Emergence of the Coexistence of mcr-1, blaNDM-5, and blaCTX-M-55 in Kl | IDR – Dove Medical Press

Introduction

Klebsiella pneumoniae (K. pneumoniae) is an opportunistic pathogen and the leading cause of healthcare-associated infections.1 Multidrug-resistant (MDR) K. pneumoniae isolates are rapidly spreading, thus limiting the choice of antimicrobial agents for empiric treatment of infections caused by these microorganisms; hence, this is a public health challenge.2

Polymyxins are last-resort antibiotics used to treat infections caused by carbapenem-resistant K. pneumoniae (CRKP).3 The two polymyxins currently in clinical use are polymyxin B and colistin (polymyxin E). They have similar antibacterial activity, but their structures differ by only one amino acid.4

The antibacterial effect of polymyxins on gram-negative bacteria is mainly a two-step mechanism comprising initial binding to and permeabilization of the outer membrane, followed by the destruction of cytoplasmic membrane.5 Notably, with the increase in the clinical use of polymyxins, polymyxin resistance has emerged and is rising rapidly. Polymyxin-resistant K. pneumoniae often spread in different hospital wards, making clinical treatment more difficult.6 The previously reported mechanisms of polymyxin resistance are chromosomally mediated and involve the regulation of two-component regulatory systems (eg, pmrAB, phoPQ, and its negative regulator, mgrB, in the case of K. pneumoniae), leading to the modification of lipid A (phosphoethanolamine or 4-amino-4-arabinose) or in rare cases, the complete loss of the lipopolysaccharide.7

Researchers reported the first plasmid-mediated polymyxin resistance mechanism, mcr-1, in Enterobacteriaceae in China. This warranted immediate worldwide attention, and mcr-1 has since been detected in Enterobacteriaceae from animals, food, and healthy people outside of China, including in Europe and the USA.8 Recently, some countries have also reported that mcr-1 and blaNDM-5 genes coexist in Escherichia coli strains,9,10 which is a serious challenge to treatment efforts.

This study assessed the current status of polymyxin resistance in CRKP isolates and investigated the possible coexistence of mcr-1 and -lactamase genes in K. pneumoniae in Nanchang, China.

From January 2018 to June 2019, a total of 107 nonduplicate CRKP isolates were isolated from hospitalized patients in different clinical departments in a tertiary teaching hospital in Nanchang, China. Different specimens were collected, and the K. pneumoniae isolates were identified using a VITEK-2 automated platform (bioMerieux, Marcy lEtoile, France). E. coli ATCC 25922 was used as a control strain.

The susceptibility of the K. pneumoniae clinical isolates to antimicrobials was determined using gram-negative susceptibility cards (AST-GN-16) on the VITEK system (bioMerieux, Marcy lEtoile, France) following the manufacturers instructions; the results were interpreted according to the Clinical and Laboratory Standards Institute (CLSI) standards.11 The MICs of polymyxin B for CRKP were further determined using the microdilution broth method according to the European Committee on Antimicrobial Susceptibility Testing (EUCAST) guidelines.12 A total of 16 antimicrobial agents were tested, including carbapenems (imipenem), -lactam/-lactamase inhibitor complexes (amoxicillin/clavulanic acid and piperacillintazobactam), monocyclic -lactam (aztreonam), cephalosporins (cefoxitin, cefepime, cefazolin, and ceftriaxone), aminoglycosides (gentamicin and amikacin), fluoroquinolones (levofloxacin and ciprofloxacin), folate metabolic pathway inhibitors (sulfamethoxazole), tetracyclines (tobramycin and tigecycline), and polymyxin B. E. coli ATCC 25922 was used as a control.

The carbapenemases produced by CRKP isolates were determined using a modified carbapenem inactivation test (mCIM) recommended by CLSI.11 In addition, a double-disc synergy test was performed to confirm the presence of metallo--lactamases (MBLs).11 The carbapenemase (blaKPC, blaGES, blaNDM, blaIMP, blaVIM, blaOXA-48, blaSIM, blaSPM, blaSME, and blaGIM), extended-spectrum -lactamase (ESBLs; blaTEM, blaDHA, blaSHV, blaCMY-II, and blaCTX-M), and polymyxin B (mcr-1 to mcr-8) resistance genes were detected using polymerase chain reaction (PCR) and DNA sequencing as described previously.2,13

Multilocus sequence typing (MLST) was performed on the polymyxin B-resistant K. pneumoniae isolates by amplifying and sequencing seven housekeeping genes (gapA, infB, mdh, pgi, phoE, rpoB, and tonB) according to a previously described protocol. Sequence types (STs) were assigned using the online database.

Pulsed-field gel electrophoresis (PFGE) was performed to analyze the phylogenetic relatedness of the polymyxin B-resistant K. pneumoniae isolates. Genomic DNA was digested by XbaI for 4 h at 37 C. Electrophoresis was performed for 19 h at 14 C, at an angle of 120, with switch times of 4 and 40 s at 6 V/cm using the CHEF III system (Bio-Rad Laboratories, Hercules, CA, USA). The Salmonella H9812 strain was used as the size marker. Analysis of the PFGE patterns using the Dice similarity coefficient was performed using the Bionumerics software (Applied Maths, Sint-Martens-Latem, Belgium). Clusters were defined as DNA patterns sharing more than 80% similarity.

A donor isolate, N816, was cultured in lysogeny broth (LB), and an azide-resistant E. coli J53 strain was used as the recipient. Transconjugants were selected on LB agar plates with 2 mg/L of polymyxin B or imipenem and 150 mg/L of sodium azide. Multiple attempts to transfer blaNDM-5 plasmid failed. Plasmid DNA was extracted from N816, transferred to competent E. coli DH5, and screened on LB agar plates with 2 mg/L imipenem. After the experiment, the transconjugant (JN816) and transformant (ZN816) were obtained and verified using PCR with previously described primers. Antimicrobial susceptibility testing was subsequently performed on JN816 and ZN816.

Genomic DNA was extracted from JN816 and ZN816 with the Qiagen Midi kit (Qiagen, Hilden, Germany) and sequenced with an Illumina HiSeq 2000 sequencer following a paired-end 2100-bp protocol.14 The raw data were mapped to a reference sequence found on the CLC genomics workbench version 8.0. Sequence comparison and alignment were performed using MEGA 5.01.15

Among the 107 K. pneumoniae isolates, 15 (14.0%) were resistant to polymyxin B according to EUCAST 7.0 guidelines.12 The antimicrobial resistance rates of these isolates are shown in Table 1. These isolates were resistant to imipenem, aztreonam, cefazolin, cefepime, cefoxitin, ceftriaxone, ciprofloxacin, and sulfamethoxazole. The resistance rates of isolates for amikacin, gentamicin, tobramycin, and tigecycline were 46.7, 60.0, 53.3, and 13.3%, respectively.

Table 1 Antimicrobial Resistance Profiling of 15 Carbapenem-Resistant Klebsiella pneumoniae Isolates

Twelve of the 15 polymyxin B-resistant isolates were confirmed as carbapenemase producers as determined using the mCIM assay, among which two isolates had a positive result for the double-disc synergy test, indicating that they also produced an MBL. In addition, 10 CRKP isolates were positive for blaKPC-2, and two were positive for blaNDM. Other carbapenemase genes including blaGES, blaIMP, blaVIM, blaOXA-48, blaSIM, blaSPM, blaSME, and blaGIM were not detected in any of the tested isolates. In addition to blaKPC-2, all isolates were positive for blaSHV, and eight (53.3%) were positive for the ESBL gene, blaCTX-M-65. Only one CRKP isolate was positive for mcr-1, blaNDM-5, blaCTX-M-55, and blaSHV-27 (Figure 1).

Figure 1 Pulsed-field gel electrophoresis results for 15 carbapenem-resistant Klebsiella pneumoniae isolates.

Among the 15 CRKP isolates, five STs were identified, including ST11 (11 isolates), and one isolate each in ST34, ST334, ST485, and a novel ST. The PFGE results showed that the 15 isolates were divided into nine different PFGE clusters; cluster A (4; 26.7%), cluster C (3; 20.0%), and cluster D (2; 13.3%). Each of the remaining six isolates were classified as singletons (Figure 1).

The two CRKP N816 plasmids harboring mcr-1 and blaCTX-M-55, designated as pMCR-1-N816 and pCTX-M-55-N816, respectively, were successfully transferred into the recipient strain (J53) via filter mating conjugation. We confirmed the presence of mcr-1 and blaCTX-M-55 genes in these plasmids. The antimicrobial resistance patterns of CRKP N816 and its transconjugant are shown in Table 2. The blaNDM-5-harboring plasmid of CRKP N816, designated as pNDM-5-N816, was electroporated into E. coli DH5 as described previously. Growth was observed only on plates with imipenem 2 mg/L, and the transformants were screened for the presence of blaNDM-5 using PCR, and blaNDM-5 was located on the plasmid. The antimicrobial resistance patterns of CRKP N816 and its transformants are shown in Table 2.

Table 2 Minimum Inhibitory Concentrations of Antimicrobials Against N816, JN816, ZN816, J53, and DH5

pMCR-1-N816 is 33309 base pairs (bp) long, with an average guanine-cytosine (GC) content of 41.84%. It has 41 predicted open reading frames (ORFs) and belongs to the IncX4 incompatibility group. A BLAST search of the plasmid sequences against the GenBank database showed that pMCR-1-N816 is similar (with 100% query coverage and >98.0% nucleotide identity) to pKP15450-MCR-1, an IncX4-type plasmid carrying mcr-1 among K. pneumoniae isolates in China (Figure 2). Plasmid pCTX-M-55-N816 is 76526-bp in length, with an average GC content of 51.93%. It has 105 predicted ORFs, and a BLAST search of the plasmid sequences against the GenBank database showed that pCTX-M-55-N816 is similar to pKP32558-4, with 89% query coverage and >98.0% nucleotide identity (Figure 3). Furthermore, pNDM-5-N816 is 46286-bp in length, with an average GC content of 46.63%, 59 predicted ORFs, and belongs to the IncX3 incompatibility group. A BLAST search showed that pNDM-5-N816 is similar to pNDM5-LDR, an IncX3-type plasmid carrying blaNDM-5 among K. pneumoniae isolates in China, with 100% query coverage and >99.9% nucleotide identity (Figure 4).

