The role of natural killer (NK) cell expansion and activation on the in vivo anti-tumor activity of immunomodulatory drug (IMiD) CC4047 plus rituximab in a severe combined immunodeficiency (SCID) lymphoma mouse model

2004 ◽  
Vol 22 (14_suppl) ◽  
pp. 2556-2556
Author(s):  
N. M. Reddy ◽  
F. J. Hernandez-Ilizaliturri ◽  
M. S. Czuczman
1991 ◽  
Vol 173 (5) ◽  
pp. 1053-1063 ◽  
Author(s):  
R M Welsh ◽  
J O Brubaker ◽  
M Vargas-Cortes ◽  
C L O'Donnell

The activation, proliferation, and antiviral properties of natural killer (NK) cells were examined in severe combined immunodeficiency (SCID) mice to determine the influence of mature T or B cells on virus-induced NK cell functions and to more conclusively determine the antiviral properties of prototypical CD3- NK cells. NK cells were activated to high levels of cytotoxicity 3 d after infection of mice with lymphocytic choriomeningitis virus (LCMV) or murine cytomegalovirus (MCMV). Analyses of spleen leukocytes from LCMV-infected mice by a variety of techniques indicated that the NK cells proliferated and increased in number during infection. Propidium iodide staining of the DNA of cycling cells revealed that the great majority of proliferating spleen leukocytes 3 d after LCMV infection was of the NK cell phenotype (CD3-, Ig-, Mac-1+, CZ1+, 50% Thy-1+), in contrast to uninfected mice, whose proliferating cells were predominantly of other lineages. Analyses of the NK cell responses over a 2 wk period in control CB17 mice infected with MCMV indicated a sharp rise in serum interferon (IFN) and spleen NK cell activity early (days 3-5) in infection, followed by sharp declines at later stages. In SCID mice the IFN levels continued to rise over a 10-d period, whereas the NK cell response peaked on day 3-5 and gradually tapered. In contrast to the immunocompetent CB17 mice, SCID mice did not clear the MCMV infection and eventually succumbed. SCID mice, again in contrast to immunocompetent CB17 mice, also failed to clear infections with LCMV and Pichinde virus (PV); these mice, infected as adults, did not die but instead developed long-term persistent infections. Depletion of the NK cells in vivo with antiserum to asialo GM1 rendered both SCID and CB17 control mice much more sensitive to MCMV infection, as shown by titers of virus in organs and by survival curves. In contrast, similar depletions of NK cells did not enhance the titers of the NK cell-resistant virus, LCMV. Two variants of PV, one sensitive to NK cells and the other selected for resistance to NK cells by in vivo passage, were also tested in NK cell-depleted SCID mice. The NK-sensitive PV replicated to higher titers in NK cell-depleted SCID mice, whereas the titers of the NK cell-resistant PV were the same, whether or not the mice had NK cells. These experiments support the concept that CD3- prototypical NK cells mediate resistance to NK cell-sensitive viruses via a mechanism independent of antiviral or "natural" antibody.(ABSTRACT TRUNCATED AT 400 WORDS)


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4550-4550 ◽  
Author(s):  
Xiaomei Wang ◽  
Wei-Chun Chang ◽  
Daniel L. Jasinski ◽  
Jan L. Medina ◽  
Ming Zhang ◽  
...  

