SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "WFRF:(Yin Wen 1993 ) "

Sökning: WFRF:(Yin Wen 1993 )

  • Resultat 1-9 av 9
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Pinto, Soraia, et al. (författare)
  • Nanoparticles targeting the intestinal Fc receptor enhance intestinal cellular trafficking of semaglutide
  • 2024
  • Ingår i: Journal of Controlled Release. - : Elsevier BV. - 0168-3659 .- 1873-4995. ; 366, s. 621-636
  • Tidskriftsartikel (refereegranskat)abstract
    • Semaglutide is the first oral glucagon-like peptide-1 (GLP-1) analog commercially available for the treatment of type 2 diabetes. In this work, semaglutide was incorporated into poly(lactic-co-glycolic acid)-poly(ethylene glycol) (PLGA-PEG) nanoparticles (NPs) to improve its delivery across the intestinal barrier. The nanocarriers were surface-decorated with either a peptide or an affibody that target the human neonatal Fc receptor (hFcRn), located on the luminal cell surface of the enterocytes. Both ligands were successfully conjugated with the PLGA-PEG via maleimide-thiol chemistry and thereafter, the functionalized polymers were used to produce semaglutide-loaded NPs. Monodisperse NPs with an average size of 170 nm, neutral surface charge and 3% of semaglutide loading were obtained. Both FcRn-targeted NPs exhibited improved interaction and association with Caco-2 cells (cells that endogenously express the hFcRn), compared to non-targeted NPs. Additionally, the uptake of FcRn-targeted NPs was also observed to occur in human intestinal organoids (HIOs) expressing hFcRn through microinjection into the lumen of HIOs, resulting in potential increase of semaglutide permeability for both ligand-functionalized nanocarriers. Herein, our study demonstrates valuable data and insights that the FcRn-targeted NPs has the capacity to promote intestinal absorption of therapeutic peptides.
  •  
2.
  •  
3.
  • Rinne, Sara S., et al. (författare)
  • Targeting Tumor Cells Overexpressing the Human Epidermal Growth Factor Receptor 3 with Potent Drug Conjugates Based on Affibody Molecules
  • 2022
  • Ingår i: Biomedicines. - : MDPI AG. - 2227-9059. ; 10:6
  • Tidskriftsartikel (refereegranskat)abstract
    • Increasing evidence suggests that therapy targeting the human epidermal growth factor receptor 3 (HER3) could be a viable route for targeted cancer therapy. Here, we studied a novel drug conjugate, ZHER3-ABD-mcDM1, consisting of a HER3-targeting affibody molecule, coupled to the cytotoxic tubulin polymerization inhibitor DM1, and an albumin-binding domain for in vivo half-life extension. ZHER3-ABD-mcDM1 showed a strong affinity to the extracellular domain of HER3 (K-D 6 nM), and an even stronger affinity (KD 0.2 nM) to the HER3-overexpressing pancreatic carcinoma cell line, BxPC-3. The drug conjugate showed a potent cytotoxic effect on BxPC-3 cells with an IC50 value of 7 nM. Evaluation of a radiolabeled version, [99mTc]Tc-ZHER3-ABD-mcDM1, showed a relatively high rate of internalization, with a 27% internalized fraction after 8 h. Further in vivo evaluation showed that it could target BxPC-3 (pancreatic carcinoma) and DU145 (prostate carcinoma) xenografts in mice, with an uptake peaking at 6.3 +/- 0.4% IA/g at 6 h post-injection for the BxPC-3 xenografts. The general biodistribution showed uptake in the liver, lung, salivary gland, stomach, and small intestine, organs known to express murine ErbB3 naturally. The results from the study show that ZHER3-ABD-mcDM1 is a highly potent and selective drug conjugate with the ability to specifically target HER3 overexpressing cells. Further pre-clinical and clinical development is discussed.
  •  
4.