Figure 2 Structure of plasmid pMCR-1-N816 carrying mcr-1 from Klebsiella pneumoniae N816.

Figure 3 Structure of plasmid pCTX-M-55-N816 carrying blaCTX-M-55 from Klebsiella pneumoniae N816.

Figure 4 Structure of plasmid pNDM-5-N816 carrying blaNDM-5 from Klebsiella pneumoniae N816.

Carbapenems are the choice of treatment for infections caused by MDR K. pneumoniae. However, with the emergence of carbapenemase-producing bacteria, carbapenem resistance is increasing. The most common carbapenemase gene is blaKPC-2.16 Since the first discovery of the carbapenem resistance gene, blaNDM-1, in New Delhi, India,17 this gene and its multiple subtypes have been gradually discovered and reported worldwide. Moreover, the emergence of MBL-producing drug-resistant bacteria poses a great challenge for the treatment of drug-resistant bacterial infections. China reported a CRKP strain carrying the blaNDM-1 gene in 2013.18

Polymyxins have been used for many years in veterinary medicine, and nowadays, in human medicine, as a last resort for the treatment of MDR infections, especially CRKP. Thus, the increase in carbapenemase-producing Enterobacteriaceae has resulted in increased use of polymyxins with the inevitable risk of emerging polymyxin resistance.19,20 In this study, 107 CRKP isolates were tested for antimicrobial susceptibility; 15 (14.0%) of them were resistant to polymyxin B. The resistance rates of CRKP isolates to polymyxin B reported in Brazil and other European countries are 15.5% and 36%, respectively.20,21 The differences in antimicrobial resistance rates may be related to the different levels of antimicrobial usage in different countries.22

We found that 15 isolates were resistant to broad-spectrum antibiotics. Sequencing analysis showed that in addition to the blaKPC-2 gene, one or more other kinds of -lactamase genes (such as blaCTX-M, blaSHV, and blaTEM) were identified in these KPC-producing K. pneumoniae strains, with 53% (8/15) of the strains carrying the ESBL gene, blaCTX-M; this is consistent with previous reports.23 Consistent with other studies, the isolates were also more resistant to quinolones and trimethoprim/sulfamethoxazole. Quite often, plasmids carrying ESBL genes also carry other drug-resistant genes including quinolone and trimethoprim/sulfamethoxazole resistance genes.24

The drug susceptibility results of this study showed CRKP has a low resistance to amikacin, possibly because amikacin has only been used for a short time in this region or owing to the presence of restorative mutations. It may also be because of the aminoglycoside-modifying enzymes produced when amikacin is used to treat CRKP; the 16S rRNA gene targeted by amikacin is prone to mutations, resulting in a decrease in the activity of the enzyme to hydrolyze it.25 Although the resistance rate of CRKP to tigecycline is also low in this study, Its FDA approved uses include complicated skin/skin structure infections, complicated intra-abdominal infections, and community-acquired bacterial pneumonia, treatment of these infections limits its use.26 Studies have shown that polymyxin combined with amikacin has obvious synergistic and additive effects, and the MICs of this combination therapy are significantly lower than those of monotherapy.27 Polymyxin and amikacin may be sensitive to each other and as they target multiple proteins through different mechanisms to inhibit biofilm formation and increase membrane permeability, a synergistic effect to inhibit CRKP isolates is exhibited.28

Among the 15 polymyxin B-resistant CRKP isolates, most of the strains carried the blaKPC-2 gene, which is primarily responsible for carbapenem resistance, and this is consistent with our previous report.16 Among these strains, we only detected one isolate positive for mcr-1 gene. This strain was isolated from a blood culture specimen of a 71-year-old male patient and was resistant to multiple drugs, including polymyxin B, but not amikacin. This isolate also had blaCTX-M-55, blaNDM-5 and blaSHV-27 genes. Our experiments and sequencing results show that these mcr-1, blaCTX-M-55, and blaNDM-5 genes do not appear to be on the same plasmid, and the blaSHV-27 gene was found on the bacterial chromosome. Consistent with the above experimental results, the MICs of the corresponding antibiotic of the conjugants and transformants were altered accordingly (Table 2). The other 14 isolates did not harbor the mcr-1 gene. Other drug resistance mechanisms may be related to specific mutations within the genes encoding LPS-modifying enzymes, resulting in increased levels of the intrinsic regulator RamA and hyperproduction of CPS.29,30 However, this needs further testing. Unlike reports in other countries where the mcr-1 and blaNDM-5 genes were found to coexist in E. coli,9,10 this is the first time that K. pneumoniae has been reported to harbor both mcr-1, blaNDM5, and blaCTX-M-55 genes. What is even more worrying is that the plasmids in which these three genes are located have the ability to transfer horizontally. Thus, the bacteria may develop more serious drug resistance and lead to a state where there will be no treatment options. However, PFGE and MLST results of this isolate indicated that it had a different homology from the other 14 polymyxin B-resistant K. pneumoniae isolates, indicating that this type of bacteria did not have an outbreak. This isolate is not part of the most common ST (ST11) in China,16 but is a rare ST485 isolate. There is no report of an outbreak caused by K. pneumoniae ST485 at home and abroad. The patient had no history of foreign travel in the inquiry; thus, we infer that the occurrence of this isolate is a rare phenomenon in this area, but continuous monitoring to prevent the spread of this type of bacteria, which may cause more serious drug resistance, is warranted.

It has long been believed that polymyxin is mediated by chromosomes.19 Until 2015, Chinese scholars reported that the plasmid-mediated polymyxin resistance gene mcr-1 was found in Enterobacteriaceae isolated from humans and animals.3 Since then, people have a new understanding of the mechanism of polymyxin resistance. The emergence of a new type of drug resistance mechanism immediately set off a research boom among microbiologists worldwide. After sequencing and analysis of the plasmid obtained in this study, it was found that the isolate contained three plasmids of different sizes (33, 46, and 76 kb), which carried the mcr-1, blaCTX-M-55, and blaNDM-5 genes, which also verified the results of our previous conjugation, transformation, and drug susceptibility experiments. Further analysis of the data obtained using sequencing revealed that the similarity between the plasmid carrying mcr-1 and the plasmid pKP15450-MCR-1 was 98.77%. The mcr-1 gene at the starting point of the plasmid is approximately 3826 to 5451 bp, which encodes 541 amino acids. Analysis of its upstream and downstream genes showed that there are no common insert elements, indicating that the mcr-1 gene on this type of plasmid is more prone to horizontal transfer. The plasmid carrying the blaCTX-M-55 gene has a similarity of 99.96% with the plasmid pKP32558-4. The start site of the blaCTX-M-55 gene is approximately 2052 to 2927 bp, which encodes 291 amino acids and has an inserted transposon IS431 around the gene. The above two plasmids can be successfully joined to the same strain at the same time, indicating that they are compatible with each other. The plasmid carrying the blaNDM-5 gene has a similarity of 98.4% with the plasmid pNDM5-LDR. The start site of the blaNDM-5 gene is approximately 5783 to 6595 bp, which encodes 270 amino acids and has an inserted transposon, IS1086 and IS5H, upstream of the gene. The IS1086 and IS5H sequences indicate that the plasmid can transfer horizontally and confirm the results of previous experimental research. The resistance gene expression and transferability of this isolate have been further verified, which may lead to serious drug resistance.

In conclusion, the present study demonstrated for the first time the coexistence of mcr-1, blaNDM5, and blaCTX-M-55 in a K. pneumoniae ST485 isolate. Therefore, treatment strategies and monitoring should be implemented to limit the widespread of isolates containing mcr-1, blaNDM5, and blaCTX-M-55.

As the Klebsiella pneumoniae clinical isolate in this study was part of the routine hospital laboratory procedure, we have confirmed that the isolate has no identifiable patient data, the Second Affiliated Hospital of Nanchang University Medical Research Ethics Committee exempted this research for review.

This study was supported by a grant from the Department of Science and Technology of Jiangxi Province (20181BBG70030), the Jiangxi Natural Science Foundation (No.20181BAB205066), Science and Technology Plan of Jiangxi Provincial Health Commission (NO.20195211).

All authors contributed to data analysis, drafting or revising the article, gave final approval of the version to be published, and agree to be accountable for all aspects of the work.

The authors declare that they have no conflict of interest.

1. Arato V, Raso MM, Gasperini G, et al. Prophylaxis and treatment against Klebsiella pneumoniae: current insights on this emerging anti-microbial resistant global threat. Int J Mol Sci. 2021;22(8):4042. doi:10.3390/ijms22084042

2. Zhan L, Wang S, Guo Y, et al. Outbreak by hypermucoviscous Klebsiella pneumoniae ST11 isolates with carbapenem resistance in a tertiary hospital in China. Front Cell Infect Microbiol. 2017;7:182. doi:10.3389/fcimb.2017.00182

3. Chavda B, Lv J, Hou M, et al. Coidentification of mcr-4.3 and blaNDM-1 in a clinical Enterobacter cloacae isolate from China. Antimicrob Agents Chemother. 2018;62(10):e0064918. doi:10.1128/AAC.00649-18

4. Hussein M, Han ML, Zhu Y, et al. Metabolomics study of the synergistic killing of polymyxin B in combination with amikacin against polymyxin-susceptible and -resistant pseudomonas aeruginosa. Antimicrob Agents Chemother. 2019;64(1):e0158719. doi:10.1128/AAC.01587-19

5. Moffatt JH, Harper M, Harrison P, et al. Colistin resistance in Acinetobacter baumannii is mediated by complete loss of lipopolysaccharide production. Antimicrob Agents Chemother. 2010;54(12):49714977. doi:10.1128/AAC.00834-10

6. Bonura CMC, Bernardo FD, Aleo A, et al. Ongoing spread of colistin-resistant Klebsiella pneumoniae in different wards of an acute general hospital, Italy, June to December 2011. Euro Surveill. 2012;17(33):20248. doi:10.1186/1475-2875-11-277

7. Liu YY, Wang Y, Walsh TR, et al. Emergence of plasmid-mediated colistin resistance mechanism MCR-1 in animals and human beings in China: a microbiological and molecular biological study. Lancet Infect Dis. 2016;16(2):161168. doi:10.1016/s1473-3099(15)00424-7

8. Quan J, Li X, Chen Y, et al. Prevalence of mcr-1 in Escherichia coli and Klebsiella pneumoniae recovered from bloodstream infections in China: a multicentre longitudinal study. Lancet Infect Dis. 2017;17(4):400410. doi:10.1016/s1473-3099(16)30528-x

9. Yang RS, Feng Y, Lv XY, et al. Emergence of NDM-5- and MCR-1-producing Escherichia coli clones ST648 and ST156 from a Single Muscovy Duck (Cairina moschata). Antimicrob Agents Chemother. 2016;60(11):68996902. doi:10.1128/AAC.01365-16

10. Zhang Y, Liao K, Gao H, et al. Decreased fitness and virulence in ST10 Escherichia coli harboring blaNDM-5 and mcr-1 against a ST4981 strain with blaNDM-5. Front Cell Infect Microbiol. 2017;7:242. doi:10.3389/fcimb.2017.00242

11. CLSI. Performance Standards for Antimicrobial Susceptibility Testing. 28th. CLSI supplement M100. Wayne, PA: Clinical and Laboratory Standard Institute; 2018.

