Cancer Immunotherapy Using Natural Killer Cells: Successes and Challenges of the Field

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Cancer Immunology and Immunotherapy".

Deadline for manuscript submissions: closed (15 January 2020) | Viewed by 65405

Special Issue Editor


E-Mail Website
Guest Editor
School of Dentistry and Medicine, University of California, Los Angeles, CA, USA
Interests: natural killer cells in cancer immune system
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Renewed interest in adoptive NK cell therapy has generated quite a lot of enthusiasm among NK cell biologists in recent years. The demonstration of the ability of NK cells to target cancer stem cells and poorly differentiated tumors that lack or express very low levels of MHC class I has afforded these cells a very unique and indispensable role in cancer immunotherapy. It is also known that cancer patients have significantly suppressed NK cell numbers and function, and indeed, inactivation of NK function has been shown to occur at the pre-neoplastic stages of tumorigenesis, making these cells as one of the most important immune effectors responsible for the curtailment or inhibition of cancer.

However, limitation in the expansion of NK cells to sufficient numbers for clinical use, inability to obtain functional NK cells from different cellular sources, and lack of adequate understanding of the immunobiology of these cells have traditionally hampered the progress in the field of NK cell Immunotherapy. NK and T cells have to operate within a very complex and immunosuppressive tumor microenvironment. To this end, tumor infiltrating fat cells and fibroblasts which constitute important components of stromal cells in addition to MDSCs and Tregs may not only shape the numbers and function of NK cells but also other immune effectors such as CD8+ T cells. Indeed, recent work from our laboratory indicates that obesity in combination with genetic alterations in the pancreas is a driving force for NK cell immunosuppression at the pre-neoplastic stage of tumorigenesis.

Therefore, strategies should be designed to allow maintenance of good NK expansion and function in cancer patients, since NK cells are known to limit the expansion of tumor-associated macrophages (TAMs), Tregs, and MDSCs and result in the removal of CSCs/undifferentiated tumors, all of which are the hallmarks of aggressive tumors. Large numbers of functionally competent NK cells can be combined with other immunotherapeutic strategies, such as oncolytic viruses, ADCC-inducing antibodies, check point inhibitors, CAR-T, CAR-NK, TIL, and chemotherapeutic and radiotherapeutic strategies for the ultimate goal of tumor eradication.

This collection of reviews and original articles presented in this volume highlights the significant advances made in the field of NK cells and potential new directions which should help to advance the field and bring us to the finish line. After all, we should not forget that the beneficiaries are all those who have valiantly fought against cancer and are looking up to the scientific community for treatments that are not only effective in curing their cancer but also allowing them to enjoy a good quality of life.

Prof. Dr. Anahid Jewett
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • NK cells
  • NK-CAR
  • Differentiation
  • Cancer stem cells
  • NK immunotherapy

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

24 pages, 5921 KiB  
Article
Natural Killer Cell Activation Receptor NKp30 Oligomerization Depends on Its N-Glycosylation
by Ondřej Skořepa, Samuel Pazicky, Barbora Kalousková, Jan Bláha, Celeste Abreu, Tomáš Ječmen, Michal Rosůlek, Alexander Fish, Arthur Sedivy, Karl Harlos, Jan Dohnálek, Tereza Skálová and Ondřej Vaněk
Cancers 2020, 12(7), 1998; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12071998 - 21 Jul 2020
Cited by 12 | Viewed by 4266
Abstract
NKp30 is one of the main human natural killer (NK) cell activating receptors used in directed immunotherapy. The oligomerization of the NKp30 ligand binding domain depends on the length of the C-terminal stalk region, but our structural knowledge of NKp30 oligomerization and its [...] Read more.
NKp30 is one of the main human natural killer (NK) cell activating receptors used in directed immunotherapy. The oligomerization of the NKp30 ligand binding domain depends on the length of the C-terminal stalk region, but our structural knowledge of NKp30 oligomerization and its role in signal transduction remains limited. Moreover, ligand binding of NKp30 is affected by the presence and type of N-glycosylation. In this study, we assessed whether NKp30 oligomerization depends on its N-glycosylation. Our results show that NKp30 forms oligomers when expressed in HEK293S GnTI cell lines with simple N-glycans. However, NKp30 was detected only as monomers after enzymatic deglycosylation. Furthermore, we characterized the interaction between NKp30 and its best-studied cognate ligand, B7-H6, with respect to glycosylation and oligomerization, and we solved the crystal structure of this complex with glycosylated NKp30, revealing a new glycosylation-induced mode of NKp30 dimerization. Overall, this study provides new insights into the structural basis of NKp30 oligomerization and explains how the stalk region and glycosylation of NKp30 affect its ligand affinity. This furthers our understanding of the molecular mechanisms involved in NK cell activation, which is crucial for the successful design of novel NK cell-based targeted immunotherapeutics. Full article
Show Figures

