scholarly journals Blockade of Ubiquitin Receptor Rpn13 in Plasmacytoid Dendritic Cells Enhances Anti-Myeloma Immunity

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3098-3098
Author(s):  
Arghya Ray ◽  
Yan Song ◽  
Ting DU ◽  
Dharminder Chauhan ◽  
Kenneth C. Anderson

Introduction Although proteasome inhibitor (PI) based combination therapies achieve remarkable responses multiple myeloma (MM), emergence of PI resistance is common. The mechanism(s) of PI-resistance include tumor-intrinsic factors such as mutations of the 20S proteasomal subunits, and/or tumor-extrinsic cellular components in the BM microenvironment. Interactions of BM accessory cells, immune effector cells, and tumor cells confer both drug-resistance and immune suppression in MM. For example, we showed that interactions of MM plasmacytoid dendritic cells (pDCs) with MM cells and with T/NK cells both confer immune suppression via immune checkpoints, as well as trigger MM cell growth by inducing secretion of MM cell growth factors. We recently reported that targeting proteasome-associated ubiquitin receptor Rpn13 triggers cytotoxicity and overcomes tumor-intrinsic PI-resistance in MM (Song et al, Leukemia 2016;30:1877). Here we utilized our co-culture models of patient pDCs, T cells, NK cells, and autologous MM cells to characterize the immune sequelae of Rpn13 inhibition. Methods Analysis of pDCs activation Purified patient-pDCs (n =7) were treated with Rpn13 inhibitor RA190 (0.05 µM) for 24h, followed by multicolor staining using fluorophore-conjugated Abs against pDC activation/maturation markers CD80, CD83, and CD86. Transient transfections Purified MM patient pDCs were transfected with Rpn13-siRNA using TransIT-X2 transfection Kit,and analyzed for alterations in maturation markers. CTL/NK activity assays Purified MM-BM CD8+ T- or NK-cells (n = 8) were co-cultured with autologous BM-pDCs (pDC:T/NK; 1:10 ratio) for 3 days, in the presence or absence of Rpn13 inhibitor RA190 (100 nM). After washing, cells were cultured for 24h with autologous MM cells pre-stained with CellTracker/CellTrace Violet (10 T/NK:1 MM), followed by 7-AAD staining and quantification of CTL-or NK cell-mediated MM cell lysis by FACS. Results 1) RA190 triggers significant upregulation of maturation markers CD80, CD83, and CD86 on MM-pDCs (fold change vs untreated: CD80: 1.2; p = 0.007; CD83: 2.15; p = 0.006; CD86: 1.4; p = 0.003). In contrast, bortezomib-treated pDCs showed no significant upregulation of these markers. 2) Similar to pharmacological inhibition of Rpn13 with RA190, Rpn13-siRNA increased CD80 (1.76-fold), CD83 (3.12-fold), and CD86 (2.28-fold) expression on MM pDCs (p<0.01). Of note, both RA190 and bortezomib block protein degradation via proteasome, but only RA190 activates pDCs. 3) RA190 treatment increases pDC-induced MM-specific CD8+ CTL activity, as well as NK cell-mediated cytolytic activity against autologous tumor cells, evidenced by decreased viable patient MM cells. 4) Treatment of MM-pDCs with RA190 increases expression of calnexin, a molecular chaperone protein of endoplasmic reticulum which regulates immune co-stimulatory molecules, immune-regulatory signaling, and restores the ability of pDCs to induce proliferation of MM-specific CTLs or NK cells. These findings were also confirmed using pDC cell line CAL-1. Conclusions Our prior findings showed that inhibition of UbR Rpn13 overcomes intrinsic PI-resistance in MM cells. Here we show that targeting Rpn13 also triggers anti-MM immune responses. Rpn13 blockade therefore represents a novel therapeutic approach to overcome both PI-resistance and immune suppression in MM. Disclosures Chauhan: C4 Therapeutics.: Equity Ownership; Stemline Therapeutics: Consultancy. Anderson:Takeda: Consultancy, Speakers Bureau; Celgene: Consultancy, Speakers Bureau; Janssen: Consultancy, Speakers Bureau; Bristol-Myers Squibb: Other: Scientific Founder; Oncopep: Other: Scientific Founder; Amgen: Consultancy, Speakers Bureau; Sanofi-Aventis: Other: Advisory Board.

