Combination Therapy of a Defucosylated Anti-HM1.24 Monoclonal Antibody Plus Lenalidomide Significantly Enhances Myeloma Cell Cytotoxicity Via Antibody-Dependent Cellular Cytotoxicity

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.

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.


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 ◽  
2006 ◽  
Vol 108 (8) ◽  
pp. 2648-2654 ◽  
Author(s):  
Julie A. Bowles ◽  
Siao-Yi Wang ◽  
Brian K. Link ◽  
Barrett Allan ◽  
Gregory Beuerlein ◽  
...  

Abstract Growing evidence indicates that the affinity of monoclonal antibodies (mAbs) for CD16 (FcγRIII) plays a central role in the ability of the mAb to mediate antitumor activity. We evaluated how CD16 polymorphisms, and mAb with modified affinity for target antigen and CD16, affect natural killer (NK) cell phenotype when CD20+ malignant B cells were also present. The mAb consisted of rituximab (R), anti-CD20 with enhanced affinity for CD20 (AME-B), and anti-CD20 with enhanced affinity for both CD20 and CD16 (AME-D). Higher concentrations of mAb were needed to induce CD16 modulation, CD54 up-regulation, and antibody-dependent cellular cytotoxicity (ADCC) on NK cells from subjects with the lower affinity CD16 polymorphism. The dose of mAb needed to induce NK activation was lower with AME-D irrespective of CD16 polymorphism. At saturating mAb concentrations, peak NK activation was greater for AME-D. Similar results were found with measurement of CD16 modulation, CD54 up-regulation, and ADCC. These data demonstrate that cells coated with mAb with enhanced affinity for CD16 are more effective at activating NK cells at both low and saturating mAb concentrations irrespective of CD16 polymorphism, and they provide further evidence for the clinical development of such mAbs with the goal of improving clinical response to mAb.


2019 ◽  
Vol 203 (12) ◽  
pp. 3126-3135 ◽  
Author(s):  
A. Robin Temming ◽  
Steven W. de Taeye ◽  
Erik L. de Graaf ◽  
Louise A. de Neef ◽  
Gillian Dekkers ◽  
...  

2009 ◽  
Vol 83 (17) ◽  
pp. 8705-8712 ◽  
Author(s):  
Qingquan Liu ◽  
Yongtao Sun ◽  
Suzannah Rihn ◽  
Anne Nolting ◽  
Peter Nicholas Tsoukas ◽  
...  

ABSTRACT Increasing evidence suggests that NK cells not only are critical in the initial host defense against pathogens but also may contribute to continued protection from human immunodeficiency virus type 1 (HIV-1) disease progression. NK cell cytolysis can be induced directly through diverse receptor families or can be induced indirectly through Fc receptors by antibodies mediating antibody-dependent cellular cytotoxicity (ADCC). ADCC has been implicated in both protection from simian immunodeficiency virus infection and slower progression of HIV-1 disease. ADCC activity declines with advancing infection, and yet the underlying mechanism for this dysfunction has not been defined, nor has it been determined whether the activity can be reconstituted. Here we demonstrate that NK cell-mediated ADCC is severely compromised in chronic HIV infection. The potency of ADCC function was directly correlated with baseline FcγRIIIa receptor (CD16) expression on NK cells. CD16 expression was negatively influenced by elevated expression of a group of enzymes, the matrix metalloproteinases (MMPs), normally involved in tissue/receptor remodeling. Inhibition of MMPs resulted in increased CD16 expression and augmented ADCC activity in response to antibody-coated target cells. These data suggest that MMP inhibitors may improve NK cell-mediated ADCC, which may provide subjects with an opportunity to harness the cytolytic power of NK cells through naturally occurring nonneutralizing HIV-specific antibodies.


2020 ◽  
Vol 88 (3) ◽  
Author(s):  
Jacqueline Moebius ◽  
Rajan Guha ◽  
Mary Peterson ◽  
Kaveh Abdi ◽  
Jeff Skinner ◽  
...  