12. The European Committee on Antimicrobial susceptibility testing breakpoint tables for interpretation of MICs and zone diameters. Version 6.0,2016. Available from: http://www.eucast.org. Accessed March 30,2016.

13. Yang F, Shen C, Zheng X, et al. Plasmid-mediated colistin resistance gene mcr-1 in Escherichia coli and Klebsiella pneumoniae isolated from market retail fruits in Guangzhou, China. Infect Drug Resist. 2019;12:385389. doi:10.2147/idr.s194635

14. Chen L, Hu H, Chavda KD, et al. Complete sequence of a KPC-producing IncN multidrug-resistant plasmid from an epidemic Escherichia coli sequence type 131 strain inChina. Antimicrob Agents Chemother. 2014;58(4):24222425. doi:10.1128/aac.02587-13

15. Wang Z, Li M, Shen X, et al. Outbreak of blaNDM-5-harboring Klebsiella pneumoniae ST290 in a tertiary hospital in China. Microb Drug Resist. 2019;25(10):14431448. doi:10.1089/mdr.2019.0046

16. Hu L, Liu Y, Deng L, et al. Outbreak by ventilator-associated ST11 K. pneumoniae with co-production of CTX-M-24 and KPC-2 in a SICU of a tertiary teaching hospital in central China. Front Microbiol. 2016;7:1190. doi:10.3389/fmicb.2016.01190

17. Yong D, Toleman MA, Giske CG, et al. Characterization of a new metallo-beta-lactamase gene, bla(NDM-1), and a novel erythromycin esterase gene carried on a unique genetic structure in Klebsiella pneumoniae sequence type 14 from India. Antimicrob Agents Chemother. 2009;53(12):50465054. doi:10.1128/AAC.00774-09

18. Hu L, Zhong Q, Tu J, et al. Emergence of blaNDM-1 among Klebsiella pneumoniae ST15 and novel ST1031 clinical isolates in China. Diagn Microbiol Infect Dis. 2013;75(4):373376. doi:10.1016/j.diagmicrobio.2013.01.006

19. Mendes AC, Novais A, Campos J, et al. mcr-1 in carbapenemase-producing Klebsiella pneumoniae with hospitalized patients, Portugal, 20162017. Emerg Infect Dis. 2018;24(4):762766. doi:10.3201/eid2404.171787

20. Wang R, van Dorp L, Shaw LP, et al. The global distribution and spread of the mobilized colistin resistance gene mcr-1. Nat Commun. 2018;9(1):19. doi:10.1038/s41467-018-03205-z

21. Di Tella D, Tamburro M, Guerrizio G, et al. Molecular epidemiological insights into colistin-resistant and carbapenemases-producing clinical Klebsiella pneumoniae. Infect Drug Resist. 2019;Volume 12:37833795. doi:10.2147/idr.s226416

22. European Centre for Disease P., Control, A. European Food Safety. ECDC/EFSA/EMA second joint report on the integrated analysis of the consumption of antimicrobial agents and occurrence of antimicrobial resistance in bacteria from humans and food-producing animals: Joint Interagency Antimicrobial Consumption and Resistance Analysis (JIACRA) Report. EFSA J. 2017;15(7):e04872. doi:10.2903/j.efsa.2017.4872

23. Liu H, Lin H, Sun Z, et al. Distribution of beta-lactamase genes and genetic context of bla KPC-2 in clinical carbapenemase-producing Klebsiella pneumoniae Isolates. Infect Drug Resist. 2021;14:237247. doi:10.2147/IDR.S290434

24. Xiao Y, Hang Y, Chen Y, et al. A retrospective analysis of risk factors and patient outcomes of bloodstream infection with extended-spectrum beta-lactamase-producing Escherichia coli in a Chinese tertiary hospital. Infect Drug Resist. 2020;13:42894296. doi:10.2147/IDR.S269989

25. Zhang J, Yu L, Fu Y, et al. Tigecycline in combination with other antibiotics against clinical isolates of carbapenem-resistant Klebsiella pneumoniae in vitro. Ann Palliat Med. 2019;8(5):622631. doi:10.21037/apm.2019.09.11

26. Gong J, Su D, Shang J, et al. Efficacy and safety of high-dose tigecycline for the treatment of infectious diseases: a meta-analysis. Medicine. 2019;98(38):e17091. doi:10.1097/MD.0000000000017091

27. Yu L, Zhang J, Fu Y, et al. Synergetic effects of combined treatment of colistin with meropenem or amikacin on carbapenem-resistant Klebsiella pneumoniae in vitro. Front Cell Infect Microbiol. 2019;9:422. doi:10.3389/fcimb.2019.00422

28. Ayoub Moubareck C. Polymyxins and bacterial membranes: a review of antibacterial activity and mechanisms of resistance. Membranes. 2020;10(8):181. doi:10.3390/membranes10080181

29. Poirel L, Jayol A, Nordmann P. Polymyxins: antibacterial activity, susceptibility testing, and resistance mechanisms encoded by plasmids or chromosomes. Clin Microbiol Rev. 2017;30(2):557596. doi:10.1128/CMR.00064-16

30. Petrosillo N, Taglietti F, Granata G. Treatment options for colistin resistant Klebsiella pneumoniae: present and future. J Clin Med. 2019;8(7):934. doi:10.3390/jcm8070934

More:
Emergence of the Coexistence of mcr-1, blaNDM-5, and blaCTX-M-55 in Kl | IDR - Dove Medical Press

Your Healthy Family: New gene therapy providing hope for those with rare genetic disorders – KOAA.com Colorado Springs and Pueblo News

COLUMBUS, OHIO A novel method of gene therapy is helping children born with a rare genetic disorder called AADC deficiency that causes severe physical and developmental disabilities. The study, led by researchers at The Ohio State University Wexner Medical Center and The Ohio State University College of Medicine, offers new hope to those living with incurable genetic and neurodegenerative diseases.

Research findings are published online in the journal Nature Communications.

This study describes the findings from the targeted delivery of gene therapy to midbrain to treat a rare deadly neurodevelopmental disorder in children with a neurogenetic disease, aromatic L-amino acid decarboxylase (AADC) deficiency characterized by deficient synthesis of dopamine and serotonin.

Only about 135 children worldwide are known to be missing the enzyme that produces dopamine in the central nervous system, which fuels pathways in the brain responsible for motor function and emotions. Without this enzyme, children lack muscle control, and are usually unable to speak, feed themselves or even hold up their head. They also suffer from seizure-like episodes called oculogyric crises that can last for hours.

Remarkably, these episodes are the first symptom to disappear after gene therapy surgery, and they never return, said study co-author Dr. Krystof Bankiewicz, professor of neurological surgery at Ohio State College of Medicine who leads the Bankiewicz Lab. In the months that follow, many patients experience life-changing improvements. Not only do they begin laughing and have improved mood, but many are able to begin speaking and even walking. They are making up for the time they lost during their abnormal development.

The directed gene therapy in seven children ages 4 to 9 who were infused with the viral vector resulted in dramatic improvement of symptoms, motor function and quality of life. Six children were treated at UCSF Benioff Childrens Hospital in San Francisco and one at Ohio State Wexner Medical Center. This therapeutic modality promises to transform the treatment of AADC deficiency and other similar disorders of the brain in the future, Bankiewicz said.

During the gene therapy surgery, physicians infuse a benign virus programmed with specific DNA into precisely targeted areas of the brain. The infusion is delivered extremely slowly as surgeons monitor exactly how it spreads within the brain using real-time MRI imaging.

Really, what we're doing is introducing a different code to the cell, said Dr. James Brad Elder, director of neurosurgical oncology at Ohio State Wexner Medical Centers Neurological Institute. And we're watching the whole thing happen live. So we continuously repeat the MRI and we can see the infusion blossom within the desired nucleus.

Researchers believe this same method of gene therapy can be used to treat other genetic disorders as well as common neurodegenerative diseases, such as Parkinsons and Alzheimers disease. Clinical trials are underway to test this procedure in others living with debilitating and incurable neurological conditions.

The directed gene therapy, in these patients, resulted in dramatic improvement of symptoms, motor function and quality of life. This therapeutic modality promises to transform the treatment of AADC deficiency and other similar disorders of the brain in the future.

The findings described in this study are the culmination of decades of work by teams from multiple academic institutions, including University of California San Francisco, Washington University in St. Louis, Medical Neurogenetics Laboratory in Atlanta, St. Louis Childrens Hospital and Nationwide Childrens Hospital in Columbus, Ohio.

The research was supported by the National Institute of Neurological Disorders and Stroke and foundational grants, including the AADC Research Trust, the Pediatric Neurotransmitter Disease Association and funding from The Ohio State University.

This work provides a framework for the treatment of other human nervous system genetic diseases. Its our hope that this will be first of many ultra-rare and other neurologic disorders that will be treated with gene therapy in a similar manner, Bankiewicz said.

