scholarly journals Lymphokine-activated killer cells from normal and lymphoma subjects are cytotoxic for cells coated with antibody derivatives displaying human Fc gamma

Blood ◽  
1988 ◽  
Vol 72 (6) ◽  
pp. 1985-1991
Author(s):  
RJ Dearman ◽  
FK Stevenson ◽  
M Wrightham ◽  
TJ Hamblin ◽  
MJ Glennie ◽  
...  

Lymphokine-activated killer (LAK) cells were successfully generated in all cases from blood mononuclear cells obtained from six patients with lymphoma. The LAK cells from three of these patients and from five normal adult donors were tested for their effector abilities in antibody-dependent cellular cytotoxicity (ADCC) against guinea pig leukemic lymphocytes coated with various antiidiotype antibodies. Cells from all the donors behaved similarly. Mouse monoclonal antibodies of IgG1, IgG2a, and IgG2b isotypes invoked no ADCC. However, substantial ADCC was invoked by the chimeric antibody FabFc, in which Fab'gamma from mouse antiidiotype is thioether-bonded to human normal Fc gamma. Similar results were obtained on testing LAK cells from a normal donor against uncultured human lymphoma targets coated with native or chimeric antiidiotype. The ADCC invoked by the mouse-human chimeric antibodies appears to depend on the human Fc gamma they display and not on the univalency of the derivatives used. The findings imply that LAK technology could usefully augment serotherapy that uses antibody derivatives displaying human Fc gamma.

Blood ◽  
1988 ◽  
Vol 72 (6) ◽  
pp. 1985-1991 ◽  
Author(s):  
RJ Dearman ◽  
FK Stevenson ◽  
M Wrightham ◽  
TJ Hamblin ◽  
MJ Glennie ◽  
...  

Abstract Lymphokine-activated killer (LAK) cells were successfully generated in all cases from blood mononuclear cells obtained from six patients with lymphoma. The LAK cells from three of these patients and from five normal adult donors were tested for their effector abilities in antibody-dependent cellular cytotoxicity (ADCC) against guinea pig leukemic lymphocytes coated with various antiidiotype antibodies. Cells from all the donors behaved similarly. Mouse monoclonal antibodies of IgG1, IgG2a, and IgG2b isotypes invoked no ADCC. However, substantial ADCC was invoked by the chimeric antibody FabFc, in which Fab'gamma from mouse antiidiotype is thioether-bonded to human normal Fc gamma. Similar results were obtained on testing LAK cells from a normal donor against uncultured human lymphoma targets coated with native or chimeric antiidiotype. The ADCC invoked by the mouse-human chimeric antibodies appears to depend on the human Fc gamma they display and not on the univalency of the derivatives used. The findings imply that LAK technology could usefully augment serotherapy that uses antibody derivatives displaying human Fc gamma.


PEDIATRICS ◽  
1977 ◽  
Vol 59 (1) ◽  
pp. 22-28
Author(s):  
Steven L. Shore ◽  
Henry Milgrom ◽  
Phyllis A. Wood ◽  
André J. Nahmias

The functional adequacy of antibody-dependent cellular cytotoxicity (ADCC) in the human neonate was evaluated in a 51Cr release assay which employs tissue culture cells acutely infected with type 1 or type 2 herpes simplex virus (HSV) as targets. Two aspects of ADCC were assessed: cytotoxic effector activity in cord blood mononuclear cells (MC) and the ability of the antibody mediating ADCC to pass the placenta. Although effector cell activity was readily detected in all 13 cord blood specimens tested, cord blood MC showed moderately reduced cytotoxic activity when compared with blood MC from normal adults at the same effector cell:target cell ratio. This finding suggests that effector cells in cord blood make up a reduced proportion of the total circulating MC population and may be of relevance to the newborns increased susceptibility to HSV infection. On the other hand, the number of MC in cord blood was found to be almost twice that of adult blood, suggesting that the absolute number of ADCC effector cells in cord blood was within the adult range. The antibody mediating ADCC to HSV-infected cells was shown to be transferred (quantitatively across the placenta, providing further evidence that it is an IgG immunoglobulin.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1833-1833
Author(s):  
Takeshi Harada ◽  
Shuji Ozaki ◽  
Asuka Oda ◽  
Hiroe Amou ◽  
Shiro Fujii ◽  
...  