ABSTRACT Natural killer (NK) cells are key effector cells of innate resistance capable of destroying tumors and virus-infected cells through cytotoxicity and rapid cytokine production. The control of NK cell responses is complex and only partially understood. PD-1 is an inhibitory receptor that regulates T cell function, but a role for PD-1 in regulating NK cell function is only beginning to emerge. Here, we investigated PD-1 expression on NK cells in children and adults in Mali in a longitudinal analysis before, during, and after infection with Plasmodium falciparum malaria. We found that NK cells transiently upregulate PD-1 expression and interleukin-6 (IL-6) production in some individuals during acute febrile malaria. Furthermore, the percentage of PD-1 expressing NK cells increases with age and cumulative malaria exposure. Consistent with this, NK cells of malaria-naive adults upregulated PD-1 following P. falciparum stimulation in vitro. Additionally, functional in vitro studies revealed that PD-1 expression on NK cells is associated with diminished natural cytotoxicity but enhanced antibody-dependent cellular cytotoxicity (ADCC). These data indicate that PD-1+ NK cells expand in the context of chronic immune activation and suggest that PD-1 may contribute to skewing NK cells toward enhanced ADCC during infections such as malaria.


Blood ◽  
1979 ◽  
Vol 54 (3) ◽  
pp. 573-580
Author(s):  
G Fernandes ◽  
T Garrett ◽  
M Nair ◽  
D Straus ◽  
RA Good ◽  
...  

Leukemic blasts from patients with acute nonlymphoid leukemia were examined for the presence of Ig, receptors for IgGFc, and for their capacity to mediate antibody-dependent cellular cytotoxicity (ADCC) against chicken red blood cells (RBC) coated with IgG and spontaneous cell-mediated cytotoxicity (SCMC) against cells of K562 cell line. Leukemic blasts from acute myeloblastic leukemia (AML) patients lacked both Fc receptors and Ig on their surface, had no SCMC activity and majority, but not all of them, lacked ADCC activity. Leukemic blasts from patients with acute monocytic leukemia (AMOL) had Fc receptors, and 50% had IgG on their surface. IgG was cytophilic and appeared not to be directed against cell-surface antigens. This antibody did not interfere with the ADCC activity of leukemic cells. Leukemic blasts from majority of patients with AMOL mediated ADCC, but had no SCMC activity. An association between ADCC and presence of Fc receptor was observed.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3099-3099
Author(s):  
Mara N Zeissig ◽  
Duncan R Hewett ◽  
Krzysztof M Mrozik ◽  
Vasilios Panagopoulos ◽  
Monika Engelhardt ◽  
...  

