scholarly journals A novel therapeutic strategy for skeletal disorders: Proof of concept of gene therapy for X-linked hypophosphatemia

2021 ◽  
Vol 7 (44) ◽  
Author(s):  
Volha V. Zhukouskaya ◽  
Louisa Jauze ◽  
Séverine Charles ◽  
Christian Leborgne ◽  
Stéphane Hilliquin ◽  
...  
2020 ◽  
Vol 318 (5) ◽  
pp. H1296-H1307 ◽  
Author(s):  
Carlos J. Munoz ◽  
Ivan S. Pires ◽  
Jin Hyen Baek ◽  
Paul W. Buehler ◽  
Andre F. Palmer ◽  
...  

This study highlights the apoHb-Hp complex as a novel therapeutic strategy to attenuate the adverse systemic and microvascular responses to intravascular Hb and heme exposure. In vitro and in vivo Hb exchange and heme transfer experiments demonstrated proof-of-concept Hb/heme ligand transfer to apoHb-Hp. The apoHb-Hp complex reverses Hb- and heme-induced systemic hypertension and microvascular vasoconstriction, preserves microvascular blood flow, and functional capillary density. In summary, the unique properties of the apoHb-Hp complex prevent adverse systemic and microvascular responses to Hb and heme-albumin exposure and introduce a novel therapeutic approach to facilitate simultaneous removal of extracellular Hb and heme.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1701-1701 ◽  
Author(s):  
Mattias Hofmans ◽  
Barbara Depreter ◽  
Tim Lammens ◽  
Helene Cave ◽  
Christian Flotho ◽  
...  

BACKGROUND Juvenile myelomonocytic leukemia (JMML) is a rare and aggressive myelodysplastic/myeloproliferative disorder of early childhood. Hematopoietic stem cell transplantation results in long-term overall survival of only 50-60% and is fraught with frequent relapse and toxicity. Consequently, there is need to develop novel treatments. Evidence is growing that non-coding RNAs can serve as alternative therapeutic targets. Long non-coding RNAs (lncRNAs) are a recently discovered class of RNAs, with a minimum length of 200 nucleotides, of which perturbation can impact the cancer phenotype in vivo. Recently, we documented for the first time the lncRNA landscape in 44 JMML patients using microarrays and have associated lncRNA expression with clinical and molecular characteristics. Also, we demonstrated that JMML patients exhibit a distinct lncRNA expression profile compared to healthy controls and we identified 15 lncRNAs overexpressed in JMML patients. AIM The aim of this study is to identify overexpressed lncRNAs in JMML and to provide proof-of-concept for lncRNA perturbation as novel therapeutic strategy in this disease. METHODS To further identify overexpressed lncRNAs in JMML, total RNA from isolated mononuclear cell preparations from 19 previously untreated JMML patients (median age: 2.02 years) and 3 normal pediatric bone marrow (NPBM) samples from healthy controls (siblings screened for transplantation) was sequenced (HiSeq, Illumina). Differential gene expression (adjusted P-value < 0.05 and FDR < 0.10) was performed with DESEQ2 and EdgeR (R Bioconductor). Functional analysis of differentially expressed lncRNAs was performed through a lncRNA-mRNA interaction network (lncPath, R Bioconductor). A subset of overexpressed lncRNAs, based on fold change ≥2 and low to absent expression in NPBM, were subsequently validated by quantitative reverse-transcriptase PCR (qPCR). Antisense LNA™ GapmeRs (Qiagen), potent antisense oligonucleotides used for highly efficient inhibition of mRNA and lncRNA function, were designed for a subset of significantly overexpressed lncRNAs. As no JMML cell lines are available, cell lines from other pediatric and adult haematopoietic malignancies were selected based on publically available RNA-seq data and lncRNA expression was verified on qPCR. The effect of GapmeR treatment on lncRNA expression and cell viability was tested with qPCR and flow cytometry viability assays (7-AAD and annexin V staining). RESULTS Total RNA sequencing additionally revealed presence of 122 upregulated and 47 downregulated lncRNAs, most of which not present on the microarray. JMML patients clustered together and divergent from healthy controls on principal component analysis using only lncRNAs. Different pathways associated with RAS-MAPK signaling, cancer development, DNA metabolism and cell cycle were identified as being synergistically regulated by these differentially expressed lncRNAs through a lncRNA-mRNA interaction network. Forty-two lncRNAs (55 different transcripts) showing overexpression in JMML were validated with qPCR and in 13 transcripts from 11 lncRNAs a significantly higher expression could be observed in JMML patients compared to NPBM (Figure 1A), thus providing potential therapeutic targets. A minimum of four different antisense LNA GapmeRs were subsequently designed for each of these validated lncRNAs, overexpressed in JMML. For each lncRNA, one or more hematopoietic cell lines, showing expression of that lncRNA, could be identified and validated with qPCR. In vitro incubation of these cell lines with GapmeRs against lncRNA with expression in these cell lines, revealed concentration-dependent molecular knockdown in ≥ 2 GapmeRs for 4/5 lncRNAs (Figure 1B). For two targets, lnc-THADA4-1 and lnc-ACOT9, additional cell viability assays were performed and an effect on cell viability could be observed in GapmeR treated wells (Figure 1C). CONCLUSION Using microarray and total RNA sequencing, we documented the lncRNA landscape in JMML, and identified deregulated lncRNAs associated with key processes in JMML pathogenesis. We further provided proof-of-concept that knockdown of these lncRNAs using LNA GapmeRs can be a feasible therapeutic strategy in vitro in hematopoietic cell lines. Subsequently, safety and efficacy of this novel therapeutic strategy needs to be validated in JMML xenograft models in vivo. Figure 1 Disclosures Niemeyer: Celgene: Consultancy.


