Establishment of Bioluminescence Imaging Based Leukemia In Vivo Model for Preclinical Testing

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4879-4879
Author(s):  
Myoung Woo Lee ◽  
Hye Jin Kim ◽  
Dae Seong Kim ◽  
Meong Hi Son ◽  
Soo Hyun Lee ◽  
...  

Abstract Abstract 4879 Background. A hematological malignant animal model is an essential tool for evaluating efficacy of anti-cancer drugs and elucidating underlying mechanism of leukemogenesis. Intraperitoneal (IP) and intravenous (IV) xenograft of acute lymphoblastic leukemia (ALL) cells have limited capacity as in vivo anti-cancer drug screening system. Purpose. In this study, we aimed to establish an ALL animal model using NOD/SCID mouse and evaluate efficiency and sensitivity of the model as a preclinical drug screening system. Materials and Methods. Firefly luciferase (fLuc)-gene introduced ALL (ALL/fLuc) cell line and patient-originated ALL cells were transplanted into a tibia of NOD/SCID mouse. We conducted a comparative analysis of intra-bone marrow (IBMT) transplanted leukemia model with IP and IV transplantation of leukemic cells. Results. IBMT of ALL/fLuc cells effectively established a bioluminescent leukemia NOD/SCID mouse model. Upon comparison of IBMT model with IP and IV transplantation models, infusing identical number of ALL/fLuc cells into NOD/SCID mice resulted in IBMT model with evaluable bioluminescent signal, but not in IP and IV models. In IBMT model, bioluminescent signals of ALL/fLuc cells emitted from peripheral blood, tibia and infiltrated organs indicated that leukemia model was established. The changes in these signals' strength reflected dose-dependent cytotoxic effects of vincristine, which allowed leukemia model with evaluable bioluminescent signal to be utilized as a preclinical drug screening system. IBMT leukemia model was also established using primary ALL cells that can provide additional insights for the development of leukemia therapeutics. Conclusion. IBMT of ALL/fLuc cells enables development of leukemia mouse model with the greater bioluminescent sensitivity than IP and IV in NOD/SCID to evaluate candidate for development of anti-cancer drug. Disclosures: No relevant conflicts of interest to declare.

2020 ◽  
Vol 74 (3) ◽  
pp. 187-196
Author(s):  
Jasmina Stojkovska ◽  
Jovana Zvicer ◽  
Milena Milivojevic ◽  
Isidora Petrovic ◽  
Milena Stevanovic ◽  
...  

Development of drugs is a complex, time- and cost-consuming process due to the lack of standardized and reliable characterization techniques and models. Traditionally, drug screening is based on in vitro analysis using two-dimensional (2D) cell cultures followed by in vivo animal testing. Unfortunately, application of the obtained results to humans in about 90 % of cases fails. Therefore, it is important to develop and improve cell-based systems that can mimic the in vivo-like conditions to provide more reliable results. In this paper, we present development and validation of a novel, user-friendly perfusion bioreactor system for single use aimed for cancer research, drug screening, anti-cancer drug response studies, biomaterial characterization, and tissue engineering. Simple design of the perfusion bioreactor provides direct medium flow at physiological velocities (100?250 ?m s-1) through samples of different sizes and shapes. Biocompatibility of the bioreactor was confirmed in short term cultivation studies of cervical carcinoma SiHa cells immobilized in alginate microfibers under continuous medium flow. The results have shown preserved cell viability indicating that the perfusion bioreactor in conjunction with alginate hydrogels as cell carriers could be potentially used as a tool for controlled anti-cancer drug screening in a 3D environment.


PLoS ONE ◽  
2019 ◽  
Vol 14 (4) ◽  
pp. e0215080 ◽  
Author(s):  
Jang Ho Cho ◽  
Ju-Sun Kim ◽  
Seung Tae Kim ◽  
Jung Yong Hong ◽  
Joon Oh Park ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4518-4518
Author(s):  
Takuya Matsunaga ◽  
Kazunori Kato ◽  
Maki Tanaka ◽  
Yukari Masuta ◽  
Kageaki Kuribayashi ◽  
...  

