Recognition of Myeloma by MAGE-A3 Specific Cytotoxic T Lymphocytes Induced by Treatment with Azacitidine and MGCD0103

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3674-3674
Author(s):  
Amberly Moreno-Bost ◽  
Susann Szmania ◽  
Katie Stone ◽  
Jumei Shi ◽  
Tarun K. Garg ◽  
...  

Abstract Demethylating agents and histone deacetylase inhibitors (HDACi) are epigenetic modulators that can induce re-expression of tumor suppressor and cell cycle proteins that have been silenced through aberrant hypermethylation associated with tumoral transformation. Azacitidine (Aza) is a cytosine analogue that primarily affects RNA during transcription. However, DNA is also a target for demethylation during replication thereby increasing gene re-expression. Treatment with HDACi, such as MGCD0103 (MGC), can synergize with demethylating agents to boost the epigenetic effects of either drug used alone. Our gene expression profiling data shows that the cancer-testis antigen MAGE-A3 is expressed in 31% of myeloma patients at diagnosis and the frequency of expression is increased at relapse to 49% (n=51 paired samples, p<0.001, unpublished data). However, expression of MAGE-A3 is often heterogeneous. We hypothesized that the combination of Aza and MGC could induce MAGE-A3 expression, thus facilitating killing of myeloma cells by MAGE-A3 specific CTLs isolated from a HLA-A68 positive patient post MAGE-A3 protein vaccination (J Immunother2007; 30:847). The MAGE-A3 negative myeloma cell line LP1 was first transfected with HLA-A68. MAGE-A3 protein production was optimized by dose finding and time course experiments using Aza alone or Aza and MGC sequentially. Induction of MAGE-A3 RNA expression was assessed by real time PCR and protein expression by Western blotting. 51Cr-release assays were used to measure killing of Aza/MGC treated cell lines by MAGE-A3 specific CTLs. MAGE-A3 RNA expression was detected in LP1-A68 treated with 500nM Aza for 3 days and expression was enhanced by sequential treatment with 1mM MGC for 1 day when compared to Aza treatment alone. However, protein expression was low. In an effort to optimize protein production, we increased the time of treatment with 500nM Aza to 5 days and with 500nM MGC to 2 days. After this sequential treatment, protein was clearly expressed (Figure 1) and LP1-A68 cells were killed by MAGE-A3 specific CTLs (specific lysis: 70% ± 9% at E:T ratio of 5:1), whilst untreated controls only showed background killing (specific lysis: 12% ± 5%) (Figure 2). Repeat experiments are in progress to verify these results. 500nM Aza in vitro is comparable to a clinically achievable in vivo dose of 12.5mg/m2 (Leukemia2008; 22:965). 500nM MGC is comparable to a 280mg/m2in vivo dose (Blood2006; 108: 1954). Additional titration experiments with MGC will be tested to achieve clinically relevant concentrations in vivo that can induce MAGE-A3 expression. In conclusion, epigenetic modulation by Aza and MGC can enhance MAGE-A3 expression and result in increased killing by MAGE-A3 specific CTLs. Hypomethylating agents and HDACi may be useful to sensitize tumor cells to immune effectors. Figure 1. Treatment with 50nM Aza and/or sequential MGC at 500 nM induces de nono expression of MAGE-A3 protein in the myeloma a cell line transfectant LP1 A68. Figure 1. Treatment with 50nM Aza and/or sequential MGC at 500 nM induces de nono expression of MAGE-A3 protein in the myeloma a cell line transfectant LP1 A68. Figure 2. Lysis of LP-1 A68 Aza/MGC treated targets by MAGE-A3 specific CTL effective. Figure 2. Lysis of LP-1 A68 Aza/MGC treated targets by MAGE-A3 specific CTL effective.

2021 ◽  
Vol 41 (1) ◽  
Author(s):  
Bolun Wang ◽  
Haohui Guo ◽  
Tianxiang Geng ◽  
Kening Sun ◽  
Liang Zhang ◽  
...  