2001 ◽  
Vol 120 (5) ◽  
pp. A685-A685
Author(s):  
B SINGH ◽  
V MALMSTROM ◽  
F POWRIE

2020 ◽  
Vol 26 ◽  
Author(s):  
Maryam Dashtiahangar ◽  
Leila Rahbarnia ◽  
Safar Farajnia ◽  
Arash Salmaninejad ◽  
Arezoo Gowhari Shabgah ◽  
...  

: The development of recombinant immunotoxins (RITs) as a novel therapeutic strategy has made a revolution in the treatment of cancer. RITs are resulting from the fusion of antibodies to toxin proteins for targeting and eliminating cancerous cells by inhibiting protein synthesis. Despite indisputable outcomes of RITs regarding inhibiting multiple cancer types, high immunogenicity has been known as the main obstacle in the clinical use of RITs. Various strategies have been proposed to overcome these limitations, including immunosuppressive therapy, humanization of the antibody fragment moiety, generation of immunotoxins originated from endogenous human cytotoxic enzymes, and modification of the toxin moiety to escape the immune system. This paper devoted to reviewing recent advances in the design of immunotoxins with lower immunogenicity.


PLoS ONE ◽  
2019 ◽  
Vol 14 (3) ◽  
pp. e0214250 ◽  
Author(s):  
Amanda H. Kahn-Kirby ◽  
Akiko Amagata ◽  
Celine I. Maeder ◽  
Janet J. Mei ◽  
Steve Sideris ◽  
...  

2021 ◽  
pp. 153537022098676
Author(s):  
Jing Qian ◽  
Mo Yang ◽  
Qiang Feng ◽  
Xin-Yan Pan ◽  
Li-Lin Yang ◽  
...  

Ras gene mutation or overexpression can lead to tumorigenesis in multiple kinds of cancer, including glioma. However, no drugs targeting Ras or its expression products have been approved for clinical application thus far. Adenoviral gene therapy is a promising method for the treatment of glioma. In this study, the human glioma cell line U251 was co-cultured with recombinant adenovirus KGHV500, and the anti-tumor effects of KGHV500 were determined by MTT, scratch test, Transwell invasion, and apoptosis assays. Then, KGHV500 was delivered via the intravenous injection of CIK cells into glioma xenografts. Tumor volume, ki67 proliferation index, apoptosis levels, and anti-p21Ras scFv expression were tested to evaluate targeting ability, anti-tumor efficacy, and safety. We found that the KGHV500 exhibited anti-tumor activity in U251 cells and increased the intracellular expression of anti-p21Ras scFv compared with that in the control groups. CIK cells delivered KGHV500 to U251 glioma cell xenografts and enhanced anti-tumor activity against glioma xenografts compared to that produced by the control treatment. In conclusion, targeting Ras is a useful therapeutic strategy for gliomas and other Ras-driven cancers, and the delivery of anti-p21Ras scFv by recombinant adenovirus and CIK cells may play an essential role in the therapy of Ras-driven cancers. Impact statement For glioma treatment, gene therapy/virotherapy approach is a promising candidate. The Ras gene is reported to play a vital role in the RAS/RAF/mitogen-activated protein kinase (MAPK) pathway in gliomas. Thus, targeting the Ras gene should be a reasonable potential therapeutic method for glioma. In the present study, we used cytokine-induced killer (CIK) cells as secondary vectors to systemically deliver recombinant adenovirus KGHV500 to glioma xenografts and investigated the anti-tumor efficiency of recombinant adenovirus KGHV500 in vitro and in vivo. Our results expand evidence that targeting Ras is a useful and potential therapeutic strategy for gliomas. We believe that anti-p21Ras scFv delivered by recombinant adenovirus and CIK cells may play an important role in the therapy of Ras-driven cancers.


Pharmaceutics ◽  
2020 ◽  
Vol 12 (6) ◽  
pp. 549
Author(s):  
Laura Garcia-Perez ◽  
Anita Ordas ◽  
Kirsten Canté-Barrett ◽  
Pauline Meij ◽  
Karin Pike-Overzet ◽  
...  

Recent clinical trials using patient’s own corrected hematopoietic stem cells (HSCs), such as for primary immunodeficiencies (Adenosine deaminase (ADA) deficiency, X-linked Severe Combined Immunodeficiency (SCID), X-linked chronic granulomatous disease (CGD), Wiskott–Aldrich Syndrome (WAS)), have yielded promising results in the clinic; endorsing gene therapy to become standard therapy for a number of diseases. However, the journey to achieve such a successful therapy is not easy, and several challenges have to be overcome. In this review, we will address several different challenges in the development of gene therapy for immune deficiencies using our own experience with Recombinase-activating gene 1 (RAG1) SCID as an example. We will discuss product development (targeting of the therapeutic cells and choice of a suitable vector and delivery method), the proof-of-concept (in vitro and in vivo efficacy, toxicology, and safety), and the final release steps to the clinic (scaling up, good manufacturing practice (GMP) procedures/protocols and regulatory hurdles).


Sign in / Sign up

Export Citation Format

Share Document