siRNA for the Ig Light Chain Constant Region Reduces Light Chain Production and Secretion By Human Plasma Cells and in a Murine Xenograft Model

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5722-5722
Author(s):  
Xun Ma ◽  
Ping Zhou ◽  
Monika Pilichowska ◽  
Chakra P Chaulagain ◽  
Sandy Wong ◽  
...  

Abstract Background Ig light chain (LC) diseases such as AL amyloidosis and monoclonal light-chain deposition disease are caused by pathologic free LC. Treatment is aimed at eliminating LC production but success is limited. RNA interference (RNAi) can stop LC production but the diversity of LC variable region sequences poses a challenge that targeting consensus sequences in the constant region (CR) of LC mRNA may overcome (Blood 2014;123:3440). We have developed siRNA pools designed to target the κ or λ LC CR mRNA in human plasma cells and impair LC production and secretion, and have shown that the pool targeting the λ LC CR can do so, and can also trigger a terminal unfolded protein response in clones producing intact Ig due to intracellular accumulation of unpaired heavy chains (ibid). Here we report the results of continued in vitro and in vivo testing of these pools in patient specimens and in a murine xenograft model. Methods Pools of siRNA for the κ or λ LC CR (si[IGLCκCR], si[IGLCλCR]) were custom produced with a non-target control (si[-]). They were introduced in vitro into human plasma cells by an optimized streptolysin O-based method (SLO) and in a NOD.SCID xenograft flank plasmacytoma model by in vivo electroporation as per Gene Therapy 2011;18:1150. In vitro we evaluated LC gene expression, production and secretion at 24 hours in human myeloma cell lines and CD138-selected specimens from patients with plasma cell neoplasms, using real-time PCR (qPCR) for LC mRNA, flow cytometry for intracellular LC mean fluorescence intensity (MFI) and ELISA (Bethyl Laboratories) for LC secretion in 24-hour suspension cultures (106 cells/ml). In vivo we inoculated each of the flanks of NOD.SCID mice with 107 human myeloma cells (ALMC-1 or ALMC-2). When plasmacytomas were 0.5cm3 we injected si[IGLCλCR] or si[-] one time to each flank plasmacytoma respectively, allowing each mouse to serve as its own control. Two days later, the mice were sacrificed and the plasmacytomas excised for qPCR for λ LC mRNA and serum was obtained to measure human λ LC levels by ELISA. Results We have previously described results with siRNA targeting the λ LC CR in human cell lines that make λ LC (ALMC-1, ALMC-2, EJM, OPM2, MM.1S, and MM.1R) and in 16 AL λ patient specimens. We demonstrated significant decreases in LC mRNA, intracellular LC MFI, and λ LC secretion by cell lines (Blood 2014;123:3220); moreover, transcriptional profiling indicated minimal off-target effects (ibid; Supplement). We now report that in vitro secretion of λ LC by CD138-selected plasma cells from AL patients (n=3, newly diagnosed λ) treated with si[IGLCλCR] was reduced by 65% from a mean of 3.1 to 1.0µg/ml and that the residual λ LC mRNA was 49% of control. Similarly we treated κ LC secreting human myeloma cell lines with si[IGLCκCR] and si[-] (IM9, H929, JJN-3, and ARH77). By qPCR the residual κ LC mRNA was 13%, by flow cytometry the MFI was reduced by a median of 67.3% (22.5-90.8), and by ELISA mean κ LC secretion was reduced from 3.7 to 0.8µg/ml (P = 0.055, paired t test). We treated CD138-selected κ patient samples (AL 3, LCDD 1, MM 6) in the same way. By qPCR the residual κ LC mRNA was 57% control, by flow cytometry the MFI was reduced by a median of 37.5% (14-69.8), and by ELISA secretion was reduced from 9.4 to 6.5µg/ml (P = 0.02, paired t test). In the murine dual-flank xenograft model employing λ secreting cells, by qPCR there was a reduction in λ LC mRNA with si[IGLCλCR] treatment in 13 of 16 mice (ALMC-1 11/114, ALMC-2 2/2). In these 14, the median λ LC expression was 66% of control (range, 17-97). In 6/13 the average reduction in λ LC expression was 59%. Of note, measurable levels of human λ LC were found in the blood of all mice at sacrifice. Conclusion With one pool of siRNA targeting the constant region of the κ or λ LC we can significantly reduce production and secretion of LC by clonal human plasma cells, including patient cells, and also reduce the expression of LC in xenograft plasmacytomas in vivo. Two methods of siRNA delivery have been employed in this work thus far, SLO and in vivo electroporation, neither of which require endosomal escape. The specificity of the siRNA pools for plasma cell LC genes and the possible receptivity of plasma cells to RNAi are important positive aspects of this work. Further pre-clinical development of Ig LC CR RNAi employing lipid-based nanoparticle platforms is warranted in order to optimize cell-specific delivery, delivery efficiency and siRNA targeting. Disclosures No relevant conflicts of interest to declare.

