Home>>Natural Products>>DFO (9H-1,8-Diazafluoren-9-one)

DFO (9H-1,8-Diazafluoren-9-one) Sale

(Synonyms: 1,8-二氮杂-9-芴酮,DFO) 目录号 : GC30151

DFO (9H-1,8-Diazafluoren-9-one) 是一种荧光染料,可与氨基酸反应形成高荧光衍生物(激发 470 nm;发射 570 nm)。

DFO (9H-1,8-Diazafluoren-9-one) Chemical Structure

Cas No.:54078-29-4

规格 价格 库存 购买数量
50mg
¥536.00
现货
100mg
¥893.00
现货

电话:400-920-5774 Email: sales@glpbio.cn

Customer Reviews

Based on customer reviews.

Sample solution is provided at 25 µL, 10mM.

102

客户使用产品发表文献 2

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Fingermark detection[1]:

Preparation method( Collection of samples)

As porous surface for the fingerprints deposition, it was selected white office paper in sheets of A4 size with a paper weight of 75 g/m2. Two female and three male subjects were chosen to deposit the fingerprints. Each subject deposited by rotation the fingerprints of all fingers on four different sheets of paper, two for each hand, divided into 30 quadrants (5 columns 6 rows). On the first sheet, natural fingermarks were collected; on the second sheet charged fingerprints were deposited after running the fingers on the areas of maximum secretion of the face, such as the forehead, nose or chin. This procedure was repeated six times, waiting half an hour between two subsequent depositions, to ensure the regeneration of natural secretions on the hands of the donor. The columns of the sheet corresponded to the days in which the analyses were made and the rows corresponded to the different solutions with which the fingerprints were treated. Once the deposition was completed, the sheets were cut to separate the quadrants with only one fingerprint deposited. Each quadrant was then immerged in a different solution (S1, S2, S3 or SD ( DFO (9H-1,8-Diazafluoren-9-one) solution)) within a plastic tray until it was completely soaked, then air-dried for a few seconds and finally placed in an oven for 3 min at 170 ℃.

DFO solution preparation

the solution of DFO (9H-1,8-Diazafluoren-9-one) was prepared: DFO solution (SD): 0.0625 g of DFO, 7.5 mL of methanol, 5 mL of glacial acetic acid, 70 mL of HFE-71DE, 180 mL of HFE-7100.

Sample handling

Tried to simulate the initial condition under which a natural fingermark is deposited on the paper. As done during the qualitative analysis, the quadrants of paper were immersed in the IND-Zn and DFO (9H-1,8-Diazafluoren-9-one) solutions, air-dried for a few seconds and placed in an oven at 170 ℃ for 3 min. Once pulled out of the oven, all the paper samples had assumed a pink color, indicating that the reaction had occurred. Subsequently, the quadrants were further cut into squares all having the same size and each of them was placed in an Eppendorf tube containing the carrier solvent of the solution in which they were previously immersed. So, for the samples treated with S1 it was used as extraction solvent the petroleum ether, for those treated with S2, S3, and SD, it was used the HFE-7100. Then, the tubes were centrifuged for 2 min at 10,000 rpm to complete the extraction process and make the sample ready to be transferred in the fluorometer and analyzed.

Applications

DFO (9H-1,8-Diazafluoren-9-one) shows orange fluorescent fingerprints. DFO, which is up to date the enhancing reagent most used in the forensic laboratories of the RIS, confirmed its excellent qualities from the point of view both of the fluorescence intensity and of the definition of the fingerprints enhanced, showing performances sufficiently effective even after the 3 months of shelf-life expected.

References:

[1].Marriott C, Lee R, et,al. Evaluation of fingermark detection sequences on paper substrates. Forensic Sci Int. 2014 Mar;236:30-7. doi: 10.1016/j.forsciint.2013.12.028. Epub 2014 Jan 6. PMID: 24529772.

产品描述

DFO (9H-1,8-Diazafluoren-9-one) is a fluorescent dye that reacts with amino acids to form highly fluorescent derivatives (excitation wavelength ~470 nm;Emission wavelength ~570 nm) is used to detect potential fingerprints on paper, and has a very high sensitivity and is widely used in sweat fingerprint identification[1].

It reacts with the amino acids contained in the natural secretions giving a pale red/magenta product, which has also a strong luminescence at room temperature without need of further treatment (such as the addition of metal salts). In order to start the reaction, it is necessary to apply heat using an oven or a hot plate at temperatures that go from 100 to 180 ¡校nd for times not exceeding 20 min[1].DFO (9H-1,8-Diazafluoren-9-one) are used in the forensic field to enhance latent fingerprints deposited on porous surfaces due to the formation of fluorescent products by reacting with the amino acids present in the papillary exudate[2]. A DFO (9H-1,8-Diazafluoren-9-one) formulation in a mixture of HFE7100 and trans-1,2-dichloroethylene is an effective replacement for the CFC 113-based formulation[3].

