Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Peter Preiser
- School of Biological Sciences, Nanyang Technological University, Singapore.,Singapore-MIT Alliance for Research and Technology, Infectious Disease Interdisciplinary Research Group, Singapore
| | - Jianzhu Chen
- Singapore-MIT Alliance for Research and Technology, Infectious Disease Interdisciplinary Research Group, Singapore.,Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| |
Collapse
|
32
|
Doherty E, Rouce RH. Primed to Kill: CTV-1 Stimulated Haploidentical Natural Killer Cells for Consolidation of AML. Biol Blood Marrow Transplant 2018; 24:1533-1535. [PMID: 29933070 DOI: 10.1016/j.bbmt.2018.06.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 06/11/2018] [Indexed: 10/28/2022]
Affiliation(s)
- Erin Doherty
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas
| | - Rayne H Rouce
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas.
| |
Collapse
|
33
|
Fehniger TA, Miller JS, Stuart RK, Cooley S, Salhotra A, Curtsinger J, Westervelt P, DiPersio JF, Hillman TM, Silver N, Szarek M, Gorelik L, Lowdell MW, Rowinsky E. A Phase 1 Trial of CNDO-109-Activated Natural Killer Cells in Patients with High-Risk Acute Myeloid Leukemia. Biol Blood Marrow Transplant 2018; 24:1581-1589. [PMID: 29597002 DOI: 10.1016/j.bbmt.2018.03.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/22/2018] [Indexed: 12/24/2022]
Abstract
Natural killer (NK) cells are an emerging immunotherapy approach to acute myeloid leukemia (AML); however, the optimal approach to activate NK cells before adoptive transfer remains unclear. Human NK cells that are primed with the CTV-1 leukemia cell line lysate CNDO-109 exhibit enhanced cytotoxicity against NK cell-resistant cell lines. To translate this finding to the clinic, CNDO-109-activated NK cells (CNDO-109-NK cells) isolated from related HLA-haploidentical donors were evaluated in a phase 1 dose-escalation trial at doses of 3 × 105 (n = 3), 1 × 106 (n = 3), and 3 × 106 (n = 6) cells/kg in patients with AML in first complete remission (CR1) at high risk for recurrence. Before CNDO-109-NK cell administration, patients were treated with lymphodepleting fludarabine/cyclophosphamide. CNDO-109-NK cells were well tolerated, and no dose-limiting toxicities were observed at the highest tested dose. The median relapse-free survival (RFS) by dose level was 105 (3 × 105), 156 (1 × 106), and 337 (3 × 106) days. Two patients remained relapse-free in post-trial follow-up, with RFS durations exceeding 42.5 months. Donor NK cell microchimerism was detected on day 7 in 10 of 12 patients, with 3 patients having evidence of donor cells on day 14 or later. This trial establishes that CNDO-109-NK cells generated from related HLA haploidentical donors, cryopreserved, and then safely administered to AML patients with transient persistence without exogenous cytokine support. Three durable complete remissions of 32.6 to 47.6+ months were observed, suggesting additional clinical investigation of CNDO-109-NK cells for patients with myeloid malignancies, alone or in combination with additional immunotherapy strategies, is warranted.
Collapse
Affiliation(s)
- Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Robert K Stuart
- Department of Hematology and Oncology, Medical University of South Carolina, Charleston, South Carolina
| | - Sarah Cooley
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Amandeep Salhotra
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Julie Curtsinger
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Peter Westervelt
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | | | | | - Michael Szarek
- Department of Epidemiology and Biostatistics, SUNY Downstate School of Public Health, Brooklyn New York
| | | | - Mark W Lowdell
- Department of Hematology, Royal Free Hospital, UCL Medical School, London, United Kingdom
| | | |
Collapse
|
34
|
Abstract
Translating cellular therapy from the laboratory to the clinic is a complicated process that involves scale-up of procedures to generate clinically relevant cell numbers, adaptation to reagents and equipment that are qualified for human use, establishing parameters of safety for reagents and equipment that are not already qualified for human use, codifying these processes into standards of practice and rules of conduct, and obtaining approval from regulatory bodies based on those codified standards and rules. As the laws and regulations that apply to cellular therapy will vary by time and geography, this chapter reviews some common key principles for the manufacturing of NK cells for human use that will need to be considered within the constraints of local policies and regulations.