Cancers ◽  
2021 ◽  
Vol 13 (7) ◽  
pp. 1577
Author(s):  
Matteo Tanzi ◽  
Michela Consonni ◽  
Michela Falco ◽  
Federica Ferulli ◽  
Enrica Montini ◽  
...  

The limited efficacy of Natural Killer (NK) cell-based immunotherapy results in part from the suboptimal expansion and persistence of the infused cells. Recent reports suggest that the generation of NK cells with memory-like properties upon in vitro activation with defined cytokines might be an effective way of ensuring long-lasting NK cell function in vivo. Here, we demonstrate that activation with IL-12, IL-15 and IL-18 followed by a one-week culture with optimal doses of Interleukin (IL-2) and IL-15 generates substantial numbers of memory-like NK cells able to persist for at least three weeks when injected into NOD scid gamma (NSG) mice. This approach induces haploidentical donor-derived memory-like NK cells that are highly lytic against patients’ myeloid or lymphoid leukemia blasts, independent of the presence of alloreactive cell populations in the donor and with negligible reactivity against patients’ non-malignant cells. Memory-like NK cells able to lyse autologous tumor cells can also be generated from patients with solid malignancies. The anti-tumor activity of allogenic and autologous memory-like NK cells is significantly greater than that displayed by NK cells stimulated overnight with IL-2, supporting their potential therapeutic value both in patients affected by high-risk acute leukemia after haploidentical hematopoietic stem cell transplantation and in patients with advanced solid malignancies.


Pharmaceutics ◽  
2021 ◽  
Vol 13 (4) ◽  
pp. 525
Author(s):  
Kwang-Soo Kim ◽  
Dong-Hwan Kim ◽  
Dong-Hyun Kim

Among various immunotherapies, natural killer (NK) cell cancer immunotherapy using adoptive transfer of NK cells takes a unique position by targeting tumor cells that evade the host immune surveillance. As the first-line innate effector cell, it has been revealed that NK cells have distinct mechanisms to both eliminate cancer cells directly and amplify the anticancer immune system. Over the last 40 years, NK cell cancer immunotherapy has shown encouraging reports in pre-clinic and clinic settings. In total, 288 clinical trials are investigating various NK cell immunotherapies to treat hematologic and solid malignancies in 2021. However, the clinical outcomes are unsatisfying, with remained challenges. The major limitation is attributed to the immune-suppressive tumor microenvironment (TME), low activity of NK cells, inadequate homing of NK cells, and limited contact frequency of NK cells with tumor cells. Innovative strategies to promote the cytolytic activity, durable persistence, activation, and tumor-infiltration of NK cells are required to advance NK cell cancer immunotherapy. As maturing nanotechnology and nanomedicine for clinical applications, there is a greater opportunity to augment NK cell therapeutic efficacy for the treatment of cancers. Active molecules/cytokine delivery, imaging, and physicochemical properties of nanoparticles are well equipped to overcome the challenges of NK cell cancer immunotherapy. Here, we discuss recent clinical trials of NK cell cancer immunotherapy, NK cell cancer immunotherapy challenges, and advances of nanoparticle-mediated NK cell therapeutic efficacy augmentation.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3004-3004
Author(s):  
Kathrin Meinhardt ◽  
Ruth Bauer ◽  
Irena Kroeger ◽  
Julia Schneider ◽  
Franziska Ganss ◽  
...  