References:
[1]. Pounds, C. Anthony et al. "The Use of 1,8-Diazafluoren-9-one (DFO) for the Fluorescent Detection of Latent Fingerprints on Paper. A Preliminary Evaluation." Journal of Forensic Sciences 35 (1990): 169-175.
[2]. D'Elia V, Materazzi S, et,al. Evaluation and comparison of 1,2-indanedione and 1,8-diazafluoren-9-one solutions for the enhancement of latent fingerprints on porous surfaces. Forensic Sci Int. 2015 Sep;254:205-14. doi: 10.1016/j.forsciint.2015.07.036. Epub 2015 Jul 30. PMID: 26254628.
[3]. Sarah Merrick; Sarah J. Gardner; et,al. Hewlett An operational trial of ozone-friendly DFO and 1,2-indandione formulations for latent fingerprint detection. Journal of Forensic Identification Volume: 52 Issue: 5 Dated: September/October 2002 Pages: 595-605

Chemical Properties

Cas No. 54078-29-4 SDF
别名 1,8-二氮杂-9-芴酮,DFO
Canonical SMILES O=C1C2=NC=CC=C2C3=CC=CN=C31
分子式 C11H6N2O 分子量 182.18
溶解度 DMSO : 20 mg/mL (109.78 mM);Water : < 0.1 mg/mL (insoluble) 储存条件 Store at -20°C
General tips 请根据产品在不同溶剂中的溶解度选择合适的溶剂配制储备液;一旦配成溶液,请分装保存,避免反复冻融造成的产品失效。
储备液的保存方式和期限:-80°C 储存时,请在 6 个月内使用,-20°C 储存时,请在 1 个月内使用。
为了提高溶解度,请将管子加热至37℃,然后在超声波浴中震荡一段时间。
Shipping Condition 评估样品解决方案:配备蓝冰进行发货。所有其他可用尺寸:配备RT,或根据请求配备蓝冰。

溶解性数据

制备储备液
1 mg 5 mg 10 mg
1 mM 5.4891 mL 27.4454 mL 54.8908 mL
5 mM 1.0978 mL 5.4891 mL 10.9782 mL
10 mM 0.5489 mL 2.7445 mL 5.4891 mL
  • 摩尔浓度计算器

  • 稀释计算器

  • 分子量计算器

质量
=
浓度
x
体积
x
分子量
 
 
 
*在配置溶液时,请务必参考产品标签上、MSDS / COA(可在Glpbio的产品页面获得)批次特异的分子量使用本工具。

计算

动物体内配方计算器 (澄清溶液)

第一步:请输入基本实验信息(考虑到实验过程中的损耗,建议多配一只动物的药量)
给药剂量 mg/kg 动物平均体重 g 每只动物给药体积 ul 动物数量
第二步:请输入动物体内配方组成(配方适用于不溶于水的药物;不同批次药物配方比例不同,请联系GLPBIO为您提供正确的澄清溶液配方)
% DMSO % % Tween 80 % saline
计算重置

Research Update

"

Development of [89Zr]DFO-elotuzumab for immunoPET imaging of CS1 in multiple myeloma

Eur J Nucl Med Mol Imaging.2021 May;48(5):1302-1311.PMID: 33179150DOI: 10.1007/s00259-020-05097-y

Purpose: Multiple myeloma (MM) is a bone marrow malignancy that remains mostly incurable. Elotuzumab is an FDA-approved therapeutic monoclonal antibody targeted to the cell surface glycoprotein CS1, which is overexpressed in MM cells. Identifying patients who will respond to CS1-targeted treatments such as elotuzumab requires the development of a companion diagnostic to assess the presence of CS1. Here, we evaluated [89Zr]DFO-elotuzumab as a novel PET tracer for imaging CS1 expression in preclinical MM models. Methods: Conjugation of desferrioxamine-p-benzyl-isothiocyanate (DFO-Bz-NCS) to elotuzumab enabled zirconium-89 radiolabeling. MM.1S-CG cells were intravenously injected in NOD SCID gamma (NSG) mice. Small animal PET imaging with [89Zr]DFO-elotuzumab (1.11 MBq/mouse, 7 days post-injection), [89Zr]DFO-IgG (1.11 MBq/mouse, 7 days post-injection), and [18F]FDG (7-8 MBq, 1 h post-injection) was performed. Additionally, biodistribution of [89Zr]DFO-elotuzumab post-imaging at 7 days was also done. In vivo specificity of [89Zr]DFO-elotuzumab was further evaluated with a blocking study and ex vivo autoradiography. Results: [89Zr]DFO-elotuzumab was produced with high specific activity (56 ± 0.75 MBq/nmol), radiochemical purity (99% ± 0.5), and yield (93.3% ± 1.5). Dissociation constant of 40.4 nM and receptor density of 126 fmol/mg was determined in MM.1S-CG cells. Compared to [89Zr]DFO-IgG, [89Zr]DFO-elotuzumab localized with a significantly higher standard uptake value in tumor-bearing bone tissue (8.59 versus 4.77). Blocking with unlabeled elotuzumab significantly reduced (P < 0.05) uptake of [89Zr]DFO-elotuzumab in the bones. Importantly, while [18F]FDG demonstrated similar uptake in the bone and muscle, [89Zr]DFO-elotuzumab showed > 3-fold enhanced uptake in bones. Conclusion: These data demonstrate the feasibility of [89Zr]DFO-elotuzumab as a companion diagnostic for CS1-targeted therapies.

Head-to-head comparison of DFO* and DFO chelators: selection of the best candidate for clinical 89Zr-immuno-PET

Eur J Nucl Med Mol Imaging.2021 Mar;48(3):694-707.PMID: 32889615DOI: 10.1007/s00259-020-05002-7

Abstract Purpose: Almost all radiolabellings of antibodies with 89Zr currently employ the hexadentate chelator desferrioxamine (DFO). However, DFO can lead to unwanted uptake of 89Zr in bones due to instability of the resulting metal complex. DFO*-NCS and the squaramide ester of DFO, DFOSq, are novel analogues that gave more stable 89Zr complexes than DFO in pilot experiments. Here, we directly compare these linker-chelator systems to identify optimal immuno-PET reagents. Methods: Cetuximab, trastuzumab and B12 (non-binding control antibody) were labelled with 89Zr via DFO*-NCS, DFOSq, DFO-NCS or DFO*Sq. Stability in vitro was compared at 37 °C in serum (7 days), in formulation solution (24 h ± chelator challenges) and in vivo with N87 and A431 tumour-bearing mice. Finally, to demonstrate the practical benefit of more stable complexation for the accurate detection of bone metastases, [89Zr]Zr-DFO*-NCS and [89Zr]Zr-DFO-NCS-labelled trastuzumab and B12 were evaluated in a bone metastasis mouse model where BT-474 breast cancer cells were injected intratibially. Results: [89Zr]Zr-DFO*-NCS-trastuzumab and [89Zr]Zr-DFO*Sq-trastuzumab showed excellent stability in vitro, superior to their [89Zr]Zr-DFO counterparts under all conditions. While tumour uptake was similar for all conjugates, bone uptake was lower for DFO* conjugates. Lower bone uptake for DFO* conjugates was confirmed using a second xenograft model: A431 combined with cetuximab. Finally, in the intratibial BT-474 bone metastasis model, the DFO* conjugates provided superior detection of tumour-specific signal over the DFO conjugates. Conclusion: DFO*-mAb conjugates provide lower bone uptake than their DFO analogues; thus, DFO* is a superior candidate for preclinical and clinical 89Zr-immuno-PET.

Light-Induced Radiosynthesis of 89Zr-DFO-Azepin-Onartuzumab for Imaging the Hepatocyte Growth Factor Receptor

J Nucl Med .2020 Jul;61(7):1072-1078. PMID: 31924725DOI: 10.2967/jnumed.119.237180

