scholarly journals Glycoprotein Targeted CAR-NK Cells for the Treatment of SARS-CoV-2 Infection

2021 ◽  
Vol 12 ◽  
Author(s):  
Ilias Christodoulou ◽  
Ruyan Rahnama ◽  
Jonas W. Ravich ◽  
Jaesung Seo ◽  
Sergey N. Zolov ◽  
...  

H84T-Banana Lectin (BanLec) CAR-NK cells bind high mannose glycosites that decorate the SARS-CoV-2 envelope, thereby decreasing cellular infection in a model of SARS-CoV-2. H84T-BanLec CAR-NK cells are innate effector cells, activated by virus. This novel cellular agent is a promising therapeutic, capable of clearing circulating SARS-CoV-2 virus and infected cells. Banana Lectin (BanLec) binds high mannose glycans on viral envelopes, exerting an anti-viral effect. A point mutation (H84T) divorces BanLec mitogenicity from antiviral activity. SARS-CoV-2 contains high mannose glycosites in proximity to the receptor binding domain of the envelope Spike (S) protein. We designed a chimeric antigen receptor (CAR) that incorporates H84T-BanLec as the extracellular moiety. Our H84T-BanLec CAR was devised to specifically direct NK cell binding of SARS-CoV-2 envelope glycosites to promote viral clearance. The H84T-BanLec CAR was stably expressed at high density on primary human NK cells during two weeks of ex vivo expansion. H84T-BanLec CAR-NK cells reduced S-protein pseudotyped lentiviral infection of 293T cells expressing ACE2, the receptor for SARS-CoV-2. NK cells were activated to secrete inflammatory cytokines when in culture with virally infected cells. H84T-BanLec CAR-NK cells are a promising cell therapy for further testing against wild-type SARS-CoV-2 virus in models of SARS-CoV-2 infection. They may represent a viable off-the-shelf immunotherapy for patients suffering from COVID-19.

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2803-2803
Author(s):  
Ilias Christodoulou ◽  
Ruyan Rahnama ◽  
Wesley J. Ravich ◽  
Jaesung Seo ◽  
Sergey Zolov ◽  
...  