Abstract Abstract 3004 Clinical studies exploiting the impact of natural killer (NK) cells in allogeneic hematopoietic stem cell transplantation (HSCT) have provided promising results. It is known that NK cells are a heterogeneous population and can be divided into functionally distinct NK cell subpopulations. Murine NK cells can be separated along their expression of CD27 and CD11b and CD117 (c-kit). However, the functional relevance of distinct NK cell subsets in graft-versus-host-disease (GVHD) has not been investigated in detail so far. We have established different protocols for ex vivo isolation and expansion of murine NK cell subpopulations. These NK subsets were further analyzed in vitro and in vivo in an allogeneic murine GVHD model. Here we report on different genomic, phenotypic and functional properties of 4 NK cell subsets. Our data clearly demonstrate that CD27+ NK cells revealed the highest IFN-g production upon coculture with tumor cells and/or IL-2. Interestingly, the CD11b+ NK cells express multiple genes of cytotoxic pathways and develop the highest cytotoxic capacity towards tumor cells. We observed up to 60% tumor lysis by CD27- CD11b+ NK cells compared to 40–45% by CD27+ CD11b+, about 25% by CD27+ CD11b- and 10% by c-kit+ CD11b- NK cells at an effector-target ratio of 5:1, respectively. Furthermore, the CD11b+ NK cell subset significantly reduced T cell proliferation induced by allogeneic dendritic cells in mixed lymphocytes reactions. Next, we analyzed the migratory capacity and tissue-specific homing of FACS-sorted NK cell subsets by adoptive transfer of congeneic CD45.1+ and Luc+ NK cell subpopulations in autologous and allogeneic bone marrow transplantation. Of interest, FACS analysis and in vivo imaging showed that CD11b+ NK cells migrated to peripheral GVHD target organs, whereas CD27+ NK cells preferentially homed to the bone marrow. Finally, this study addressed for the first time the role of distinct NK cell subpopulations in the development of GVHD in a fully MHC mismatched HSCT mouse model. Importantly, we identified the CD11b+ NK cell population as the NK cell subset that significantly diminished GVHD. In vivo imaging of Luc+CD11b+ NK cells revealed that this subset migrates to the colonic tissue to prevent development of GVHD colitis as shown by colonoscopy. In summary, our comparative study outlines that only CD11b+ NK cells, migrating to the peripheral GVHD target organs and providing the most efficient cytolytic capacity directed against allogeneic dendritic cells, protect against GVHD. These new insights are highly relevant for the selection of optimal NK cell subsets in the field of cellular immunotherapy. Disclosures: No relevant conflicts of interest to declare.


2002 ◽  
Vol 195 (3) ◽  
pp. 343-351 ◽  
Author(s):  
Guido Ferlazzo ◽  
Ming L. Tsang ◽  
Lorenzo Moretta ◽  
Giovanni Melioli ◽  
Ralph M. Steinman ◽  
...  

During the innate response to many inflammatory and infectious stimuli, dendritic cells (DCs) undergo a differentiation process termed maturation. Mature DCs activate antigen-specific naive T cells. Here we show that both immature and mature DCs activate resting human natural killer (NK) cells. Within 1 wk the NK cells increase two– to fourfold in numbers, start secreting interferon (IFN)-γ, and acquire cytolytic activity against the classical NK target LCL721.221. The DC-activated NK cells then kill immature DCs efficiently, even though the latter express substantial levels of major histocompatibility complex (MHC) class I. Similar results are seen with interleukin (IL)-2–activated NK cell lines and clones, i.e., these NK cells kill and secrete IFN-γ in response to immature DCs. Mature DCs are protected from activated NK lysis, but lysis takes place if the NK inhibitory signal is blocked by a human histocompatibility leukocyte antigen (HLA)-A,B,C–specific antibody. The NK activating signal mainly involves the NKp30 natural cytotoxicity receptor, and not the NKp46 or NKp44 receptor. However, both immature and mature DCs seem to use a NKp30 independent mechanism to act as potent stimulators for resting NK cells. We suggest that DCs are able to control directly the expansion of NK cells and that the lysis of immature DCs can regulate the afferent limb of innate and adaptive immunity.


2005 ◽  
Vol 175 (9) ◽  
pp. 5790-5798 ◽  
Author(s):  
Béatrice Le Maux Chansac ◽  
Alessandro Moretta ◽  
Isabelle Vergnon ◽  
Paule Opolon ◽  
Yann Lécluse ◽  
...  