Introduction:Multiple myeloma (MM) disease progression is dependent on the ability of the MM plasma cells (PC) to leave the bone marrow (BM), re-enter the peripheral blood (PB) and disseminate to other BM sites. Previous studies show that expression of CXCL12 by BM stromal cells is crucial for MM PC retention within the BM. However, the mechanisms which overcome this retention signal enabling MM PC egress and dissemination via the PB are poorly understood. Previous studies in haematopoietic progenitor cells have demonstrated that CCL3 overcomes the CXCL12 retention signal to drive mobilisation to the PB (Lord et al. Blood 1995). Here, we examined the role of the CCL3 chemokine receptor CCR1 in driving MM PC dissemination. Methods and results: Initially, we assessed the expression of CCR1 protein on CD138+CD38++CD45loCD19- PC from 28 MM, 8 MGUS and 2 SMM patients by flow cytometry. Results show CCR1 expression is significantly increased in newly diagnosed MM compared with premalignant MGUS and SMM patients (p=0.03; CCR1 MFI mean±SEM, MGUS: 53.0±33.6; SMM: 37.6±8.9 MM: 250.9±71.6). Furthermore, CCR1 expression on PB MM PC positively correlated with PB MM PC numbers (p=0.03; n=11 patients). To identify mechanistically how CCR1 may promote dissemination, the effect of CCL3 on the response to CXCL12 in human myeloma cell lines (HMCL) was assessed in vitro. The migration of RPMI-8226 and OPM2 cells was induced by CCL3 or CXCL12 chemoattractant in a transwell assay. Notably, pre-treatment of RPMI-8226 or OPM2 with CCL3 abrogated migration towards CXCL12 and blocked F-actin remodelling in response to CXCL12 in vitro. These findings suggest that CCL3 can desensitise cells to exogenous CXCL12, providing a potential mechanism facilitating loss of the CXCL12 retention signal. To confirm whether CCR1 is required for driving MM PC dissemination, homozygous CCR1 knockout (KO) cells were generated using a lentiviral CRISPR/Cas9 system in OPM2 cells. CCR1-KO OPM2 cells were confirmed to have no detectable CCR1 expression by flow cytometry and could no longer migrate towards CCL3 in vitro. Empty vector (EV) or CCR1-KO OPM2 MM PC were injected into the tibia of immune-compromised NOD-scidgamma (NSG) mice. After 4 weeks, primary tumour within the injected tibia and disseminated tumour in the PB and the contralateral tibia and femur was assessed by flow cytometry. We found that mice bearing CCR1-KO cells have a 45.5% decrease in primary tumour growth (p=0.008; % GFP+ of total mononuclear cells, EV: 77.2±17.2; CCR1-KO: 42.1±24.4), a 97.8% reduction in PB MM PC (p<0.0001; EV: 1.39±0.7; CCR1-KO: 0.03±0.046) anda 99.9% reduction in BM tumour dissemination (p<0.0001; EV: 49.5±17; CCR1-KO: 0.019±0.013), compared with controls. In a supportive study, CCR1 was expressed in the murine MM cell line 5TGM1 using lentiviral transduction. 5TGM1-CCR1 cells were confirmed to express CCR1 by qPCR and were able to migrate towards CCL3 in vitro. 5TGM1-CCR1 or EV cells were injected into the tibiae of C57BL/KaLwRij mice and allowed to initiate systemic MM disease for 3.5 weeks. Importantly, while 55% of control mice exhibited disseminated tumours, this increased to 92% with CCR1 expression (p<0.0001; n=12/group). These data suggest that CCR1 expression on MM PC may play an important role in MM PC dissemination. To determine whether therapeutic inhibition of CCR1 prevents dissemination, the effect of a small molecule CCR1 inhibitor, CCR1i, was assessed in vivo. OPM2 EV or RPMI-8226 cells were injected into the tibia of NSG mice and, after 3 days, mice were treated with CCR1i (15mg/kg) or vehicle twice daily by oral gavage for 25 days. OPM2-inoculated CCR1i-treated mice had 66.1% lower PB MM PC (p<0.0001; % GFP+ of total mononuclear cells, vehicle: 23.9±7.2; CCR1i: 8.1±3.8) and a 22.1% reduction in BM dissemination (p=0.0002; vehicle: 78.1±4.8;CCR1i: 60.8±7.1) compared with controls. Similarly, CCR1i treatment reduced BM dissemination by 59.6% in RPMI-8226 bearing mice (p<0.0001; % GFP+ of total mononuclear cells, vehicle: 0.86±0.15; CCR1i: 0.26±0.05). This suggests that CCR1 inhibition can slow tumour dissemination in vivo. Conclusion:This study identified CCR1 as a novel driver of MM PC dissemination in vivo, at least in part by overcoming the CXCL12 retention signal. Importantly, this study demonstrated for the first time that targeting CCR1 can be a viable therapeutic strategy to limit dissemination and potentially slow disease progression. Disclosures Croucher: Trovagene: Employment.


Sign in / Sign up

Export Citation Format

Share Document