Abstract Background Natural Killer (NK) lymphocytes possess innate anti-tumor activity that has the potential to be used as an allogeneic cell therapy due to reduced GvHD risk relative to αβ T cells. Despite their potential, adoptive NK cell immunotherapies have been limited by poor expansion in vivo. Using our previously developed Chimeric Antigen Receptor-T cell (CAR-T) strategy that relies on rimiducid-based dimerization of inducible MyD88/CD40 (iMC) to regulate T cell expansion and survival, we demonstrate that iMC can also be applied to NK cell growth and anti-tumor efficacy in vitro and in vivo. Moreover, a rapamycin-inducible Caspase-9 (iRC9) was used to provide an orthogonally regulated safety switch. Methods and Results CD56+ NK cells were isolated from peripheral blood of human donors, stimulated overnight with IL-15 then activated by seeding with K562 erythroleukemia target cells. NK cells were then transduced with γ-retrovirus encoding control iRC9-2A-ΔCD19, iRC9-2A-ΔCD19-2A-iMC (dual-switch NK) or iRC9-2A-IL-15-2A-ΔCD19-2A-iMC (dual-switch/IL-15 NK). ΔCD19 marked transduced cells in 50:50 cocultures with untransduced NK cells. NK cells containing only iRC9 grew at the same rate as untransduced cells, but iMC-expressing NK cells displayed enhanced growth that was further augmented by 1 nM rimiducid treatment. In cocultures with THP1 acute myeloid leukemia cells at increasing Target:Effector (T:E) ratios, presence (P < 0.001, two way ANOVA) and activation (P <0.001) of iMC increased tumor killing activity. Inflammatory cytokine and chemokine production was also dramatically (10 to 1000-fold) elevated by the expression and activation of iMC in NK cells in the presence and absence of THP1 tumor target. To study in vivo anti-tumor activity, immunodeficient NSG mice were engrafted with dual-switch NK cells with or without autocrine IL-15 expression in the presence or absence of THP-1 tumor targets. When tumor was present, unstimulated iMC with IL-15 or activation of iMC without IL-15 expression supported modest NK cell expansion, but rimiducid stimulation of iMC plus autocrine IL-15 showed enhanced NK expansion in vivo. Furthermore, in tumor-free animals only dual-switch/IL-15 NK cells with weekly rimiducid stimulation expanded and persisted in vivo (up to 7 weeks). Cotransduction of a first generation CD123-targeted CAR to produce dual-switch/IL-15 CD123CAR-NK cells led to rimiducid-dependent control of THP1 tumor outgrowth in vivo beyond 40 days. Conversely, temsirolimus-mediated activation of the iRC9 safety switch rapidly (< 24 hours) ablated dual-switch NK cells in vivo. Conclusions Inducible MyD88/CD40 is an activation switch that supports NK cell expansion, persistence and anti-tumor activity. When paired with autocrine IL-15 expression, this platform supports NK expansion and persistence in vivo, and AML tumoricidal activity that can be further activated by target-specific CAR expression. Moreover, the fast-acting, orthogonally regulated proapoptotic switch, iRC9, mitigates the risk of off-tumor targeting. Therefore, we describe a novel, regulated NK cell platform that solves many of the challenges of NK cell-based therapy and should be amenable to a readily translatable off-the-shelf cellular therapy for malignancies. Disclosures Wang: Bellicum Pharmaceuticals: Employment, Equity Ownership. Chang:Bellicum Pharmaceuticals: Employment, Equity Ownership. Jasinski:Bellicum Pharmaceuticals: Employment, Equity Ownership. Medina:Bellicum Pharmaceuticals: Employment, Equity Ownership. Zhang:Bellicum Pharmaceuticals: Employment, Equity Ownership. Foster:Bellicum: Employment, Equity Ownership. Spencer:Bellicum Pharmaceuticals: Employment, Equity Ownership. Bayle:Bellicum Pharmaceuticals: Employment, Equity Ownership.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 827-827 ◽  
Author(s):  
Sarah Cooley ◽  
Purvi Gada ◽  
David McKenna ◽  
Valarie McCullar ◽  
Susan Fautsch ◽  
...  