Abstract Aseptic loosening following periprosthetic osteolysis is the primary complication that limits the lifetime of total joint arthroplasty (TJA). The wear particles trigger a chronic inflammation response in the periprosthetic tissue and turn over the bone balance to bone resorption. The present study aimed to investigate the possible effect and mechanism of strontium ranelate (SR), a clinically safe drug for osteoporosis, on particle-induced periprosthetic osteolysis. Thirty-six female C57BL/6j mice underwent tibial Ti-nail implantation to establish an animal model of aseptic loosening. After 12 weeks, micro-CT results showed that strontium ranelate could inhibit periprosthetic bone resorption. In vitro, Ti particles were used to stimulate RAW264.7 cell line to collect conditioned medium, and co-culture MC3T3-E1 cell line with conditioned medium to establish a cell model of aseptic loosening. The results of alkaline phosphatase (ALP) detection, immunofluorescence, and flow cytometry demonstrated that strontium ranelate could regulate the expression of OPG/RANKL, promote differentiation and mineralization, and inhibit apoptosis in osteoblasts. Moreover, we revealed that SR’s exerted its therapeutic effect by down-regulating sclerostin, thereby activating the Wnt/β-catenin signal pathway. Therefore, this research suggests that strontium ranelate could be a potential drug for the prevention and treatment of particle-induced aseptic loosening post-TJA.


2002 ◽  
Vol 227 (6) ◽  
pp. 398-401 ◽  
Author(s):  
Jianping Qiu ◽  
Lonny R. Levin ◽  
Jochen Buck ◽  
Marcus M. Reidenberg

Gossypol, a polyphenolic, aldehyde-containing constituent of cottonseed, produced partial responses (>50% reduction in tumor size) in some patients with advanced cancer and suppressed sperm as an antifertility agent for men. This action in vivo and its novel side effect profile suggest a specific mechanism of the action of gossypol. Using the random homozygous knockout approach of Li and Cohen (1), we developed a cell line resistant to killing by gossypol, but sensitive to methotrexate and doxorubicin. It showed stereospecific resistance to killing by (–) gossypol (ED50 4.9 μM) compared with wild type (ED50 2.0 μM). The resistant and wild-type cells were equally sensitive to (+) gossypol (ED50 8.8 and 8.4 μM, respectively), methotrexate, and doxyrubicin. We conclude that gossypol affects cells by a stereospecific pathway for (–) gossypol, possibly related to its selective effects, and a nonstereospecific pathway for (+) gossypol and higher concentrations of (-) gossypol. Further knowledge about the stereospecific pathway may lead to new therapeutic drugs.


1998 ◽  
Vol 88 (12) ◽  
pp. 1367-1371 ◽  
Author(s):  
Y. D. Chen ◽  
T. A. Chen

In an attempt to develop maize plants with resistance to corn stunt spi-roplasma (CSS), a single-chain Fv fragment (scFv) gene that was constructed from antibodies with strong inhibitory activity against CSS, was expressed in a cell line of maize. However, plants regenerated from this transgenic cell line showed no distinct resistance to CSS infection under the greenhouse conditions. The affinity and functionality of scFv in vivo and the locations of CSS and expressed scFv in maize could be a part of the contributing factors affecting this result. Progress of expressing antibodies in plants for plant pathogen resistance is also discussed.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3173-3173
Author(s):  
Yukio Kondo ◽  
Xingmin Feng ◽  
Yoshihisa Kumano ◽  
Shinji Nakao