Collapse
|
35
|
Granzin M, Wagner J, Köhl U, Cerwenka A, Huppert V, Ullrich E. Shaping of Natural Killer Cell Antitumor Activity by Ex Vivo Cultivation. Front Immunol 2017; 8:458. [PMID: 28491060 PMCID: PMC5405078 DOI: 10.3389/fimmu.2017.00458] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 04/04/2017] [Indexed: 01/11/2023] Open
Abstract
Natural killer (NK) cells are a promising tool for the use in adoptive immunotherapy, since they efficiently recognize and kill tumor cells. In this context, ex vivo cultivation is an attractive option to increase NK cells in numbers and to improve their antitumor potential prior to clinical applications. Consequently, various strategies to generate NK cells for adoptive immunotherapy have been developed. Here, we give an overview of different NK cell cultivation approaches and their impact on shaping the NK cell antitumor activity. So far, the cytokines interleukin (IL)-2, IL-12, IL-15, IL-18, and IL-21 are used to culture and expand NK cells. The selection of the respective cytokine combination is an important factor that directly affects NK cell maturation, proliferation, survival, distribution of NK cell subpopulations, activation, and function in terms of cytokine production and cytotoxic potential. Importantly, cytokines can upregulate the expression of certain activating receptors on NK cells, thereby increasing their responsiveness against tumor cells that express the corresponding ligands. Apart from using cytokines, cocultivation with autologous accessory non-NK cells or addition of growth-inactivated feeder cells are approaches for NK cell cultivation with pronounced effects on NK cell activation and expansion. Furthermore, ex vivo cultivation was reported to prime NK cells for the killing of tumor cells that were previously resistant to NK cell attack. In general, NK cells become frequently dysfunctional in cancer patients, for instance, by downregulation of NK cell activating receptors, disabling them in their antitumor response. In such scenario, ex vivo cultivation can be helpful to arm NK cells with enhanced antitumor properties to overcome immunosuppression. In this review, we summarize the current knowledge on NK cell modulation by different ex vivo cultivation strategies focused on increasing NK cytotoxicity for clinical application in malignant diseases. Moreover, we critically discuss the technical and regulatory aspects and challenges underlying NK cell based therapeutic approaches in the clinics.
Collapse
Affiliation(s)
- Markus Granzin
- Clinical Research, Miltenyi Biotec Inc., Gaithersburg, MD, USA
| | - Juliane Wagner
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents Medicine, Hospital of the Goethe University, Frankfurt, Germany.,LOEWE Center for Cell and Gene Therapy, Cellular Immunology, Goethe University, Frankfurt, Germany
| | - Ulrike Köhl
- Institute of Cellular Therapeutics, Integrated Research and Treatment Center Transplantation, Hannover Medical School, Hannover, Germany
| | - Adelheid Cerwenka
- Innate Immunity Group, German Cancer Research Center, Heidelberg, Germany.,Division of Immunbiochemistry, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Volker Huppert
- R&D Reagents, Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Evelyn Ullrich
- Division for Stem Cell Transplantation and Immunology, Department for Children and Adolescents Medicine, Hospital of the Goethe University, Frankfurt, Germany.,LOEWE Center for Cell and Gene Therapy, Cellular Immunology, Goethe University, Frankfurt, Germany
| |
Collapse
|
36
|
Pal M, Schwab L, Yermakova A, Mace EM, Claus R, Krahl AC, Woiterski J, Hartwig UF, Orange JS, Handgretinger R, André MC. Tumor-priming converts NK cells to memory-like NK cells. Oncoimmunology 2017; 6:e1317411. [PMID: 28680749 PMCID: PMC5486172 DOI: 10.1080/2162402x.2017.1317411] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 03/17/2017] [Accepted: 04/03/2017] [Indexed: 12/25/2022] Open
Abstract
Fascinating earlier evidence suggests an intrinsic capacity of human natural killer (NK) cells to acquire adaptive immune features in the context of cytomegalovirus (CMV) infection or pro-inflammatory cytokine stimulation. Since the role of memory NK cells in cancer has so far remained elusive and adoptive NK cell transfer in relapsing pediatric acute B cell precursor leukemia (BCP-ALL) patients awaits improvement, we asked the question whether tumor-priming could promote the generation of memory NK cells with enhanced graft-vs.-leukemia (GvL) reactivity. Here, we provide substantial evidence that priming of naive human NK cells with pediatric acute B cell leukemia or acute myeloid leukemia specimens induces a functional conversion to tumor-induced memory-like (TIML)-NK cells displaying a heightened tumor-specific cytotoxicity and enhanced perforin synthesis. Cell cycles analyses reveal that tumor-priming sustainably alters the balance between NK cell activation and apoptosis in favor of survival. In addition, gene expression patterns differ between TIML- and cytokine-induced memory-like (CIML)-NK cells with the magnitude of regulated genes being distinctly higher in TIML-NK cells. As such, the tumor-induced conversion of NK cells triggers the emergence of a so far unacknowledged NK cell differentiation stage that might promote GvL effects in the context of adoptive cell transfer.