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4357-4357
Author(s):  
Arghya Ray ◽  
Ting DU ◽  
Nina N. Nupponen ◽  
Fredrik Lehmann ◽  
Jakob Lindberg ◽  
...  

Abstract Introduction Melphalan flufenamide (Melflufen; Oncopeptides AB) is a novel enzyme-activated analogue of melphalan that enables a more rapid and higher intracellular accumulation of melphalan in tumor cells than is achievable by direct exposure to equimolar doses of melphalan. Our preclinical study showed that melflufen is a more potent anti-myeloma (MM) agent than melphalan, overcomes drug-resistance, and induces synergistic anti-MM activity in combination with bortezomib, lenalidomide, or dexamethasone (Chauhan et al, Clinical Cancer Res 2013;19:3019). However, the effect of melflufen on the immunosuppressive and tumor-promoting MM-host bone marrow (BM) accessory cells such as immunologically dysfunctional plasmacytoid dendritic cells (pDCs; CD123/IL-3Rα) remains unclear. Here, we utilized our coculture models of pDCs, T-, and NK cells with autologous patient MM cells to examine whether a combination of melflufen and immune checkpoint inhibitor anti-PD-L1 Ab, or daratumumab (anti-CD38 Ab), restores anti-MM immunity. Methods MM patient BM and PB samples (N=10; obtained after informed consent), and cell lines were used for the study. Minimally cytotoxic concentration of melflufen (0.1 µM) was used to assess immune functions. CTL/NK activity assays MM CD8 + T- or NK-cells were cultured with autologous pDCs (1:10 pDC:T/NK ratio) with melflufen (0.1 μM) alone, and with anti-PD-L1 (5 μg/ml) or anti-CD38 (0.5 μg/ml) Abs for 3-5 days; cells were washed to remove the drugs, and then cultured for another 24h with pre-stained target MM cells (10:1 E/T ratio; T/NK:MM), followed by quantification of viable MM cells by flow. Results 1) Both MM tumor cells and pDCs showed higher PD-L1 and CD38 levels vs normal plasma cells; 2) Treatment of MM patient total BM mononuclear cells or purified MM cells with melflufen (0.1 µM) increased PD-L1 expression on MM cells (1.84-fold, treated vs untreated; p&lt;0.05). Importantly, treatment of MM cells with melflufen and anti-PD-L1 Abs enhanced anti-MM cytotoxicity; 3) Combination of melflufen and anti-PD-L1 Ab triggers activation of CD3 + T cells, evidenced by an increase in CD69 expression on CD3 + T cells (1.15-fold, treated vs untreated, p&lt;0.05); 4) Combination of melflufen and anti-PD-L1 Ab induced a more robust autologous MM-specific CD8 + cytotoxic T lymphocyte (CTL) activity than melflufen alone (% MM lysis: melflufen: 20%; melflufen plus anti-PD-L1 Ab: 60%; n=5; p=0.013); 5) Meflufen and anti-PD-L1 also triggered pDC-induced NK cell-mediated MM-specific cytolytic activity (p&lt;0.05); and finally, 6) Low doses of melflufen and anti-CD38 Abs enhanced pDC-induced NK cell-mediated MM-specific cytolytic activity (%Viability: melflufen: 75%; melflufen + anti-CD38 Ab: 12.5%; n=4; p=0.001). Conclusions The combination of melflufen and anti-PD-L1 increases pDC-induced T- and NK cell-mediated cytolytic activities against MM. Moreover, combined melflufen and anti-CD38 Abs modestly enhance pDC-induced NK cell-mediated MM-specific cytolytic activity. Our preclinical data suggest targeting PD-L1 in combination with melflufen as well as support an ongoing clinical trial of melflufen with anti-CD38 Abs to enhance anti-MM immunity. Disclosures Nupponen: Oncopeptides AB: Consultancy. Lehmann: Oncopeptides AB: Current Employment. Lindberg: Oncopeptides: Current Employment, Current equity holder in publicly-traded company, Divested equity in a private or publicly-traded company in the past 24 months, Other: Travel, Accommodations, Expenses; Camurus: Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accommodations, Expenses; Affibody: Membership on an entity's Board of Directors or advisory committees. Gullbo: Oncopeptides AB: Consultancy. Richardson: Takeda: Consultancy, Research Funding; Celgene/BMS: Consultancy, Research Funding; Janssen: Consultancy; Sanofi: Consultancy; Protocol Intelligence: Consultancy; Karyopharm: Consultancy, Research Funding; GlaxoSmithKline: Consultancy; Regeneron: Consultancy; AstraZeneca: Consultancy; Secura Bio: Consultancy; AbbVie: Consultancy; Oncopeptides: Consultancy, Research Funding; Jazz Pharmaceuticals: Consultancy, Research Funding. Chauhan: C4 Therapeutics: Current equity holder in publicly-traded company; Oncopeptides: Consultancy; Stemline Therapeutics: Consultancy. Anderson: Janssen: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Sanofi-Aventis: Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Millenium-Takeda: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Scientific Founder of Oncopep and C4 Therapeutics: Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company; AstraZeneca: Membership on an entity's Board of Directors or advisory committees; Mana Therapeutics: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1779-1779
Author(s):  
Matthias Peipp ◽  
Christian Kellner ◽  
Andreas Günther ◽  
Andreas Humpe ◽  
Roland Repp ◽  
...  