Abstract We have previously shown that adoptive transfer of haploidentical natural killer (NK) cells can induce remissions in 27% of patients with refractory or relapsed acute myeloid leukemia (AML) [Miller et al., Blood 2005, 105 (8)]. Aiming to optimize NK cell expansion, which we hypothesize is required for therapeutic efficacy, we tested additional CD56-positive selection (N=10) versus the CD3-depletion method used for our earlier NK cell infusions (N=10). Donor-derived NK cells were not measurable immediately after infusion. Successful in vivo NK cell expansion, defined as >100 donor-derived NK cells/ml at 14 days (by VNTR chimerism and flow cytometry) was not improved with CD56-selection (11% vs. 11%; mean 131±3 NK cells/ml), and was associated with poorer outcomes (10% vs. 27% AML remissions). Because the remissions induced by adoptive NK cell transfer were not durable, we added a CD34+ stem cell infusion to create a nonmyeloablative haploidentical transplantation protocol for older and less fit patients. We also added radiation to the NK cell-based preparative regimen to further improve NK cell expansion. The lymphodepleting chemoradiation plus NK cell preparative regimen included fludarabine 25 mg/m2 × 5 (day -18 through day -14), cyclophosphamide 60 mg/kg × 2 (days -16 and -15), and 200 cGy of total body irradiation (twice a day on day -13). The NK cell product, prepared by cliniMACS (Miltenyi) CD3-depletion of a single leukapheresis collection from a haploidentical related donor, was incubated overnight in 1000 U/ml IL-2 and then infused on day -12 followed by 6 doses subcutaneous IL-2 (10 million units) given every other day to promote in vivo NK cell expansion. The mean NK cell dose was 1.85 × 107 cells/kg and the mean CD3+ cell dose was 4.8 × 104 cells/kg (maximum permitted 3 × 105 CD3+ cells/kg). A CD34-selected filgrastim-mobilized peripheral blood graft from the same donor (target dose >3 × 106 CD34 cells/kg) was given with Thymoglobulin 3 mg/kg days 0, +1 and +2 as the only additional immunosuppression. In the 13 patients treated to date a significantly higher rate of NK cell expansion (75% [9/12 evaluable]; mean 607±184 NK cells/ml) was achieved compared to the adoptive NK cell transfer regimen, which did not include radiation. Plasma IL-15, which is critical for NK expansion, was highest on day -12 (the NK infusion day) after the preparative regimen (64 ± 8 pg/ml [day -12] vs. 6 ± 1 pg/ml [baseline pre-chemo]; p <.0001). This adoptive NK cell plus allograft protocol led to 66% of relapsed or refractory AML patients (8/12 evaluable) clearing leukemia by day -1, with only one late relapse (day +93). Patients who did not clear leukemia (N=4) did not engraft, and it was not evaluable in 3 patients with early (pre-day +13) treatment related mortality (TRM). All others (N=6), engrafted quickly (defined by an absolute neutrophil count >500/ml and 100% donor chimerism: median 17 days [range 11–31]). None developed graft vs. host disease (GVHD), but infections were common (3 fatal EBV/PTLD; 1 Fusarium). To prevent EBV reactivation NK products are now CD19 depleted and patients receive prophylactic Rituxan to prevent PTLD. The other deaths were due to persistent disease (N=4) or relapse (N=1). One patient is alive in remission beyond day +100. No clear associations between killer immunoglobulinlike receptor (KIR) ligand mismatch between donor and recipient were detected. In this series of patients with refractory AML, addition of haploidentical NK cells to a nonmyeloablative haploidentical transplantation yields NK cell expansion in a majority of patients, achievement of complete remission, and quick engraftment without GVHD. This is a promising platform upon which to add other strategies aimed at improving disease free survival in patients with refractory AML.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 355-355 ◽  
Author(s):  
Sarah Cooley ◽  
Bree Foley ◽  
Michael R Verneris ◽  
David McKenna ◽  
Xianghua Luo ◽  
...  