Abstract Introduction: Banana Lectin (BanLec) is a glycoprotein-binding lectin derived from banana fruit that has antiviral activity. BanLec binds high mannose glycans expressed on the viral envelopes of HIV, Ebola, influenza, and coronaviruses. BanLec mitogenicity can be divorced from antiviral activity via a single amino acid change (H84T). The SARS-CoV-2 spike (S) protein is decorated with high mannose N-glycosites that are in close proximity to the viral receptor binding domain (RBD). Our goal was to use the H84T-BanLec as the extracellular targeting domain of a chimeric antigen receptor (CAR). We hypothesized that engineering NK cells to express an H84T-BanLec CAR would specifically direct antiviral cytotoxicity against SARS-CoV-2. Methods: H84T-BanLec was synthesized and added to a 4-1BB.ζ CAR by subcloning into an existing retroviral vector. To modify primary human NK cells, CD3-depleted peripheral blood mononuclear cells were first activated with lethally irradiated feeder cells (K562.mbIL15.4-1BBL), then transduced with transiently produced replication incompetent γ-retrovirus carrying the H84T-BanLec.4-1BB.ζ CAR construct. Vector Copy Number (VCN) per cell was measured and CAR protein expression detected with Western blotting. 293T cells were engineered to express human ACE2 (hACE2.293T), the binding receptor for SARS-CoV-2. CAR expression on NK cells and SARS-CoV-2 S-protein binding to hACE2.293T were measured using FACS. S-protein pseudotyped lentivirus carrying a firefly Luciferase (ffLuc) reporter was produced. Viral infectivity was measured using bioluminescence (BL) detection in virally transduced cells. H84T-BanLec CAR NK cells were added to our S-protein pseudotyped lentiviral infectivity assay and degree of inhibited transduction was measured. NK cell activation was assessed with detection of IFNγ and TNFα secretion using ELISA. Results: A median of 4.5 integrated H84T-BanLec CAR copies per cell was measured (range 3.5-7.45, n=4). The CAR was detected by Western blot in NK cell lysates using antibodies to TCRζ and H84T-BanLec. Surface expression of the CAR on primary NK cells was recorded on day 4 after transduction (median [range], 67.5% CAR-positive [64.7-75%], n=6; Fig. 1). CAR expression was maintained on NK cells in culture for 14 days (58.9% CAR-positive [43.6-66.7%], n=6; Fig. 1). ACE2 expression and binding of recombinant S-proteins to hACE2 on hACE2.293T but not parental 293Ts was verified. S-protein pseudotyped lentiviral transduction of hACE2.293T was confirmed with increase in BL from baseline across diminishing viral titer (n=3; Fig. 2). Control 293T cells without hACE2 expression were not transduced, confirming specificity of viral binding and entry dependent on hACE2 (n=3; Fig. 2). S-protein pseudoviral infectivity of hACE2.293T cells was inhibited by both H84T-BanLec CAR-NK and unmodified NK cells, with enhanced inhibition observed in the CAR-NK condition (mean % pseudovirus infectivity +/- SEM of hACE2.293T in co-cultures with unmodified NK vs. H84T-BanLec CAR-NK; 65 +/-11% vs 35%+/- 6% for 1:1 effector-to-target ratio, p=0.05; 78 +/-3% vs 68%+/- 3% for 1:2.5 effector-to-target ratio, p=0.03; n=6; Fig.3). Both unmodified and H84T-BanLec CAR-NK cells were stimulated to secrete inflammatory mediators when co-cultured with pseudoviral particles and virally infected cells. CAR-NK cells showed overall higher cytokine secretion both at baseline and with viral stimulation. Conclusions: A glycoprotein binding H84T-BanLec CAR was stably expressed on the surface of NK cells. CAR-NK cells are activated by SARS-CoV-2 S-pseudovirus and virally infected cells. Viral entry into hACE2 expressing cells was inhibited by H84T-BanLec CAR-NK cells. Translation of H84T-BanLec CAR-NK cells to the clinic may have promise as an effective cellular therapy for SARS-CoV-2 infection. Figure 1 Figure 1. Disclosures Markovitz: University of Michigan: Patents & Royalties: H84T BanLec and of the H84T-driven CAR construct. Bonifant: Merck, Sharpe, Dohme: Research Funding; BMS: Research Funding; Kiadis Pharma: Research Funding.


Biomedicines ◽  
2021 ◽  
Vol 9 (5) ◽  
pp. 557
Author(s):  
Xuewen Deng ◽  
Hiroshi Terunuma ◽  
Mie Nieda

Natural killer (NK) cells are cytotoxic immune cells with an innate capacity for eliminating cancer cells and virus- infected cells. NK cells are critical effector cells in the immunosurveillance of cancer and viral infections. Patients with low NK cell activity or NK cell deficiencies are predisposed to increased risks of cancer and severe viral infections. However, functional alterations of human NK cells are associated with lifestyles and aging. Personal lifestyles, such as cigarette smoking, alcohol consumption, stress, obesity, and aging are correlated with NK cell dysfunction, whereas adequate sleep, moderate exercise, forest bathing, and listening to music are associated with functional healthy NK cells. Therefore, adherence to a healthy lifestyle is essential and will be favorable for immunosurveillance of cancer and viral infections with healthy NK cells.


Vaccines ◽  
2021 ◽  
Vol 9 (11) ◽  
pp. 1363
Author(s):  
Elena V. Abakushina ◽  
Liubov I. Popova ◽  
Andrey A. Zamyatnin ◽  
Jens Werner ◽  
Nikolay V. Mikhailovsky ◽  
...  