Abstract Antibody-dependent cell-mediated cytotoxicity (ADCC) represents a major effector function of many therapeutic antibodies. Thus, enhancing ADCC is a promising approach to further improve antibody therapy. Here, the CD20-specific immunoligands ULBP2:7D8 and B7-H6:7D8, which engage the stimulatory NK cell receptors natural killer group 2 member D (NKG2D) and NKp30, respectively, were compared for their abilities to boost ADCC in an attempt to design an effective antibody combination strategy. The immunoligands are designed as single chain molecules, with a single chain fragment variable (scFv) of the CD20 antibody 7D8 fused to UL16-binding protein (ULBP) 2 or B7 homologue 6 (B7-H6), which are ligands of the activating NK cell receptors NKG2D and NKp30, respectively. By binding to lymphoma cells the immunoligands designated as ULBP2:7D8 and B7-H6:7D8 mimicked an induced self phenotype and thereby triggered NK cells to kill lymphoma and leukemia cells. Both immunoligands augmented ADCC by NK cells synergistically when combined with the lymphoma-directed antibodies rituximab or daratumumab recognizing CD20 and CD38, respectively. Antibody combinations with ULBP2:7D8 resulted in higher cytotoxicity (up to 10-fold lower EC50-values) in comparison to combinations with B7-H6:7D8, which in individual experiments failed to boost ADCC. Thus, NK cells were triggered more efficiently when NKG2D rather than NKp30 was co-ligated together with FcγRIIIA. Although a combination of ULBP2:7D8 and B7-H6:7D8 produced synergistic effects, no significant improvements were obtained by combining the three agents rituximab, B7-H6:7D8 and ULBP2:7D8. Enhancement of ADCC by the immunoligands was also achieved when NK cells from lymphoma or leukemia patients were analyzed as effector cells. ULBP2:7D8 in particular increased lysis not only of allogeneic but also of autologous tumor cells. In summary, co-targeting of NKG2D was more effective in promoting NK cell-mediated ADCC than co-ligation of NKp30 and may represent a promising approach to further enhance the efficacy of therapeutic antibodies. Based on these results we propose a ‘dual-dual-targeting’ concept by co-targeting of two surface antigens on tumor cells and concomitant engagement of two different activating NK cell receptors. Disclosures van de Winkel: Genmab BV: Employment, Patents & Royalties. Parren:Genmab: Employment, Equity Ownership.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3507-3507
Author(s):  
Dharminder Chauhan ◽  
Mohan Brahmandam ◽  
Ajita Singh ◽  
Giada Bianchi ◽  
Klaus Podar ◽  
...  