Abstract Bone marrow (BM) minimal residual disease causes relapse after chemotherapy in AML. We have previously reported that VLA4-positive leukemic cells acquired resistance to drug-induced apoptosis through the PI-3K/AKT/Bcl-2 signaling pathway, which is activated by the interaction of VLA4 and fibronectin on BM stromal cells. This resistance was negated by mouse-anti-human VLA4Ab (mouse VLA4Ab). In human leukemia SCID mouse model, we demonstrated a 100% survival rate with combination of mouse VLA4 Ab and AraC, while with AraC alone, only slight prolongation of survival was attained. In clinical study, overall survival at 5 years was 90% for 10 VLA4− patients and 25% for 15 VLA4+ patients (Matsunaga T et al, Nature Med 2003, 9, 1158–1165). In the present study, to perform the translational research, we first examined the myelosuppressive effect of the combination of rat-anti-mouse VLA4 Ab and AraC in C57/BL6 mice, and found that CBC data were almost the same as those of the mice treated with AraC alone. We next produced humanized chimeric-anti-human VLA4 Ab (chimeric-VLA4Ab), and examined its efficacy in combination with anti-cancer drugs in vitro and in vivo (human leukemia SCID mouse model). Chimeric-VLA4Abs were produced as follows: (i) total RNA of mouse VLA4 Abs were extracted from two hybridomas (SG/17 and SG/73), (ii) cDNA were synthesized by reverse transcriptase, (iii) variable region gene of mouse VLA4 Abs were amplified by 5′RACE method, (iv) TA cloning of amplified gene was performed, (v) sequence of mouse VLA4Abs gene was determined, (v) cloned variable region gene of mouse VLA4 Abs and constant region gene of human IgG1 were inserted into expression vector, and the expression vector was transfected into 293T cells, (vi) supernatant of the 293T cells was collected and purified to obtain chimeric-VLA4 Abs. The effects of chimeric-VLA4 Abs thus obtained in in vitro and in vivo (human leukemia SCID mouse model) were comparable to those of mouse VLA4 Abs. To perform the clinical study, we are presently producing the GMP-graded chimeric-VLA4 Abs.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3691-3691
Author(s):  
Takuo Mizukami ◽  
Kazuya Takizawa ◽  
Jumpei Yamazaki ◽  
Wakako Kuribayashi ◽  
Madoka Kuramitsu ◽  
...  

Abstract Adult T cell leukemia (ATL) is a lymphoproliferative disorder caused by infection with HTLV-I. Although various chemotherapies have shown significant complete remission rates, most of the treated patients relapse. These data indicate the existence of leukemic stem cells (LSCs) and a specific niche that regulates stemness and protects LSCs from various chemotherapies. We have reported in previous studies that the ATL-LSCs isolated from a Tax-transgenic (Tax-Tg) mouse are enriched in the CD117+/CD38–/CD71– fraction of the lymphoma, and LSCs have the potential to reproduce the original tumor when transplanted into a NOD/SCID mouse (Yamazaki et al., Blood, 2009). However, the niche of ATL-LSCs is still unclear. To identify the ATL-LSC niche in vivo, we performed a homing assay. Splenic lymphoma cells isolated from a Tax-Tg mouse were GFP transduced by a lentivirus, and then sorted GFP+ cells were transplanted intra-peritoneally into a non-irradiated NOD/SCID mouse. The homing of GFP+ cells to tissues was assessed by flow cytometry (FCM) at 16 hours and 3, 7, 14 and 21 days after transplantation. As a result, GFP+ lymphoma cells were first detected in the spleen and BM at 16 hours after transplantation. No GFP+ lymphoma cells were detected in the thymus and LN. Interestingly, more than 60% of first colonized cells in the spleen and BM at 16 hours after transplantation were AT-LSCs (GFP+/CD117+ cells). From day 3 to 7, more than 40% of colonizing cells in the BM and spleen were ATL-LSCs. To identify the specific niche of ATL-LSCs in the BM, we performed imaging analysis of ATL-LSCs. ATL-LSCs (GFP+/CD117+ and CD38–/CD71–/CD117+ cells) were mainly localized near the endosteal region of trabecular bone in the BM. We found that ATL-LSCs were also attached to the reticular cells in the trabecular bone. In addition, we found the number of osteoclast was significantly increased at the trabecular region. Increasing number of osteoclasts correlates the increased the serum calcium concentration and decreased the mass of trabecular bone. FCM analysis and in vitro differentiation assay confirmed that the number of osteoclast precursors was increased in the ATL BM. To clarify the role of osteoclast in the ATL BM, we treated osteoclast inhibitor Zoledronic acid (ZOL) to the ATL mouse model. As a result, ZOL itself significantly reduced the number of GFP+ ATL cells in the BM. When we treated ZOL with anti cancer drug, GFP+ ATL cells were dramatically reduced in the BM and extend the mouse survival rate significantly despite anti cancer drug does not reduced the number of ATL cells itself. In addition, abnormal trabecular bone morphology was completely recovered in the treated mouse. These data suggest that osteoclast may have a function to support leukemic stem cell niche. To clarify the key signals to induce osteoclast in ATL BM, we checked the expression of RANKL and PTHrP. We found that RANKL was up-regulated both in the lymphoma cell and stromal cells in the bone marrow.   In this study, we found that ATL-LSC niche is located at the trabecular bone region in the BM and osteoclasts have a role to support ATL cell and develop LSCs niche in a mouse model of ATL. We conclude that osteoclast have a potential therapeutic target in the mouse model of ATL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2725-2725
Author(s):  
Salvia Jain ◽  
Luigi Scotto ◽  
Enrica Marchi ◽  
Matko Kalac ◽  
Jennifer Amengual ◽  
...  

