Targeted Radioimmunotherapy Using Anti-CD45 Antibody-Mediated Delivery Of An In Vivo Nanoparticle-Based Alpha Generator In a Xenograft Model Of Human Leukemia

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2908-2908
Author(s):  
Aimee L. Kenoyer ◽  
Alexandra H. Hernandez ◽  
Mazyar Shadman ◽  
Mark McLaughlin ◽  
David Robertson ◽  
...  

Abstract Introduction Acute Myeloid Leukemia (AML) relapse rates remain high despite treatment with combination chemotherapy and hematopoietic cell transplantation. New targeted treatment modalities including radioimmunotherapy (RIT) have been developed to reduce relapse. However, minimally toxic means of targeting AML cells for delivery of radionuclides may not be optimal for all therapeutically favorable isotopes. Alpha (α) particles have a higher linear energy transfer and shorter path length in contrast to beta (β) particles, thus leading to the promising exploration of α-particle RIT for AML. Treatment efficacies may be further improved by the use of in vivo α-generators such as 225Ac, which emits a total of 4 α-particles upon decay. However, targeted therapy utilizing traditional 225Ac chelates poses a challenge since the α-emitting daughter radionuclides while short-lived, are difficult to retain near the targeted malignant cells and may be released into circulation, leading to unwanted non-specific toxicities such as to the renal or hepato-biliary systems. Methods To improve the therapeutic potential of 225Ac-based RIT for AML and reduce toxicities we have developed an anti-CD45 antibody (Ab)-conjugated gold-coated lanthanide phosphate nanoparticle that contains multiple gadolinium (Gd) shells designed to sequester 225Ac in the core and retain the α-daughters within the nanoparticle. Previous work demonstrated the in vivo retention of the 221Fr daughter radionuclide of over 90% for at least 3 weeks past the core nanoparticle synthesis. In this work the nanoparticle core was first synthesized using LaCl3 and GdCl3 using 177Lu as a radioactive surrogate for 225Ac. Four consecutive GdPO4 layers were added to the lanthanide core, followed by NaAuCl4 to form a metallic gold coat. Anti-human CD45 Ab (BC8) was separately labeled with 125I and then conjugated to the gold coat of the nanoparticle via a polyethylene glycol linker. This dual-labeled approach allowed for verification of stability of the Ab-nanoparticle. A competitive binding flow cytometry assay was used to measure the efficiency of the Ab conjugation to the nanoparticle and determine the concentration of Ab in solution. In vivo targeting of the Ab-nanoparticle was initially tested in athymic nude mice bearing human AML xenografts by injection with 50 µCi 177Lu-anti-CD45 Ab-nanoparticle at 100 µg per dose of Ab injected. Results Nanoparticle-BC8 dual-labeled with 177Lu and 125I bound effectively in vitro to human AML cells (HEL) with an increase in mean fluorescence index (MFI) of 9.2-fold compared to non-binding isotype control. Labeled nanoparticle-BC8 also effectively bound to human Burkitt’s lymphoma Ramos and Raji cells with an increased MFI of 86- and 36-fold compared to control, respectively. A 96-well plate-based assay for cell binding to test radioactive conjugates was also employed to verify that anti-CD45 Ab-nanoparticle remained stable in vitro. Non-conjugated BC8 effectively blocked binding of 125I-BC8-conjugated 177Lu-nanoparticle to Ramos cells in comparison with control with a reduction in binding by 21.3 % with respect to 125I-BC8 (p = 0.016) and 20.2 % with respect to the 177Lu-NP core (p = 0.026). 125I-labeled nanoparticle-BC8 conjugate bound effectively in vivo to human leukemia xenografts. Favorable targeting to sites of disease was seen by 4 hours post-injection, with 18.0 ±2.9 % injected dose per gram of targeting Ab in the tumor. Conclusion Combined nanoparticle-antibody therapy is a promising, novel approach to target malignant cells. Antibody-mediated delivery of α-particle generators represents a potential solution for the difficulties of safe and effective targeting using 225Ac, largely nonspecific toxicities due to dispersal of α-particle daughters. We have shown that 177Lu is a useful surrogate for 225Ac for preliminary characterization assays, and that 177Lu is retained over time in the nanoparticle core. We have shown in vitro targeting of leukemia and lymphoma cells and have made strides towards obtaining a favorable biodistribution in a model of human AML. However, challenges remain as liver uptake by nanoparticles cleared through the reticuloendothelial system is unfavorable and may cause dose-limiting toxicities. Future work will further characterize nanoparticle-Ab conjugates and progress toward studies involving 225Ac for AML therapy. Disclosures: Press: Genentech, inc.: Consultancy, Research Funding.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3729-3729
Author(s):  
Heather Gilbert ◽  
John Cumming ◽  
Josef T. Prchal ◽  
Michelle Kinsey ◽  
Paul Shami