Collapse
Affiliation(s)
- Marina Pal
- University Children´s Hospital, Dep. of Pediatric Hematology and Oncology, Eberhard Karls University, Tuebingen, Germany
| | - Lisa Schwab
- University Children´s Hospital, Dep. of Pediatric Hematology and Oncology, Eberhard Karls University, Tuebingen, Germany
| | - Anastasiya Yermakova
- Center for Human Immunobiology, Feigin Center, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Emily M. Mace
- Center for Human Immunobiology, Feigin Center, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Rainer Claus
- Department of Medicine, Division of Hematology and Oncology, University of Freiburg Medical Center, Freiburg, Germany
| | - Ann-Christin Krahl
- University Children´s Hospital, Dep. of Pediatric Hematology and Oncology, Eberhard Karls University, Tuebingen, Germany
| | - Jeanette Woiterski
- University Children´s Hospital, Dep. of Pediatric Hematology and Oncology, Eberhard Karls University, Tuebingen, Germany
| | - Udo F. Hartwig
- University Medical Center, 3rd Department of Medicine Hematology, Internal Oncology and Pneumology, Johannes Gutenberg-University, Mainz, Germany
| | - Jordan S. Orange
- Center for Human Immunobiology, Feigin Center, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Rupert Handgretinger
- University Children´s Hospital, Dep. of Pediatric Hematology and Oncology, Eberhard Karls University, Tuebingen, Germany
| | - Maya C. André
- University Children´s Hospital, Dep. of Pediatric Hematology and Oncology, Eberhard Karls University, Tuebingen, Germany
- University Children´s Hospital, Department of Pediatric Intensive Care, Basel, Switzerland
| |
Collapse
|
37
|
Domogala A, Blundell M, Thrasher A, Lowdell MW, Madrigal JA, Saudemont A. Natural killer cells differentiated in vitro from cord blood CD34 + cells are more advantageous for use as an immunotherapy than peripheral blood and cord blood natural killer cells. Cytotherapy 2017; 19:710-720. [PMID: 28428057 DOI: 10.1016/j.jcyt.2017.03.068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/06/2017] [Accepted: 03/13/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND AIMS Natural killer (NK) cells have the potential to become a successful immunotherapy as they can target malignant cells without being direct effectors of graft-versus-host disease. Our group has previously shown that large numbers of functional NK cells can be differentiated in vitro from umbilical cord blood (CB) CD34+ cells. To produce a clinically relevant and effective immunotherapy, we hypothesized that it is essential that the NK cells are able to proliferate and persist in vivo while maintaining an optimal activation status and killing capacity. METHODS We evaluated the proliferation capacity, telomere length and terminal differentiation markers expressed by NK cells differentiated in vitro. We also determined how their cytotoxicity compared with peripheral blood (PB) NK cells and CBNK cells when targeting patient acute myeloid leukemia (AML) blasts and solid tumor cell lines. RESULTS We found that the differentiated NK cells could respond to interleukin-2 and proliferate in vitro. Telomere length was significantly increased, whereas CD57 expression was significantly reduced compared with PBNK cells. The cytotoxicity of the differentiated NK cells was equivalent to that of the PBNK and CBNK cell controls, and priming consistently led to higher levels of killing of patient leukemic blasts and solid tumor cell lines in vitro. Interestingly, this activation step was not required to observe killing of patient AML blasts in vivo. CONCLUSION We are able to generate NK cells from CBCD34+ cells in high numbers, allowing for multiple infusions of highly cytotoxic NK cells that have potential to further proliferate in vivo, making them a desirable product for application as an immunotherapy in the clinic.
Collapse
Affiliation(s)
- Anna Domogala
- Anthony Nolan Research Institute, London, UK; University College London, London, UK
| | | | | | | | - J Alejandro Madrigal
- Anthony Nolan Research Institute, London, UK; University College London, London, UK
| | - Aurore Saudemont
- Anthony Nolan Research Institute, London, UK; University College London, London, UK.
| |
Collapse
|
38
|
Baggio L, Laureano ÁM, Silla LMDR, Lee DA. Natural killer cell adoptive immunotherapy: Coming of age. Clin Immunol 2017; 177:3-11. [DOI: 10.1016/j.clim.2016.02.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 02/06/2016] [Accepted: 02/09/2016] [Indexed: 11/26/2022]
|
39
|
Nocturne G, Boudaoud S, Ly B, Pascaud J, Paoletti A, Mariette X. Impact of anti-TNF therapy on NK cells function and on immunosurveillance against B-cell lymphomas. J Autoimmun 2017; 80:56-64. [PMID: 28214146 DOI: 10.1016/j.jaut.2017.02.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/03/2017] [Accepted: 02/07/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is associated with an increased risk of lymphoma linked to activity of the disease. Immunosuppressive drugs have been suspected to induce an additional risk. Since, NK cells have been recently shown to participate to anti-lymphoma immunosurveillance, we aimed to assess if anti-TNF might impact their anti-lymphoma activity. METHODS NK cells have been assessed ex vivo in patients with RA treated with methotrexate (MTX) with or without anti-TNF. Phenotype has been studied by flow cytometry and function has been assessed after NKp30-cross linking. NK have been cultured 6 days in presence of anti-TNF, TNF-R inhibitors or controls and phenotype has been studied. Then cytotoxicity against 2 B non-Hodgkin lymphoma cell lines [Farage (EBV+) and SU-DHL4 (EBV-)] was assessed. RESULTS Exposure to anti-TNF was associated with a decreased activation of NK cells. NK cells exhibited an impaired function in patients treated with anti-TNF compared to patients treated with MTX alone as assessed by the percentage of degranulation (20.9% [18.5-32.9] vs 31.3% [21.5-49.1], p = 0.04) and a decreased IFN-γ secretion ((17.4% [8.9-25.9] vs to 29.7% [22.5-43.1], p = 0.007). In vitro, exposure to anti-TNF impaired NK cells function and impacted negatively anti-lymphoma activity. These effects may be the consequence of inhibition of TNFR1 signaling. CONCLUSIONS Thus, even if meta-analysis of randomized controlled trials and of registries have not demonstrated to date an increased risk of lymphoma with anti-TNF, cautious must be pursued concerning this possible side effect in patients with long-term anti-TNF exposure.
Collapse
Affiliation(s)
- Gaetane Nocturne
- INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud, Le Kremlin-Bicêtre, France; Department of Rheumatology, AP-HP, Hôpitaux Universitaires Paris-Sud, Le Kremlin-Bicêtre, France.
| | - Saida Boudaoud
- INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud, Le Kremlin-Bicêtre, France
| | - Bineta Ly
- INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud, Le Kremlin-Bicêtre, France
| | - Juliette Pascaud
- INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud, Le Kremlin-Bicêtre, France
| | - Audrey Paoletti
- INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud, Le Kremlin-Bicêtre, France
| | - Xavier Mariette
- INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud, Le Kremlin-Bicêtre, France; Department of Rheumatology, AP-HP, Hôpitaux Universitaires Paris-Sud, Le Kremlin-Bicêtre, France
| |
Collapse
|
40
|
Luna JI, Grossenbacher SK, Murphy WJ, Canter RJ. Targeting Cancer Stem Cells with Natural Killer Cell Immunotherapy. Expert Opin Biol Ther 2016; 17:313-324. [PMID: 27960589 DOI: 10.1080/14712598.2017.1271874] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Standard cytoreductive cancer therapy, such as chemotherapy and radiotherapy, are frequently resisted by a small portion of cancer cells with 'stem-cell' like properties including quiescence and repopulation. Immunotherapy represents a breakthrough modality for improving oncologic outcomes in cancer patients. Since the success of immunotherapy is not contingent on target cell proliferation, it may also be uniquely suited to address the problem of resistance and repopulation exerted by cancer stem cells (CSCs). Areas covered: Natural killer (NK) cells have long been known for their ability to reject allogeneic hematopoietic stem cells, and there are increasing data demonstrating that NK cells can selectively identify and lyse CSCs. The authors review the current knowledge of CSCs and NK cells and highlight recent studies that support the concept that NK cells are capable of targeting CSC in solid tumors, especially in the context of combination therapy simultaneously targeting non-CSCs and CSCs. Expert opinion: Unlike cytotoxic cancer treatments, NK cells can target and eliminate quiescent/non-proliferating cells such as CSCs, and these enigmatic cells are an important source of relapse and metastasis. NK targeting of CSCs represents a novel and potentially high impact method to capitalize on the intrinsic therapeutic potential of NK cells.
Collapse
Affiliation(s)
- Jesus I Luna
- a Department of Dermatology , University of California Davis School of Medicine , Sacramento , CA USA
| | - Steven K Grossenbacher
- a Department of Dermatology , University of California Davis School of Medicine , Sacramento , CA USA
| | - William J Murphy
- a Department of Dermatology , University of California Davis School of Medicine , Sacramento , CA USA.,b Department of Internal Medicine , University of California Davis Medical Center , Sacramento , CA USA
| | - Robert J Canter
- c Division of Surgical Oncology, Department of Surgery , University of California Davis School of Medicine , Sacramento , CA USA
| |
Collapse
|
41
|
Hayashizaki K, Kimura MY, Tokoyoda K, Hosokawa H, Shinoda K, Hirahara K, Ichikawa T, Onodera A, Hanazawa A, Iwamura C, Kakuta J, Muramoto K, Motohashi S, Tumes DJ, Iinuma T, Yamamoto H, Ikehara Y, Okamoto Y, Nakayama T. Myosin light chains 9 and 12 are functional ligands for CD69 that regulate airway inflammation. Sci Immunol 2016; 1:eaaf9154. [PMID: 28783682 DOI: 10.1126/sciimmunol.aaf9154] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/24/2016] [Indexed: 12/12/2022]
Abstract
Recent decades have witnessed a rapid worldwide increase in chronic inflammatory disorders such as asthma. CD4+ T helper 2 cells play critical roles in the pathogenesis of allergic airway inflammation, and CD69 expression on activated CD4 T cells is required to induce allergic inflammation in tissues. However, how CD69 mechanistically controls allergic inflammation remains poorly defined. In lymphoid tissues, CD69 regulates cellular retention through inhibition of S1P1 expression and requires no specific ligands to function. In contrast, we show herein that myosin light chain (Myl) 9 and Myl12 are new functional ligands for CD69. Blockade of CD69-Myl9/12 interaction ameliorates allergic airway inflammation in ovalbumin-induced and house dust mite-induced mouse models of asthma. Within the inflamed mouse airways, we found that the expression of Myl9/12 was increased and that platelet-derived Myl9/12 localized to the luminal surface of blood vessels and formed intravascular net-like structures. Analysis of nasal polyps of eosinophilic chronic rhinosinusitis patients revealed that Myl9/12 expression was increased in inflammatory lesions and was distributed within net-like structures in the intravascular space. In addition, we detected Myl9/12 in perivascular spaces where many CD69+ cells were positioned within Myl9/12 structures. Thus, CD69-Myl9/12 interaction is a key event in the recruitment of activated CD69+ T cells to inflamed tissues and could be a therapeutic target for intractable airway inflammatory diseases.