Figure 1

23 pages, 4470 KiB  
Article
Probiotic-Treated Super-Charged NK Cells Efficiently Clear Poorly Differentiated Pancreatic Tumors in Hu-BLT Mice
by Kawaljit Kaur, Anna Karolina Kozlowska, Paytsar Topchyan, Meng-Wei Ko, Nick Ohanian, Jessica Chiang, Jessica Cook, Phyu Ou Maung, So-Hyun Park, Nicholas Cacalano, Changge Fang and Anahid Jewett
Cancers 2020, 12(1), 63; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12010063 - 24 Dec 2019
Cited by 37 | Viewed by 4532
Abstract
Background and Aims: We have previously demonstrated that the stage of differentiation of tumors has profound effect on the function of NK cells, and that stem-like/poorly differentiated tumors were preferentially targeted by the NK cells. Therefore, in this study we determined the role [...] Read more.
Background and Aims: We have previously demonstrated that the stage of differentiation of tumors has profound effect on the function of NK cells, and that stem-like/poorly differentiated tumors were preferentially targeted by the NK cells. Therefore, in this study we determined the role of super-charged NK cells in immune mobilization, lysis, and differentiation of stem-like/undifferentiated tumors implanted in the pancreas of humanized-BLT (hu-BLT) mice fed with or without AJ2 probiotics. The phenotype, growth rate and metastatic potential of pancreatic tumors differentiated by the NK cells (NK-differentiated) or patient derived differentiated or stem-like/undifferentiated pancreatic tumors were investigated. Methods: Pancreatic tumor implantation was performed in NSG and hu-BLT mice. Stage of differentiation of tumors was determined using our published criteria for well-differentiated tumors exhibiting higher surface expression of MHC- class I, CD54, and PD-L1 (B7H1) and lower expression of CD44 receptors. The inverse was seen for poorly-differentiated tumors. Results: Stem-like/undifferentiated pancreatic tumors grew rapidly and formed large tumors and exhibited lower expression of above-mentioned differentiation antigens in the pancreas of NSG and hu-BLT mice. Unlike stem-like/undifferentiated tumors, NK-differentiated MP2 (MiaPaCa-2) tumors or patient-derived differentiated tumors were not able to grow or grew smaller tumors, and were unable to metastasize in NSG or hu-BLT mice, and they were susceptible to chemotherapeutic drugs. Stem-like/undifferentiated pancreatic tumors implanted in the pancreas of hu-BLT mice and injected with super-charged NK cells formed much smaller tumors, proliferated less, and exhibited differentiated phenotype. When differentiation of stem-like tumors by the NK cells was prevented by the addition of antibodies to IFN-γ and TNF-α, tumors grew rapidly and metastasized, and they remained resistant to chemotherapeutic drugs. Greater numbers of immune cells infiltrated the tumors of NK-injected and AJ2-probiotic bacteria-fed mice. Moreover, increased IFN-γ secretion in the presence of decreased IL-6 was seen in tumors resected and cultured from NK-injected and AJ2 fed mice. Tumor-induced decreases in NK cytotoxicity and IFN-γ secretion were restored/increased within PBMCs, spleen, and bone marrow when mice received NK cells and were fed with AJ2. Conclusion: NK cells prevent growth of pancreatic tumors through lysis and differentiation, thereby curtailing the growth and metastatic potential of stem-like/undifferentiated-tumors. Full article
Show Figures