Abstract Abstract 3729 Poster Board III-665 Mantle cell lymphoma (MCL) is a well defined B-cell non-Hodgkin lymphoma characterized by a translocation that juxtaposes the BCL1 gene on chromosome 11q13, which encodes cyclin D1 (CD1), next to the immunoglobulin heavy chain gene promoter on chromosome 14. The resulting constitutive overexpression of CD1 leads to a deregulated cell cycle and activation of cell survival mechanisms. In addition, the gene which encodes GST-n, an enzyme that has been implicated in the development of cancer resistance to chemotherapy, is also located on chromosome 11q13 and is often coamplified along with the BCL1 gene in MCL (1). These two unique biological features of MCL - the overproduction of cyclin D1 and GST-n – may be involved in the carcinogenesis, tumor growth and poor response of this disease to treatment, and they offer potential mechanisms for targeted anti-cancer therapy. Nitric oxide (NO) is a biologic effector molecule that contributes to a host's immune defense against microbial and tumor cell growth. Indeed, NO is potently cytotoxic to tumor cells in vitro (2–4). However, NO is also a potent vasodilator and induces hypotension, making the in vivo administration of NO very difficult. To use NO in vivo requires agents that selectively deliver NO to the targeted malignant cells. A new compound has recently been developed that releases NO upon interaction with glutathione in a reaction catalyzed by GST-n. JS-K seeks to exploit known GST-n upregulation in malignant cells by generating NO directly in cancer cells, and it has been shown to decrease the growth and increase apoptosis in vitro in AML cell lines, AML cells freshly isolated from patients, multiple myeloma cell lines, hepatoma cells and prostate cancer cell lines (3, 5–7). JS-K also decreases tumor burden in NOD/SCID mice xenografted with AML and multiple myeloma cells (5, 7). Importantly, JS-K has been used in cytotoxic doses in the mouse model without significant hypotension. To evaluate whether JS-K treatment has anti-tumor activity in MCL, the human MCL cell lines Jeko1, Mino, Granta and Hb-12 were grown with media only, with JS-K at varying concentrations and with DMSO as an appropriate vehicle control. For detection of apoptotic cells, cell-surface staining was performed with FITC-labeled anti–Annexin V and PI. Cell growth was evaluated using the Promega MTS cytotoxicity assay. Results show that JS-K (at concentrations up to 10 μM) inhibits the growth of MCL lines compared to untreated controls, with an average IC50 of 1 μM. At 48 hours of incubation, all cell lines showed a significantly greater rate of apoptosis than untreated controls. A human MCL xenograft model was then created by subcutaneously injecting two NOD/SCID IL2Rnnull mice with luciferase-transfected Hb12 cells. Seven days post-injection, one of the mice was treated with JS-K at a dose of 4 μmol/kg (expected to give peak blood levels of around 17 mM in a 20 g mouse). Injections of JS-K were given intravenously through the lateral tail vein 3 times a week. The control mouse was injected with an equivalent volume of micellar formulation (vehicle) without active drug. The Xenogen bioluminescence imaging clearly showed a difference in tumor viability, with a significantly decreased signal in the JS-K treated mouse. Our studies demonstrate that JS-K markedly decreases cell proliferation and increases apoptosis in a concentration- and time-dependent manner in mantle cells in vitro. In a xenograft model of mantle cell lymphoma, treatment with JS-K results in decreased tumor viability. Proposed future research includes further defining the molecular basis of these treatment effects; using this therapy in combination with other cancer treatments both in vitro and in vivo; and studying JS-K treatment in MCL patients. Disclosures: Shami: JSK Therapeutics: Founder, Chief Medical Officer, Stockholder.