In the last decade, an impressive advance was achieved in adoptive cell therapy (ACT), which has improved therapeutic potential and significant value in promising cancer treatment for patients. The ACT is based on the cell transfer of dendritic cells (DCs) and/or immune effector cells. DCs are often used as vaccine carriers or antigen-presenting cells (APCs) to prime naive T cells ex vivo or in vivo. Cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells are used as major tool effector cells for ACT. Despite the fact that NK cell immunotherapy is highly effective and promising against many cancer types, there are still some limitations, including insignificant infiltration, adverse conditions of the microenvironment, the immunosuppressive cellular populations, and the low cytotoxic activity in solid tumors. To overcome these difficulties, novel methods of NK cell isolation, expansion, and stimulation of cytotoxic activity should be designed. In this review, we discuss the basic characteristics of DC vaccines and NK cells as potential adoptive cell preparations in cancer therapy.


2019 ◽  
Vol 18 (1) ◽  
Author(s):  
Kristina S. Burrack ◽  
Geoffrey T. Hart ◽  
Sara E. Hamilton

Abstract Natural killer (NK) cells are important innate effector cells that are well described in their ability to kill virally-infected cells and tumors. However, there is increasing appreciation for the role of NK cells in the control of other pathogens, including intracellular parasites such as Plasmodium, the cause of malaria. NK cells may be beneficial during the early phase of Plasmodium infection—prior to the activation and expansion of antigen-specific T cells—through cooperation with myeloid cells to produce inflammatory cytokines like IFNγ. Recent work has defined how Plasmodium can activate NK cells to respond with natural cytotoxicity, and inhibit the growth of parasites via antibody-dependent cellular cytotoxicity mechanisms (ADCC). A specialized subset of adaptive NK cells that are negative for the Fc receptor γ chain have enhanced ADCC function and correlate with protection from malaria. Additionally, production of the regulatory cytokine IL-10 by NK cells prevents overt pathology and death during experimental cerebral malaria. Now that conditional NK cell mouse models have been developed, previous studies need to be reevaluated in the context of what is now known about other immune populations with similarity to NK cells (i.e., NKT cells and type I innate lymphoid cells). This brief review summarizes recent findings which support the potentially beneficial roles of NK cells during Plasmodium infection in mice and humans. Also highlighted are how the actions of NK cells can be explored using new experimental strategies, and the potential to harness NK cell function in vaccination regimens.


2020 ◽  
Vol 4 (10) ◽  
pp. 2297-2307 ◽  
Author(s):  
Chiara Barberi ◽  
Claudia De Pasquale ◽  
Alessandro Allegra ◽  
Giacomo Sidoti Migliore ◽  
Daniela Oliveri ◽  
...  

Abstract Natural killer (NK) cells represent innate effector cells potentially able to play a role during the immune response against multiple myeloma (MM). To better define the distribution and the specific properties of NK cell subsets during MM disease, we analyzed their features in the bone marrow and peripheral blood of newly diagnosed MM patients. Our findings revealed that, in both compartments, NK cells were more abundant than in healthy donors. Among total MM-NK cells, a significant increase of CD94lowCD56dim NK cell subset was observed, which already appears in clinical precursor conditions leading to MM, namely monoclonal gammopathy of undetermined significance and smoldering MM, and eventually accumulates with disease progression. Moreover, a consistent fraction of CD94lowCD56dim NK cells was in a proliferation phase. When analyzed for their killing abilities, they represented the main cytotoxic NK cell subset against autologous MM cells. In vitro, MM cells could rapidly induce the expansion of the CD94lowCD56dim NK cell subset, thus reminiscent of that observed in MM patients. Mechanistically, this accumulation relied on cell to cell contacts between MM and NK cells and required both activation via DNAM-1 and homophilic interaction with CD56 expressed on MM cells. Considering the growing variety of combination treatments aimed at enhancing NK cell-mediated cytotoxicity against MM, these results may also be informative for optimizing current immunotherapeutic approaches.