Methods that provide rapid access to radiolabeled antibodies are vital in the development of diagnostic and radiotherapeutic agents for PET or radioimmunotherapy. The human hepatocyte growth factor receptor (c-MET) signaling pathway is dysregulated in several malignancies, including gastric cancer, and is an important biomarker in drug discovery. Here, we used a photoradiochemical approach to produce 89Zr-radiolabeled onartuzumab (a monovalent, antihuman c-MET antibody), starting directly from the fully formulated drug (MetMAb). Methods: Simultaneous 89Zr-radiolabeling and protein conjugation was performed in one-pot reactions containing 89Zr-oxalate, the photoactive chelate desferrioxamine B (DFO)-aryl azide (DFO-ArN3), and MetMAb to give 89Zr-DFO-azepin-onartuzumab. As a control, 89Zr-DFO-benzyl Bn-isothiocyanate Bn-NCS-onartuzumab was prepared via a conventional two-step process using prepurified onartuzumab and DFO-Bn-NCS. Radiotracers were purified by using size-exclusion methods and evaluated by radiochromatography. Radiochemical stability was studied in human serum, and immunoreactivity was determined by cellular binding assays using MKN-45 gastric carcinoma cells. PET imaging at multiple time points (0-72 h) was performed on female athymic nude mice bearing subcutaneous MKN-45 xenografts. Biodistribution experiments were performed after the final image was obtained. The tumor specificity of 89Zr-DFO-azepin-onartuzumab was assessed in vivo by competitive inhibition (blocking) studies. Results: Initial photoradiosynthesis experiments produced 89Zr-DFO-azepin-onartuzumab in less than 15 min, with an isolated decay-corrected radiochemical yield (RCY) of 24.8%, a radiochemical purity of approximately 90%, and a molar activity of approximately 1.5 MBq nmol-1 Reaction optimization improved the radiochemical conversion of 89Zr-DFO-azepin-onartuzumab to 56.9% ± 4.1% (n = 3), with isolated RCYs of 41.2% ± 10.6% (n = 3) and radiochemical purity of more than 90%. Conventional methods produced 89Zr-DFO-Bn-NCS-onartuzumab with an isolated RCY of more than 97%, radiochemical purity of more than 97% and molar activity of approximately 14.0 MBq nmol-1 Both radiotracers were immunoreactive and stable in human serum. PET imaging and biodistribution studies showed high tumor uptake for both radiotracers. By 72 h, tumor and liver uptake (percentage injected dose [%ID]) reached 15.37 ± 5.21 %ID g-1 and 6.56 ± 4.03 %ID g-1, respectively, for 89Zr-DFO-azepin-onartuzumab (n = 4) and 21.38 ± 11.57 %ID g-1 and 18.84 ± 6.03 %ID g-1, respectively, for 89Zr-DFO-Bn-NCS-onartuzumab (n = 4). Blocking experiments gave a statistically significant reduction in tumor uptake (6.34 ± 0.47 %ID g-1) of 89Zr-DFO-azepin-onartuzumab (n = 4). Conclusion: The experiments demonstrated that photoradiosynthesis is a viable alternative approach for producing 89Zr-radiolabeled antibodies directly in protein formulation buffer, reducing protein aggregation and liver uptake.

Radiolabelling of the octadentate chelators DFO* and oxoDFO* with zirconium-89 and gallium-68

J Biol Inorg Chem .2020 Aug;25(5):789-796.PMID: 32661784DOI: 10.1007/s00775-020-01800-4

In recent years, clinical imaging with zirconium-89 (89Zr)-labelled monoclonal antibodies (Ab) by positron emission tomography (immunoPET) has been gaining significant importance in nuclear medicine for the diagnosis of different types of cancer. For complexation of the radiometal 89Zr and its attachment to the Ab, chelating agents are required. To date, only the hexadentate chelator desferrioxamine (DFO) is applied in the clinic for this purpose. However, there is increasing preclinical evidence that the [89Zr]Zr-DFO complex is not sufficiently stable and partly releases the radiometal in vivo due to the incomplete coordination sphere of the metal. This leads to unfavourable unspecific uptake of the osteophilic radiometal in bones, hence decreasing the signal-to-noise-ratio and leading to an increased dose to the patient. In the past, several new chelators with denticities > 6 have been published, notably the octadentate DFO derivative DFO*. DFO*, however, shows limited water solubility, wherefore an oxygen containing analogue, termed oxoDFO*, was developed in 2017. However, no data on the suitability of oxoDFO* for radiolabelling with 89Zr has yet been reported. In this proof-of-concept study, we present the first radiolabelling results of the octadentate, water-soluble chelator oxoDFO*, as well as the in vitro stability of the resulting complex [89Zr]Zr-oxoDFO* in comparison to the analogous octadentate, but less water-soluble derivative DFO* and the current "standard" chelator DFO. In addition, the suitability of DFO* and oxoDFO* for radiolabeling with the short-lived PET metal gallium-68 is discussed. The water-soluble, octadentate chelator oxoDFO* provides stable complexes with the positron emitter Zirconium-89. The radiolabelling can be performed at room temperature and neutral pH and thus, oxoDFO* represents a promising chelator for applications in immunoPET.

Electrospun artificial periosteum loaded with DFO contributes to osteogenesis via the TGF-β1/Smad2 pathway

Biomater Sci.2021 Mar 21;9(6):2090-2102. PMID: 33475652DOI: 10.1039/d0bm01304h

Deferoxamine (DFO), an iron chelator regarded as a hypoxic analogue, has been reported to be involved in angiogenesis and osteogenic differentiation. In this study, DFO was loaded into nanospheres, Then, DFO-loaded NPs and free DFO were co-encapsulated in nanofibers through coaxial electrospinning and its effects on cell viability, migration, and osteogenic differentiation, and the potential mechanisms were investigated. The results suggested that DFO maintained cell viability and promoted the migration of human mesenchymal stem cells (hMSCs) and MC3T3-E1 cells. ALP activity, calcium deposition, and expression of osteogenesis-related markers, including collagen, osteocalcin, and osteopontin, were all increased with DFO. Moreover, hypoxia-inducible factor-1α, transforming growth factor-β, and Smad2 were upregulated with DFO, which indicated activation of the TGF-β1/Smad2 signalling pathway. This may contribute to osteogenic differentiation of cells.

"