Read the original post:
Your Healthy Family: New gene therapy providing hope for those with rare genetic disorders - KOAA.com Colorado Springs and Pueblo News

Dissecting the shared genetic basis of migraine and mental disorders using novel statistical tools. – Physician’s Weekly

Migraine is three times more prevalent in people with bipolar disorder or depression. The relationship between schizophrenia and migraine is less certain although glutamatergic and serotonergic neurotransmission are implicated in both. A shared genetic basis to migraine and mental disorders has been suggested but previous studies have reported weak or non-significant genetic correlations and five shared risk loci. Using the largest samples to date and novel statistical tools, we aimed to determine the extent to which migraines polygenic architecture overlaps with bipolar disorder, depression, and schizophrenia beyond genetic correlation, and to identify shared genetic loci. Summary statistics from genome-wide association studies were acquired from large-scale consortia for migraine (n cases=59,674; n controls=316,078), bipolar disorder (n cases=20,352; n controls=31,358), depression (n cases=170,756; n controls=328,443) and schizophrenia (n cases=40,675, n controls=64,643). We applied the bivariate causal mixture model to estimate the number of disorder-influencing variants shared between migraine and each mental disorder, and the conditional/conjunctional false discovery rate method to identify shared loci. Loci were functionally characterised to provide biological insights. Univariate MiXeR analysis revealed that migraine was substantially less polygenic (2.8K disorder-influencing variants) compared to mental disorders (8.1K-12.3K disorder-influencing variants). Bivariate analysis estimated that 0.8K (0.3K), 2.1K (SD=0.1K) and 2.3K (SD=0.3K) variants were shared between bipolar disorder, depression and schizophrenia, respectively. There was also extensive overlap with intelligence (1.8K, SD=0.3K) and educational attainment (2.1K, SD=0.3K) but not height (1K, SD=0.1K). We next identified 14 loci jointly associated with migraine and depression and 36 loci jointly associated with migraine and schizophrenia, with evidence of consistent genetic effects in independent samples. No loci were associated with migraine and bipolar disorder. Functional annotation mapped 37 and 298 genes to migraine and each of depression and schizophrenia, respectively, including several novel putative migraine genes such as L3MBTL2, CACNB2, SLC9B1. Gene-set analysis identified several putative gene-sets enriched with mapped genes including transmembrane transport in migraine and schizophrenia. Most migraine-influencing variants were predicted to influence depression and schizophrenia, although a minority of mental disorder-influencing variants were shared with migraine due to the difference in polygenicity. Similar overlap with other brain-related phenotypes suggests this represents a pool of pleiotropic variants which influence vulnerability to diverse brain-related disorders and traits. We also identified specific loci shared between migraine and each of depression and schizophrenia, implicating shared molecular mechanisms and highlighting candidate migraine genes for experimental validation. The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain.

More:
Dissecting the shared genetic basis of migraine and mental disorders using novel statistical tools. - Physician's Weekly

Researchers partner with industry to create better gene therapy tools – The Hub at Johns Hopkins

ByGina Wadas

Viruses are experts at infiltrating the body, as the SARS-CoV-2 virus (and resulting COVID-19 pandemic) have amply demonstrated. But their efficiency in targeting specific and isolated cells also make them useful drug delivery vehicles, known as viral vectors.

Viral vectors are modified viruses that can act as couriers to transport therapeutic "packages" to specific diseased cells. These packages contain instructions with modified or designed DNA or RNA to correct or supplement a faulty or missing gene. For instance, the Johnson & Johnson COVID-19 vaccine uses viral vectors to transport modified genetic material from the SARS-CoV-2 virus to cells, generating an immune response.

Though viral vector-based gene therapies are among the most advanced treatments for many congenital and acquired diseases, producing them is complex and costly.

"One of the major challenges in viral vector gene therapy is how to improve the quality, purity, and cost of the manufactured viral vectors, so that we can use the smallest possible effective dose, reduce immune side effects, and lower the cost of treatments," said Hai-Quan Mao, associate director and core faculty member of the Institute for NanoBioTechnology. He is also a professor in the departments of Materials Science and Engineering and Biomedical Engineering and a core faculty member at the Translational Tissue Engineering Center.

Hai-Quan Mao

Associate director, Institute for NanoBioTechnology

To address this challenge, Mao and his team are teaming up with Nolan Sutherland, senior scientist at bluebird bio, a Cambridge, Massachusetts-based biotechnology company that develops gene therapies. The partnership started about two years ago when Yizong Hu, a biomedical engineering PhD student under the mentorship of Mao, was at an annual meeting for the American Society for Gene and Cell Therapy presenting his research on a new particle assembly technology. Sutherland heard the presentation and approached Hu to discuss the technology and its application to the production of lentiviral vectors, which are made from a family of viruses that infect people by reverse transcription of their RNA into DNA in their host cells' genome.

Sutherland thought that the Mao team's approach might help streamline transfection, a key step in producing viral vectors. During transfection, a polymer solution is combined with a mixture of DNA plasmids to form transfection particles, a cumbersome procedure involving complicated solution blending and strictly timed dosing.

Mao, Hu, and Yining Zhu, also a biomedical engineering PhD student, developed a more effective and shelf-stable formulation of DNA particles in a ready-to-dose form. They also discovered that size-controlled sub-micron particles are most effective in transfecting cells and producing viral vectors. This production method is based on the team's years of experience in controlling transfection vehicle characteristics to enhance performances and stability.

The team members validated their findings with Sutherland at bluebird bio using that company's bioreactor. They compared the new method with the industry standard, and the results showed improved vector production yield, shelf stability, handling stability, and quality control of the transfection process.

"With the drastic increase in demand for lentiviral vector-based cell therapy products ... this new technology will greatly improve the production quality, consistency, and yield of our therapeutic LVVs," Sutherland said.

The team reported its findings in Nano Letters and is scaling up production with an eye to transferring the technology to the marketplace.

"This work represents a great example how we can partner with corporate collaborators to accelerate the translation of discoveries on the bench to the industry. This type of collaboration with industry provides us opportunities to identify the technical gaps in the engineering solutions that we develop, and fine tune them to better address the real-world problems in a more targeted fashion," Mao said.

According to Sutherland, the partnership with Mao and his team has "allowed bluebird to pursue high risk/reward innovation in a space outside of its core expertise. The team has a keen eye for application to industry which has made the partnership incredibly productive."

Team members say that this new particle engineering technology will find a wide range of applications in the manufacture of a variety of viral vectors for gene and cell therapy applications.

Also contributing to the project are Jordan Green, professor in the Department of Biomedical Engineering and associate member at the INBT, and Sashank Reddy, assistant professor of plastic and reconstructive surgery at Johns Hopkins Medicine, medical director at Johns Hopkins Technology Ventures, and affiliate faculty member at the INBT.

More here:
Researchers partner with industry to create better gene therapy tools - The Hub at Johns Hopkins

GenSight’s mutation-agnostic gene therapy triggers partial vision recovery – FierceBiotech

GenSight Biologics has reported the partial recovery of the vision of a retinitis pigmentosa patient treated with its mutation-independent approach to the restoration of visual function.

Spark Therapeutics, now part of Roche, won FDA approval for its retinitis pigmentosa gene therapy Luxturna in 2017. However, the approval only covered the use of Luxturna in patients with confirmed biallelic RPE65 mutation-associated retinal dystrophy. As retinitis pigmentosa is caused by changes to more than 70 genes, it is questionable whether mutation-specific approaches can cover all patients.

Recognizing that, GenSight is developing GS030, an optogenetic therapy. The drug component of the intervention consists of a viral vector designed to cause the expression of the light-sensitive opsin ChrimsonR in retinal ganglion cells.

In a phase 1/2 clinical trial, physicians administered the gene therapy via a single intravitreal injection to the worse-seeing eye of a 58-year-old man who was diagnosed with retinitis pigmentosa 40 years ago and was unable to detect objects visually. The patient was still unable to detect objects visually after the injection.

However, the patient could perceive, locate, count and touch different objects using his treated eye while wearing a pair of goggles. The goggles detect changes in light intensity and send pulses of light to the retina in real time, effectively amplifying the signal sent to the ChrimsonR-expressing cells.

Writing in Nature Medicine, the investigators said the patient is the first reported case of partial functional recovery in a neurodegenerative disease after optogenetic therapy. The claim is based on three visual tests, the last of which was performed five months after the others, that took place in an indoor laboratory, but there is also patient-reported evidence of real-world benefits.

The patient spontaneously reported identifying crosswalks and he could count the number of white stripes. Subsequently, the patient testified to a major improvement in daily visual activities, such as detecting a plate, mug or phone, finding a piece of furniture in a room or detecting a door in a corridor but only when using the goggles, the authors of the research paper wrote.

The paper only describes the experience of one patient, and the recovery in his vision was partial. GenSight recruited six patients across the first two cohorts of the dose-escalation trial before putting enrollment on hold in response to the pandemic. Enrollment in the third cohort is now complete, and GenSight expects to recruit an extension cohort that will get the optimal dose by the end of 2021.

A clearer picture of the efficacy of GS030 will emerge as GenSight shares data on other recipients of the treatment. Other companies are pursuing the same opportunity. Novartis, for example, bought optogenetic gene therapy startup Vedere Bio for $150 million upfront last year.

Read the rest here:
GenSight's mutation-agnostic gene therapy triggers partial vision recovery - FierceBiotech

Clinical Significance of the D-Loop Gene Mutation in Mitochondrial DNA | OTT – Dove Medical Press

Introduction

Laryngeal cancer is a relatively common malignant tumor in the head and neck, accounting for 15% of malignant tumors in the body. Squamous cell carcinomas account for 9698% of laryngeal cancers. Incidences of laryngeal cancer are increasing yearly as the smoking population expands, and the environment deteriorates. In 2015, there were 26,400 new cases of laryngeal cancer in China. Among these, 14,500 people died. The male to female ratio was 9:1.1 The incidence of laryngeal cancer is high in middle-aged and older males. In recent years, more incidences of laryngeal cancer are being found in the younger population, with significant differences among different races and regions.2 In China, the incidence of laryngeal cancer is highest in the Northeast and the North. In the United States, over the past 40 years, the percentage of patients with laryngeal cancer who survive for five years or more has decreased from 66% to 63%,3 suggesting the need for innovation in detection and treatment. Mitochondria are widely involved in cellular activities, including the regulation of cell cycle, oxidative stress, and apoptosis.4 Mitochondrial DNA (mtDNA) injury is closely correlated with tumors, aging, and neurodegenerative diseases.5,6 The D-loop region is the non-coding region in mtDNA. Most of the regulatory sequences related to the mtDNA replication, transcription, and translation are found in this region, which is prone to mutation. Although there have been achievements in the field concerning the relationship between tumorigenesis and mitochondrial mutation, and some related research at home and abroad have clarified some related mechanisms, the research on the relationship between tumorigenesis and mtDNA remains unclear. Moreover, no direct relationship between the abnormal changes of mtDNA and tumorigenesis has been found so far. Although many studies have reported the relationship between mitochondrial gene mutations and various tumors, the study on the mechanism of laryngeal cancer has not been reported. The purpose of the present study was to investigate the relationship between the D-loop gene mutation in mtDNA and the clinical and pathological parameters of laryngeal cancer by gene sequencing.