2012 ◽  
Vol 49 (5) ◽  
pp. 802-810 ◽  
Author(s):  
R. C. Goupil ◽  
J. J. Bushey ◽  
J. Peters-Kennedy ◽  
J. J. Wakshlag

Canine osteosarcoma is an insidious disease with few effective treatment modalities; therefore, use of pharmacologic intervention to improve mortality or morbidity is constantly sought. The use of cyclooxygenase enzyme inhibitors has been an area of interest with limited efficacy based on retrospective examination of tumor expression and in vivo cell proliferation models. Recently, examination of dual cyclooxygenase and 5-lipoxygenase inhibitors in human and canine oncology suggests that 5-lipoxygenase inhibitors may be an effective approach in vitro and during tumor induction in rodent models. Therefore, the authors decided to examine 5-lipoxygenase expression in primary canine osteosarcoma samples and have shown that approximately 65% of osteosarcomas label positive for cytoplasmic 5-lipoxygenase. Further examination of a cell culture and xenograft model shows similar 5-lipoxygenase expression. Surprisingly, a canine 5-lipoxygenase inhibitor (tepoxalin) significantly reduced cell proliferation at physiologic doses in vitro and diminished xenograft tumor growth in nude mice, suggesting that further investigation is needed. Traditionally, 5-lipoxygense leads to production of lipid mediators, such as leukotriene B4 and 5-oxo-eicosatetraenoic acid, which, when added back to the media of tepoxalin-treated cells, did not recover cell proliferation. The lack of nuclear staining in primary and xenografted tumors and the lack of response to eicoasanoids suggest that lipid mediator production is not the primary means by which tepoxalin acts to alter proliferation. Regardless of the mechanisms involved in retarding cell proliferation, future investigation is warranted.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii90-ii91
Author(s):  
Kaijun Di ◽  
Senjie Du ◽  
Javier Lepe ◽  
Nitesh Nandwana ◽  
Bhaskar Das ◽  
...  

Abstract BACKGROUNDS Glioblastoma (GBM) is an aggressive infiltrative brain tumor, and has an extremely poor prognosis despite the use of multiple treatment modalities, including surgery, radiation, and chemotherapy. Meanwhile, mitochondrial changes represent a significant part of cancer cell biology since cancer cells must survive and adapt to challenging microenvironments, specifically in conditions where tumor growth makes oxygen and glucose scarce. As GBM is characterized by extensive hypoxia-induced phenotypic changes such as abnormal vascular proliferation and necrosis, regulation of mitochondrial function could be a novel approach for treating GBM that currently lacks effective therapies. Magmas (mitochondria-associated protein involved in granulocyte-macrophage colony-stimulating factor signal transduction) is a nuclear gene that encodes for the mitochondrial import inner membrane translocase subunit Tim16. We previously demonstrated that a novel Magmas inhibitor, BT#9, significantly exerted anti-tumor effect in glioma in vitro, and may cross the blood brain barrier in vivo, indicating that Magmas inhibitor may be a new chemotherapeutic agent for the treatment of GBM. METHODS In this study, the antitumor effect of Magmas inhibitor BT#9 was tested in an orthotopic xenograft model of human GBM. The molecular mechanism of BT#9 was investigated using glioma cell lines. RESULTS The mice were tolerated to BT#9, and there was no statistical difference in the weight of animals between the control and MTD (Maximum-tolerated Dose, 50mg/kg) groups. The immunocompromised mice, intracranially implanted with human D-54 GBM xenografts, survived significantly longer than the controls (P< 0.5) when treated with BT#9 at MTD. In vitro study showed that the MAP kinase pathways are involved in BT#9-induced tumor suppression. DISCUSSION This is the first study on the role of Magmas in glioma in vivo. Our findings suggested that Magmas plays a key role in glioma survival and targeting Magmas by Magmas inhibitor has the potential to become a therapeutic strategy in glioma patients.