Figure 1

Review

Jump to: Research

33 pages, 5071 KiB  
Review
Transcriptional Regulation of Natural Killer Cell Development and Functions
by Dandan Wang and Subramaniam Malarkannan
Cancers 2020, 12(6), 1591; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12061591 - 16 Jun 2020
Cited by 34 | Viewed by 7820
Abstract
Natural killer (NK) cells are the major lymphocyte subset of the innate immune system. Their ability to mediate anti-tumor cytotoxicity and produce cytokines is well-established. However, the molecular mechanisms associated with the development of human or murine NK cells are not fully understood. [...] Read more.
Natural killer (NK) cells are the major lymphocyte subset of the innate immune system. Their ability to mediate anti-tumor cytotoxicity and produce cytokines is well-established. However, the molecular mechanisms associated with the development of human or murine NK cells are not fully understood. Knowledge is being gained about the environmental cues, the receptors that sense the cues, signaling pathways, and the transcriptional programs responsible for the development of NK cells. Specifically, a complex network of transcription factors (TFs) following microenvironmental stimuli coordinate the development and maturation of NK cells. Multiple TFs are involved in the development of NK cells in a stage-specific manner. In this review, we summarize the recent advances in the understandings of TFs involved in the regulation of NK cell development, maturation, and effector function, in the aspects of their mechanisms, potential targets, and functions. Full article
Show Figures

Figure 1

23 pages, 1685 KiB  
Review
Tissue-Resident NK Cells: Development, Maturation, and Clinical Relevance
by Elaheh Hashemi and Subramaniam Malarkannan
Cancers 2020, 12(6), 1553; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12061553 - 12 Jun 2020
Cited by 51 | Viewed by 7153
Abstract
Natural killer (NK) cells belong to type 1 innate lymphoid cells (ILC1) and are essential in killing infected or transformed cells. NK cells mediate their effector functions using non-clonotypic germ-line-encoded activation receptors. The utilization of non-polymorphic and conserved activating receptors promoted the conceptual [...] Read more.
Natural killer (NK) cells belong to type 1 innate lymphoid cells (ILC1) and are essential in killing infected or transformed cells. NK cells mediate their effector functions using non-clonotypic germ-line-encoded activation receptors. The utilization of non-polymorphic and conserved activating receptors promoted the conceptual dogma that NK cells are homogeneous with limited but focused immune functions. However, emerging studies reveal that NK cells are highly heterogeneous with divergent immune functions. A distinct combination of several activation and inhibitory receptors form a diverse array of NK cell subsets in both humans and mice. Importantly, one of the central factors that determine NK cell heterogeneity and their divergent functions is their tissue residency. Decades of studies provided strong support that NK cells develop in the bone marrow. However, evolving evidence supports the notion that NK cells also develop and differentiate in tissues. Here, we summarize the molecular basis, phenotypic signatures, and functions of tissue-resident NK cells and compare them with conventional NK cells. Full article
Show Figures

Figure 1

33 pages, 2854 KiB  
Review
Natural Killer Cells: Tumor Surveillance and Signaling
by Lizeth G. Meza Guzman, Narelle Keating and Sandra E. Nicholson
Cancers 2020, 12(4), 952; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12040952 - 11 Apr 2020
Cited by 50 | Viewed by 9945
Abstract
Natural killer (NK) cells play a pivotal role in cancer immunotherapy due to their innate ability to detect and kill tumorigenic cells. The decision to kill is determined by the expression of a myriad of activating and inhibitory receptors on the NK cell [...] Read more.
Natural killer (NK) cells play a pivotal role in cancer immunotherapy due to their innate ability to detect and kill tumorigenic cells. The decision to kill is determined by the expression of a myriad of activating and inhibitory receptors on the NK cell surface. Cell-to-cell engagement results in either self-tolerance or a cytotoxic response, governed by a fine balance between the signaling cascades downstream of the activating and inhibitory receptors. To evade a cytotoxic immune response, tumor cells can modulate the surface expression of receptor ligands and additionally, alter the conditions in the tumor microenvironment (TME), tilting the scales toward a suppressed cytotoxic NK response. To fully harness the killing power of NK cells for clinical benefit, we need to understand what defines the threshold for activation and what is required to break tolerance. This review will focus on the intracellular signaling pathways activated or suppressed in NK cells and the roles signaling intermediates play during an NK cytotoxic response. Full article
Show Figures