From June 2013 to June 2019, 60 patients with squamous cell laryngeal carcinoma were selected from the First Affiliated Hospital of Henan University of Science and Technology. No patients received radiotherapy or chemotherapy before the operation. The age ranged between 4080, with an average age of 59.12 10.35. There were 55 males and five females. There were 15 cases in stage I, 26 in stage II, ten in stage III, and nine in stage IV (according to the 2017 UICC staging criteria). The specimens from all cases were diagnosed and screened by the pathological experts of the First Affiliated Hospital of Henan University of Science and Technology. This study was approved by the medical ethics committee of the Henan University of Science and Technology. Written informed consent was obtained from all participants. This study complied with the Declaration of Helsinki.

DNA Marker DL2000 (CB15727772 TAKARA, Japan); 10Xbuffer (Mg2+plus) (DR001AM TAKARA, Japan); dNTPs (DR001AM TAKARA, Japan); Taq DNA polymerase (DR001AM TAKARA, Japan); agarose (D1200 Beijing Solabo Technology Co., Ltd); Primers (JN0060-02 Shanghai Bioengineering Technology Co., Ltd).

Thermo high-speed centrifuge (USA); PCR instrument (ABI, USA); Gel imaging system (USA); 80C refrigerator (Japan).

The specimens resected during laryngeal cancer surgery were removed, and the central part of the specimen with cancer focus was taken as the cancer tissue. Approximately 20 mg of the cancer tissue was cut and placed in a centrifuge tube. The tissue was broken into cell suspension by a high-speed tissue homogenizer. The following procedures were operated according to the kit instructions. After a series of extraction and washing, the final purified DNA solution was placed in a 20C refrigerator for storage and further detection.

The sequence in the D-loop region in mtDNA was the target sequence with a full length of 1122 bp. The primers and probes were synthesized by the Shanghai Bioengineering Technology Service Co., Ltd. The sequences of primers were as follows: the upper primer 5-CCATTAGCACCCAAAGCTAAG-3, the lower primer 5-TGCTTTGAGGTAAGCTACA-3. The reaction system was 50 L, with 5 L of the 10 buffer (Mg2+ plus) solution, 4 L of the mixture of dNTP, 0.25 L of the DNA polymerase, and 1 L of the upper primer and 1 L of the lower primer, 1 L of the template DNA, and 37.75 L of distilled water. The reaction conditions were as follows: 94C 30s, 55C 30s, 72C 90s, with a total of 30 cycles. The electrophoresis of the PCR products was under the condition of 120V for 30 minutes. After the electrophoresis, the gel imaging system was used to take photos to confirm that the amplified fragment was the desired target fragment.

The purification of the PCR products and the determination of gene sequences were completed by the Beijing Sequencing Department of Shanghai Yingjun Biotechnology Co., Ltd. The quality of the peak map was evaluated by Chromas 2.31 software, and the samples with low quality were re-sequenced. After landing on the website (https://www.Ncbi.Nlm.Nih.gov), the Align two (or more) sequences using BLAST software was used to compare the gene sequence of laryngeal cancer tissue with the Cambridge standard sequence. Once the mutation site is identified, the Chromas software should be adopted to observe the sequencing peak and compare it with the databases reported mutation site. If there is no report, it is considered a novel mutation. At the same time, these mutation sites were compared with the sub-database of the polymorphic sites of the mtDB database. If the mutation frequency was more than 1% in all populations tested in the database, it was regarded as the single nucleotide polymorphism (SNP). The gene sequencing data was provided in the Supplementary Document.

The Prism GraphPad 5.0 software was used for statistical analysis of the results. The Pearsons or the Spearman correlation test was adopted to test the correlation between the two variables, and 2 test was used to compare the number of mutations between genders. P < 0.05 was considered statistically significant.

The length of the target gene was 1122bp after the PCR amplification of the full length of the D-loop region (Figure 1). A single bright band was demonstrated by the gel imaging instrument, and the molecular weight was determined by the standard of DL2000DNAMark.

Figure 1 Results of the mtDNA PCR product.

Among the 60 samples, 38 mutation sites had been detected in 51 samples (85%), with a total of 174 mutations. In three HV in the D-loop, 160 mutations had been detected, accounting for 91.9% of the total mutations (Table 1). Microsatellite instability existed in the D310 regions in 15 cases (25%) (Figure 2), mainly manifested as the increased insertion of poly-C. Among them, nine cases showed insertion of one base C, six cases showed insertion of two bases, and the base pair exchanges within a microsatellite were found in two cases (Figure 3).

Table 1 Sequencing Results and Analysis

Figure 2 Microsatellite instability in D310 region (insertion of base pair C).

Figure 3 Microsatellite instability in D310 region (T, C exchange).

As shown in Table 2, there was no correlation between the age, gender, tumor diameter, TNM stage, and the number of the D-loop mutations in mtDNA in the 51 specimens with mutations (P > 0.05).

Table 2 The Correlation Between the D-Loop Region Mutations in mtDNA and the Clinical and Pathological Parameters in Laryngeal Cancer (Number of Cases)

Mitochondria are the only organelles with DNA outside the nucleus in the eukaryotes, which are the main sites for oxidative phosphorylation to produce active oxygen and provide necessary energy and oxygen radicals for cell activities. When mtDNA mutation occurs, the cellular energy supply is dysfunctional, and a large number of ROS are produced. This leads to changes in cell function and even cell necrosis, thus showing a variety of clinical symptoms. Mitochondrial dysfunction may play an important role in tumor development, early diagnosis, drug resistance, prevention of recurrence, and prognosis.79 MtDNA is a closed-loop double-stranded DNA molecule composed of 16,569 base pairs. As the main non-coding region, the D-loop region of npl6024-np576 is responsible for regulating mtDNA replication and transcription.10 Sanchez-Cespedes et al11 found that 41% of head and neck squamous cell carcinoma had mutations in the D-loop region, including the deletion, insertion, and point mutation. Ha et al12 revealed that patients with head and neck tumors had the change of poly-C in the D-loop region of mtDNA. Our experiment confirmed this. In recent years, the D-loop region is agreed to be the HV of mtDNA mutations. In the present study, 91.9% of the mutation sites were concentrated in the HV region, consistent with the above view. Microsatellites are the short tandem repeats in the human genome. Microsatellite instability refers to the changes of microsatellite in length and the emergence of new microsatellite alleles in tumors due to the insertion or deletion of repeat units compared with normal tissues. Habano et al13 first proposed the mitochondrial microsatellite instability in a study on rectal cancer. In the present study, microsatellite instabilities were found in 15 cases of laryngeal cancer, mainly concentrated in the D310 region and manifested as the increase of poly-C insertion. Among these cases, nine showed insertion of one base C, six showed insertion of two bases, and base exchanges within a microsatellite were found in two cases. This was consistent with the study by Sanchez-Cespedes et al that 41% of head and neck squamous cell carcinoma had mutations in the D-loop region area.11 The HV of the D310 region may be related to its location in the intron, coder, and promoter of the gene and the repeated repair caused by slip error during replication.14 Therefore, the analysis of mtDNA mutations, especially the detection of changes in the D310 region, may play an important role in the cytological diagnosis, especially for cases with no obvious morphological changes or rare tumor cells. In conclusion, PCR amplification and direct sequencing were used in the present study to detect the D-loop gene changes in mtDNA in samples of laryngeal cancer tissue. It was found that there were a large number of mutations and microsatellite instability in the D-loop region in mtDNA of patients with laryngeal cancer, indicating that the D-loop gene mutations in mtDNA may play an important role in the development of laryngeal cancer.

We would like to acknowledge the hard and dedicated work of Dr. Kai Xi and Dr. Hao Chang that helped and contributed to us in terms of technology and software of the study.

The authors report no conflicts of interest in this work.

1. Chen W, Zheng R, Baade PD, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115132. doi:10.3322/caac.21338

2. Shin JY, Truong MT. Racial disparities in laryngeal cancer treatment and outcome: a population-based analysis of 24,069 patients. Laryngoscope. 2015;125(7):16671674. doi:10.1002/lary.25212

3. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):730. doi:10.3322/caac.21332

4. Correia-Melo C, Passos JF. Mitochondria: are they causal players in cellular senescence? Biochim Biophys Acta. 2015;1847(11):13731379. doi:10.1016/j.bbabio.2015.05.017

5. Bose A, Beal MF. Mitochondrial dysfunction in Parkinsons disease. J Neurochem. 2016;139(Suppl 1):216231. doi:10.1111/jnc.13731

6. Lee J, Chang JY, Kang YE, et al. Mitochondrial energy metabolism and thyroid cancers. Endocrinol Metab. 2015;30(2):117123. doi:10.3803/EnM.2015.30.2.117

7. Chen N, Wen S, Sun X, et al. Elevated mitochondrial DNA copy number in peripheral blood and tissue predict the opposite outcome of cancer: a meta-analysis. Sci Rep. 2016;18(6):37404. doi:10.1038/srep37404

8. Huang X, Zhou X, Hu Q, et al. Advances in esophageal cancer: a new perspective on pathogenesis associated with long non-coding RNAs. Cancer Lett. 2018;28(413):94101. doi:10.1016/j.canlet.2017.10.046

9. St John JC. Mitochondrial DNA copy number and replication in reprogramming and differentiation. Semin Cell Dev Biol. 2016;52:93101. doi:10.1016/j.semcdb.2016.01.028

10. Morandi L, Asioli S, Cavazza A, Pession A, Damiani S. Genetic relationship among atypical adenomatous hyperplasia, bronchioloalveolar carcinoma and adenocarcinoma of the lung. Lung Cancer. 2007;56(1):3542. doi:10.1016/j.lungcan.2006.11.022

11. Sanchez-Cespedes M, Parrella P, Nomoto S, et al. Identification of a mononucleotide repeat as a major target for mitochondrial DNA alterations in human tumors. Cancer Res. 2001;61(19):70157019.