Collapse
Affiliation(s)
- Koji Hayashizaki
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Motoko Y Kimura
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Koji Tokoyoda
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.,Deutsches Rheuma-Forschungszentrum Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Hiroyuki Hosokawa
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Kenta Shinoda
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Tomomi Ichikawa
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Atsushi Onodera
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Asami Hanazawa
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.,Deutsches Rheuma-Forschungszentrum Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Chiaki Iwamura
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Jungo Kakuta
- KAN Research Institute Inc., 6-8-2 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kenzo Muramoto
- KAN Research Institute Inc., 6-8-2 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Shinichiro Motohashi
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba 260-8670, Japan
| | - Damon J Tumes
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.,South Australian Health and Medical Research Institute, North Terrace, Adelaide SA 5000, Australia
| | - Tomohisa Iinuma
- Department of Otorhinolaryngology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba 260-8670, Japan
| | - Heizaburo Yamamoto
- Department of Otorhinolaryngology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba 260-8670, Japan
| | - Yuzuru Ikehara
- Department of Molecular and Tumor Pathology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba 260-8670, Japan
| | - Yoshitaka Okamoto
- Department of Otorhinolaryngology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba 260-8670, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan. .,Japan Agency for Medical Research and Development (AMED)-Core Research for Evolutionary Medical Science and Technology (CREST), AMED, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| |
Collapse
|
42
|
Austin R, Smyth MJ, Lane SW. Harnessing the immune system in acute myeloid leukaemia. Crit Rev Oncol Hematol 2016; 103:62-77. [PMID: 27247119 DOI: 10.1016/j.critrevonc.2016.04.020] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/13/2016] [Accepted: 04/28/2016] [Indexed: 12/13/2022] Open
|
43
|
Tan JT, Zhong JH, Yang Y, Mao NQ, Liu DS, Huang DM, Zhao YX, Zuo CT. Comparison of postoperative immune function in patients with thoracic esophageal cancer after video-assisted thoracoscopic surgery or conventional open esophagectomy. Int J Surg 2016; 30:155-160. [PMID: 27174507 DOI: 10.1016/j.ijsu.2016.04.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 04/26/2016] [Indexed: 12/29/2022]
Abstract
AIM The aim of the study was to compare postoperative immune function in patients with thoracic esophageal cancer (EC) after video-assisted thoracoscopic surgery (VATS) or conventional open esophagectomy. PATIENTS AND METHODS Medical records were retrospectively analyzed for 228 patients with thoracic EC treated at a single hospital using VATS (n = 52) or conventional open esophagectomy (n = 176). Proportions of CD3(+), CD4(+), CD8(+), and natural kill (NK) cells, as well as the ratio of CD4(+) to CD8(+) cells, were measured in the two groups using flow cytometry on preoperative day (PrD) 1 and postoperative days (PoD) 1 and 7. RESULTS Proportions of CD3(+), CD4(+), and NK cells as well as the CD4+/CD8+ ratio decreased significantly from PrD1 to PoD1 in both the VATS and open esophagectomy groups. In the VATS group, these parameters had returned to preoperative levels (PrD1) by PoD7. These parameters in open esophagectomy group increased from PoD1 to PoD7 but also lowered significantly to PrD1 by PoD7. The proportion of CD8(+) cells was similar between the two groups at all time points tested. CONCLUSION Patients may experience less postoperative immune suppression after VATS than after conventional open esophagectomy, and they may recover preoperative immune function more quickly.