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 14143-14143 ◽  
Author(s):  
A. Telekes ◽  
E. Rásó

14143 Background: The positive effect of the wheat germ extract Avemar has already been proved in cancer. Compared to the control group significantly longer survival times were achieved in both in vivo experiments and clinical studies. Inhibition of cell growth was also detected in K562 human leukaemia cell line in vitro. Avemar given p.o.(3 g/kg) resulted in significant increase of the survival time compared to the control group (p<0.005 Mann-Whitney) in i.v. implanted K562 xenograft model, which was practically the same as the effect of Gleevec treatment. Since, the mechanism(s) of action of Avemar is still not properly characterized a kinase expression panel in K562 in vitro model was examined. Methods: K562 cells (8x105 cell/ml), were treated with Avemar (500 μg/ml) and mRNS from 3–3 parallel samples and their appropriate controls were isolated 24, 48 hours after the treatment and 24 hours after washing the cells previously treated with Avemar for 48 hours. To determine the kinase expression pattern Kinase OpenArray™ plates were used, having over 500 kinase genes with controls in quadruplicates in each plate. Changes in expression was declared if the average value was over 1 (2-fold change in mRNA copy number) and the standard deviation was relatively small (2xSTDEV = AVERAGE). Results: We have found 16 kinases which expression has temporary or durative (maintained for 24 hour after washing) decreased (e.g.: CCL2, ABR, FLT1, EphB6, TGFa) and 30 which expression has increased (e.g.: CPT1B, IRE1, ITK, RON, LTK, EphB2, FASTK). Conclusions: Our result demonstrated that many of the kinases which expression was altered by Avemar treatment is known to participate in cell cycle, cell migration, apoptosis and signal transduction. Thus, our results might shed light on the main mechanism(s) of action of Avemar and raise the possibility to identify the active substance(es) of this natural extract. No significant financial relationships to disclose.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3749-3749 ◽  
Author(s):  
Aimee L. Kenoyer ◽  
Yukang Lin ◽  
Johnnie J. Orozco ◽  
Ethan Balkin ◽  
Don Hamlin ◽  
...  

Abstract Introduction: Acute Myeloid Leukemia (AML) has a high rate of relapse despite hematopoietic cell transplantation (HCT). Pre-targeted radioimmunotherapy (PRIT) prior to HCT offers a promising approach to reduce relapse for high-risk patients. PRIT studies with antibody-streptavidin (Ab-SAV) conjugates have shown promise, but may be limited by SAV immunogenicity. We have developed a recombinant anti-human CD45 bispecific Ab with picomolar affinity to radiolabeled DOTA chelates for targeting AML xenografts in a preclinical model. Methods: We engineered a bispecific Ab recognizing CD45 and DOTA-metal chelates. The light chain sequence from anti-human CD45 Ab (BC8) was cloned into a pFUSE vector along with a single-chain variable region developed to bind to DOTA chelates of multiple radionuclides. Ab was co-transfected into HEK 293 F cells with a pFUSE vector containing heavy chain sequence from Rituximab. Cultures were grown to extinction, and Ab purified via Protein A ion-exchange chromatography. Ab was characterized by SDS-PAGE, HPLC, and flow cytometry to determine specificity and integrity of material. Ab was tested for in vivo targeting in a xenograft model of human AML. Athymic mice bearing flank HEL tumors (30 to 100 µg) were injected with 280 µg bispecific Ab or 300 µg Ab-SAV. Twenty two hours later mice were treated with or without clearing agent (5 µg DOTAY-dextran for bispecific Ab and 50 µg NAGB for Ab-SAV groups). Two hours later mice were given Yttrium-90 (90Y)-DOTA-biotin. Blood was taken at time points over 22 hours post-injection for pharmacokinetic study; xenografts and normal tissues were taken 4, 24, and 48 hours after DOTA-biotin injection for gamma counts. Results: Bispecific Ab targeting CD45 bound to human AML cells in vitro with an increase in mean fluorescence intensity >2 logs compared to control. SDS-PAGE and HPLC demonstrated a construct of expected size. Blood clearance of 90Y-DOTA-biotin was assessed following injection of bispecific Ab with or without clearing agent and compared to mice that received BC8-SAV as the first-step reagent. Even without clearing agent, mice that had received bispecific Ab showed rapid blood clearance of 90Y-DOTA-biotin similar to that of Ab-SAV-injected mice that had received clearing agent, with 8.15 ± 0.56 % ID/g in the blood 5 minutes post-DOTA-biotin injection falling to 1.02 ± 0.62% ID/g within 4 hours and 0.40 ± 0.16 % ID/g at 20 hours. Mice that received BC8-SAV followed by clearing agent had 7.32 ± 2.0 % ID/g in the blood 5 minutes post-injection, falling to 1.13 ± 0.77 % within 4 hours and 0.73 ± 0.46 % at 20 hours. Mice receiving bispecific Ab followed by DOTAY-dextran clearing agent and 90Y-DOTA-biotin showed a rapid blood clearance with a radioactivity concentration of 8.07 ± 3.0% ID/g at 5 minutes and falling to 0.14 ± 0.03 % within 4 hours post-injection. In vivo targeting of the bispecific Ab was similar to Ab-SAV groups; within 4 hours 3.62 ± 0.91 % ID/g was detected in the tumor compared to 0.12 ± 0.05 % in the blood for mice receiving bispecific Ab, while Ab-SAV mice had a tumor value of 3.85 ± 2.5 % and blood was 1.17 ± 0.63 %. Uptake in non-target tissues was low at 4 hours post-injection compared to tumor, with liver and kidney values of 0.05 ± 0.014 % ID/g and 0.518 ± 0.09 % ID/g, respectively. Tumor retention remained steady at 24 and 48 hours with values of 4.13 ± 1.0% ID/g and 4.14 ± 2.47% ID/g respectively. Groups that received bispecific Ab as the first-step agent followed by clearing agent and then 90Y-DOTA-biotin showed slightly higher uptake in normal organs early on, but displayed higher uptake in target tissue. Tumor values at 4, 24, and 48 hours post-injection were 4.65 ± 2.4, 5.89 ± 4.2, and 3.95 ± 2.3 % ID/g, respectively. Conclusion: Recombinant bispecific Ab targeting both human CD45 and radiometal-labeled DOTA is promising as a novel therapeutic approach for AML prior to HCT. Bispecific Ab targets human AML cells in vitro and in vivo with favorable biodistribution showing specific uptake in tumors while normal organs displayed minimal uptake. Blood clearance of the bispecific Ab is rapid and comparable to that of Ab-SAV, even without the addition of a clearing agent. Future studies will assess therapeutic potential of this bispecific Ab PRIT approach in a murine model of AML. Funding Source Acknowledgment: Research reported in this publication was supported by the National Cancer Institute under award numbers R01CA109663, R01CA136639, and R01CA138720. Disclosures No relevant conflicts of interest to declare.


Cancers ◽  
2020 ◽  
Vol 12 (4) ◽  
pp. 853 ◽  
Author(s):  
Alireza Basti ◽  
Rita Fior ◽  
Müge Yalҫin ◽  
Vanda Póvoa ◽  
Rosario Astaburuaga ◽  
...  

Malfunctions of circadian clock trigger abnormal cellular processes and influence tumorigenesis. Using an in vitro and in vivo xenograft model, we show that circadian clock disruption via the downregulation of the core-clock genes BMAL1, PER2, and NR1D1 impacts the circadian phenotype of MYC, WEE1, and TP53, and affects proliferation, apoptosis, and cell migration. In particular, both our in vitro and in vivo results suggest an impairment of cell motility and a reduction in micrometastasis formation upon knockdown of NR1D1, accompanied by altered expression levels of SNAI1 and CD44. Interestingly we show that differential proliferation and reduced tumour growth in vivo may be due to the additional influence of the host-clock and/or to the 3D tumour architecture. Our results raise new questions concerning host–tumour interaction and show that core-clock genes are involved in key cancer properties, including the regulation of cell migration and invasion by NR1D1 in zebrafish xenografts.