Abstract Abstract 2725 Background: Sezary syndrome (SS) is an aggressive leukemic form of cutaneous T cell lymphoma (CTCL) and is generally considered incurable. Until now no true animal model for SS exists which could be used for the screening of novel compounds against the disease. We successfully developed a bioluminescent xenograft mouse model for SS to noninvasively monitor tumor cell engraftment and progression and to measure the effects of treatments on tumor burden. Methods: A fusion protein was constructed consisting of the monomeric mutant red fluorescent mCherry, and the synthetic-firefly Luciferase by cloning the mCherry gene into the plasmid vector pGL4.13 [luc2/SV40] (Promega) carrying the luciferase gene, thus obtaining the pGLCherryLuciferase plasmid, where the Cherry and the luciferase genes formed one open reading frame. Analysis of pGLCherryluciferase transfected human embryonic kidney 293 (HEK 293) cells via flow cytometry and luciferase activity confirmed that the cherryluciferase fusion protein retained its dual bioluminescent/fluorescent activity in vitro. The H9 cell line derived from a SS patient was transfected with this plasmid using the transfection reagent, Effectene. The transfected H9 cells were injected subcutaneously into the flank of 5–7 week old female severe combined immunodeficiency (SCID-Beige) and NOD/Shi-scid/IL-2Rγnull (NOG) mice (Taconic Laboratories, Germantown, NY). After confirmation of a bioluminescent signal, NOG mice were assigned to the vehicle control group or treatment groups, which received romidepsin (1.2 or 2.3mg/kg on day1, 4, 8 and 11, ip) or pralatrexate (30mg/kg on day1, 4, 8 and 11, ip). In vivo bioluminescence imaging over 3 weeks was performed using an IVIS imaging system (Caliper Life Sciences, Alameda, CA). Mice were anesthetized with 3% isoflurane and imaged for 1 minute following ip injection of 150 mg/kg of D-luciferin. For quantification of light intensities in time course experiments, an equal constant analysis gate was defined for all mice and individual tumor photon counts were determined using Living Image®software (Caliper Life Sciences, Alameda, CA). Results: The SCID/Beige mice did not permit any engraftment of the H9 injected cells whereas all the NOG mice exhibited tumor growth and progression. This suggests that the NOG mice are superior animal recipients for xenotransplantation of Sezary cells, potentially making them a preclinical tool to understand tumorigenesis and drugs effects in this rare malignancy. All mice bearing H9-mCherry-luc cells developed a bioluminescent signal at the site of inoculation within 7 days of injection of transfected cells. Sequential quantitative signals from bioluminescent imaging over the 3-week period were significantly lower in the mice treated with pralatexate compared with the control group (Figure 1). The mice treated with 1.2mg/kg of romidepsin had no effect on tumor suppression whereas the mice that received the higher dose of romidepsin demonstrated decrease of tumor growth suggesting dose-dependent tumor inhibition. Mice treated with pralatrexate demonstrated diminished bioluminescent signal as early as after the first dose compared to the mice treated with higher dose of romidepsin which produced a slower time to onset of activity. No mice treated with pralatrexate or with the higher dose of romidepsin exhibited toxicity. Conclusion: This novel bioluminescent xenograft mouse model of SS enables non-invasive, sensitive, quantitative evaluation of disease progression in living animals and evaluation of pharmacologic factors in real time. We are able to detect and monitor lymphoma cell growth before the presentation of clinical manifestations. Further this model recapitulates our understanding of behavior of drugs used in the treatment of lymphomas such as pralatrexate which has a rapid onset of action compared to romidepsin that has a delayed time to onset of activity. This represents the first bioluminescent animal model of human CTCL that is intended to be used to investigate novel treatment platforms in preclinical studies. This preclinical model also compliments the ongoing phase 2 trial of pralatrexate in relapsed or refractory CTCL. Further in vivo studies to evaluate synergy of promising new agents in this novel mouse model of SS have begun and will be reported. Disclosures: O'Connor: Celgene: Consultancy, Research Funding; Merck: Research Funding; Novartis: Research Funding; Spectrum: Research Funding.