Abstract The bone marrow (BM) microenvironment confers growth, survival, and drug resistance in multiple myeloma (MM) cells. Here we have characterized the role of plasmacytoid dendritic cells (pDCs) in the MM BM milieu. Immunochemistry (IHC) analysis of tissue microarrays on MM patient BM biopsies with Abs specific against pDCs (CD123) and MM cells (CD138) shows pDCs in proximity of MM cells. Quantification of pDCs obtained by direct isolation from MM patient BM aspirates or peripheral blood (PB) showed increased numbers of pDCs in MM-BM compared to PB. Freshly isolated pDCs from normal healthy donors stimulate significant growth of MM cells: 4.1 ± 0.8 fold increase {3H}-thymidine uptake in MM cells co-cultured with pDCs versus control MM cells alone, (P < 0.005). Irradiated pDCs retain their ability to trigger proliferation of MM cells; furthermore, pDC-depleted PBMCs did not trigger significant growth of MM cells, confirming a specific MM cell growth-promoting activity of pDCs. Co-culture of patient MM cells with pDCs triggered a significant growth of tumor cells, but not normal BM-derived plasma cells. Importantly, both allogeneic and autologous MM-derived pDCs induced tumor cell growth. To determine whether pDCs enhance MM cell growth in vivo, mice were implanted subcutaneously with pDCs alone, MM cells alone or pDCs + MM cells, and tumor growth was monitored over 3 weeks. A robust growth of tumor in mice receiving pDC + MM occurred within 12 days, whereas mice injected with MM cell alone showed a similar tumor growth only at day 21. We further examined the ability of pDCs to prolong ex-vivo survival of patient MM cells. Co-culture of pDCs with patient MM cells significantly increased the survival of patient tumor cells (59%, n=5 P<0.05), and IHC analysis of pDCs-MM cells co-cultures at 4 weeks confirmed that MM cells are clonal. We next examined the effect of anti-MM agents bortezomib and dexamethasone on the viability of pDCs and pDC-induced MM cell growth. Treatment of pDCs with bortezomib (20 nM) or dexamethasone (500 nM) does not significantly decrease viability of these cells (P = 0.25), higher concentrations of bortezomib (50 and 100 nM) decrease the viability of pDCs by less than 10%. Importantly, proliferation assays confirmed that pDCs triggered MM cell growth even in the presence of bortezomib, albeit to a lesser extent than without bortezomib(P<0.05). Microarray analysis showed that the pDCs-MM cells interaction triggered significant changes in transcriptional activity of genes related to growth, survival, anti-apoptosis, and migration in MM cells. Cytokine bead array analysis of supernatants from pDCs-MM cells co-cultures showed a marked increase in the secretion of MM cell growth, survival and chemotactic factors, such as IL-10, IL-6, IL-8, TNF-α, IL-1Rα, IL-1α, IL-13, IL-15, CD40L, MCP-1, MIP-1β, IP10 and VEGF. Overall, our data therefore show that pDCs predominantly localize in the MM BM and functionally interact with MM cells via cell-cell contact and subsequent cytokine secretion, allowing for MM cell growth and survival even in the presence of conventional and novel drugs. These studies will provide the basis for novel therapeutic approaches targeting pDC-MM interaction to improve patient outcome in MM.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2905-2905
Author(s):  
Dongxia Xing ◽  
Alan G Ramsay ◽  
John G. Gribben ◽  
William K Decker ◽  
Jared K Burt ◽  
...  