Figure 1

25 pages, 759 KiB  
Review
Mechanisms of Resistance to NK Cell Immunotherapy
by Christian Sordo-Bahamonde, Massimo Vitale, Seila Lorenzo-Herrero, Alejandro López-Soto and Segundo Gonzalez
Cancers 2020, 12(4), 893; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12040893 - 07 Apr 2020
Cited by 34 | Viewed by 8073
Abstract
Immunotherapy has recently been a major breakthrough in cancer treatment. Natural killer (NK) cells are suitable targets for immunotherapy owing to their potent cytotoxic activity that may target cancer cells in a major histocompatibility complex (MHC) and antigen-unrestricted manner. Current therapies targeting NK [...] Read more.
Immunotherapy has recently been a major breakthrough in cancer treatment. Natural killer (NK) cells are suitable targets for immunotherapy owing to their potent cytotoxic activity that may target cancer cells in a major histocompatibility complex (MHC) and antigen-unrestricted manner. Current therapies targeting NK cells include monoclonal antibodies that promote NK cell antibody-dependent cell-mediated cytotoxicity (ADCC), hematopoietic stem cell transplantation (HSCT), the adoptive transfer of NK cells, the redirection of NK cells using chimeric antigen receptor (CAR)-NK cells and the use of cytokines and immunostimulatory drugs to boost the anti-tumor activity of NK cells. Despite some encouraging clinical results, patients receiving these therapies frequently develop resistance, and a myriad of mechanisms of resistance affecting both the immune system and cancer cells have been reported. A first contributing factor that modulates the efficacy of the NK cell therapy is the genetic profile of the individual, which regulates all aspects of NK cell biology. Additionally, the resistance of cancer cells to apoptosis and the immunoediting of cancer cells, a process that decreases their immunogenicity and promotes immunosuppression, are major determinants of the resistance to NK cell therapy. Consequently, the efficacy of NK cell anti-tumor therapy is specific to each patient and disease. The elucidation of such immunosubversive mechanisms is crucial to developing new procedures and therapeutic strategies to fully harness the anti-tumor potential of NK cells. Full article
Show Figures

Figure 1

23 pages, 1844 KiB  
Review
You Have Got a Fast CAR: Chimeric Antigen Receptor NK Cells in Cancer Therapy
by Aline Pfefferle and Nicholas D. Huntington
Cancers 2020, 12(3), 706; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12030706 - 17 Mar 2020
Cited by 75 | Viewed by 9844
Abstract
The clinical success stories of chimeric antigen receptor (CAR)-T cell therapy against B-cell malignancies have contributed to immunotherapy being at the forefront of cancer therapy today. Their success has fueled interest in improving CAR constructs, identifying additional antigens to target, and clinically evaluating [...] Read more.
The clinical success stories of chimeric antigen receptor (CAR)-T cell therapy against B-cell malignancies have contributed to immunotherapy being at the forefront of cancer therapy today. Their success has fueled interest in improving CAR constructs, identifying additional antigens to target, and clinically evaluating them across a wide range of malignancies. However, along with the exciting potential of CAR-T therapy comes the real possibility of serious side effects. While the FDA has approved commercialized CAR-T cell therapy, challenges associated with manufacturing, costs, and related toxicities have resulted in increased attention being paid to implementing CAR technology in innate cytotoxic natural killer (NK) cells. Here, we review the current landscape of the CAR-NK field, from successful clinical implementation to outstanding challenges which remain to be addressed to deliver the full potential of this therapy to more patients. Full article
Show Figures

