Clustered nanobody–drug conjugates for targeted cancer therapy

2020 ◽  
Vol 56 (65) ◽  
pp. 9344-9347
Author(s):  
Tiantian Wu ◽  
Manman Liu ◽  
Hai Huang ◽  
Yaping Sheng ◽  
Haihua Xiao ◽  
...  

A novel clustered nanobody–drug conjugate is constructed by a site-specific ligation of a nanobody to dendrimeric-lysine, which enhances drug efficacy and reduces side-effects.

2018 ◽  
Vol 16 (1) ◽  
pp. 165-172 ◽  
Author(s):  
Hyungjun Kim ◽  
Dobeen Hwang ◽  
Minsuk Choi ◽  
Soyoung Lee ◽  
Sukmo Kang ◽  
...  

RSC Advances ◽  
2015 ◽  
Vol 5 (66) ◽  
pp. 53846-53856 ◽  
Author(s):  
Shihou Sheng ◽  
Tao Zhang ◽  
Shijie Li ◽  
Jun Wei ◽  
Guangjun Xu ◽  
...  

A traditional Chinese medicine cantharidin which was previously found to be effective on colorectal cancer cells was translated into nanoparticles for drug delivery to reduce its side effects and enhance its drug efficacy.


Nanoscale ◽  
2021 ◽  
Author(s):  
Cheng Lv ◽  
Jian Ao ◽  
Ji Wang ◽  
Man Tang ◽  
An-An Liu ◽  
...  

Targeted cancer therapy has aroused broad interests of researchers due to its accuracy in specific tumor targeting and few side effects on normal cells. In the last decades, oncolytic viral...


2020 ◽  
Author(s):  
Camille M. Le Gall ◽  
Johan M.S. van der Schoot ◽  
Iván Ramos-Tomillero ◽  
Melek Parlak Khalily ◽  
Floris J. van Dalen ◽  
...  

I.AbstractFunctionalized antibodies and antibody fragments have found applications in the fields of biomedical imaging, theragnostics, and antibody-drug conjugates (ADC). Antibody functionalization is classically achieved by coupling payloads onto lysine or cysteine residues. However, such stochastic strategies typically lead to heterogenous products, bearing a varying number of payloads. This affects bioconjugate efficacy and stability, as well as its in vivo biodistribution, and therapeutic index, while potentially obstructing the binding sites and leading to off-target toxicity. In addition, therapeutic and theragnostic approaches benefit from the possibility to deliver more than one type of cargo to target cells, further challenging stochastic labelling strategies. Thus, bioconjugation methods to reproducibly obtain defined homogenous conjugates bearing multiple different cargo molecules, without compromising target affinity, are in demand. Here, we describe a straightforward CRISPR/Cas9-based strategy to rapidly engineer hybridoma cells to secrete Fab’ fragments bearing two distinct site-specific labelling motifs, which can be separately modified by two different sortase A mutants. We show that sequential genetic editing of the heavy chain (HC) and light chain (LC) loci enables the generation of a stable cell line that secretes a dual tagged Fab’ molecule (DTFab’), which can be easily isolated in high yields. To demonstrate feasibility, we functionalized the DTFab’ with two distinct cargos in a site-specific manner. This technology platform will be valuable in the development of multimodal imaging agents, theragnostics, and next-generation ADCs.


Author(s):  
Satoka Mori ◽  
Arisa Abe ◽  
Naoto Ishikawa ◽  
Abdur Rafique ◽  
Yuji Ito

Abstract Recently, there has been an increasing interest in site-specific modifications of antibodies used in immunoassays for disease diagnosis and as antibody therapeutics, such as antibody−drug conjugates. Previously, we established a site-specific chemical conjugation system using an IgG-Fc binding chemical conjugation affinity peptide (CCAP). CCAP could be used only for the modification of human IgG owing to the lack of affinity of CCAP to rodent IgG molecules. In this study, novel CCAP reagents are proposed, which can be used for both human and mouse IgG, based on the Staphylococcus aureus protein A domain-derived affinity peptides Z34C and Z33. Compared with the activity of a conventional randomly modified antibody, mouse IgG modified using this method had favourable features in two immunoassays, demonstrating the advantages of the proposed CCAP method in preserving antibody functionality during conjugation.


2018 ◽  
Vol 10 (3) ◽  
pp. 309-319 ◽  
Author(s):  
Zhijiang Xi ◽  
Bing Zheng

Aptamer is a new-type of specific recognition molecule and is emerging as a promising therapeutic agent. The greatest advantage of aptamers is their high binding affinity and specificity towards the target. The toxicity of anticancer drugs to surrounding healthy tissues limits their clinical applications. However, nanomaterials for drug carriers can considerably improve drug efficacy while reducing toxicity, because they accumulate and release drugs at the lesion without affecting healthy tissues. As drug carriers, aptamer-conjugated nanomaterials can enhance active targeting and then release the drug into the targeted cancer cells effectively. Therefore, aptamer-conjugated nanomaterials for targeted cancer therapy constitute an evolving treatment approach with considerable potential to enhance the efficacy of cancer therapy via the delivery of therapeutic agents specific to and into the targeted tumor cells. In this paper, various aptamer-conjugated nanomaterials such as gold nanoparticles, magnetic nanoparticles, silica nanoparticles, carbon nanotubes, poly D,L-lactic-co-glycolic acid, and quantum dots for targeted cancer therapy, are reviewed and their prospect as therapeutic agents is evaluated.


mAbs ◽  
2013 ◽  
Vol 6 (1) ◽  
pp. 34-45 ◽  
Author(s):  
Siler Panowski ◽  
Sunil Bhakta ◽  
Helga Raab ◽  
Paul Polakis ◽  
Jagath R Junutula

Sign in / Sign up

Export Citation Format

Share Document