Abstract Natural killer (NK) cells play an important role in host immunity by eradicating pathogen-infected and tumor cells. Peripheral blood-derived NK cell therapy has shown promise in clinical trials for acute myeloid leukemia. Cord blood (CB) is another potentially rich source of NK cells. However NK cells isolated directly from CB have poor cytolytic activity. We investigated the mechanism for the low cytolytic activity of CB NK cells and whether the defect could be overcome by ex vivo expansion. NK cell killing of the target cells is achieved by formation of a mature immune synapse, followed by secretion of lytic granules containing perforin and granzymes. F-actin polymerization at the NK-tumor cell conjugates is a hallmark of immune synapse formation which can be detected and quantitated by confocal microscopy. We hypothesized that CB NK cells exhibit low cytotoxicity against leukemia target cells due to a defect in the formation of the immune synapse. We confirmed that unmanipulated CB NK cells exhibit low cytotoxicity against AML blasts (5% at an E:T ratio 20:1) in comparison to peripheral blood (PB) NK cells (35% at an E:T ratio 20:1). Evaluation of the natural cytotoxicity receptors (NCRs) showed normal expression of the NKp30 and the NK46 receptors. We then investigated whether this poor cytolytic capability is accompanied by poor immune synapse formation with tumor cells. Surprisingly, both CB and PB NK cells expressed comparable levels of perforin and demonstrated similar perforin polarization to the immune synapse. F-actin polarization was observed in only 12% (range 9– 21%) of the CB NK cell/K562 conjugates versus 85% (range 68–87%) of PB NK cell/K562 conjugates (P&lt;0.001). Remarkably, this impairment could be reversed by ex vivo expansion of CB NK cells with rIL-2. Expanded CB NK cells formed an increased percentage of immune synapses with K562 tumor cells 65% (range 60–71%) and primary human AML blasts 48% (range 39 –55%) which was comparable to the levels generated with PB NK cells. The present data reinforce the conclusion that formation of the activating NK cell immune synapse is required for cytotoxic activity. Furthermore, natural cytotoxicity receptors (NCR) and Kir receptors including KIR2DL1/S1 and KIR2DL2 were preserved on the expanded CB NK population. Moreover, the expanded CB NK cells were able to lyse AML targets in vitro (29% at 20:1 E:T ratio). Finally, we demonstrated that ex vivo expanded CB NK cells efficiently kill human AML in an NOD/SCID null mouse model. A 50 % reduction in AML tumor burden was documented in comparison to control groups by 6 weeks post NK infusion (P&lt;0.05). Our results suggest that ex vivo expansion of CB NK cells is a feasible and potentially effective strategy for the treatment of AML.


Cancers ◽  
2019 ◽  
Vol 11 (6) ◽  
pp. 877 ◽  
Author(s):  
Beatriz Sanchez-Correa ◽  
Isabel Valhondo ◽  
Fakhri Hassouneh ◽  
Nelson Lopez-Sejas ◽  
Alejandra Pera ◽  
...  

Natural killer (NK) cells are lymphocytes of the innate immune response characterized by their role in the destruction of tumor cells. Activation of NK cells depend on a fine balance between activating and inhibitory signals mediated by different receptors. In recent years, a family of paired receptors that interact with ligands of the Nectin/Nectin-like (Necl) family has attracted great interest. Two of these ligands, Necl-5 (usually termed CD155 or PVR) and Nectin-2 (CD112), frequently expressed on different types of tumor cells, are recognized by a group of receptors expressed on T and NK cells that exert opposite functions after interacting with their ligands. These receptors include DNAM-1 (CD226), TIGIT, TACTILE (CD96) and the recently described PVRIG. Whereas activation through DNAM-1 after recognition of CD155 or CD112 enhances NK cell-mediated cytotoxicity against a wide range of tumor cells, TIGIT recognition of these ligands exerts an inhibitory effect on NK cells by diminishing IFN-γ production, as well as NK cell-mediated cytotoxicity. PVRIG has also been identified as an inhibitory receptor that recognizes CD112 but not CD155. However, little is known about the role of TACTILE as modulator of immune responses in humans. TACTILE control of tumor growth and metastases has been reported in murine models, and it has been suggested that it negatively regulates the anti-tumor functions mediated by DNAM-1. In NK cells from patients with solid cancer and leukemia, it has been observed a decreased expression of DNAM-1 that may shift the balance in favor to the inhibitory receptors TIGIT or PVRIG, further contributing to the diminished NK cell-mediated cytotoxic capacity observed in these patients. Analysis of DNAM-1, TIGIT, TACTILE and PVRIG on human NK cells from solid cancer or leukemia patients will clarify the role of these receptors in cancer surveillance. Overall, it can be speculated that in cancer patients the TIGIT/PVRIG pathways are upregulated and represent novel targets for checkpoint blockade immunotherapy.


Sign in / Sign up

Export Citation Format

Share Document