  • Rinne, Sara, et al. (författare)
  • Targeting tumor cells overexpressing the human epidermal growth factor receptor 3 with potent drug conjugates based on affibody molecules
  • Annan publikation (övrigt vetenskapligt/konstnärligt)abstract
    • Increasing evidence suggests that therapy targeting the human epidermal growth factor receptor 3 (HER3) could be a viable route for targeted cancer therapy. Here, we have studied a novel drug conjugate, ZHER3-ABD-mcDM1, consisting of a HER3-targeting affibody molecule, coupled to the cytotoxic tubulin polymerization inhibitor DM1, and an albumin- binding domain for in vivo half-life extension. ZHER3-ABD-mcDM1 showed strong affinity to the extracellular domain of HER3 (KD 6 nM), and an even stronger affinity (KD 0.2 nM) to the HER3-overexpressing pancreatic cancer cell line, BxPC3. The drug conjugate showed a potent cytotoxic effect on BxPC3 cells with an IC50 value of 7 nM. Further in vitro evaluation of a radiolabeled version [99mTc]Tc-ZHER3-ABD-mcDM1, showed that it had a relatively high rate of internalization into HER3-expressing BxPC-3 as well as DU145 (prostate cancer) cells, with a 27% internalized fraction after 8 h. Further in vivo evaluation showed that it could specifically target BxPC-3 and DU145-derived xenografts in mice, with an uptake peaking at 6.3 ± 0.4 %ID/g at 6 h post-injection for the BxPC-3-derived xenografts. The general biodistribution showed uptake in liver, lung, salivary gland, stomach, and small intestine, organs known to express HER3 naturally. The results from the study show that ZHER3-ABD- mcDM1 is a highly potent and specific drug conjugate, that may be further developed towards HER3-targeted cancer therapy.
  •  
5.
  •  
6.
  • Yin, Wen, 1993-, et al. (författare)
  • A comparison of affibody conjugates loaded with auristatin and maytansine derived drugs
  • Annan publikation (övrigt vetenskapligt/konstnärligt)abstract
    • Auristatin and maytansine-derived drugs are cytotoxic tubulin polymerization inhibitors commonly used as payloads in drug conjugates intended for targeted cancer therapy. We have previously shown that an affibody molecule ZHER2, binding to the human epidermal growth factor receptor 2 (HER2), can be site-specifically conjugated to DM1, a maytansine- derived payload, creating the potent and specific drug conjugate, ZHER2-ABD-mcDM1, where the ABD is an albumin binding domain used for in vivo half-life extension. Here, we investigated the properties of the HER2-binding affibody molecule conjugated with the two auristatin-derived payloads, monomethyl auristatin E and F (MMAE and MMAF), in comparison with the construct with DM1. We found that the drug conjugate ZHER2-ABD- mcMMAF was more potent than ZHER2-ABD-mcDM1, with IC50 values to high-HER2 expressing cell lines ranging from 0.18 to 12 nM. By contrast the IC50 values of ZHER2-ABD- mcMMAE was considerably weaker and this construct would probably benefit from a different linker connecting the drug to the affibody fusion protein. Quantification of uptake in HER2-expressing tumors and normal organs of 99m-technetium labeled drug conjugates showed that they were predominantly cleared by the kidneys, with relatively high tumor uptake, peaking at 11.1 ± 4.1 %ID/g for ZHER2-ABD-mcMMAE at 24 h post-injection, 8.5 ± 1.5 %ID/g for ZHER2-ABD-mcMMAF at 48 h post-injection, and 7.1 ± 1.8 %ID/g for ZHER2- ABD-mcDM1 at 48 h post-injection. Most normal organs, except for the kidneys, had a relatively low uptake. In conclusion, ZHER2-ABD-mcMMAF was the best performing drug conjugate with the highest potency, and lowest uptake in liver; slightly outperforming ZHER2- ABD-mcDM1.
  •  
7.