Micromachines ◽  
2021 ◽  
Vol 12 (6) ◽  
pp. 624
Author(s):  
Qiang Liu ◽  
Tian Zhao ◽  
Xianning Wang ◽  
Zhongyao Chen ◽  
Yawei Hu ◽  
...  

Three-dimensional cultured patient-derived cancer organoids (PDOs) represent a powerful tool for anti-cancer drug development due to their similarity to the in vivo tumor tissues. However, the culture and manipulation of PDOs is more difficult than 2D cultured cell lines due to the presence of the culture matrix and the 3D feature of the organoids. In our other study, we established a method for lung cancer organoid (LCO)-based drug sensitivity tests on the superhydrophobic microwell array chip (SMAR-chip). Here, we describe a novel in situ cryopreservation technology on the SMAR-chip to preserve the viability of the organoids for future drug sensitivity tests. We compared two cryopreservation approaches (slow freezing and vitrification) and demonstrated that vitrification performed better at preserving the viability of LCOs. Next, we developed a simple procedure for in situ cryopreservation and thawing of the LCOs on the SMAR-chip. We proved that the on-chip cryopreserved organoids can be recovered successfully and, more importantly, showing similar responses to anti-cancer drugs as the unfrozen controls. This in situ vitrification technology eliminated the harvesting and centrifugation steps in conventional cryopreservation, making the whole freeze–thaw process easier to perform and the preserved LCOs ready to be used for the subsequent drug sensitivity test.


Author(s):  
Lauren Marshall ◽  
Isabel Löwstedt ◽  
Paul Gatenholm ◽  
Joel Berry

The objective of this study was to create 3D engineered tissue models to accelerate identification of safe and efficacious breast cancer drug therapies. It is expected that this platform will dramatically reduce the time and costs associated with development and regulatory approval of anti-cancer therapies, currently a multi-billion dollar endeavor [1]. Existing two-dimensional (2D) in vitro and in vivo animal studies required for identification of effective cancer therapies account for much of the high costs of anti-cancer medications and health insurance premiums borne by patients, many of whom cannot afford it. An emerging paradigm in pharmaceutical drug development is the use of three-dimensional (3D) cell/biomaterial models that will accurately screen novel therapeutic compounds, repurpose existing compounds and terminate ineffective ones. In particular, identification of effective chemotherapies for breast cancer are anticipated to occur more quickly in 3D in vitro models than 2D in vitro environments and in vivo animal models, neither of which accurately mimic natural human tumor environments [2]. Moreover, these 3D models can be multi-cellular and designed with extracellular matrix (ECM) function and mechanical properties similar to that of natural in vivo cancer environments [3].


2018 ◽  
Vol 25 (6) ◽  
pp. 677-686 ◽  
Author(s):  
Shani Avniel-Polak ◽  
Gil Leibowitz ◽  
Victoria Doviner ◽  
David J Gross ◽  
Simona Grozinsky-Glasberg

Patients with neuroendocrine neoplasms (NENs) often require systemic treatment, which is frequently limited by the emergence of drug resistance. mTOR inhibitors (mTORi), such as RAD001 (everolimus), have been shown to inhibit neoplasm progression. mTORi stimulates autophagy, a degradation pathway that might promote the survival of neoplasm cells that are exposed to anti-cancer therapy. Chloroquine (CQ), a well-known anti-malarial and anti-rheumatic drug, suppresses autophagy. Based on our previous results, we hypothesized that CQ may enhance the anti-tumorigenic effects of mTORi by inhibiting autophagy and we aimed to examine the anti-tumorigenic effect of CQ, alone or in combination with RAD001. We established a NEN subcutaneous xenograft mouse model and evaluated the effect of the drugs on tumor growth, mTOR pathway, autophagy and apoptosis. CQ alone and in combination with RAD001 significantly decreased neoplasm volume. Histopathological analysis revealed that the combination of CQ and RAD001 markedly inhibited mTOR activity and neoplasm cell growth, along with accumulation of autophagosomes and increased apoptosis. In conclusion, CQ enhances the anti-tumorigenic effect of RAD001 in vivo by inhibiting autophagy. Clinical trials addressing the effects of CQ therapy on neoplasm progression in patients with NENs, mainly in those treated with mTORi, are warranted.


Sign in / Sign up

Export Citation Format

Share Document