Toxins ◽  
2020 ◽  
Vol 12 (3) ◽  
pp. 157 ◽  
Author(s):  
Adriana Tomoko Nishiya ◽  
Marcia Kazumi Nagamine ◽  
Ivone Izabel Mackowiak da Fonseca ◽  
Andrea Caringi Miraldo ◽  
Nayra Villar Scattone ◽  
...  

Canine oral mucosal melanomas (OMM) are the most common oral malignancy in dogs and few treatments are available. Thus, new treatment modalities are needed for this disease. Bacillus anthracis (anthrax) toxin has been reengineered to target tumor cells that express urokinase plasminogen activator (uPA) and metalloproteinases (MMP-2), and has shown antineoplastic effects both, in vitro and in vivo. This study aimed to evaluate the effects of a reengineered anthrax toxin on canine OMM. Five dogs bearing OMM without lung metastasis were included in the clinical study. Tumor tissue was analyzed by immunohistochemistry for expression of uPA, uPA receptor, MMP-2, MT1-MMP and TIMP-2. Animals received either three or six intratumoral injections of the reengineered anthrax toxin prior to surgical tumor excision. OMM samples from the five dogs were positive for all antibodies. After intratumoral treatment, all dogs showed stable disease according to the canine Response Evaluation Criteria in Solid Tumors (cRECIST), and tumors had decreased bleeding. Histopathology has shown necrosis of tumor cells and blood vessel walls after treatment. No significant systemic side effects were noted. In conclusion, the reengineered anthrax toxin exerted inhibitory effects when administered intratumorally, and systemic administration of this toxin is a promising therapy for canine OMM.


Author(s):  
Zizhen Si ◽  
Lei Yu ◽  
Haoyu Jing ◽  
Lun Wu ◽  
Xidi Wang

Abstract Background Long non-coding RNAs (lncRNA) are reported to influence colorectal cancer (CRC) progression. Currently, the functions of the lncRNA ZNF561 antisense RNA 1 (ZNF561-AS1) in CRC are unknown. Methods ZNF561-AS1 and SRSF6 expression in CRC patient samples and CRC cell lines was evaluated through TCGA database analysis, western blot along with real-time PCR. SRSF6 expression in CRC cells was also examined upon ZNF561-AS1 depletion or overexpression. Interaction between miR-26a-3p, miR-128-5p, ZNF561-AS1, and SRSF6 was examined by dual luciferase reporter assay, as well as RNA binding protein immunoprecipitation (RIP) assay. Small interfering RNA (siRNA) mediated knockdown experiments were performed to assess the role of ZNF561-AS1 and SRSF6 in the proliferative actives and apoptosis rate of CRC cells. A mouse xenograft model was employed to assess tumor growth upon ZNF561-AS1 knockdown and SRSF6 rescue. Results We find that ZNF561-AS1 and SRSF6 were upregulated in CRC patient tissues. ZNF561-AS1 expression was reduced in tissues from treated CRC patients but upregulated in CRC tissues from relapsed patients. SRSF6 expression was suppressed and enhanced by ZNF561-AS1 depletion and overexpression, respectively. Mechanistically, ZNF561-AS1 regulated SRSF6 expression by sponging miR-26a-3p and miR-128-5p. ZNF561-AS1-miR-26a-3p/miR-128-5p-SRSF6 axis was required for CRC proliferation and survival. ZNF561-AS1 knockdown suppressed CRC cell proliferation and triggered apoptosis. ZNF561-AS1 depletion suppressed the growth of tumors in a model of a nude mouse xenograft. Similar observations were made upon SRSF6 depletion. SRSF6 overexpression reversed the inhibitory activities of ZNF561-AS1 in vivo, as well as in vitro. Conclusion In summary, we find that ZNF561-AS1 promotes CRC progression via the miR-26a-3p/miR-128-5p-SRSF6 axis. This study reveals new perspectives into the role of ZNF561-AS1 in CRC.


Sign in / Sign up

Export Citation Format

Share Document