Abstract Abstract 355FN2 We have previously shown that adoptive transfer of haploidentical natural killer (NK) cells can induce remissions in patients with refractory or relapsed acute myeloid leukemia (AML). We hypothesize that in vivo expansion of functional NK cells is required for therapeutic efficacy. To achieve the adequate host immune suppression required for expansion we added total body irradiation (TBI) to our lymphodepleting chemotherapy regimen, giving patients fludarabine (Flu) 25 mg/m2 × 5 days, cyclophosphamide (Cy) 60 mg/kg × 2 days, and 400 cGy of TBI. The NK cell product, a CD3- and CD19-depleted lymphapheresis from a haploidentical related donor, was incubated overnight in 1000 U/ml IL-2 and infused followed by 6 doses of alternate day subcutaneous IL-2 (10 million units) to promote in vivo expansion. Because of the increased myelosuppression following the TBI, a CD34-selected filgrastim-mobilized peripheral blood graft from the same donor (target dose >3 × 106 CD34 cells/kg) was given for hematopoietic rescue. Successful in vivo NK cell expansion was prospectively defined as >100 donor-derived NK cells/ml at 14 days after adoptive transfer (by analysis of STR chimerism, % NK and the clinical absolute lymphocyte count). In the 38 evaluable patients, robust in vivo expansion was induced in 50% (absolute donor NK count of 666 ± 134 cells/μL blood), a rate considerably higher than the 10% we observed in a cohort receiving Cy/Flu alone without TBI. Successful NK cell expansion correlated with leukemia clearance (<1% marrow blasts 14 days after NK cell infusion) and remission (leukemia free with donor neutrophil engraftment at day +30; 42 days after NK infusion). Of the 19 patients who achieved NK cell expansion, 89% cleared their leukemia compared to 42% of the non-expanders (p=0.002); and 84% achieved remission vs. 10% of non-expanders (p <.0001). The robust in vivo expansion of adoptively transferred NK cells gave us the unique opportunity to study their function. We studied blood collected from patients 14 days after NK cell infusion and compared it to paired donor samples obtained at steady state from the apheresis products prior to IL-2 stimulation. Using multi-color flow cytometry, we measured CD107a expression (a surrogate marker for NK cell cytotoxicity) on NK cells which we could subset by expression of single inhibitory killer cell immunoglobulin-like receptors (KIR) (CD158a, CD158b and CD158e) or NKG2A. We defined NK subsets as self-KIR+ or non-self KIR+ based on the cognate HLA ligands (C2, C1, Bw4) present in the donor or recipient. The bulk population of in vivo expanded donor NK cells exhibited hyperfunction with 62.4±4.4% degranulation in response to class I negative K562 targets compared to 36.6±3.0% in the donor product samples (N=15; p=0.0043). As expected, the most potent NK cells in the unstimulated donor product were the self-KIR+ subset, which expressed 39.5±3.0% CD107a after incubation with K562 (N=23) compared to either the non-self KIR+subset (13.1±4.0%, N=6; p=0.0001), or the uneducated KIR−/NKG2A− subset (12.4±5.8%, N=10; p<0.0001). Remarkably, all 3 subsets exhibited even greater degranulation activity after 14 days of in vivo expansion where they were exposed to homeostatic factors and the IL-2 administered to the patient. While all subsets expressed more CD107a, the rules of education were maintained. The subset expressing self-KIR that recognized donor HLA ligands degranulated significantly better than the non-self KIR+ subset (53.5±14.1% vs. 34.3±13.6%, p<0.01). Interestingly, the in vivo expanded NK cells with KIR recognizing cognate ligands unique to the recipient also functioned better (53.1±14.3% [recipient self KIR+] vs. 32.4±12.0% [non-self KIR+], N=25 and N=6; p<0.0045), showing that the education status of adult NK cells is dynamic, not fixed. Importantly, the KIR−/NKG2A− subset functioned better after in vivo expansion (39.5±115.3%, N=12), demonstrating that adoptively transferred NK cells can acquire function by two separate mechanisms: 1) acquisition of function through NK cell education; and 2) acquisition of function by homeostatic expansion and cytokine activation. These data suggest that while hyperfunctioning NK cells that expand in vivo after adoptive transfer partially overcome self tolerance, which may augment their anti-leukemic effects, they still follow the rules of NK cell education where self KIR+ cells kill better than non-self KIR+ cells. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 11 ◽  
Author(s):  
Meng Guo ◽  
Chen Sun ◽  
Yuping Qian ◽  
Liye Zhu ◽  
Na Ta ◽  
...  

Adoptive natural killer (NK) cell transfer has been demonstrated to be a promising immunotherapy approach against malignancies, but requires the administration of sufficient activated cells for treatment effectiveness. However, the paucity of clinical-grade to support the for large-scale cell expansion limits its feasibility. Here we developed a feeder-based NK cell expansion approach that utilizes OX40L armed NK-92 cell with secreting neoleukin-2/15 (Neo-2/15), a hyper-stable mimetic with a high affinity to IL-2Rβγ. The novel feeder cells (NK92-Neo2/15-OX40L) induced the expansion of NK cells with a 2180-fold expansion (median; 5 donors; range, 1767 to 2719) after 21 days of co-culture without added cytokines. These cells were highly cytotoxic against Raji cells and against several solid tumors in vivo. Mechanistically, NK92-Neo2/15-OX40L induced OX40 and OX40L expression on expanded NK cells and promoted the OX40-OX40L positive feedback loop, thus boosting NK cell function. Our data provided a novel NK cell expansion mechanism and insights into OX40-OX40L axis regulation of NK cell expansion.


2014 ◽  
Vol 7 ◽  
pp. MRI.S13145 ◽  
Author(s):  
Naomi S. Sta Maria ◽  
Samuel R. Barnes ◽  
Russell E. Jacobs

Natural killer (NK) cells are a crucial part of the innate immune system and play critical roles in host anti-viral, anti-microbial, and anti-tumor responses. The elucidation of NK cell biology and their therapeutic use are actively being pursued with 200 clinical trials currently underway. In this review, we outline the role of NK cells in cancer immunotherapies and summarize current noninvasive imaging technologies used to track NK cells in vivo to investigate mechanisms of action, develop new therapies, and evaluate efficacy of adoptive transfer.


In Vivo ◽  
2018 ◽  
Vol 32 (4) ◽  
pp. 771-781 ◽  
Author(s):  
IN HYE JUNG ◽  
DO HEE KIM ◽  
DA KYUNG YOO ◽  
SUN YOUNG BAEK ◽  
SEONG HOON JEONG ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document