Collapse
Affiliation(s)
- Jun-Tao Tan
- Thoracic Surgery Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, PR China
| | - Jian-Hong Zhong
- Surgery Oncology Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, PR China
| | - Yang Yang
- Hematology Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | - Nai-Qun Mao
- Thoracic Surgery Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, PR China
| | - De-Seng Liu
- Thoracic Surgery Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, PR China
| | - Ding-Ming Huang
- Thoracic Surgery Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, PR China
| | - Yong-Xiang Zhao
- Thoracic Surgery Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, PR China; Immunology Department, Guangxi Medical University, Nanning, PR China.
| | - Chuan-Tian Zuo
- Thoracic Surgery Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, PR China.
| |
Collapse
|
44
|
Rouce RH, Shaim H, Sekine T, Weber G, Ballard B, Ku S, Barese C, Murali V, Wu MF, Liu H, Shpall EJ, Bollard CM, Rabin KR, Rezvani K. The TGF-β/SMAD pathway is an important mechanism for NK cell immune evasion in childhood B-acute lymphoblastic leukemia. Leukemia 2016; 30:800-11. [PMID: 26621337 PMCID: PMC4823160 DOI: 10.1038/leu.2015.327] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 09/28/2015] [Accepted: 10/15/2015] [Indexed: 01/05/2023]
Abstract
Natural killer (NK) cells are key components of the innate immune system, providing potent antitumor immunity. Here, we show that the tumor growth factor-β (TGF-β)/SMAD signaling pathway is an important mechanism for NK cell immune evasion in childhood B-acute lymphoblastic leukemia (ALL). We characterized NK cells in 50 consecutive children with B-ALL at diagnosis, end induction and during maintenance therapy compared with age-matched controls. ALL-NK cells at diagnosis had an inhibitory phenotype associated with impaired function, most notably interferon-γ production and cytotoxicity. By maintenance therapy, these phenotypic and functional abnormalities partially normalized; however, cytotoxicity against autologous blasts remained impaired. We identified ALL-derived TGF-β1 to be an important mediator of leukemia-induced NK cell dysfunction. The TGF-β/SMAD signaling pathway was constitutively activated in ALL-NK cells at diagnosis and end induction when compared with healthy controls and patients during maintenance therapy. Culture of ALL blasts with healthy NK cells induced NK dysfunction and an inhibitory phenotype, mediated by activation of the TGF-β/SMAD signaling pathway, and abrogated by blocking TGF-β. These data indicate that by regulating the TGF-β/SMAD pathway, ALL blasts induce changes in NK cells to evade innate immune surveillance, thus highlighting the importance of developing novel therapies to target this inhibitory pathway and restore antileukemic cytotoxicity.
Collapse
Affiliation(s)
- Rayne H. Rouce
- Texas Children’s Cancer and Hematology Centers/Baylor College of Medicine, Houston
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
| | - Hila Shaim
- Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston
| | - Takuya Sekine
- Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston
| | - Gerrit Weber
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
| | - Brandon Ballard
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
| | - Stephanie Ku
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
| | - Cecilia Barese
- Program for Cell Enhancement and Technologies for Immunotherapy, and Center for Cancer and Immunology Research, Children’s National Health System, Washington, DC
| | - Vineeth Murali
- Texas Children’s Cancer and Hematology Centers/Baylor College of Medicine, Houston
| | - Meng-Fen Wu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
| | - Hao Liu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
| | - Elizabeth J. Shpall
- Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston
| | - Catherine M. Bollard
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston
- Program for Cell Enhancement and Technologies for Immunotherapy, and Center for Cancer and Immunology Research, Children’s National Health System, Washington, DC
| | - Karen R. Rabin
- Texas Children’s Cancer and Hematology Centers/Baylor College of Medicine, Houston
| | - Katayoun Rezvani
- Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston
| |
Collapse
|
45
|
Belting L, Hömberg N, Przewoznik M, Brenner C, Riedel T, Flatley A, Polić B, Busch DH, Röcken M, Mocikat R. Critical role of the NKG2D receptor for NK cell-mediated control and immune escape of B-cell lymphoma. Eur J Immunol 2015; 45:2593-601. [DOI: 10.1002/eji.201445375] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 06/02/2015] [Accepted: 06/19/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Lena Belting
- Institut für Molekulare Immunologie; Helmholtz-Zentrum München; München Germany
| | - Nadine Hömberg
- Institut für Molekulare Immunologie; Helmholtz-Zentrum München; München Germany
| | | | - Christoph Brenner
- Institut für Molekulare Immunologie; Helmholtz-Zentrum München; München Germany
| | - Tanja Riedel
- Institut für Molekulare Immunologie; Helmholtz-Zentrum München; München Germany
| | - Andrew Flatley
- Institut für Molekulare Immunologie; Helmholtz-Zentrum München; München Germany
| | - Bojan Polić
- Department of Histology and Embryology; Faculty of Medicine; University of Rijeka; Rijeka Croatia
| | - Dirk H. Busch
- Institut für Medizinische Mikrobiologie; Immunologie und Hygiene; Technische Universität München; München Germany
| | - Martin Röcken
- Universitäts-Hautklinik, Eberhard-Karls-Universität; Tübingen Germany
| | - Ralph Mocikat
- Institut für Molekulare Immunologie; Helmholtz-Zentrum München; München Germany
| |
Collapse
|
46
|
Two-Stage Priming of Allogeneic Natural Killer Cells for the Treatment of Patients with Acute Myeloid Leukemia: A Phase I Trial. PLoS One 2015; 10:e0123416. [PMID: 26062124 PMCID: PMC4465629 DOI: 10.1371/journal.pone.0123416] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 02/16/2015] [Indexed: 12/04/2022] Open
Abstract
Human Natural Killer (NK) cells require at least two signals to trigger tumor cell lysis. Absence of ligands providing either signal 1 or 2 provides NK resistance. We manufactured a lysate of a tumour cell line which provides signal 1 to resting NK cells without signal 2. The tumor-primed NK cells (TpNK) lyse NK resistant Acute Myeloid Leukemia (AML) blasts expressing signal 2 ligands. We conducted a clinical trial to determine the toxicity of TpNK cell infusions from haploidentical donors. 15 patients with high risk AML were screened, 13 enrolled and 7 patients treated. The remaining 6 either failed to respond to re-induction chemotherapy or the donor refused to undergo peripheral blood apheresis. The conditioning consisted of fludarabine and total body irradiation. This was the first UK trial of a cell therapy regulated as a medicine. The complexity of Good Clinical Practice compliance was underestimated and led to failures requiring retrospective independent data review. The lessons learned are an important aspect of this report. There was no evidence of infusional toxicity. Profound myelosuppression was seen in the majority (median neutrophil recovery day 55). At six months follow-up, three patients treated in Complete Remission (CR) remained in remission, one patient infused in Partial Remission had achieved CR1, two had relapsed and one had died. One year post-treatment one patient remained in CR. Four patients remained in CR after treatment for longer than their most recent previous CR. During the 2 year follow-up six of seven patients died; median overall survival was 400 days post infusion (range 141–910). This is the first clinical trial of an NK therapy in the absence of IL-2 or other cytokine support. The HLA-mismatched NK cells survived and expanded in vivo without on-going host immunosuppression and appeared to exert an anti-leukemia effect in 4/7 patients treated.
Collapse
|
47
|
Domogala A, Madrigal JA, Saudemont A. Natural Killer Cell Immunotherapy: From Bench to Bedside. Front Immunol 2015; 6:264. [PMID: 26089820 PMCID: PMC4453475 DOI: 10.3389/fimmu.2015.00264] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/13/2015] [Indexed: 11/13/2022] Open
Abstract
The potential of natural killer (NK) cells to target numerous malignancies in vitro has been well documented; however, only limited success has been seen in the clinic. Although NK cells prove non-toxic and safe regardless of the cell numbers injected, there is often little persistence and expansion observed in a patient, which is vital for mounting an effective cellular response. NK cells can be isolated directly from peripheral blood, umbilical cord blood, or bone marrow, expanded in vitro using cytokines or differentiated in vitro from hematopoietic stem cells. Drugs that support NK cell function such as lenalidomide and bortezomib have also been studied in the clinic, however, the optimum combination, which can vary among different malignancies, is yet to be identified. NK cell proliferation, persistence, and function can further be improved by various activation techniques such as priming and cytokine addition though whether stimulation pre- or post-injection is more favorable is another obstacle to be tackled. Here, we review the various methods of obtaining and activating NK cells for use in the clinic while considering the ideal product and drug complement for the most successful cellular therapy.
Collapse
Affiliation(s)
- Anna Domogala
- Anthony Nolan Research Institute , London , UK ; University College London , London , UK
| | - J Alejandro Madrigal
- Anthony Nolan Research Institute , London , UK ; University College London , London , UK
| | - Aurore Saudemont
- Anthony Nolan Research Institute , London , UK ; University College London , London , UK
| |
Collapse
|
48
|
Chu Y, Hochberg J, Yahr A, Ayello J, van de Ven C, Barth M, Czuczman M, Cairo MS. Targeting CD20+ Aggressive B-cell Non-Hodgkin Lymphoma by Anti-CD20 CAR mRNA-Modified Expanded Natural Killer Cells In Vitro and in NSG Mice. Cancer Immunol Res 2014; 3:333-44. [PMID: 25492700 DOI: 10.1158/2326-6066.cir-14-0114] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 11/25/2014] [Indexed: 11/16/2022]
Abstract
The prognosis is very dismal for patients with relapsed CD20(+) B-cell non-Hodgkin lymphoma (B-NHL). Facilitating the development of alternative novel therapeutic strategies is required to improve outcomes in patients with recurrent/refractory CD20(+) B-NHL. In this study, we investigated functional activities of anti-CD20 CAR-modified, expanded peripheral blood NK cells (exPBNK) following mRNA nucleofection against CD20(+) B-NHL in vitro and in vivo. CAR(+) exPBNK had significantly enhanced in vitro cytotoxicity, compared with CAR(-) exPBNK against CD20(+) Ramos (P < 0.05), Daudi, Raji, and two rituximab-resistant cell lines, Raji-2R and Raji-4RH (P < 0.001). As expected, there was no significant difference against CD20(-) RS4;11 and Jurkat cells. CD107a degranulation and intracellular IFNγ production were also enhanced in CAR(+) exPBNK in response to CD20(+) B-NHL -: specific stimulation. In Raji-Luc and Raji-2R-Luc xenografted NOD/SCID/γ-chain(-/-) (NSG) mice, the luciferase signals measured in the CAR(+) exPBNK-treated group were significantly reduced, compared with the signals measured in the untreated mice and in mice treated with the CAR(-) exPBNK. Furthermore, the CAR exPBNK-treated mice had significantly extended survival time (P < 0.001) and reduced tumor size, compared with those of the untreated and the CAR(-) exPBNK-treated mice (P < 0.05). These preclinical data suggest that ex vivo-exPBNK modified with anti-CD20 CAR may have therapeutic potential for treating patients with poor-risk CD20(+) hematologic malignancies.
Collapse
Affiliation(s)
- Yaya Chu
- Department of Pediatrics, Maria Fareri Children's Hospital, New York Medical College, Valhalla, New York
| | - Jessica Hochberg
- Department of Pediatrics, Maria Fareri Children's Hospital, New York Medical College, Valhalla, New York
| | - Ashlin Yahr
- Department of Pediatrics, Maria Fareri Children's Hospital, New York Medical College, Valhalla, New York
| | - Janet Ayello
- Department of Pediatrics, Maria Fareri Children's Hospital, New York Medical College, Valhalla, New York
| | - Carmella van de Ven
- Department of Pediatrics, Maria Fareri Children's Hospital, New York Medical College, Valhalla, New York
| | - Matthew Barth
- Department of Pediatrics, State University of New York at Buffalo, Buffalo, New York
| | - Myron Czuczman
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York. Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York
| | - Mitchell S Cairo
- Department of Pediatrics, Maria Fareri Children's Hospital, New York Medical College, Valhalla, New York. Department of Medicine, New York Medical College, Valhalla, New York. Department of Pathology, New York Medical College, Valhalla, New York. Department of Microbiology and Immunology, New York Medical College, Valhalla, New York. Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York.
| |
Collapse
|
49
|
Davies JOJ, Stringaris K, Barrett AJ, Rezvani K. Opportunities and limitations of natural killer cells as adoptive therapy for malignant disease. Cytotherapy 2014; 16:1453-1466. [PMID: 24856895 DOI: 10.1016/j.jcyt.2014.03.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 03/25/2014] [Indexed: 10/25/2022]
Abstract
Although natural killer (NK) cells can be readily generated for adoptive therapy with current techniques, their optimal application to treat malignant diseases requires an appreciation of the dynamic balance between signals that either synergize with or antagonize each other. Individuals display wide differences in NK function that determine their therapeutic efficacy. The ability of NK cells to kill target cells or produce cytokines depends on the balance between signals from activating and inhibitory cell-surface receptors. The selection of NK cells with a predominant activating profile is critical for delivering successful anti-tumor activity. This can be achieved through selection of killer immunoglobulin-like receptor-mismatched NK donors and by use of blocking molecules against inhibitory pathways. Optimum NK cytotoxicity may require licensing or priming with tumor cells. Recent discoveries in the molecular and cellular biology of NK cells inform in the design of new strategies, including adjuvant therapies, to maximize the cytotoxic potential of NK cells for adoptive transfer to treat human malignancies.
Collapse
Affiliation(s)
- James O J Davies
- Department of Haematology, Imperial College London, London, United Kingdom
| | - Kate Stringaris
- Department of Haematology, Imperial College London, London, United Kingdom
| | - A John Barrett
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Katayoun Rezvani
- Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
50
|
Clinical utility of natural killer cells in cancer therapy and transplantation. Semin Immunol 2014; 26:161-72. [PMID: 24618042 DOI: 10.1016/j.smim.2014.02.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 01/30/2014] [Accepted: 02/04/2014] [Indexed: 12/13/2022]
Abstract
Natural killer (NK) cells recognize deranged cells that display stress receptors or loss of major histocompatibility complex (MHC) class I. During development, NK cells become "licensed" only after they encounter cognate human leukocyte antigen (HLA) class I, leading to the acquisition of effector function. NK cells can be exploited for cancer therapy in several ways. These include targeting with monoclonal antibodies alone or combined with ex vivo and in vivo NK cell activation to facilitate adoptive immunotherapy using donor-derived NK cell products to induce graft-vs-tumor effects. In the adoptive transfer setting, persistence and in vivo expansion requires lymphodepleting chemotherapy to prevent rejection and provide homeostatic cytokines (such as IL-15) that activate NK cells. IL-15 has the advantage of avoiding regulatory T-cell expansion. Clinical applications are currently being tested. To enhance in vivo expansion, IL-2 has been used at low doses. However, low dose administration also leads to the stimulation of regulatory T cells. Monoclonal antibodies and bispecific killer engagers (BiKEs) may enhance specificity by targeting CD16 on NK cells to tumor antigens. Inhibition of CD16 shedding may also promote enhanced cytotoxicity. Future strategies include exploiting favorable donor immunogenetics or ex vivo expansion of NK cells from blood, progenitors, or pluripotent cells. Comparative clinical trials are needed to test these approaches.
Collapse
|