Abstract Aberrantly expressed self-antigens in leukemic cells serve as leukemia-associated-antigens (LAAs) of leukemia reactive T cells. Although such self-antigen-derived LAAs potentially induce high avidity CTLs in patients with leukemia, these CTLs usually do not persist due to apoptosis upon encountering leukemic cells. In allogeneic stem cell transplant (allo-SCT) recipients with leukemia, residual leukemic cell may sensitize donor-derived T cells by LAA in vivo and induce high avidity CTLs specific to leukemic cells. Cyclin-dependent kinase 2 (CDK2) is a cell cycle regulator protein that is aberrantly expressed in AML, MDS, ALL and MCL cells. We previously reported that CDK2-derived nonamer peptides (CDK2158: TYTHEVVTL, CDK2167: WYRQPEILL, CDK2178: KYYSTAVDI) avidly bound to HLA-A24 molecule to elicit each peptide-specific CTL from peripheral blood mononuclear cells (PBMCs) of healthy individuals (Blood. 106 (11): a3103. 2005). When we generated CDK2158-specific CD8+ T cells from an HLA identical sibling donor of a patient with AML by stimulating donor PBMCs with CDK2158-coated HLA-A24-transfected T2, CDK2158-specific CD8+ T cells preferentially killed the recipient AML cells which aberrantly expressed CDK2 proteins. The percentages of specific lysis in the 51Cr-release assay at an E/T ratio of 40 were 36.8% for AML cells and 24.8% for autologous PBMCs. To determine if CDK2-specific CD8+ T cells are present in PBMCs from allo-SCT patients, we studied 14 patients possessing HLA-A24 [6 patients with AML (2 AML-M0, 3 AML-M2, 1 AML with multilinage dysplasia), 1 with MDS, 1 with CML, 2 with ALL, 2 with MCL and 2 with RCC] using CDK2158/A24 and CDK2178/A24 multimers. Cryopreserved PBMCs obtained before and 4–73 months after allo-SCT were assayed for multimer staining. The source of graft was unrelated BM in 4, related BM in 2, related PBMC in 3 and CB in 5 patients. All CB and 1 BM graft were HLA mismatched. Seven patients were in complete remission (CR) at the time of SCT while 7 were in non-CR. Ten patients remained in CR 4–79 months after SCT. Small populations (0.13–0.77 % of lymphocyte) of CDK2158 and CDK2178-specific CTL were detectable in 5 patients (3 with AML-M2, 1 with CML and 1 with ALL) 10–73 months after SCT. All of the 5 remained in CR 11–79 months after SCT. Only one of them had active cGvHD at sampling. On the other hand, among 7 patients who did not show an increase in the proportion of neither CDK2158 nor CDK2178-specific CTLs, 2 patients with AML-M0 relapsed 4 and 5 months after SCT, respectively. Both of them had active cGvHD at sampling. CDK2-specific CTLs were also undetectable in 2 patients with RCC after allo-SCT. These data provide evidence for the first time that CDK2-specific CTLs can be induced from donor-derived T cells without peptide immunization possibly due to in vivo sentitization of donor T cells by residual leukemic cells. Vaccination with CDK2-derived peptides after allo-SCT may therefore be useful in inducing CDK2-specific CTLs and thereby preventing relapse of leukemia. Fig. Appearance of CDK2-CTLs after allo SCT in patients with leukemia in remission. Fig. Appearance of CDK2-CTLs after allo SCT in patients with leukemia in remission.


1984 ◽  
Vol 160 (1) ◽  
pp. 341-346 ◽  
Author(s):  
E S Vitetta ◽  
R J Fulton ◽  
J W Uhr

In vitro killing of the human Daudi cell line by either univalent [F(ab')] or divalent (IgG) forms of rabbit anti-human Ig (RAHIg) coupled to ricin A chain can be specifically potentiated by a "piggyback" treatment with ricin B chain coupled to goat anti-rabbit Ig (GARIg). When cells are treated with univalent immunotoxin (IT) [F(ab') RAHIg-A] and then cultured, IT can be detected on the cell surface for at least 5 h, since GARIg-B can still enhance killing at this time. These results provide a strategy for in vivo use of A chain- and B chain-containing IT.


1973 ◽  
Vol 138 (2) ◽  
pp. 364-372 ◽  
Author(s):  
M. Hatanaka ◽  
R. Klein ◽  
C. W. Long ◽  
R. Gilden

Tumorigenic and nontumorigenic mutants induced by a single 5'-bromodeoxyuridine (BrdU) treatment of a nonproducer (NP) tumorigenic cell line were isolated and characterized. Among the cloned derivatives were examples of virus-free and sarcoma virus-producing cell lines. Oncogenicity did not correlate with production of virus or ease of rescue of the sarcoma genome. All lines, including nononcogenic derivatives, retained the sarcoma genome. Phenotypic reversion of some cell mutants was observed after in vivo inoculation or long term in vitro cultivation. The M-50T cell line, obtained from a tumor induced by M-50 cells, had a sarcoma genome rescuable by direct superinfection; this was only achieved with parental M-50 cells by a cell fusion rescue technique. The M-43-2T cell, obtained from a single small static tumor induced by otherwise nononcogenic M-43-2 cells, shed sarcoma virus and became tumorigenic. M-58-4-48 became tumorigenic after passage 48 of the M-58-4 line, which was originally nontumorigenic. These observations of phenotypic reversion demonstrate that the presence of the sarcoma gene in cells is an essential but not sufficient condition of tumorigenesis.


2022 ◽  
Vol 12 (1) ◽  
Author(s):  
Warren Fiskus ◽  
Steffen Boettcher ◽  
Naval Daver ◽  
Christopher P. Mill ◽  
Koji Sasaki ◽  
...  