2014 ◽  
Vol 89 (5) ◽  
pp. 2906-2917 ◽  
Author(s):  
Zeguang Wu ◽  
Christian Sinzger ◽  
Johanna Julia Reichel ◽  
Marlies Just ◽  
Thomas Mertens

ABSTRACTHuman cytomegalovirus (HCMV) transmission within the host is important for the pathogenesis of HCMV diseases. Natural killer (NK) cells are well known to provide a first line of host defense against virus infections. However, the role of NK cells in the control of HCMV transmission is still unknown. Here, we provide the first experimental evidence that NK cells can efficiently control HCMV transmission in different cell types. NK cells engage different mechanisms to control the HCMV transmission both via soluble factors and by cell contact. NK cell-produced interferon gamma (IFN-γ) suppresses HCMV production and induces resistance of bystander cells to HCMV infection. The UL16 viral gene contributes to an immune evasion from the NK cell-mediated control of HCMV transmission. Furthermore, the efficacy of the antibody-dependent NK cell-mediated control of HCMV transmission is dependent on a CD16-158V/F polymorphism. Our findings indicate that NK cells may have a clinical relevance in HCMV infection and highlight the need to consider potential therapeutic strategies based on the manipulation of NK cells.IMPORTANCEHuman cytomegalovirus (HCMV) infects 40% to 100% of the human population worldwide. After primary infection, mainly in childhood, the virus establishes a lifelong persistence with possible reactivations. Most infections remain asymptomatic; however, HCMV represents a major health problem since it is the most frequent cause of infection-induced birth defects and is responsible for high morbidity and mortality in immunocompromised patients. The immune system normally controls the infection by antibodies and immune effector cells. One type of effector cells are the natural killer (NK) cells, which provide a rapid response to virus-infected cells. NK cells participate in viral clearance by inducing the death of infected cells. NK cells also secrete antiviral cytokines as a consequence of the interaction with an infected cell. In this study, we investigated the mechanisms by which NK cells control HCMV transmission, from the perspectives of immune surveillance and immune evasion.


2021 ◽  
Vol 12 ◽  
Author(s):  
Natasha Mupeta Kaweme ◽  
Fuling Zhou

Natural killer (NK) cells are prominent cytotoxic and cytokine-producing components of the innate immune system representing crucial effector cells in cancer immunotherapy. Presently, various NK cell-based immunotherapies have contributed to the substantial improvement in the reconstitution of NK cells against advanced-staged and high-risk AML. Various NK cell sources, including haploidentical NK cells, adaptive NK cells, umbilical cord blood NK cells, stem cell-derived NK cells, chimeric antigen receptor NK cells, cytokine-induced memory-like NK cells, and NK cell lines have been identified. Devising innovative approaches to improve the generation of therapeutic NK cells from the aforementioned sources is likely to enhance NK cell expansion and activation, stimulate ex vivo and in vivo persistence of NK cells and improve conventional treatment response of myeloid leukemia. The tumor-promoting properties of the tumor microenvironment and downmodulation of NK cellular metabolic activity in solid tumors and hematological malignancies constitute a significant impediment in enhancing the anti-tumor effects of NK cells. In this review, we discuss the current NK cell sources, highlight ongoing interventions in enhancing NK cell function, and outline novel strategies to circumvent immunosuppressive factors in the tumor microenvironment to improve the efficacy of NK cell-based immunotherapy and expand their future success in treating myeloid leukemia.


2019 ◽  
Author(s):  
Domenico Viola ◽  
Ada Dona ◽  
Enrico Caserta ◽  
Estelle Troadec ◽  
Emine Gulsen Gunes ◽  
...  