  • Yin, Wen, 1993-, et al. (författare)
  • Comparison of HER2-targeted affibody conjugates loaded with auristatin-and maytansine-derived drugs
  • 2023
  • Ingår i: Journal of Controlled Release. - : Elsevier. - 0168-3659 .- 1873-4995. ; 355, s. 515-527
  • Tidskriftsartikel (refereegranskat)abstract
    • Treatment with antibody drug conjugates targeting receptors over-expressed on cancer cells is well established for clinical use in several types of cancer, however, resistance often occurs motivating the development of novel drugs. We have recently investigated a drug conjugate consisting of an affibody molecule targeting the human epidermal growth factor receptor 2 (HER2), fused to an albumin-binding domain (ABD) for half-life extension, loaded with the cytotoxic maytansine derivative DM1. In this study, we investigated the impact of the cytotoxic payload on binding properties, cytotoxicity and biodistribution by comparing DM1 with the auristatins MMAE and MMAF, as part of the drug conjugate. All constructs had specific and high affinity binding to HER2, human and mouse albumins with values in the low- to sub-nM range. ZHER2-ABD-mcMMAF demonstrated the most potent cytotoxic effect on several HER2-over-expressing cell lines. In an experimental therapy study, the MMAFbased conjugate provided complete tumor regression in 50% of BALB/c nu/nu mice bearing HER2-overexpressing SKOV3 tumors at a 2.9 mg/kg dose, while the same dose of ZHER2-ABD-mcDM1 provided only a moderate anti-tumor effect. A comparison with the non-targeting ZTaq-ABD-mcMMAF control demonstrated HER2-targeting specificity. In conclusion, a combination of potent cytotoxicity in vitro, with minimal uptake in normal organs in vivo, and efficient delivery to tumors provided a superior anti-tumor effect of ZHER2-ABDmcMMAF, while maintaining a favorable toxicity profile with no observed adverse effects.
  •  
8.
  • Yin, Wen, 1993- (författare)
  • Engineering of alternative scaffold derived drug conjugates and fusion-toxins for targeted cancer therapy
  • 2022
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • Cancer is a major health problem, with premature death for many patients, and with a high monetary cost to society. The disease is complex to treat due to the many different types, but also because the disease-causing cells, that needs to be removed, are very similar to the normal cells. Traditional chemotherapy and radiotherapy therefore have a narrow therapeutic window. Targeted cancer therapy aims to precisely deliver a drug to the tumor tissue by recognizing biomarkers that are only, or at least mostly, associated with the cancer cells. Drug conjugates and fusion-toxins are two types of targeted drugs. They both consist of a cancer-targeting protein, a linker and a cytotoxic payload. The efficacy of drug conjugates and fusion toxins are determined by their targeting accuracy and cytotoxic potency. Optimization of drug conjugates and fusion-toxins concerns selecting the best combination of the three components for the best efficacy. In my studies I have worked with targeting the human epidermal growth factor receptor 2 and 3, HER2 and HER3, respectively. I have used affibody molecules: ZHER2 and ZHER3, and an albumin binding domain?derived affinity protein targeting HER2, ADAPT6. For in vivo half-life extension, I have used an albumin binding domain, ABD. As payloads, I have used the cytotoxic drugs DM1, MMAE, and MMAF, as well as a truncated version of Pseudomonas exotoxin A, PE25. In paper I, the goal was to investigate the architecture of the tumor targeting part of affibody-derived drug conjugates. Seven different affibody constructs with different number and position of the affibody and ABD-domains were designed and evaluated, in vitro and in vivo. They differed in their effect on cell proliferation, where particularly the constructs with two affibody domains increased the rate of proliferation for the SKOV-3 cell line with high expression of HER2. In vivo, the constructs with one affibody domain had a longer blood retention and lower hepatic uptake compared to the constructs with two affibody domains. Two constructs were selected for characterization as drug conjugates, one that promoted cell proliferation strongly, and one that had only a minor effect on cell proliferation. The conjugates were investigated as drugs for treatment of mice carrying HER2- overexpressing SKOV-3 tumors. The results showed that the affibody drug conjugate ZHER2-ABD-mcDM1 was the