12. Ha PK, Tong BC, Westra WH, et al. Mitochondrial C-tract alteration in premalignant lesions of the head and neck: a marker for progression and clonal proliferation. Clin Cancer Res. 2002;8(7):22602265.

13. Habano W, Sugai T, Nakamura SI, Uesugi N, Yoshida T, Sasou S. Microsatellite instability and mutation of mitochondrial and nuclear DNA in gastric carcinoma. Gastroenterology. 2000;118(5):835841. doi:10.1016/S0016-5085(00)70169-7

14. Lin CS, Lee HT, Lee MH, et al. Role of mitochondrial DNA copy number alteration in human renal cell carcinoma. Int J Mol Sci. 2016;17(6):814. doi:10.3390/ijms17060814

Visit link:
Clinical Significance of the D-Loop Gene Mutation in Mitochondrial DNA | OTT - Dove Medical Press

GenSight Biologics Announces Nature Medicine Case Report Showing Visual Recovery after GS030 Optogenetic Treatment – Business Wire

PARIS--(BUSINESS WIRE)--Regulatory News:

GenSight Biologics (Paris:SIGHT) (Euronext: SIGHT, ISIN: FR0013183985, PEA-PME eligible), a biopharma company focused on developing and commercializing innovative gene therapies for retinal neurodegenerative diseases and central nervous system disorders, today announced that the highly-regarded journal Nature Medicine has published the first case report of partial recovery of visual function in a blind patient with late stage retinitis pigmentosa (RP). The subject is a participant in the ongoing PIONEER Phase I/II clinical trial of GenSight Biologics GS030 optogenetic therapy. Published in the May issue under the title Partial recovery of visual function in a blind patient after optogenetic therapy, the paper* is the first peer-reviewed documentation of visual recovery after a blind patient was treated with optogenetic therapy.

These are truly groundbreaking findings that move the promise of optogenetics another step from therapeutic concept to clinical use, commented Bernard Gilly, Co-Founder and Chief Executive Officer of GenSight. These could not have occurred without the close collaboration we enjoyed with our partners at the Institut de la Vision, the Institute of Ophthalmology Basel and Streetlab. We are especially grateful to the patients who are participating in our trial, whose experiences and input will help us design the next stage of GS030s clinical development. We will now accelerate the GS030 program to make it our second product to reach the market after LUMEVOQ.

Optogenetic therapies combine cellular expression of light-sensitive opsins with light stimulation using a medical device. GS030 uses an optimized viral vector (GS030-DP) to express the light-sensitive opsin ChrimsonR in retinal ganglion cells and proprietary light-stimulating goggles (GS030-MD) to project the right wavelength and intensity of light onto the treated retina. GS030-DP is administered via an intravitreal injection.

It was breathtaking to witness the first recovery of some visual function in a blind patient, commented Dr. Botond Roska, MD, PhD, last and co-corresponding author and a pioneer in the field of optogenetic vision restoration. Dr. Roska is Founding Director of the Institute of Molecular and Clinical Ophthalmology Basel (IOB) in Switzerland and a Co-Founder of GenSight. We have worked on optogenetic therapy in the lab for 16 years and now seeing the proof of concept in a patient is a unique experience, he said. I am most grateful to have shared this long journey with Jos Sahel, a fellow founder of GenSight; the dedicated team at GenSight; and our other collaborators.

The subject in the case report, who had been diagnosed with RP 40 years prior to enrollment, had such low visual acuity that prior to receiving GS030, he could only perceive light. His gene therapy injection was followed four and a half months later by training on the use of the GS030-MD device. Seven months after the start of his training, he began to report signs of visual improvement. Visual function tests showed he acquired the ability to perceive, locate, count and touch objects when his treated eye was stimulated with the GS030-MD goggles. Without the goggles, he could not perform the tasks.

While the patient performed vision-oriented tasks, recordings were taken using extracranial multi-channel electroencephalography (EEG), a non-invasive technique that provides a readout of neuronal activity across the cortex. The EEG signals suggest that the act of carrying out the visual perception tests was accompanied by neurophysiological activity in the visual cortex.

In addition, the patient also reported significant improvements in his ability to conduct day-to-day activities such as navigating in outdoor and indoor environments and detecting household objects and furniture.

Watching a patient benefit for the first time from this trial using optogenetics to treat blindness has been a uniquely rewarding experience, commented Dr. Jos-Alain Sahel, MD, PhD, lead and co-corresponding author, Co-Founder of GenSight, and Founder of the Institut de la Vision (Sorbonne-Universit/Inserm/CNRS), Paris, France. Dr. Sahel is also Director of Institut Hospitalo-Universitaire FOReSIGHT, Paris, France, and Distinguished Professor and Chairman of the Department of Ophthalmology at the University of Pittsburgh School of Medicine and UPMC (University of Pittsburgh Medical Center), USA. He added, Being able to take part in bringing this new scientific approach to the clinic reflects the long-term collaboration with Botond Roska, the scientists of the Vision Institute, our clinicians, the Streetlab and psychophysics teams, and GenSight.

A video of the patient performing the tests, which was submitted as supplementary material to Nature Medicine, can be viewed at http://www.gensight-biologics.com.

Key Opinion Leader Webcast: June 4, 2021 at 2:00 PM CEST/8:00 AM EDT

Dr. Sahel and Dr. Roska will discuss the case report on a KOL webcast dedicated to Optogenetics and GS030 and hosted by GenSight Biologics.

Details will be announced at a later date.

Context

RP is the leading cause of inherited blindness and is caused by mutations in more than 71 different genes.a By using gene therapy to induce light sensitivity in unaffected retinal ganglion cells, GS030 overcomes the challenge among genetics-based treatments of exclusively addressing a specific underlying mutation and thus offers a treatment that is independent of the underlying pathogenic mutation.

PIONEER is the Phase I/II first-in-human, multi-center, open-label dose-escalation clinical trial to evaluate the safety and tolerability of GS030 in subjects with late-stage RP. A total of 12 to 18 subjects are planned to be enrolled. Three cohorts with three subjects each will be administered an increasing dose of GS030-DP via a single intravitreal injection in their worse-seeing eye. An extension cohort will receive the highest tolerated dose. A Data Safety Monitoring Board (DSMB) reviews the safety data of all treated subjects in each cohort and makes recommendations before the next cohort is enrolled. The primary outcome analysis will be the safety and tolerability at one year post-injection.

In line with the PIONEER protocol, the subject received the lowest dose (5.0E10 vector genomes) of GS030-DP in his worse-seeing eye. Four and a half months after injection, the patient began systematic training at Streetlab, a specialized visual rehabilitation facility, to learn how to use the light-stimulating goggles. The timing of the training was based on the estimated time it takes for the expression of light-sensitive opsin to stabilize in foveal ganglion cells.

Highlights of Visual Function Findings from Case Report

In the first visual test, the subject was asked to perceive, locate, and touch a single object placed in front of him on a white table. The subject had no success without the goggles. When the subjects treated eye was stimulated by the GS030-MD goggles, his ability to perceive, locate, and touch an object depended on the size of the object, with a significantly higher rate of successful trials with a large object (a notebook; 92%) than with the smaller object (a staple box; 36%). The success rate was similar for objects at different contrasts, suggesting that even objects at lower contrasts generated enough retinal activity for perception. Finally, the success rate was similar for the different tasks of perceiving, locating, and touching, suggesting that once the object was perceived, the patient could coordinate his motor system with the percept.

The second visual test required the subject to perceive, count, and locate two or three tumblers of different contrasts placed in front of him on a white table. As in the first test, the subject had no success without the goggles. When the subjects treated eye was stimulated by the GS030-MD goggles, the patient perceived, correctly counted, and located the objects in the majority (58-63%) of the trials. As in the first test, the success rate was similar for objects of different contrasts.

In the third visual test, the patient had to assess the presence or absence of a tumbler on a white table. The success rate with the goggles stimulating the treated eyes was statistically significantly higher than without the goggles (41% vs. 6%; p < 0.001).

Highlights of Safety Findings from Case Report

In-depth ocular examinations were performed regularly before and after injection, and potential intraocular inflammation was monitored according to international guidelines of the Standardization of Uveitis Nomenclature (SUN) Working Group.b Both eyes of the subject showed no intraocular inflammation and no changes in the anatomy of the retina; there were no ocular or systemic adverse events over the 84 weeks of assessment.

The subject tested the light-stimulating goggles three times before being injected with the gene therapy. On each of these occasions, he reported no change of vision or photophobia.

Detailed findings can be found at https://www.nature.com/articles/s41591-021-01351-4.