AbstractTreatment with Menin inhibitor (MI) disrupts the interaction between Menin and MLL1 or MLL1-fusion protein (FP), inhibits HOXA9/MEIS1, induces differentiation and loss of survival of AML harboring MLL1 re-arrangement (r) and FP, or expressing mutant (mt)-NPM1. Following MI treatment, although clinical responses are common, the majority of patients with AML with MLL1-r or mt-NPM1 succumb to their disease. Pre-clinical studies presented here demonstrate that genetic knockout or degradation of Menin or treatment with the MI SNDX-50469 reduces MLL1/MLL1-FP targets, associated with MI-induced differentiation and loss of viability. MI treatment also attenuates BCL2 and CDK6 levels. Co-treatment with SNDX-50469 and BCL2 inhibitor (venetoclax), or CDK6 inhibitor (abemaciclib) induces synergistic lethality in cell lines and patient-derived AML cells harboring MLL1-r or mtNPM1. Combined therapy with SNDX-5613 and venetoclax exerts superior in vivo efficacy in a cell line or PD AML cell xenografts harboring MLL1-r or mt-NPM1. Synergy with the MI-based combinations is preserved against MLL1-r AML cells expressing FLT3 mutation, also CRISPR-edited to introduce mtTP53. These findings highlight the promise of clinically testing these MI-based combinations against AML harboring MLL1-r or mtNPM1.


2019 ◽  
Vol 93 (13) ◽  
Author(s):  
Diem-Lan Vu ◽  
Albert Bosch ◽  
Rosa M. Pintó ◽  
Enric Ribes ◽  
Susana Guix

ABSTRACT MLB astroviruses were identified 10 years ago in feces from children with gastroenteritis of unknown etiology and have been unexpectedly detected in severe cases of meningitis/encephalitis, febrile illness of unknown etiology, and respiratory syndromes. The aim of this study was to establish a cell culture system supporting MLB astrovirus replication. We used two clinical strains to infect several cell lines, an MLB1 strain from a gastroenteritis case, and an MLB2 strain associated with a neurologic infection. Efforts to propagate the viruses in the Caco-2 cell line were unsuccessful. In contrast, we identified two human nonintestinal cell lines, Huh-7 and A549, permissive for both genotypes. After serial passages in the Huh-7.5 cell line, the adapted strains were able to establish persistent infections in the Huh-7.5, Huh-7AI, and A549 cell lines, with high viral loads (up to 10 log10 genome copies/ml) detected by quantitative reverse transcription-PCR (RT-qPCR) in the culture supernatant. Immunofluorescence assays demonstrated infection in about 10% of cells in persistently infected cultures. Electron microscopy revealed particles of 32 to 33 nm in diameter after negative staining of cell supernatants and capsid arrays in ultrathin sections with a particularly high production in Huh-7.5 cells. Interferon (IFN) expression by infected cells and effect of exogenous IFN varied depending on the type of infection and the cell line. The availability of a cell culture system to propagate MLB astroviruses represents a key step to better understand their replicative cycle, as well as a source of viruses to conduct a wide variety of basic virologic studies. IMPORTANCE MLB astroviruses are emerging viruses infecting humans. More studies are required to determine their exact epidemiology, but several reports have already identified them as the cause of unexpected clinical diseases, including severe neurologic diseases. Our study provides the first description of a cell culture system for the propagation of MLB astroviruses, enabling the study of their replicative cycle. Moreover, we demonstrated the unknown capacity of MLB astrovirus to establish persistent infections in cell culture. Whether these persistent infections are also established in vivo remains unknown, but the clinical consequences would be of high interest if persistence was confirmed in vivo. Finally, our analysis of IFN expression provides some trails to understand the mechanism by which MLB astroviruses can cause persistent infections in the assayed cultures.


2014 ◽  
Vol 32 (15_suppl) ◽  
pp. 5046-5046 ◽  
Author(s):  
Robert J. van Soest ◽  
Ellen S. de Morrée ◽  
Corrina M.A. de Ridder ◽  
Herman Burger ◽  
Erik A.C. Wiemer ◽  
...  

1965 ◽  
Vol 20 (4) ◽  
pp. 290-292 ◽  
Author(s):  
Ernst Hadorn

The occurrence and the manifestation of a cell line is described which had suddenly and irreversibly lost the potency for forming bristles on any part of the adult cuticle after culturing in vivo over a period of more than one year. On the other hand, it is shown that the cells maintained the capacity for the differentiation of the region-specific ground pattern which consists of hairs and other cuticular structures and which characterize antennae, head parts, legs, wings and the thorax. The aristae are not affected by the change which initiated the bristle-less cell line. Thus it is concluded that the aristae are formations belonging to the ground pattern. A general developmental factor which is indispensible for and common to all bristles regard-less of their organspecific structure is postulated.


Sign in / Sign up

Export Citation Format

Share Document