most efficient drug. ZHER2-ABD- mcDM1 suppressed tumor growth and extended the median survival time of the mice from 37 to 63 days. In paper II, the goal was to investigate the properties of affibody or ADAPT- based fusion-toxins, targeting the HER2 receptor. The results showed that utilizing ZHER2 as targeting domain, to carry the toxic peptide PE25 to HER2 overexpressing cells, was better than using a dual-HER2 binder, consisting of ZHER2 and ADAPT6. Furthermore, the results showed that PE25?based fusion toxins with high affinity to HER2 do not necessarily increase the cytotoxic effect beyond a certain point in affinity for the receptor. In paper III, the first HER3-targeting affibody drug conjugate was produced and studied. It was designed using the optimal format from paper I, i.e. affibody-ABD-mcDM1. The results showed a drug conjugate with a potent cytotoxic effect on the pancreatic cancer cell line, BxPC3, with an IC50 value of 6 nM. In mice, injection of a radiolabeled version of ZHER3-ABD-mcDM1, showed uptake in implanted BxPC3 tumors peaking at 6.3 ± 0.4 %ID/g at 6 h post-injection. The general biodistribution showed uptake in liver, lung, salivary gland, stomach, and small intestine, organs known to express HER3 naturally. Collectively, the results show that ZHER3-ABD-mcDM1 is a highly potent and specific drug conjugate, that motivates further development, and possibly investigation of its functionality in experimental therapy of HER3 overexpressing tumors in mice, and later humans. In paper IV, the goal was to investigate different cytotoxic drugs as part of an affibody-based drug conjugate targeting HER2. The optimal architecture from paper I, was employed also here, ZHER2-ABD-drug. The drugs tested were the tubulin polymerization inhibitors DM1, MMAE, and MMAF. ZHER2-ABD-mcMMAF had the most potent cytotoxic effect, with an IC50 value of 0.18 nM for SKBR3 cells with high HER2 expression. ZHER2-ABD- mcMMAE did not perform well in the cytotoxicity experiment. A better linker connecting the drug to the protein is probably required. ZHER2-ABD- mcMMAF was the best performing drug conjugate with the highest potency, and lowest uptake in liver; slightly outperforming ZHER2-ABD-mcDM1. In conclusion, the four papers cover generation and optimization of drug conjugates and fusion-toxin based on affibody molecules and ADAPTs. In the papers, new variants with desirable properties were identified and characterized. 
  •  
9.
  • Yin, Wen, 1993-, et al. (författare)
  • The Influence of Domain Permutations of an Albumin-Binding Domain-Fused HER2-Targeting Affibody-Based Drug Conjugate on Tumor Cell Proliferation and Therapy Efficacy
  • 2021
  • Ingår i: Pharmaceutics. - : MDPI AG. - 1999-4923. ; 13:11, s. 1974-1974
  • Tidskriftsartikel (refereegranskat)abstract
    • Human epidermal growth factor receptor 2 (HER2) is a clinically validated target for breast cancer therapy. Previously, a drug-fused HER2-targeting affinity protein construct successfully extended the survival of mice bearing HER2-expressing xenografts. The aim of this study was to evaluate the influence of the number and positioning of the protein domains in the drug conjugate. Seven HER2-targeting affibody-based constructs, including one or two affibody molecules (Z) with or without an albumin-binding domain (ABD), namely Z, Z-ABD, ABD-Z, Z-Z, Z-Z-ABD, Z-ABD-Z, and ABD-Z-Z, were evaluated on their effects on cell growth, in vivo targeting, and biodistribution. The biodistribution study demonstrated that the monomeric constructs had longer blood retention and lower hepatic uptake than the dimeric ones. A dimeric construct, specifically ABD-Z-Z, could stimulate the proliferation of HER2 expressing SKOV-3 cells in vitro and the growth of tumors in vivo, whereas the monomeric construct Z-ABD could not. These two constructs demonstrated a therapeutic effect when coupled to mcDM1; however, the effect was more pronounced for the non-stimulating Z-ABD. The median survival of the mice treated with Z-ABD-mcDM1 was 63 days compared to the 37 days for those treated with ABD-Z-Z-mcDM1 or for the control animals. Domain permutation of an ABD-fused HER2-targeting affibody-based drug conjugate significantly influences tumor cell proliferation and therapy efficacy. The monomeric conjugate Z-ABD is the most promising format for targeted delivery of the cytotoxic drug DM1.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-9 av 9

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy