Early Clinical Experience of aAPC Activated NK-DLI Following Allogeneic PBSCT in Pediatric Solid Tumor Patients.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3013-3013
Author(s):  
Kristin Baird ◽  
Terry J Fry ◽  
Melinda Merchant ◽  
Kelly Richards ◽  
Cynthia Delbrook ◽  
...  

Abstract Abstract 3013 Background: Allogeneic Hematopoietic Stem Cell Transplant (alloHSCT) is effective in some hematologic malignancies and studies of alloHSCT for ultra high-risk pediatric solid tumors have shown some promise. Preclinical data demonstrates that activated NK cells readily kill pediatric solid tumors and leukemias, large numbers of activated NK cells can be generated ex vivo using artificial APCs (aAPCs) and the post-transplant period may be favorable for expansion and survival of adoptively transferred NK cells. Methods: We initiated a Phase I trial to assess feasibility and toxicity of escalating doses of donor-derived activated NK cell donor lymphocyte infusions (NK-DLI) on days 7 and 35 days following HLA-matched T cell depleted PBSCT in children and young adults with ultra-high risk solid tumors and leukemias. Donors underwent a single apheresis for filgrastim mobilized PBSC. The product was T cell depleted and CD34 and CD56 selected prior to cryopreservation, and the CD56+ fraction was cultured for 9–11 days with a K562 based aAPC expressing 4-1BBL and IL-15Ra plus rhIL-15 to generate the NK-DLI. NK-DLI consistently expressed high levels of natural cytotoxicity receptors NKp44 and NKp46 and mediated potent cytotoxicity ex vivo. T cell addback to the CD34 selected graft was performed to administer a T cell dose of 0.8–1.4 × 10e4 T cells/kg. Except for two pilot patients where NK-DLI was infused following engraftment, NK-DLI were administered ∼Day 7 and 35 post-transplant. Patients received a reduced intensity preparative regimen (fludarabine 30 mg/m2 and cyclophosphamide, 1200 mg/m2 on days –6 to -3; and melphalan 100 mg/m2 on day -2), and no prophylactic immunosuppression. Results: Seven patients with sarcomas were transplanted with NK-DLI infusion (1×10e5 CD56+ cells/kg/dose). Engraftment was brisk (median platelet recovery at 8d and neutrophil recovery at 9d). Patients had full donor myeloid chimerism by day 14 and all but one had >80% lymphoid chimerism by day 28. Patient #1 received NK-DLI on day +24 following alloHSCT and experienced aGVHD within 24 hours of NK-DLI with bullous skin rash and voluminous diarrhea. Prior to infusion the patient had what appeared to be a viral rash and fevers (skin biopsy negative for GVHD), but in retrospect likely GVHD, which was exacerbated by NK-DLI. Patient #2 received NK-DLI on Day +15 with no adverse effects. Subsequent patients enrolled onto cohort 1 with planned NK-DLI at days 7 and 35 (+7). Two patients developed aGVHD (rash and diarrhea), one with a cytokine-type reaction (fevers, hypotension) within 48 hours of NK-DLI infusion and one with aGVHD starting 21 days post NK-DLI. All 3 patients with matched unrelated donors experienced aGVHD after NK-DLI, whereas the 5 patients with matched sibling donors had no apparent adverse reactions nor GVHD. Persistence/engraftment of infused NK-DLI cannot be definitively determined, however, mean NK cell counts measured pre-alloHSCT and day +28 are 165/mm3 and 668/mm3 in the present cohort (n=6) compared to 149/mm3 and 395/mm3 in a historical alloHSCT population that did not receive NK-DLI (n=24). Antitumor effects via PET imaging were observed in patients with measurable disease and those without disease at the time of transplant have remained NED. Conclusions: Ex-vivo, aAPC expanded NK-DLIs do not interfere with stem cell engraftment but may contribute to or induce aGVHD, particularly in patients with matched, unrelated donors. Preliminary results suggest that aAPC NK-DLI expand in vivo and mediate antitumor effects following allogeneic PBSCT. Disclosures: No relevant conflicts of interest to declare.

Cancers ◽  
2021 ◽  
Vol 13 (11) ◽  
pp. 2796
Author(s):  
Aicha E. Quamine ◽  
Mallery R. Olsen ◽  
Monica M. Cho ◽  
Christian M. Capitini

Treatment of metastatic pediatric solid tumors remain a significant challenge, particularly in relapsed and refractory settings. Standard treatment has included surgical resection, radiation, chemotherapy, and, in the case of neuroblastoma, immunotherapy. Despite such intensive therapy, cancer recurrence is common, and most tumors become refractory to prior therapy, leaving patients with few conventional treatment options. Natural killer (NK) cells are non-major histocompatibility complex (MHC)-restricted lymphocytes that boast several complex killing mechanisms but at an added advantage of not causing graft-versus-host disease, making use of allogeneic NK cells a potential therapeutic option. On top of their killing capacity, NK cells also produce several cytokines and growth factors that act as key regulators of the adaptive immune system, positioning themselves as ideal effector cells for stimulating heavily pretreated immune systems. Despite this promise, clinical efficacy of adoptive NK cell therapy to date has been inconsistent, prompting a detailed understanding of the biological pathways within NK cells that can be leveraged to develop “next generation” NK cell therapies. Here, we review advances in current approaches to optimizing the NK cell antitumor response including combination with other immunotherapies, cytokines, checkpoint inhibition, and engineering NK cells with chimeric antigen receptors (CARs) for the treatment of pediatric solid tumors.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A163-A163
Author(s):  
Yui Harada ◽  
Yoshikazu Yonemitsu

BackgroundCancer immunotherapy has been established as a new therapeutic category since the recent success of immune checkpoint inhibitors and a type of adoptive immunotherapy, namely chimeric antigen receptor-modified T cells (CAR-T). Although CAR-T demonstrated impressive clinical results, serious adverse effects (cytokine storm and on-target off-tumor toxicity) and undefined efficacy on solid tumors are important issues to be solved. We’ve developed a cutting-edge, simple, and feeder-free method to generate highly activated and expanded human NK cells from peripheral blood (US9404083, PCT/JP2019/012744, PCT/JP2020/012386), and have been conducting further investigation why our new type of NK cells, named as GAIA-102, are so effective to kill malignant cells.MethodsCryopreserved PBMCs purchased from vendors were mixed and processed by using LOVO and CliniMACS® Prodigy (automated/closed systems). CD3+ and CD34+ cells were depleted, and the cells were cultured with high concentration of hIL-2 and 5% UltraGRO® for 14 days in our original closed system. Then, we confirmed the expression of surface markers, CD107a mobilization and cell-mediated cytotoxicity against various tumor cells and normal cells with or without monoclonal antibody drugs in vitro and antitumor effects against peritoneal dissemination model using SKOV3 in vivo.ResultsImportantly, we’ve found that our GAIA-102 exhibited CD3-/CD56bright/CD57- immature phenotype that could kill various tumor cells efficiently from various origins, including Raji cells that was highly resistant to NK cell killing. More importantly, massive accumulation, retention, infiltration and sphere destruction by GAIA-102 were affected neither by myeloid-derived suppressor cells nor regulatory T-lymphocytes. GAIA-102 was also effective in vivo to murine model of peritoneal dissemination of human ovarian cancer; thus, these findings indicate that GAIA-102 has a potential to be an ‘upward compatible’ modality over CAR-T strategy, and would be a new and promising candidate for adoptive immunotherapy against solid tumors.ConclusionsWe now just started GMP/GCTP production of this new and powerful NK cells and first-in-human clinical trials in use of GAIA-102 will be initiated on 2021.Ethics ApprovalThe animal experiments were reviewed and approved by the Institutional Animal Care and Use Committee of Kyushu University (approval nos. A30-234-0 and A30-359-0).


Cancers ◽  
2020 ◽  
Vol 12 (9) ◽  
pp. 2417 ◽  
Author(s):  
Tram N. Dao ◽  
Sagar Utturkar ◽  
Nadia Atallah Lanman ◽  
Sandro Matosevic

Among natural killer (NK) cell receptors, the T-cell immunoglobulin and mucin-containing domain (TIM-3) has been associated with both inhibitory and activating functions, depending on context and activation pathway. Ex vivo and in vitro, expression of TIM-3 is inducible and depends on activation stimulus. Here, we report that TIM-3 expression can be downregulated on NK cells under specific conditions. When NK cells are exposed to cancer targets, they synergize with stimulation conditions to induce a substantial decrease in TIM-3 expression on their surface. We found that such downregulation occurs following prior NK activation. Downregulated TIM-3 expression correlated to lower cytotoxicity and lower interferon gamma (IFN-γ) expression, fueling the notion that TIM-3 might function as a benchmark for human NK cell dysfunction.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 341-341
Author(s):  
Lucila Kerbauy ◽  
Mecit Kaplan ◽  
Pinaki P Banerjee ◽  
Francesca Lorraine Wei Inng Lim ◽  
Ana Karen Nunes Cortes ◽  
...  

Abstract Chimeric antigen receptors to redirect T cell specificity against tumor antigens have shown remarkable clinical responses against CD19+ malignancies. However, the manufacture of an engineered autologous T cell product is expensive and cumbersome. Natural killer (NK) cells provide an alternative source of immune effectors for the treatment of cancer. NK cell cytolytic function can be directed towards specific targets by exploiting their ability to mediate antibody-dependent cellular cytotoxicity (ADCC) through the NK cell Fc receptor, CD16 (FcγRIIIa). AFM13 is a tetravalent bispecific antibody construct based on Affimed's ROCK™ platform. AFM13 is bispecific for CD30 and CD16A, designed for the treatment of CD30 expressing malignancies. It binds CD16A on the surface of NK cells, thus activating and recruiting them to CD30 expressing tumor cells and mediating subsequent tumor cell killing. Since autologous NK effector function is impaired in many patients with malignancies, we propose to overcome this by the use of allogeneic NK cells in combination with AFM13. Cord blood (CB) is a readily available ("off-the-shelf") source of allogeneic NK cells that can be expanded to large, highly functional therapeutic doses. The feasibility and safety of therapy with allogeneic ex vivo expanded CB-derived NK cells have been shown by our group and others. In this study, we hypothesized that we can redirect the specificity of NK cells against CD30+ malignancies by preloading ex vivo activated and expanded CB-derived NK cells with AFM13 prior to adoptive infusion. Briefly, mononuclear cells were isolated from fresh or frozen CB units by ficoll density gradient centrifugation. CD56+ NK cells were cultured with rhIL-12, rhIL-18 and rhIL-15 for 16 hrs, followed by ex vivo expansion with rhIL-2 and irradiated (100 Gy) K562-based feeder cells expressing membrane-bound IL-21 and CD137-ligand (2:1 feeder cell:NK ratio). After 14 days, NK cells were loaded with serial dilutions of AFM13 (0.1, 1, 10 and 100 mg/ml). After washing twice with PBS, we tested the effector function of AFM13-loaded NK-cells (AFM13-NK) compared to expanded CB-NK cells without AFM13 against Karpas-299 (CD30 positive) and Daudi (CD30 negative) lymphoma cell lines by 51Cr release and intracellular cytokine production assays. AFM13-NK cells killed Karpas-299 cells more effectively at all effector:target ratios tested than unloaded NK cells (Figure 1) and produced statistically more INFγ and CD107a (P=0.0034; P=0.0031 respectively, n=4). In contrast, AFM13-NK cells and unloaded NK cells exerted similar cytotoxicity against Daudi cells. Next, we established the optimal concentration of AFM13 for loading (determined to be 100 μg/ml) and the optimal incubation time to obtain maximal activity (1 h) in a series of in vitro experiments. We also confirmed that the activity of AFM13-NK cells against Karpas-299 cells remains stable for at least 72h post-wash (Figure 2). Additionally, we characterized the phenotype of AFM13-NK vs. unloaded NK cells by flow cytometry using monoclonal antibodies against 22 markers, including markers of activation, inhibitory receptors, exhaustion markers and transcription factors. Compared to unloaded NK cells, AFM13-NK cells expressed higher levels of CD25, CD69, TRAIL, NKp44, granzyme B and CD57, consistent with an activated phenotype. We next tested the in vivo anti-tumor efficacy of AFM13-NK cells in an immunodeficient mouse model of FFluc-Karpas-299. Briefly, six groups of NOD/SCID/IL2Rγc null mice (n=5 per group) were transplanted by tail-vein injection with 1 x 10e5 FFluc-transduced Karpas cells. Group 1 and 6 received tumor alone or tumor + AFM13 and served as a control. Groups 2-4 receive Karpas FFLuc with either expanded NK cells or AFM13-NK cells (NK cells loaded with AFM13) or expanded NK cells and AFM13 injected separately. Group 5 received AFM13-NK cells without tumor. Initial studies confirm the antitumor activity of AFM13-NK cells. In summary, we have developed a novel premixed product, comprised of expanded CB-NK cells loaded with AFM13 to 'redirect' their specificity against CD30+ malignancies. The encouraging in vitro and in vivo data observed in this study, provide a strong rationale for a clinical trial to test the strategy of an off-the-shelf adoptive immunotherapy with AFM13-loaded CB-NK cells in patients with relapsed/refractory CD30+ malignancies. Disclosures Champlin: Sanofi: Research Funding; Otsuka: Research Funding. Koch:Affimed GmbH: Employment. Treder:Affimed GmbH: Employment. Shpall:Affirmed GmbH: Research Funding. Rezvani:Affirmed GmbH: Research Funding.


Cancers ◽  
2019 ◽  
Vol 11 (7) ◽  
pp. 1040 ◽  
Author(s):  
Ziqing Chen ◽  
Ying Yang ◽  
Lisa L. Liu ◽  
Andreas Lundqvist

The immune system plays a crucial role to prevent local growth and dissemination of cancer. Therapies based on activating the immune system can result in beneficial responses in patients with metastatic disease. Treatment with antibodies targeting the immunological checkpoint axis PD-1 / PD-L1 can result in the induction of anti-tumor T cell activation leading to meaningful long-lasting clinical responses. Still, many patients acquire resistance or develop dose-limiting toxicities to these therapies. Analysis of tumors from patients who progress on anti-PD-1 treatment reveal defective interferon-signaling and antigen presentation, resulting in immune escape from T cell-mediated attack. Natural killer (NK) cells are innate lymphocytes that can kill tumor cells without prior sensitization to antigens and can be activated to kill tumor cells that have an impaired antigen processing and presentation machinery. Thus, NK cells may serve as useful effectors against tumor cells that have become resistant to classical immune checkpoint therapy. Various approaches to activate NK cells are being increasingly explored in clinical trials against cancer. While clinical benefit has been demonstrated in patients with acute myeloid leukemia receiving haploidentical NK cells, responses in patients with solid tumors are so far less encouraging. Several hurdles need to be overcome to provide meaningful clinical responses in patients with solid tumors. Here we review the recent developments to augment NK cell responses against solid tumors with regards to cytokine therapy, adoptive infusion of NK cells, NK cell engagers, and NK cell immune checkpoints.


Blood ◽  
2009 ◽  
Vol 113 (24) ◽  
pp. 6120-6127 ◽  
Author(s):  
Andreas Lundqvist ◽  
Hisayuki Yokoyama ◽  
Aleah Smith ◽  
Maria Berg ◽  
Richard Childs

Abstract Ligation of inhibitory receptors renders natural killer (NK) cells inactive against autologous tumors. Recently, the proteasome inhibitor bortezomib was shown to sensitize tumors to autologous NK-cell cytotoxicity in vitro. Here, we show bortezomib augments the antitumor effects of syngeneic NK-cell infusions in tumor-bearing animals; this effect is further enhanced in regulatory T cell (Treg cell)–depleted hosts. In vitro, bortezomib-treated tumors had higher tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) and perforin/granzyme-mediated caspase-8 activity, which enhanced their susceptibility to NK-cell lysis. Bioluminescence imaging of mice with established tumors showed treatment with bortezomib and syngeneic NK cells reduced tumor growth and prolonged survival compared with controls receiving bortezomib or NK cells alone. In contrast, tumor progression was not delayed when animals received bortezomib and perforin-deficient NK cells, showing drug-induced augmentation in NK-cell cytotoxicity was mediated through perforin/granzyme. Furthermore, tumor growth was slower in bortezomib-treated recipients when host Treg cells were eradicated with anti-CD25 antibody before infusing NK cells compared with mice without Treg-cell ablation (tumor doubling time, 16.7 vs 4.9 days, respectively; P = .02). These findings suggest that depletion of Treg cells followed by bortezomib-induced tumor sensitization to autologous NK cells could be used as a novel strategy to treat cancer.


Author(s):  
Emily Cluff ◽  
Carina C. Magdaleno ◽  
Emyly Fernandez ◽  
Trenton House ◽  
Srividya Swaminathan ◽  
...  

AbstractNatural killer (NK) cells are cytotoxic innate lymphocytes that are specialized to kill tumor cells. NK cells are responsive to the primary cytokine IL-2 in the tumor microenvironment (TME), to activate its effector functions against tumors. Despite their inherent ability to kill tumor cells, dysfunctional NK cells observed within advanced solid tumors are associated with poor patient survival. Hypoxia in the TME is a major contributor to immune evasion in solid tumors that could contribute to impaired NK cell function. HIF-1α is a nodal regulator of hypoxia in driving the adaptive cellular responses to changes in oxygen concentrations. Whether HIF-1α is expressed in hypoxic NK cells in the context of IL-2 and whether its expression regulates NK cell effector function are unclear. Here, we report that freshly isolated NK cells from human peripheral blood in hypoxia could not stabilize HIF-1α protein coincident with impaired anti-tumor cytotoxicity. However, ex vivo expansion of these cells restored HIF-1α levels in hypoxia to promote antitumor cytotoxic functions. Similarly, the human NK cell line NKL expressed HIF-1α upon IL-2 stimulation in hypoxia and exhibited improved anti-tumor cytotoxicity and IFN-γ secretion. We found that ex vivo expanded human NK cells and NKL cells required the concerted activation of PI3K/mTOR pathway initiated by IL-2 signaling in combination with hypoxia for HIF-1α stabilization. These findings highlight that HIF-1α stabilization in hypoxia maximizes NK cell effector function and raises the prospect of NK cells as ideal therapeutic candidates for solid tumors.


Cancers ◽  
2019 ◽  
Vol 11 (10) ◽  
pp. 1534 ◽  
Author(s):  
Sooyeon Oh ◽  
Joo-Ho Lee ◽  
KyuBum Kwack ◽  
Sang-Woon Choi

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.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A405-A405
Author(s):  
David Hong ◽  
Sandip Patel ◽  
Manish Patel ◽  
Kimberly Musni ◽  
Marlisa Anderson ◽  
...  

BackgroundFT500 is an investigational, off-the-shelf NK cell cancer immunotherapy derived from a human clonal master iPSC line, a renewable cell source from which innate effector cells can be mass produced and made available off-the-shelf for broad patient access and multiple dose administration. FT500 has potent innate cellular cytotoxicity as compared to NK cells sourced from healthy donors and has been shown to synergize with T cells and anti-PD-1 blockade in preclinical studies.1MethodsFT500 is being investigated in a Phase I clinical trial as monotherapy and in combination with immune checkpoint inhibitors (ICIs) in patients with advanced solid tumors and lymphomas (ClinicalTrials.gov: NCT03841110). Treatment consists of 2 days lympho conditioning (fludarabine 25 mg/m2 and cyclophosphamide 300 mg/m2) followed by 2 cycles of 3 once weekly doses of FT500 as monotherapy or combined with 1 of 3 approved ICIs (nivolumab, pembrolizumab, or atezolizumab) in patients who have failed prior ICI therapy. Key clinical and translational readouts include FT500 safety and tolerability, including immune mediated toxicities and anti-product immunogenicity.Results15 patients with relapsed/refractory disease following a median of 4 prior therapies were treated in dose escalation, including 9 with FT500 monotherapy (3 with 1×108 cells, 6 with 3×108 cells) and 6 with FT500 (3 each with 1×108 and 3×108 cells) combined with ICI. No dose limiting toxicities, Grade ³3 related adverse events (AEs), Grade ³3 related serious AEs, or related AEs leading to treatment discontinuation were reported. No graft-versus-host disease (GvHD), cytokine release syndrome (CRS), or neurotoxicity (NT) was observed. The most common treatment-emergent AEs in >3 patients were nausea (9), fatigue (7), constipation, decreased appetite, decreased lymphocyte count, decreased white blood cell count (5 each), anemia, and decreased neutrophil count (4 each). Nine of 13 efficacy-evaluable solid tumor patients had best response of stable disease by iRECIST. One patient with classical Hodgkin lymphoma (cHL) refractory to prior experimental anti-PD-1 therapy had a 58% reduction in target lesions size following FT500 plus ICI. No evidence of robust B- or T-cell mediated anti product responses was observed despite endogenous immune cell recovery following lympho-conditioning.ConclusionsAdministration of 6 doses of up to 3×108 FT500 cells is safe and tolerable without evidence of GvHD, CRS, NT, or host immune rejection. Enrollment of advanced non-small cell lung cancer and cHL patients at 3×108 FT500 cells per dose combined with ICI is ongoing.Ethics ApprovalThis study is being conducted in accordance with the Declaration of Helsinki and was approved by all Institutional Review Boards from each clinical site participating in the study. Specific approval numbers can be provided upon request.ReferenceCichocki F, Bjordahl R, Gaidarova S, Mahmood S, Rogers P, Ge MQ, Kaufman DS, Cooley S, Valamehr B, Miller JS. iPSC-derived NK cells and anti-PD-1 antibody synergize to enhance T cell cytokine and cytolytic responses against multiple tumors. Blood 2018;132(Supplement 1):730.


2019 ◽  
Vol 11 ◽  
pp. 175883591989162 ◽  
Author(s):  
Naomi C. Bosch ◽  
Reinhard E. Voll ◽  
Caroline J. Voskens ◽  
Stefanie Gross ◽  
Barbara Seliger ◽  
...  

Background: In therapeutic cancer vaccination, monocyte-derived dendritic cells (moDCs) efficiently activate specific T-cell responses; however, optimizing the activation of innate immune cells could support and improve the antitumor effects. A major disadvantage of moDCs matured with the standard cytokine cocktail (consisting of IL-1β, IL-6, TNFα, and PGE2) is their inability to secrete IL-12p70. IL-12 prominently activates natural killer (NK) cells, which are crucial in innate antitumor immunity, as they act as helper cells for the induction of a cytotoxic T lymphocyte (CTL) response and are also able to directly kill the tumor. Methods: Previously we have shown that triggering the NF-κB pathway in moDCs by transfection of mRNA encoding constitutively active IKKβ (caIKKβ) led to IL-12p70 secretion and improved the dendritic cells’ capability to activate and expand CTLs with a memory-like phenotype. In this study, we examined whether such dendritic cells could activate autologous NK cells. Results: moDCs matured with the standard cytokine cocktail followed by transfection with the caIKKβ-RNA were able to activate autologous NK cells, detected by the upregulation of CD54, CD69, and CD25 on the NK cells, their ability to secrete IFNγ, and their high lytic activity. Moreover, the ability of NK-cell activation was not diminished by simultaneous T-cell activation. Conclusion: The capacity of caIKKβ-DCs to activate both the adaptive and innate immune response indicates an enhanced potential for clinical efficacy.


Sign in / Sign up

Export Citation Format

Share Document