Figure 1

21 pages, 1945 KiB  
Review
Exosomes: Versatile Nano Mediators of Immune Regulation
by Qi Li, Helei Wang, Hourong Peng, Ting Huyan and Nicholas A. Cacalano
Cancers 2019, 11(10), 1557; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers11101557 - 14 Oct 2019
Cited by 68 | Viewed by 5155
Abstract
One of many types of extracellular vesicles (EVs), exosomes are nanovesicle structures that are released by almost all living cells that can perform a wide range of critical biological functions. Exosomes play important roles in both normal and pathological conditions by regulating cell-cell [...] Read more.
One of many types of extracellular vesicles (EVs), exosomes are nanovesicle structures that are released by almost all living cells that can perform a wide range of critical biological functions. Exosomes play important roles in both normal and pathological conditions by regulating cell-cell communication in cancer, angiogenesis, cellular differentiation, osteogenesis, and inflammation. Exosomes are stable in vivo and they can regulate biological processes by transferring lipids, proteins, nucleic acids, and even entire signaling pathways through the circulation to cells at distal sites. Recent advances in the identification, production, and purification of exosomes have created opportunities to exploit these structures as novel drug delivery systems, modulators of cell signaling, mediators of antigen presentation, as well as biological targeting agents and diagnostic tools in cancer therapy. This review will examine the functions of immunocyte-derived exosomes and their roles in the immune response under physiological and pathological conditions. The use of immunocyte exosomes in immunotherapy and vaccine development is discussed. Full article
Show Figures

Figure 1

19 pages, 266 KiB  
Review
Natural Killer Cell Therapy: A New Treatment Paradigm for Solid Tumors
by Sooyeon Oh, Joo-Ho Lee, KyuBum Kwack and Sang-Woon Choi
Cancers 2019, 11(10), 1534; https://0-doi-org.brum.beds.ac.uk/10.3390/cancers11101534 - 11 Oct 2019
Cited by 55 | Viewed by 7734
Abstract
In treatments of solid tumors, adoptive transfer of ex vivo expanded natural killer (NK) cells has dawned as a new paradigm. Compared with cytotoxic T lymphocytes, NK cells take a unique position targeting tumor cells that evade the host immune surveillance by down-regulating [...] Read more.
In treatments of solid tumors, adoptive transfer of ex vivo expanded natural killer (NK) cells has dawned as a new paradigm. Compared with cytotoxic T lymphocytes, NK cells take a unique position targeting tumor cells that evade the host immune surveillance by down-regulating self-antigen presentation. Recent findings highlighted that NK cells can even target cancer stem cells. The efficacy of allogeneic NK cells has been widely investigated in the treatment of hematologic malignancies. In solid tumors, both autologous and allogeneic NK cells have demonstrated potential efficacy. In allogeneic NK cell therapy, the mismatch between the killer cell immunoglobulin-like receptor (KIR) and human leukocyte antigen (HLA) can be harnessed to increase the antitumor activity. However, the allogeneic NK cells cause more adverse events and can be rejected by the host immune system after repeated injections. In this regard, the autologous NK cell therapy is safer. This article reviews the published results of clinical trials and discusses strategies to enhance the efficacy of the NK cell therapy. The difference in immunophenotype of the ex vivo expanded NK cells resulted from different culture methods may affect the final efficacy. Furthermore, currently available standard anticancer therapy, molecularly targeted agents, and checkpoint inhibitors may directly or indirectly enhance the efficacy of NK cell therapy. A recent study discovered that NK cell specific genetic defects are closely associated with the tumor immune microenvironment that determines clinical outcomes. This finding warrants future investigations to find the implication of NK cell specific genetic defects in cancer development and treatment, and NK cell deficiency syndrome should be revisited to enhance our understanding. Overall, it is clear that NK cell therapy is safe and promises a new paradigm for the treatment of solid tumors. Full article
Back to TopTop