2014 ◽  
Vol 2014 ◽  
pp. 1-8 ◽  
Author(s):  
Rayelle Itoua Maïga ◽  
Jennifer Lemieux ◽  
Annie Roy ◽  
Carl Simard ◽  
Sonia Néron

The in vitro CD40-CD154 interaction promotes human B lymphocytes differentiation into plasma cells. Currently, CD138 is the hallmark marker enabling the detection of human plasma cells, both in vitro and in vivo; its presence can be monitored by flow cytometry using a specific antibody. We have developed a culture system allowing for the differentiation of memory B lymphocytes. In order to detect the newly formed plasma cells, we have compared their staining using five anti-CD138 monoclonal antibodies (mAbs). As a reference, we also tested human cell lines, peripheral blood mononuclear cells, and bone marrow samples. The five anti-CD138 mAbs stained RPMI-8226 cells (>98%) with variable stain index (SI). The highest SI was obtained with B-A38 mAb while the lowest SI was obtained with DL-101 and 1D4 mAbs. However, the anti-CD138 mAbs were not showing equivalent CD138+cells frequencies within the generated plasma cells. B-A38, B-B4, and MI-15 were similar (15–25%) while DL-101 mAb stained a higher proportion of CD138-positive cells (38–42%). DL-101 and B-A38 mAbs stained similar populations in bone marrow samples but differed in their capacity to bind toCD138highandCD138locell lines. In conclusion, such cellular fluctuations suggest heterogeneity in human plasma cell populations and/or in CD138 molecules.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3100-3100 ◽  
Author(s):  
Ken Flanagan ◽  
Muntasir M Majumder ◽  
Romika Kumari ◽  
Juho Miettinen ◽  
Ana Slipicevic ◽  
...  

Background: Immunoglobulin light-chain (AL) amyloidosis is a rare disease caused by plasma cell secretion of misfolded light chains that assemble as amyloid fibrils and deposit on vital organs including the heart and kidneys, causing organ dysfunction. Plasma cell directed therapeutics, aimed at preferentially eliminating the clonal population of amyloidogenic cells in bone marrow are expected to reduce production of toxic light chain and alleviate deposition of amyloid thereby restoring healthy organ function. Melphalan flufenamide ethyl ester, melflufen, is a peptidase potentiated alkylating agent with potent toxicity in myeloma cells. Melflufen is highly lipophilic, permitting rapid cellular uptake, and is subsequently enzymatically cleaved by aminopeptidases within cells resulting in augmented intracellular concentrations of toxic molecules, providing a more targeted and localized treatment. Previous data demonstrating multiple myeloma plasma cell sensitivity for melflufen suggests that the drug might be useful to directly eliminate amyloidogenic plasma cells, thereby reducing the amyloid load in patients. Furthermore, the increased intracellular concentrations of melflufen in myeloma cells indicates a potential reduction in systemic toxicity in patients, an important factor in the fragile amyloidosis patient population. To assess potential efficacy in amyloidosis patients and to explore the mechanism of action, we examined effects of melflufen on amyloidogenic plasma cells invitro and invivo. Methods: Cellular toxicity and apoptosis were measured in response to either melflufen or melphalan in multiple malignant human plasma cell lines, including the amyloidosis patient derived light chain secreting ALMC-1 and ALMC-2 cells, as well as primary bone marrow cells from AL amyloidosis patients, using annexin V and live/dead cell staining by multicolor flow cytometry, and measurement of cleaved caspases. Lambda light chain was measured in supernatant by ELISA, and intracellular levels were detected by flow cytometry. To assess efficacy of melflufen in vivo, the light chain secreting human myeloma cell line, JJN3, was transduced with luciferase and adoptively transferred into NSG mice. Cell death in response to melflufen or melphalan was measured by in vivo bioluminescence, and serum light chain was monitored. Results: Melflufen demonstrated increased potency against multiple myeloma cell lines compared to melphalan, inducing malignant plasma cell death at lower doses on established light chain secreting plasma cell lines. While ALMC-1 cells were sensitive to both melphalan and melflufen, the IC50 for melphalan at 960 nM was approximately 3-fold higher than melflufen (334 nM). However, ALMC-2 cells were relatively insensitive to melphalan (12600 nM), but maintained a 100-fold increase in sensitivity to melflufen (121 nM). Furthermore, while 40% of primary CD138+ plasma cells from patients with diagnosed AL amyloidosis responded to melflufen treatment in vitro, only 20% responded to melphalan with consistently superior IC50 values for melflufen (Figure 1). Light chain secreting cell lines and AL amyloidosis patient samples were further analyzed by single cell sequencing. We further examined differential effects on apoptosis and the unfolded protein response in vitro in response to either melflufen or melphalan. This is of particular interest in amyloidosis, where malignant antibody producing plasma cells possess an increased requirement for mechanisms to cope with the amplified load of unfolded protein and associated ER stress. As AL amyloidosis is ultimately a disease mediated by secretion of toxic immunoglobulin, we assessed the effects of melflufen on the production of light chain invitro, measuring a decrease in production of light chain in response to melflufen treatment. Finally, we took advantage of a recently described adoptive transfer mouse model of amyloidosis to assess the efficacy of melflufen and melphalan in eliminating amyloidogenic clones and reducing the levels of toxic serum light chain in vivo. Conclusions: These findings provide evidence that melflufen mediated toxicity, previously described in myeloma cells, extends to amyloidogenic plasma cells and further affects the ability of these cells to produce and secrete toxic light chain. This data supports the rationale for the evaluation of melflufen in patients with AL amyloidosis. Figure 1 Disclosures Flanagan: Oncopeptides AB: Employment. Slipicevic:Oncopeptides AB: Employment. Holstein:Celgene: Consultancy; Takeda: Membership on an entity's Board of Directors or advisory committees; Adaptive Biotechnologies: Membership on an entity's Board of Directors or advisory committees; GSK: Consultancy; Genentech: Membership on an entity's Board of Directors or advisory committees; Sorrento: Consultancy. Lehmann:Oncopeptides AB: Employment. Nupponen:Oncopeptides AB: Employment. Heckman:Celgene: Research Funding; Novartis: Research Funding; Oncopeptides: Research Funding; Orion Pharma: Research Funding.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1419-1419
Author(s):  
Soraya Wuilleme-Toumi ◽  
Nelly Robillard ◽  
Patricia Gomez-Bougie ◽  
Philippe Moreau ◽  
Steven Le Gouill ◽  
...  

Abstract Multiple Myeloma (MM) is a fatal malignancy of B-cell origin characterized by the accumulation of plasma cells within the bone marrow. The expression of the pro-survival members of the Bcl-2 family has been shown to be a key process in the survival of myeloma cells. More particularly, Mcl-1 expression turned out to be critical for their survival. Indeed, knockdown of Mcl-1 by antisenses induces apoptosis in myeloma cells. Finally, Mcl-1 was found to be the only anti-apoptotic Bcl-2 family member which level of expression was modified by cytokine treatment of myeloma cells. For these reasons, we have evaluated the expression of Mcl-1 in vivo in normal, reactive and malignant plasma cells (PC) i.e., myeloma cells from 55 patients with MM and 20 human myeloma cell lines using flow cytometry. We show that Mcl-1 is overexpressed in MM in comparison with normal bone marrow PC. Forty-seven percent of patients with MM at diagnosis (p=.017) and 80% at relapse (p=.014 for comparison with diagnosis) overexpress Mcl-1. Of note, only myeloma cell lines but not reactive plasmocytoses have abnormal Mcl-1 expression, although both plasmocyte expansion entities share similar high proliferation rates (>20%). Of interest, Bcl-2 as opposed to Mcl-1, does not discriminate malignant from normal PC. This shows that the overexpression of Mcl-1 is clearly related to malignancy rather than to proliferation. It will be important to know whether the overexpression of Mcl-1 is related to an abnormal response to cytokines like Interleukin-6 or to mutations of the promoter of the Mcl-1 gene as already described in B chronic lymphocytic leukemia. Finally, level of Mcl-1 expression is related to disease severity, the highest values being correlated with the shortest event-free survival (p=.01). In conclusion, Mcl-1 which has been shown to be essential for the survival of human myeloma cells in vitro is overexpressed in vivo in MM and correlates with disease severity. Mcl-1 represents a major therapeutical target in MM.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3208-3208
Author(s):  
Xun Ma ◽  
Ping Zhou ◽  
Adin Kugelmass ◽  
Denis Toskic ◽  
Melissa Warner ◽  
...  

Abstract INTRODUCTION : Despite advances in therapy, patients with relapsed AL amyloidosis die of resistant disease. New therapies are needed. siRNA directed at the constant regions of Ig light chains (LC) reduces LC mRNA and protein from patient cells, from human myeloma and AL cell lines, and in a flank plasmacytoma model with in vivo electroporation (Blood 2014;123:3440; Gene Ther 2016;23:727). To deliver siRNA in vivo, we first tested a series of biodegradable lipidoid nanoparticles (LPN) generated through Michael addition of aliphatic acrylates containing disulfide bonds responsive to intracellular glutathione that enhance siRNA transit from endosome to cytoplasm, and identified the 8B-3 LPN as safe and active in vitro. To provide an in vivo model, we tested RPMI8226, ALMC-1, NCI929 and JJN3 human myeloma reporter cell lines stably expressing FFL and GFP in NOD scid γ (NSG) mice using different routes of inoculation. We sought an optimal xenograft model that would provide reliable tumor-take, brief latency for circulating LC, rapid short-term increase in LC levels, measurable β2-microglobulin (β2M) levels and ease of administration of multiple injections of LPN. The NSG JJN3 intraperitoneal (IP) model met these standards. This model not only enables timely testing of this siRNA approach but also provides the significant challenge of rapid tumor growth. We now report the results in this model of delivery by the 8B-3 LPN of siIGKC targeting κ LC production. MATERIALS AND METHODS: LPN/siRNA are formulated using a microfluid based mixer (NanoAssemblr, Precision Nanosystems, Inc), and are controlled for size by varying the relative flow rates of lipid and solvent. For in vivo delivery, cholesterol, DOPE and PEGylated co-lipids are used to form stable LPN with siNT or siIGKC (Dharmacon). Standard QA metrics are applied to each lot of 8B-3/siRNA and lot-to-lot checks for cell viability and in vitro FFL knockdown are performed. Coated loaded 8B-3 LPN are ~100nM in diameter and were tested in vitro at 8B-3:siRNA ratios of 10:1, 5:1 and 1.5:1. The 1.5:1 ratio was superior; cell viability was unaffected and κ LC reduction was 84%. NSG mice with JJN3 tumor implants (107 cells IP on day 1) are injected IP with 200μL 8B-3:siRNA (1.5:1 ratio) mixed with 400μL PBS once daily on days 5, 6 and 7. Luciferin imaging is obtained on day 5 and blood is obtained on day 5 (pre-injection) and on day 8 for ELISA for human κ LC and β2M. RESULTS : Current JJN3 cells make only κ LC without IgA (Br J Haematol 1999;106:669) and in vitro at 48 hours after a single exposure to 8B-3/siIGKC we see reductions of 84% and 25% in κ LC and β2M secretion with no change in cell viability. Neither κ LC nor β2M reductions are seen with 8B3/siNT. The NSG JJN3 IP model has a 90% tumor-take and a 5-day LC latency. IP xenograft CD138+ cells are found in liver (subcapsular) and spleen. On day 5, the mice have median serum levels (Q1-Q3) of κ LC and β2M of 2.37μg/mL (1.68-3.32) and 1.56ρg/mL (0.58-5.38), values that strongly correlate (r=0.76, P<<0.01), as do the day 5 κ LC and FLUX values (r=0.88, P<<0.01). In 3 cohorts of 10 mice each, 5 siNT and 5 siIGKC per cohort, there were no differences in day 5 κ LC, FLUX and β2M, or in day 8 β2M, between the siNT and siIGKC groups (Table 1). On day 8 after 3 IP injections, the ratio of the medians of κ LCday 8/κ LCday 5 x 100% was lower in siIGKC mice (161% versus 264%) and trends towards significant reductions in κ LC with siIGKC were observed (Table 1). In a paired comparison of the means of the groups in the 3 cohorts the 33% reduction in κ LC with siIGKC was significant (Table 1). On day 8 there were no differences in the weights or behaviour of the mice. CONCLUSIONS : We have previously shown that siIGKC, a pool of siRNA directed at consensus sequences in the κ LC constant region gene, can significantly reduce κ LC production in clonal plasma cells from patients, in human myeloma cell lines, and in vivo in a flank plasmacytoma xenograft model. In this work, we show that 8B-3 is a promising LPN for delivery of siRNA to human plasma cells and, when loaded with siIGKC, can with relative safety significantly reduce circulating κ LC in the NSG JJN3 IP model after 3 daily IP injections despite rapid tumor growth. We also show the utility of the NSG JJN3 IP model for the study of κ LC directed therapies. Extensive work lies ahead to identify and optimize a lead candidate for delivery of siRNA to human bone marrow plasma cells in vivo and to begin systematic pre-clinical safety studies. Disclosures Ma: Tufts Medical Center: Patents & Royalties: Patent: 9593332. Wang:Tufts University Medical School: Patents & Royalties: Patent: 9765022. Xu:Tufts University School of Medicine: Patents & Royalties: Patent: 9765022. Comenzo:Tufts Medical Center: Patents & Royalties: Patent: 9593332.


Biomolecules ◽  
2022 ◽  
Vol 12 (1) ◽  
pp. 107
Author(s):  
Giorgio Santoni ◽  
Consuelo Amantini ◽  
Federica Maggi ◽  
Oliviero Marinelli ◽  
Matteo Santoni ◽  
...  

Multiple myeloma (MM) is a haematological B cell malignancy characterised by clonal proliferation of plasma cells and their accumulation in the bone marrow. The aim of the present study is the evaluation of biological effects of Ibrutinib in human MM cell lines alone or in combination with different doses of Bortezomib. In addition, the relationship between the expression of TRPML2 channels and chemosensitivity of different MM cell lines to Ibrutinib administered alone or in combination with Bortezomib has been evaluated. By RT-PCR and Western blot analysis, we found that the Ibrutinib-resistant U266 cells showed lower TRPML2 expression, whereas higher TRPML2 mRNA and protein levels were evidenced in RPMI cells. Moreover, TRPML2 gene silencing in RPMI cells markedly reverted the effects induced by Ibrutinib alone or in combination with Bortezomib suggesting that the sensitivity to Ibrutinib is TRPML2 mediated. In conclusion, this study suggests that the expression of TRPML2 in MM cells increases the sensitivity to Ibrutinib treatment, suggesting for a potential stratification of Ibrutinib sensitivity of MM patients on the basis of the TRPML2 expression. Furthermore, studies in vitro and in vivo should still be necessary to completely address the molecular mechanisms and the potential role of TRPML2 channels in therapy and prognosis of MM patients.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3213-3213
Author(s):  
Georgia Rose Stewart ◽  
Simon Tazzyman ◽  
Darren Lath ◽  
Jenny Down ◽  
John A Snowden ◽  
...  

Abstract Multiple myeloma is a largely incurable disease and despite current therapies achieving good initial responses, patients frequently relapse. Therefore, new approaches are required that not only reduce the tumour load, but also prevent the growth of residual disease. One such approach is the use of oncolytic viruses. We developed an oncolytic adenovirus that utilizes transcriptional control of E1A under the myeloma-specific promoter CS1 (ADCE1A). We hypothesised that ADCE1A would be myeloma-specific, inducing tumour oncolysis and preventing tumour regrowth. A panel of myeloma cell lines (JJN3, L-363, OPM-2, U-266, RMPI-8226, NCI-H929, KMS-11, KMS-12-BM) were assessed for expression of CS1 by flow cytometry (FC). The same panel were treated with ADCE1A and cell death was monitored after 72 hours using FC and propidium iodide staining. Apoptosis was assessed following ADCE1A infection using annexin V staining and analysed by FC at 6 and 24 hours post treatment. CD138+ plasma cells from bone marrow aspirates were obtained from myeloma and plasma cell leukaemia patients and from the peripheral blood from healthy donors. The CD138+ and CD138- populations from these samples were treated with ADCE1A and cell death was monitored after 4 days using FC and propidium iodide staining. Myeloma cell regrowth was assessed after bortezomib (0.56-2.81nM) or bortezomib in combination with ADCE1A treatment using cell counts. Viral efficacy was tested in a xenograft model of myeloma, where 5 weeks after tumour cell injection (106 U266 cells intravenously), mice were treated with ADCE1A (1x107 pfu, 2x/wk) or control (PBS) for 3 weeks. Tumour burden was measured ex vivo in bone marrow flushes of the long bones by FC. CS1 was expressed in all myeloma cell lines, except KMS-12-BM. ADCE1A infected, replicated and caused oncolysis in JJN-3, L-363, OPM-2, U-266, RPMI-8226, NCI-H929, and KMS-11 myeloma cell lines. However, KMS-12-BM had the lowest sensitivity to ADCE1A. This correlated with CS1 expression, as CS1 was not expressed at the protein level in this cell line. Apoptosis, as detected by annexin V staining, was found to be a cell death mechanism involved in ADCE1A oncolysis. Importantly, ADCE1A induced oncolysis in primary patient malignant CD138+ plasma cells, but not in the non-malignant CD138- bone marrow mononuclear population from these patients. Additionally, ADCE1A had no effect on cell death in non-malignant CD138+ plasma cells and non-malignant CD138- peripheral blood mononuclear population from healthy donors. ADCE1A prevented regrowth of myeloma cell lines following treatment with bortezomib in vitro. In the U266 xenograft model, tumour load was significantly reduced (p<0.05) compared to control treated mice. In summary, ADCE1A has potential clinical efficacy as shown by preclinical models and patient tumour samples. Additionally, ADCE1A was able to stop tumour cell regrowth after chemotherapy in vitro, therefore, the use of oncolytic adenoviruses to target minimal residual disease may be a novel yet promising approach for the treatment of myeloma. Disclosures Snowden: Jannssen/J&J: Other: Speaker fees; Jazz & Sanofi: Other: Speaker fees at ASH.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3487-3487
Author(s):  
Michael Timm ◽  
Linda Wellik ◽  
Teresa Kimlinger ◽  
Jessica Haug ◽  
Michael Kline ◽  
...  

Abstract Background: Multiple myeloma remains incurable with current approaches and newer therapies are needed to improve the outcome of these patients. While monoclonal antibody based therapies have been successful in some of the hematological malignancies, such approaches have had limited efficacy in the setting of myeloma. Thymoglobulin (polyclonal rabbit antithymocyte globulin, Genzyme) (Thymo) has been extensively evaluated in the setting of allogeneic blood and marrow transplantation and solid organ transplants. Given the polyclonal nature of this product, with antibodies against different B cell antigens, we evaluated the in vitro and in vivo activity of Thymo in myeloma. Methods: MM cell lines were cultured in RPMI 1640 containing 10% fetal bovine serum supplemented with L-Glutamine, penicillin, and streptomycin. The KAS-6/1 cell line was also supplemented with 1 ng/ml IL-6. Cytotoxicity following drug treatment was measured using the MTT viability assay. Apoptosis was measured by flow cytometry using Annexin V/PI in cell lines and Apo 2.7 in primary patient plasma cells. Shifts in expression of a variety of different B cell and plasma cell antigens were examined on several different myeloma cell lines following Thymo treatment in order to identify the potential antigenic targets. In vivo activity of thymo was evaluated in a SCID plasmacytoma model injected with RPMI myeloma cell lines. Results: rATG was cytotoxic in vitro to several MM cell lines (RPMI 8226, U266, OPM1, OPM2) including the IL-6 dependent cell line Kas6/1 with LC50 of around 1 mg/mL. Additionally, thymo was cytotoxic MM cell lines resistant to conventional agents such as doxorubicin (Dox40), melphalan (LR5) and dexamethasone (MM1R). Thymo induced apoptosis in MM cell lines and in patient derived primary myeloma cells. When tested in combination with other anti-myeloma agents an additive effect was seen with doxorubicin, PS341 and melphalan. Using competitive flow cytometry, we identified CD138, CD38, Cd45, CD126, CD49d (VLA4), as well as CD20 as antigens likely to be targeted by Thymo. Tumor bearing mice injected with Thymo at two different doses (5 mg/kg and 10 mg/kg for five days) had significantly delayed tumor growth compared to non-injected mice, and this translated into a better survival for these mice. Mice receiving 10 mg/kg dose had a slower tumor growth compared to 5 mg/kg dose (Figure). Conclusions: Thymoglobulin has promising in vitro and in vivo activity in the setting of myeloma. These studies will provide the rational for future clinical development of this agent in myeloma alone or in combination with other agents. Based on these results, we are in the process of initiating a clinical trial combining Thymo with Melphalan. Figure Figure


Cancers ◽  
2021 ◽  
Vol 13 (14) ◽  
pp. 3487
Author(s):  
Yu-Ling Lu ◽  
Ming-Hsien Wu ◽  
Yi-Yin Lee ◽  
Ting-Chao Chou ◽  
Richard J. Wong ◽  
...  

Differentiated thyroid cancer (DTC) patients are usually known for their excellent prognoses. However, some patients with DTC develop refractory disease and require novel therapies with different therapeutic mechanisms. Targeting Wee1 with adavosertib has emerged as a novel strategy for cancer therapy. We determined the effects of adavosertib in four DTC cell lines. Adavosertib induces cell growth inhibition in a dose-dependent fashion. Cell cycle analyses revealed that cells were accumulated in the G2/M phase and apoptosis was induced by adavosertib in the four DTC tumor cell lines. The sensitivity of adavosertib correlated with baseline Wee1 expression. In vivo studies showed that adavosertib significantly inhibited the xenograft growth of papillary and follicular thyroid cancer tumor models. Adavosertib therapy, combined with dabrafenib and trametinib, had strong synergism in vitro, and revealed robust tumor growth suppression in vivo in a xenograft model of papillary thyroid cancer harboring mutant BRAFV600E, without appreciable toxicity. Furthermore, combination of adavosertib with lenvatinib was more effective than either agent alone in a xenograft model of follicular thyroid cancer. These results show that adavosertib has the potential in treating DTC.


2020 ◽  
Vol 15 (1) ◽  
pp. 871-883
Author(s):  
Jinshan Zhang ◽  
Dan Rao ◽  
Haibo Ma ◽  
Defeng Kong ◽  
Xiaoming Xu ◽  
...  

AbstractBackgroundOsteosarcoma is a common primary malignant bone cancer. Long noncoding RNA small nucleolar RNA host gene 15 (SNHG15) has been reported to play an oncogenic role in many cancers. Nevertheless, the role of SNHG15 in the doxorubicin (DXR) resistance of osteosarcoma cells has not been fully addressed.MethodsCell Counting Kit-8 assay was conducted to measure the half-maximal inhibitory concentration value of DXR in osteosarcoma cells. Western blotting was carried out to examine the levels of autophagy-related proteins and GDNF family receptor alpha-1 (GFRA1). Quantitative reverse transcription-polymerase chain reaction was performed to determine the levels of SNHG15, miR-381-3p, and GFRA1. The proliferation of osteosarcoma cells was measured by MTT assay. The binding sites between miR-381-3p and SNHG15 or GFRA1 were predicted by Starbase bioinformatics software, and the interaction was confirmed by dual-luciferase reporter assay. Murine xenograft model was established to validate the function of SNHG15 in vivo.ResultsAutophagy inhibitor 3-methyladenine sensitized DXR-resistant osteosarcoma cell lines to DXR. SNHG15 was upregulated in DXR-resistant osteosarcoma tissues and cell lines. SNHG15 knockdown inhibited the proliferation, DXR resistance, and autophagy of osteosarcoma cells. MiR-381-3p was a direct target of SNHG15, and GFRA1 bound to miR-381-3p in osteosarcoma cells. SNHG15 contributed to DXR resistance through the miR-381-3p/GFRA1 axis in vitro. SNHG15 depletion contributed to the inhibitory effect of DXR on osteosarcoma tumor growth through the miR-381-3p/GFRA1 axis in vivo.ConclusionsSNHG15 enhanced the DXR resistance of osteosarcoma cells through elevating the autophagy via targeting the miR-381-3p/GFRA1 axis. Restoration of miR-381-3p expression might be an underlying therapeutic strategy to overcome the DXR resistance of osteosarcoma.


2021 ◽  
Author(s):  
Evelyn M. Mrozek ◽  
Vineeta Bajaj ◽  
Yanan Guo ◽  
Izabela Malinowska ◽  
Jianming Zhang ◽  
...  

Inactivating mutations in either TSC1 or TSC2 cause Tuberous Sclerosis Complex, an autosomal dominant disorder, characterized by multi-system tumor and hamartoma development. Mutation and loss of function of TSC1 and/or TSC2 also occur in a variety of sporadic cancers, and rapamycin and related drugs show highly variable treatment benefit in patients with such cancers. The TSC1 and TSC2 proteins function in a complex that inhibits mTORC1, a key regulator of cell growth, which acts to enhance anabolic biosynthetic pathways. In this study, we identified and validated five cancer cell lines with TSC1 or TSC2 mutations and performed a kinase inhibitor drug screen with 197 compounds. The five cell lines were sensitive to several mTOR inhibitors, and cell cycle kinase and HSP90 kinase inhibitors. The IC50 for Torin1 and INK128, both mTOR kinase inhibitors, was significantly increased in three TSC2 null cell lines in which TSC2 expression was restored.  Rapamycin was significantly more effective than either INK128 or ganetespib (an HSP90 inhibitor) in reducing the growth of TSC2 null SNU-398 cells in a xenograft model. Combination ganetespib-rapamycin showed no significant enhancement of growth suppression over rapamycin. Hence, although HSP90 inhibitors show strong inhibition of TSC1/TSC2 null cell line growth in vitro, ganetespib showed little benefit at standard dosage in vivo. In contrast, rapamycin which showed very modest growth inhibition in vitro was the best agent for in vivo treatment, but did not cause tumor regression, only growth delay.


Sign in / Sign up

Export Citation Format

Share Document