*About the paper:

Partial recovery of visual function in a blind patient after optogenetic therapy

Authors:

Jos-Alain Sahel1,2,3,4, Elise Boulanger-Scemama3,4, Chlo Pagot5, Angelo Arleo1, Francesco Galluppi6, Joseph N Martel2, Simona Degli Esposti7, Alexandre Delaux1, Jean-Baptiste de Saint Aubert1, Caroline de Montleau5, Emmanuel Gutman5, Isabelle Audo1,3, Jens Duebel1, Serge Picaud1, Deniz Dalkara1, Laure Blouin6, Magali Taiel6, Botond Roska8,9

Affiliations:

1 Sorbonne Universit, INSERM, CNRS, Institut de la Vision, Paris, France

2 Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, USA

3 INSERM-Centre d'Investigation Clinique 1423, Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris, France

4 Dpartement d'Ophtalmologie, Fondation Ophtalmologique Rothschild, Paris, France

5 Streetlab, Institut de la Vision, Paris, France

6 GenSight Biologics, Paris, France

7 NIHR Moorfields Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, United Kingdom

8 Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland

9 Department of Ophthalmology, University of Basel, Basel, Switzerland

References:

About GenSight Biologics

GenSight Biologics S.A. is a clinical-stage biopharma company focused on developing and commercializing innovative gene therapies for retinal neurodegenerative diseases and central nervous system disorders. GenSight Biologics pipeline leverages two core technology platforms, the Mitochondrial Targeting Sequence (MTS) and optogenetics, to help preserve or restore vision in patients suffering from blinding retinal diseases. GenSight Biologics lead product candidate, LUMEVOQ (GS010; lenadogene nolparvovec), has been submitted for marketing approval in Europe for the treatment of Leber Hereditary Optic Neuropathy (LHON), a rare mitochondrial disease affecting primarily teens and young adults that leads to irreversible blindness. Using its gene therapy-based approach, GenSight Biologics product candidates are designed to be administered in a single treatment to each eye by intravitreal injection to offer patients a sustainable functional visual recovery.

About GS030

GS030 leverages GenSight Biololgics optogenetics technology platform, a novel approach to restore vision in blind patients using a combination of ocular gene therapy and tailored light-activation of treated retinal cells. The gene therapy, which is delivered via a single intravitreal injection, introduces a gene encoding for a light-sensitive protein (ChrimsonR-tdT) into retinal ganglion cells, making them responsive to light and bypassing photoreceptors killed off by diseases such as retinitis pigmentosa (RP). Because ChrimsonR-tdT is activated by high intensities of amber light, a wearable medical device is needed to stimulate the treated retina. The optronic lightstimulating goggles (GS030-MD) encode the visual scene in real-time and project a light beam with a specific wavelength and intensity onto the treated retina. Treatment with GS030 requires patients to wear the external wearable device in order to enable restoration of their visual function. With the support of the Institut de la Vision in Paris and the team of Dr. Botond Roska at the Friedrich Miescher Institute in Basel, GenSight is investigating GS030 as therapy to restore vision in patients suffering from late-stage RP. GenSights optogenetics approach is independent of the specific genetic mutations causing blindness and has potential applications in other diseases of the retina in which photoreceptors degenerate, like dry agerelated macular degeneration (dry-AMD). GS030 has been granted Orphan Drug Designation in the United States and Europe.

About Optogenetics

Optogenetics is a biological technique that involves the transfer of a gene encoding for a light sensitive protein to cause neuronal cells to respond to light stimulation. As a neuromodulation method, it can be used to modify or control the activities of individual neurons in living tissue and even in-vivo, with a very high spatial and temporal resolution. Optogenetics combines (1) the use of gene therapy methods to transfer a gene into target neurons with (2) the use of optics and electronics (optronics) to deliver the light to the transduced cells. Optogenetics holds clinical promise in the field of vision impairment or degenerative neurological disorders.

About Retinitis Pigmentosa

Retinitis pigmentosa (RP) is a family of orphan genetic diseases caused by multiple mutations in numerous genes involved in the visual cycle. Over 100 genetic defects have been implicated. RP patients generally begin experiencing vision loss in their young adult years, with progression to blindness by age 40. RP is the most widespread hereditary cause of blindness in developed nations, with a prevalence of about 1.5 million people throughout the world. In Europe and the United States, about 350,000 to 400,000 patients suffer from RP, and every year between 15,000 and 20,000 new patients with RP lose sight. There is currently no curative treatment for RP.

About the PIONEER Phase I/II trial

PIONEER is a first-in-man, multi-center, open label dose-escalation study to evaluate the safety and tolerability of GS030 in 12-18 subjects with late-stage retinitis pigmentosa. GS030 combines a gene therapy (GS030-DP) administered via a single intravitreal injection with a wearable optronic visual stimulation device (GS030-MD). Eligible patients in the first three cohorts are those affected by end-stage non-syndromic RP with no light perception (NLP) or light perception (LP) levels of visual acuity. The extension cohort will include patients with hand motion (HM) and counting fingers (CF) levels of visual acuity.

As per protocol, three cohorts with three subjects each will be administered an increasing dose of GS030-DP via a single intravitreal injection in their worse-seeing eye. An extension cohort will receive the highest tolerated dose. The DSMB will review the safety data of all treated subjects in each cohort and will make recommendations before a new cohort receives the next dose. The primary outcome analyses will be on the safety and tolerability at one year post-injection. PIONEER is being conducted in three centers in the United Kingdom, France and the United States.

See the original post here:
GenSight Biologics Announces Nature Medicine Case Report Showing Visual Recovery after GS030 Optogenetic Treatment - Business Wire

Decibel Therapeutics Expands World-Class Scientific Advisory BoardAdds leaders with expertise in hearing loss and balance disorders, combined with…

BOSTON, May 24, 2021 (GLOBE NEWSWIRE) -- Decibel Therapeutics (Nasdaq: DBTX), a clinical-stage biotechnology company dedicated to discovering and developing transformative treatments to restore and improve hearing and balance, today announced the appointment of leading experts in gene therapy, cochlear development and molecular genetics to its Scientific Advisory Board (SAB).

These additions to our SAB bring a deep understanding of the genetic roots of hearing loss and balance disorders, and we look forward to working closely with them as we progress our pipeline of gene therapies and regenerative medicines, said Joe Burns, Ph.D., Vice President, Discovery.

We value their insights and tremendous experience in the translational considerations for the development of gene therapies as Decibel works to address the vast unmet needs in hearing and balance disorders, said John Lee, Chief Development Officer.

Career summaries of the Scientific Advisory Board appointees:

Connie Cepko, Ph.D., is the Bullard Professor of Genetics and Neuroscience at Harvard Medical School and a Howard Hughes Medical Institute Investigator. She trained in virology with Dr. Phillip Sharp at MIT for a Ph.D., and later with Dr. Richard Mulligan at the MIT Whitehead Institute. She helped develop retroviral vectors for transduction into the central nervous system (CNS) for lineage analysis and for studies of gene function in vivo. Her laboratory has focused on the mechanisms of cell fate determination in the CNS with focus on retina through the analysis of progenitor and stem cells. More recently, she has been studying the mechanisms of photoreceptor death in diseases that cause blindness, such as retinitis pigmentosa and macular degeneration, and is developing gene therapies to avert photoreceptor death in order to prevent vision loss.

GuangpingGao, Ph.D.,is an internationally recognized researcher who played a key role in the discovery and characterization of a new family of adeno-associated virus (AAV) serotypes to advance the gene therapy field. He has published extensively in the field, with more than 300 papers, and holds more than 191 patents, with hundreds more pending. The Penelope Booth Rockwell Professor in Biomedical Research at the University of Massachusetts Medical School, Dr. Gao is an elected fellow of both the U.S. National Academy of Inventors and the American Academy of Microbiology. He is the Past President of the American Society of Gene and Cell Therapy. Dr. Gao co-founded Voyager Therapeutics, Adrenas Therapeutics and Aspa Therapeutics to develop AAV-based gene therapies for rare diseases.

Matthew Kelley, Ph.D., directs the Laboratory of Cochlear Development in the Intramural Program at the National Institute on Deafness and Other Communication Disorders, National Institutes of Health. A widely published and well-respected research scientist, he focuses on the cellular and molecular development of the mammalian cochlea. Dr. Kelley has long been an active member and is the past President of the Association for Research in Otolaryngology (ARO).

Glenn Pierce, M.D., Ph.D.,is Entrepreneur-in-Residence at Third Rock Ventures. He has 30 years of experience in drug discovery and developmentwith a particular focus on tissue regeneration, gene therapy and hematologyand has contributed to the development of six marketed products.As the former Chief Medical Officer, Hemophilia Therapeutic Area at Biogen, he led work culminating in multiple regulatory approvals for hemophilia therapeutics. Dr. Pierce has served in multiple leadership roles for the National Hemophilia Foundation as well as on advisory boards for the U.S. Food and Drug Administration and the U.S. Department of Health and Human Services. He has co-authored more than 150 scientific papers and has received more than 15 patents. Dr. Pierce serves on the Boards of Directors of Global Blood Therapeutics and Voyager Therapeutics.

Dinah Sah, Ph.D., is an accomplished drug developer and R&D leader with over 25 years of experience in research and drug development in the biotechnology industry. Most recently, she was Chief Scientific Officer at Voyager Therapeutics, joining soon after its start in 2014. Prior to Voyager, Dr. Sah was Vice President of Research at Alnylam, overseeing many of the research programs during her seven-year tenure. She has successfully led multiple research and preclinical programs toward and into clinical development across new modalities, including the CNS-focused AAV programs at Voyager and the groundbreaking novel class of RNAi therapeutics developed at Alnylam.

About Decibel TherapeuticsDecibel Therapeutics is a clinical-stage biotechnology company dedicated to discovering and developing transformative treatments to restore and improve hearing and balance, one of the largest areas of unmet need in medicine. Decibel has built a proprietary platform that integrates single-cell genomics and bioinformatic analyses, precision gene therapy technologies and expertise in inner ear biology. Decibel is leveraging its platform to advance gene therapies designed to selectively replace genes for the treatment of congenital, monogenic hearing loss and to regenerate inner ear hair cells for the treatment of acquired hearing and balance disorders. Decibels pipeline, including its lead gene therapy program, DB-OTO, to treat congenital, monogenic hearing loss, is designed to deliver on our vision of a world in which the privileges of hearing and balance are available to all. For more information about Decibel Therapeutics, please visitwww.decibeltx.comor follow us onTwitter.

Investor Contact:Julie SeidelStern Investor Relations, Inc.julie.seidel@sternir.com212-362-1200

Media Contact:Chris RaileyTen Bridge CommunicationsChris@tenbridgecommunications.com617-834-0936

Excerpt from:
Decibel Therapeutics Expands World-Class Scientific Advisory BoardAdds leaders with expertise in hearing loss and balance disorders, combined with...

Chutes & LaddersAnother Novartis exec answers the biotech call, this time for gene therapy startup Tevard – FierceBiotech

Welcome to this week's Chutes & Ladders, our roundup of hirings, firings and retirings throughout the industry. Please send the good wordor the badfrom your shop to Fraiser Kansteiner, and we will feature it here at the end of each week.