Abstract Abstract 1833 Multiple myeloma (MM) is a B-cell malignancy characterized by the accumulation of neoplastic plasma cells in the bone marrow. During the last decade, treatment of MM has been improved by incorporating bortezomib, thalidomide, and lenalidomide (LEN) into conventional cytotoxic and transplantation regimens in newly diagnosed and relapsed/refractory MM patients. However, MM still remains incurable despite the implementation of these new treatment options, so more efficacious therapies are needed to further improve the prognosis of MM. Monoclonal antibody (mAb)-based immunotherapy has recently become an alternative strategy for treatment of cancers. Our previous studies have shown that HM1.24 (CD317) is selectively expressed on terminally differentiated normal and neoplastic plasma cells and, moreover, expressed on the side population of MM cells that represents MM cancer stem cells. We have previously generated a humanized mAb (AHM) specific to HM1.24 for the treatment of MM. AHM carries an Fc region derived from human IgG1-k and exhibits the ability to induce antibody-dependent cellular cytotoxicity (ADCC) against human MM cells in the presence of human effector cells. To improve the efficacy of AHM, we have developed a defucosylated mAb (YB-AHM) with a higher affinity to Fc gamma RIII. LEN is a structural analog of thalidomide with more potent immunomodulatory activities. Several studies have shown that LEN activates NK cell function and enhances NK cell-mediated lysis of both MM cell lines and patient MM cells in vitro. Here, we evaluated the efficacy of combination therapy of YB-AHM and LEN. First, we investigated whether LEN stimulates the expression of HM1.24 on MM cells. LEN alone did not affect HM1.24 expression, but in the presence of peripheral blood mononuclear cells (PBMCs) LEN augmented the expression of HM1.24 in MM cell lines and primary MM cells. In PBMCs, expression levels of CD56 increased after stimulation with LEN. These results suggest that LEN might augment the ADCC activity by enhancing HM1.24 antigen and NK activity. Next, we evaluated ADCC activity of YB-AHM against RPMI 8226 cells by using flow cytometric PKH-26 assay. When we used PBMCs from healthy donors (n=5) as effectors, ADCC activity of YB-AHM was increased in an E:T ratio-dependent manner. Importantly, YB-AHM induced significantly higher ADCC activity compared with AHM (24±6% vs 11±7%, p<0.05; mAb, 100 ng/mL; E:T ratio, 10). Treatment of PBMCs with LEN (3 micro M for 2 days) slightly enhanced ADCC activity of AHM (12±5%) and YB-AHM (30±6%). In PBMCs from MM patients (n=11), YB-AHM induced ADCC activity (36±15%) that was further enhanced by treatment with LEN (45±15%). To evaluate the efficacy of this combination therapy in a more physiological manner, we assessed the efficacy of YB-AHM using total bone marrow mononuclear cells (BMMCs) from MM patients that contained both MM cells and effector cells. BMMCs were stimulated with LEN (3 micro M) for 2 days and further incubated with YB-AHM for 24 hours. Cytotoxicity was evaluated by the number of CD38-positive MM cells in total BMMCs using flow cytometry. YB-AHM plus LEN significantly reduced the number of MM cells (10.3%) compared to YB-AHM alone (21.6%) in patient No.1. Finally, RPMI 8226 cells were co-cultured with YB-AHM and LEN-stimulated PBMCs from MM patients, and MM colony formation was examined using methylcellulose assay. Colony formation of RPMI 8226 was significantly suppressed by YB-AHM and LEN-stimulated PBMCs compared to control (14±8 vs 49±10 colonies, p<0.01), suggesting that this combination therapy can target MM cancer stem cells. Thus, these results indicate that combining defucosylated HM1.24 mAb with immunomodulatory drugs provides a novel therapeutic strategy in patients with MM. Disclosures: No relevant conflicts of interest to declare.


2002 ◽  
Vol 30 (4) ◽  
pp. 487-490 ◽  
Author(s):  
L. G. Presta ◽  
R. L. Shields ◽  
A. K. Namenuk ◽  
K. Hong ◽  
Y. G. Meng

The binding sites on human IgG1 for human Fcγ receptor (FcγR) I, FcγRIIa, FcγRIIb, FcγRIIIa and neonatal FcR have been mapped. A common set of IgG1 residues is involved in binding to all FcγRs, while FcγRII and FcγRIII utilize distinct sites outside this common set. In addition to residues which abrogated binding to the FcγR, several positions were found which improved binding only to specific FcγRs or simultaneously improved binding to one type of FcγR and reduced binding to another type. Selected IgG1 variants with improved binding to FcγRIIIa were then tested in an in vitro antibody-dependent cellular cytotoxicity (ADCC) assay and showed an enhancement in ADCC when either peripheral blood mononuclear cells or natural killer cells were used.


Blood ◽  
1991 ◽  
Vol 77 (11) ◽  
pp. 2389-2395 ◽  
Author(s):  
JS Miller ◽  
C Verfaillie ◽  
P McGlave

Abstract We have generated a homogeneous population of recombinant interleukin-2 (rIL-2)-stimulated effector cells termed adherent lymphokine-activated killer cells (A-LAK) from peripheral blood mononuclear cells (PBMNC) of 14 normal individuals and tested the effect of A-LAK cells on autologous hematopoietic bone marrow (BM) progenitor growth. Enrichment of A-LAK from PBMNC depended on the propensity of A-LAK precursors to adhere to plastic and proliferate in the presence of rIL-2. The resultant population had the morphologic appearance of large granular lymphocytes, and the majority of cells (73% +/- 4%) expressed the CD56+/CD3- phenotype associated with rIL-2-stimulated natural killer (NK) cells. The A-LAK population had potent lytic activity in chromium release assays against both NK-sensitive (K562) and NK-resistant (Raji) targets. When BM mononuclear cells (BMMNC) were coincubated with autologous A-LAK and rIL-2 (1,000 U/mL) added at the start of culture, dose-dependent suppression of burst-forming unit-erythroid (BFU-E) and colony-forming unit mix (CFU-MIX) colony growth was observed at effector to target ratios (E:T) ranging from 0.25:1 to 5:1 (maximal suppression BFU-E = 85% +/- 6%; CFU-MIX = 95% +/- 3%). This suppression was rIL-2 dose-dependent, and no suppression was seen in the absence of rIL-2. Depletion of BM monocytes and T lymphocytes did not alter A-LAK suppression of progenitors coincubated with A-LAK cells. Addition of polyclonal neutralizing antibodies against both interferon-gamma (IFN- gamma) and tumor necrosis facto alpha (TNF-alpha) to the coincubation culture completely abrogated the suppressive effect of A-LAK on BFU-E and CFU-MIX colony growth while each neutralizing antibody used alone had intermediate effects. In contrast to coincubation studies, 36 hours of preincubation of A-LAK cells with autologous BM (E:T 2.2:1) and rIL- 2 (1,000 U/mL) followed by plating of preincubated BM cells in hematopoietic progenitor culture produced significant suppression of day 14 BFU-E (47% +/- 5%), but spared the more primitive CFU-MIX (7% +/- 9%), suggesting a divergent effect of A-LAK cells on hematopoietic progenitors at different stages of differentiation. Addition of neutralizing antibodies against IFN-gamma and TNF-alpha in preincubation failed to abrogate the suppressive effect of A-LAK on BFU- E colony growth, suggesting that this suppression occurs by a different mechanism than that seen in coincubation studies. Previous studies have demonstrated that the A-LAK population has cytotoxic and proliferative advantages over other killer cell populations.(ABSTRACT TRUNCATED AT 400 WORDS)


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1839-1839
Author(s):  
Takeshi Harada ◽  
Shuji Ozaki ◽  
Asuka Oda ◽  
Masami Iwasa ◽  
Shiro Fujii ◽  
...  

Abstract Abstract 1839 The implementation of hematopoietic stem cell transplantation and new agents such as bortezomib, thalidomide, and lenalidomide (Len) has dramatically improved survival in patients with multiple myeloma (MM). However, most MM patients eventually relapse after achieving of complete response. The existence of cancer stem cells is suggested to cause the relapse and considered as an important therapeutic target. We have demonstrated that HM1.24 (CD317) is selectively over-expressed on neoplastic plasma cells and that a defucosylated humanized monoclonal antibody (mAb) specific to HM1.24 (YB-AHM) is able to induce potent antibody-dependent cellular cytotoxicity (ADCC) against human MM cells in the presence of human effector cells. On the other hand, Len, an immunomodulatory drug, has been shown to activate NK cells to enhance their ADCC activity. Recently, we have reported that “side population (SP)” cells expelling a Hoechst 33342 dye represent a fraction with cancer stem cell-like property in MM cells. Moreover, we have found that MM cancer stem-like cells over-express pluripotency-associated transcription factors such as Sox2 and Nanog, and that these factors are useful for evaluating the potential of MM cancer stem-like cells. Here, we evaluated the efficacy of the combinatory therapy of YB-AHM and Len on MM cancer stem-like cells. We first examined the expression levels of the target molecule, HM1.24 on SP fraction of MM cells. Although SP cells expressed higher levels of ABC transporter ABCG2 compared with main population (MP) cells, HM1.24 was highly expressed in both SP and MP fractions in MM cells. We next examined the inhibitory effect of YB-AHM and Len on clonogenic activity of MM cell lines. RPMI 8226, U266, and OPM-2 cells were pre-incubated with YB-AHM (0.1 μg/ml) and Len (3 μM)-stimulated peripheral blood mononuclear cells (PBMCs) at an effector/target (E/T) ratio of 10 for 4 hours, and then were cultured into H4034 methylcellulose medium. Colony formation of MM cells was examined after 14 days. Treatment with YB-AHM and Len-stimulated PBMCs significantly suppressed the colony formation of MM cell lines compared with the no-treatment group (4±5 vs 62±2 colonies/well in RPMI 8226, p<0.01; 21±4 vs 43±8 colonies/well in U266, p<0.05; and 16±2 vs 84±4 colonies/well in OPM-2, p<0.01), suggesting that the combination therapy can target clonogenic MM cells. The mRNA expression levels of Sox2 and Nanog on MM cell lines and primary MM cells were also decreased when treated with YB-AHM (0.1 μg/ml) and Len-stimulated PBMCs (E/T=10) for 24 hours. Notably, this combination therapy decreased the mRNA expression of these transcription factors even in bortezomib-resistant MM cell line, OPM-2/BTZ. Finally, we examined the cytotoxic activity of YB-AHM plus Len using patients' bone marrow mononuclear cells (BMMCs) containing both target MM cells and autologous effector cells. BMMCs were stimulated with Len (3 μM) for 48 hours and further incubated with YB-AHM (0.1 μg/ml) for 24 hours. The cytotoxic activity was evaluated by counting CD38-positive MM cells in total BMMCs using flow cytometry. When we examined BMMCs from 10 MM patients, the combination of YB-AHM plus Len significantly induced MM cell death compared with controls (mean cytotoxicity, 46±23% vs 7±10%, p<0.01). Collectively, these results demonstrate that defucosylated HM1.24 mAb and Len in combination induce ADCC to eradicate MM cancer stem-like cells in bone marrow environment. Therefore, this combination might provide a novel therapeutic strategy targeting clonogenic drug-resistant clones in MM. Disclosures: Nakamura: Janssen Pharmaceutical K.K.: Honoraria. Abe:Janssen Pharmaceutical K.K.: Honoraria, Research Funding. Iida:Janssen Pharmaceutical K.K.: Honoraria.


Sign in / Sign up

Export Citation Format

Share Document