AbstractDaratumumab (Dara), a multiple myeloma (MM) therapy, is an antibody against the surface receptor CD38, which is expressed not only on plasma cells but also on NK cells and monocytes. Correlative data have highlighted the immune-modulatory role of Dara, despite the paradoxical observation that Dara regimens decrease the frequency of total NK cells. Here we show that, despite this reduction, NK cells play a pivotal role in Dara anti-MM activity. CD38 on NK cells is essential for Dara-induced immune modulation, and its expression is restricted to NK cells with effector function. We also show that Dara induces rapid CD38 protein degradation associated with NK cell activation, leaving an activated CD38-negative NK cell population. CD38+ NK cell targeting by Dara also promotes monocyte activation, inducing an increase in T cell costimulatory molecules (CD86/80) and enhancing anti-MM phagocytosis activity ex-vivo and in vivo. In support of Dara’s immunomodulating role, we show that MM patients that discontinued Dara therapy because of progression maintain targetable unmutated surface CD38 expression on their MM cells, but retain effector cells with impaired cellular immune function. In summary, we report that CD38+ NK cells may be an unexplored therapeutic target for priming the immune system of MM patients.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5389-5389
Author(s):  
Susann Szmania ◽  
Amy D Greenway ◽  
Joshuah D Lingo ◽  
Katie Stone ◽  
Junaid Khan ◽  
...  

Abstract CS1 is an ideal target for multiple myeloma (MM) therapy as it is highly expressed on MM while having a very limited expression profile in normal tissues.  Elotuzumab (elo), a humanized monoclonal antibody (mAb) targeting CS1, has an acceptable safety profile and clinical activity in relapsed/refractory MM when combined with the immune modulator lenalidomide (len) and low dose dexamethasone (dex).  The primary mechanism of action for elo is NK cell-mediated antibody-dependent cellular cytotoxicity.  Here we report on a patient who was given elo/len via a single patient IND 6 months after receiving therapy with ex vivo activated auto-ENK cells and low dose IL2 as previously described (Szmania et al, Blood ASH Annual Meeting Abstracts 2012;120:1912).  The patient had relapsing GEP70 high-risk MM with cytogenetic abnormalities and had failed multiple lines of prior therapy including 3 auto-PBSC transplants and further salvage treatments including len, bortezomib, pomalidomide and carfilzomib.   Although ENK cell therapy did not induce a response, subsequent disease progression was slow.  IV elo was started 187 days after ENK cell infusion, and given every 14 days at the currently studied dose of 10mg/kg.  Len at 15mg/day was given on days 1-21 of a 28-day cycle.  Dex premedication (p.o. 38mg; IV, 10mg) was added after a grade 2 infusion reaction was observed to elo dose #1. While on the ENK cell protocol this patient had a dramatic increase in circulating NK cell counts peaking 9 days after infusion (6300 NK/µL, a 48-fold increase from baseline). Although still in the high range, NK cell levels at the time of elo treatment had normalized somewhat (539 NK/ml), and the cell surface expression of key activating receptors was consistent with a resting phenotype.  NK cell count remained stable after the first dose of elo (530 NK/ml) but subsequently dipped to 179 NK/µL after elo dose #2.   Since dex has been reported to affect NK cell counts, it is important to note that an additional dose was taken prior to elo dose #2 due to a travel delay (in total 66 mg of dex was taken on this occasion).  Circulating NK cells (collected pre-elo and 11, 25, and 57 days after elo dose #1) had similar low activity against auto-MM collected prior to elo treatment (effector:target ratio 10:1, 0-5% specific lysis) and killing against MM collected after 5 elo doses was only modestly increased (3-12%).  However, the same circulating NK cells exhibited significantly increased cytolytic ability when additional elo (10µg/mL) was added during the in vitro E:T co-incubation (3-11 fold increase in killing over isotype control, p=0.0008) suggesting that the MM targets were not saturated with mAb. Bound mAb may have been reduced in part during target cell isolation and freeze/thaw.  Freshly prepared auto-ENK cells exhibited an activated immunophenotype and induced significantly higher killing of pre-elo MM (45%) compared to non-expanded NK.  ENK killing was higher still against MM collected after 5 doses of elo (61%).  When elo was present during the assay, ENK demonstrated the most effective killing of auto-MM, reaching levels  equivalent to that of the NK sensitive target K562 (85% vs. 82% lysis).  Successful mAb therapy for MM is now moving forward as target antigens with selective, high and homogeneous expression, such as CS1, are identified.  However, the activity of responding effector cells is a critical issue to consider.  Inadequate NK cell count and activity level has been reported in MM and steroids typically given to debulk and preempt mAb-induced infusion reactions may exacerbate this problem.  Immunomodulatory agents given to enhance immune cell activity are not sufficient to reverse the negative effect of steroids.  We have previously shown that large doses of highly activated auto-ENK cells can be safely infused and that these cells expand further after infusion.  In this study we show that ENK cells have significant activity in vitro against auto-MM and that elo further enhances this activity.  Combination therapy incorporating saturating doses of mAb followed by infusion of NK effector cells with optimized activity against auto-MM is an innovative approach that warrants investigation.  Infusing highly activated effector cells after dex/elo may be one way to reap the benefits of combining these modalities while circumventing steroid-induced immune suppression. Disclosures: Barlogie: Celgene: Consultancy, Honoraria, Research Funding; Myeloma Health, LLC: Patents & Royalties.


2021 ◽  
Author(s):  
Siqin Duan ◽  
Xinfeng Xu ◽  
Jinshen Wang ◽  
Liwen Huang ◽  
Jie Peng ◽  
...  

The complete eradication of human immunodeficiency virus type 1 (HIV-1) is blocked by latent reservoirs in CD4 + T cells and myeloid lineage cells. Toll-like receptors (TLRs) can induce the reversal of HIV-1 latency and trigger the innate immune response. To the best of our knowledge, there is little evidence show the “killing” effect of TLR1/2 agonists but only with a small “shock” potential. To identify a new approach for eradicating the HIV latent reservoir, we evaluated the effectiveness of SMU-Z1, a novel TLR1/2 small molecule agonist, in the “shock and kill” strategy. The results showed that SMU-Z1 can not only enhance latent HIV-1 transcription in ex vivo peripheral blood mononuclear cells (PBMCs) from aviremic HIV-1-infected donors receiving combined antiretroviral therapy (cART) but also in cells of myeloid-monocytic origin in vitro targeting the nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. Interestingly, activation marker CD69 was significantly upregulated in NK cells, B cells, and monocytes 48 hours after SMU-Z1 treatment. Furthermore, SMU-Z1 was able to activate T cells without global T cell activation, as well as increase NK cell degranulation and interferon-gamma (IFN-γ) production which further block HIV-1-infected CD4 lymphocytes. In summary, the present study found that SMU-Z1 can both enhance HIV-1 transcription and promote NK cell-mediated inhibition of HIV-1-infected autologous CD4 + T cells. These findings indicate that novel TLR1/2 agonist SMU-Z1 is a promising latency-reversing agent (LRA) for eradication of HIV-1 reservoirs. IMPORTANCE Multiple in vivo studies have shown that many LRAs implemented in the “shock and kill” approach could activate viral transcription but could not induce “killing” effectively. Therefore, a dual function LRA is needed for elimination of HIV-1 reservoirs. We previously developed a small molecule TLR1/2 agonist, SMU-Z1, and demonstrated that it could upregulate NK cells and CD8 + T cells with immune adjuvant and anti-tumor properties in vivo . In the present study, SMU-Z1 can activate innate immune cells without global T cell activation, induce production of proinflammatory and antiviral cytokines, and enhance the cytotoxic function of NK cells. We showed that SMU-Z1 displayed dual potential ex vivo in the “shock” of exposure of HIV-1 latently infected cells and in the “kill” of clearance of infected cells, which is critical for effective use in combination with therapeutic vaccines or broadly neutralizing antibody treatments aimed at curing AIDS.


Sign in / Sign up

Export Citation Format

Share Document