TevardBiosciencesGopi Shanker, Ph.D., lands at the startup as chief scientific officer.

Shanker's departure marks something of a trend for Novartis. In late April,Jeff Engelman, M.D., Ph.D.,left the Novartis Institutes for Biomedical Research (NIBR) to stand up the new oncology biotechTreelineBiosciences. Just a few days later, fellow Novartis cancer R&D execPeter Hammerman, M.D., Ph.D., leftto start as chief scientific officer of molecular machines biotech MOMA Therapeutics.Shanker, for his part, will take up the post ofchief scientific officer at gene therapy startup TevardBiosciences. After heading upneuroscience atNIBR, Shanker will nowlead the preclinical scientific programs at the transfer RNA-based gene therapystartup.A few months back, Tevard forged a pact withZogenix to seek out and work on gene therapies for Dravet syndrome, a rare form of epilepsy, and other genetic epilepsies.Fierce Biotech

Blueprint MedicinesPercy Carter, Ph.D., takes up the helm of chief scientific officer.

Carter is in good company at Blueprint, where he joins former Bristol Myers Squibb compatriotFouad Namouni, M.D., who became president of R&D last fall. Carter, who joinedBristol Myers Squibb in 2001and eventually rose through the ranks tosenior vice president and head of discovery, willspearhead research and preclinical development as the new chief scientific officer at Blueprint. More recently, Carter held posts atJohnson & Johnson, where he was global head of discovery sciences in the healthcare giant's Janssen unit, andFibroGen, where he was in the CSO seat for less than a year. He joins Blueprint a little over a year after the company's first approval. In January 2020,Ayvakit,kinase inhibitor, became the first targeted treatmentfor patients with a type of stomach cancer called gastrointestinal stromal tumor who carry a (PDGFR) exon 18 mutation.Fierce Biotech

IovanceBiotherapeuticsMariaFardis, Ph.D.,is hitting the exit.

Fardis, who's also Iovance's president and a director, alertedthe company on Tuesday that she wasresigning topursue other opportunities," Iovance quietly revealed in a Wednesday securities filing. Her exit coincides withadditional data requests on potency assays for Iovance'slead candidate lifileucelfrom the FDA.Iovancewill continue validating the potency assays and expects to submit further data and meet with the FDA in the second half of 2021.The setback pushesIovancesbiologics license application submission into the first half of 2022,the company said. The "optics" of Fardis departure are "challenging" and raise "questions about whether something else is behind the change or whether the Board chose to make the change, Jefferies analysts wrote to clients this week. Long term,we think the drug works [] and it will eventually get to market," the Jefferies team added.IovancesBoard will appoint general counsel Frederick Vogt, Ph.D., as interim president and CEO. The company will immediately kick off the huntfor a successor.Fierce Biotech

>Passage Bio has bidadieu to chief medical officer Gary Romano, M.D., Ph.D. The gene therapy startup and Romanomutually agreed they should part waysTuesday, and Romano hit the exit that same day.Chief Financial Officer Richard Morris is also leaving the company on May 30, but Passage Bio described the situation differently: It said Morris' departure was not the result of any disagreement regarding any matter relating to the Companys operations, financial statements, internal controls, auditors, policies, or practices. Passage didn't say whether Romano's departure was the resultof a disagreement.Fierce Biotech

>Merck KGaA has slottedChris Round into the role ofpresident at its North America healthcare unit, replacing Andrew Patterson, who'll step into the newly minted role of chief marketing officer.EMD Serono is hoping to cash in with three promising drugsits Pfizer-partnered immuno-oncology medBavencio, multiple sclerosis treatment Mavenclad and Tepmetko for non-small cell lung cancer.Round will be based in Rockland, Massachusetts. He served for 20 years at Merck & Co. before arriving at Merck KGaA in 2017 to spearheadcommercial operations in Asia, the Middle East and Africa.Fierce Pharma

>Clinical Ink, a global clinical trial technology firm, has summoned Janette Morgan as EVP, general manager, Europe, the Middle East and Africa. Morgan joins after a 13-year run at Medidata, where she most recently served as vice president, global partner initiatives & partner business management. In that role, she was in charge ofglobal leadership and management of Medidatas Partner Study-by-Study business.Release

>Xenter, hot on the heels of last week's $12 million series A, has enlistedScott Heuler as senior vice president of global sales and marketing, whileGary Baldwin signs onas chief technology officer in Xenter'shospital technologies business unit.Baldwin previously led R&D, engineering and product teams for multiple organizations. Heuler, for his part, has held seniorsales and marketing leadership roles with NeuroOne Medical Technologies, Guidant Corporation and U.S. Surgical Corporation, Xenter said.Release

> Clinical-stage immuno-oncology companyMedicenna has tapped Mann Muhsin, M.D., as chief medical officer. With more than 20 years experience in medical practice and drug development, Muhsin signs on with "an outstanding track record of innovation in oncology and immuno-oncology trial design," Medicenna said in a release. Muhsin kicked off his clinical research career atPICR phase I unit, where he conducted more than 17 clinical trials for international sponsors likeAstraZeneca,Hoffmann La Roche, Merck, Novartis, Eli Lilly, Johnson & Johnsonand Bayer. He then went on to lead early clinical development programs at Johnson & Johnson's Janssen unit.Release

>CDMO and clinical supply services company Experic is putting David Wood in charge as chairman and chief executive officer. Over a 30-year run in the industry, Wood has lead organizations and teams atCaptek Softgel International, Catalent Pharma Solutions, Cardinal Health PTS and Becton Dickinson. He's taking the reins from Jeffrey McMullen, one of Experic's founders and its former chairman and CEO. McMullen took on aninterim CEOrole in 2019 after the untimely passing of the executive previously managing the company.Release

>NeuBase Therapeutics, looking to push its first drug candidate into the clinic next year, will welcomeSandra Rojas-Caro, M.D., as chief medical officer onMay 24. At NeuBase, she'll be in charge of thepreclinical and clinical development, medical, and regulatory strategy of NeuBases pipeline. She most recently worked at Gemini Therapeutics, where she held the same post. Before that, she was CMO atAeglea BioTherapeutics.Release

>Rain Therapeutics, working on precision oncology therapeutics, has lifted co-founderRobert Doebele, M.D., Ph.D., into the role ofpresident and chief scientific officer. Doebele co-founded Rain with chairman and CEOAvanish Vellanki in 2017. Before he joined the company full-time in October 2020, Doebele served asassociate professor of medicine in the division of medical oncology at the University of Colorado School of Medicine, director of the Thoracic Oncology Research Initiative at the University of Colorado Cancer Center and as principal investigator for the University of Colorado Lung Cancer Specialized Program of Research Excellence.Release

> Precision oncology firm Fore Biotherapeutics is building out its leadership team with the addition ofStacie Peacock Shepherd, M.D., Ph.D., as chief medical officer and Nora Brennan as chief financial officer. Shepherd will oversee clinical strategy and operational advancement of Fore's lead precision oncology program, FORE8394, and she will also helpexpand the company's clinical pipeline in the coming months. Brennan, for her part, previously served as CFO atTELA Bio, and held the same role at Xeris Pharmaceuticals before that.Release

>Cybrexa Therapeutics has summonedStephen Basso as its chief financial officer. Before joining the Cybrexa team, Basso served as senior vice president of finance atInozyme Pharma. He also served as vice president, North America commercial finance, global G&A at Alexion Pharmaceuticals, and as director, financial planning and analysis at Pfizer.Release

>Celularity, developing off-the-shelf cell therapiesderived from the postpartum human placenta, has signed onBradley Glover, Ph.D., as EVP and chief technology officer. At Celularity, Glover will oversee all aspects of the company's technical operations, includingprocess development, quality, manufacturing, supply chain, IT, facilities and engineering, and more. He joins the team from Kite Pharma, where he wore a number of executive hats in corporate development and technical operations.He rose through the ranks at Genentech and Roche before that.Release

> Early-stage allogeneic cell therapy company Appia Bio has enlisted Qi Wei, Ph.D. as senior vice president and head of technical operations. Wei is bringingsome serious cell and gene therapy know-how to the fold. He's had stints atNovartis, Gilead's Kite, Eli Lilly, TCR2 Therapeutics and Geneception.He had a hand in the development of several CAR-Ts, including Novartis' Kymriah and Kite and Gilead's Tecartus.Release

>Aerovate Therapeutics, working on meds for patients with rare cardiopulmonary disease, has handed the CEO torch toTimothy Noyes. Noyes was also elected to the company's board of directors in April. He previously served as president and chief executive ofProteon Therapeutics. He previously served aschief operating officer of Trine Pharmaceuticals and held the position of president of Genzymes renal division following its acquisition of GelTex Pharmaceuticals. Noyes started his industry run at Merck & Co.Release

> Biopharma product and portfolio strategy planner Prescient has appointed John Crowley as director of the company's intelligence and insight business, whileSugandh Sharma enters the role of senior director in the same business unit. Before joining the fold at Prescient, Crowleyled analyst teams at Decision Resources Group, covering rare diseases in neurology, immunology and hematology, as well as infectious diseases.Release

>Cognito Therapeutics, developing a new class ofdisease-modifying digital therapeutics to treat neurodegenerative disorders likeAlzheimers disease,tapped Everett Crosland as its chief commercial officer. He recently held the same role at AppliedVR, where he was in charge ofnational launch sales, marketingand payer strategy on the first breakthrough designated virtual reality therapeutic. Prior to that, he was VP of market access & reimbursement atPear Therapeutics.Release

>Umoja Biopharma, developing an integrated, in vivo immunotherapy platform, has signed onIrena Melnikova, Ph.D., as chief financial officer. She comes over from SVB Leerink, where she was a managing director in investment banking. Before that, shewas a managing director at Burrill & Company and, prior to that, director of strategy and external innovation at Sanofi.Release

>ONI has enlistedTyler Ralston, Ph.D., as its chief technology officer. In his role, Ralston willprovide leadership, strategic vision and oversee the research and development of ONI's technology. He signs on from the 4Catalyzer incubator, where he most recently served as CTO for Tesseract. Release

> Quanterix, working ondigital protein biomarker technology to enableprecision health, tappedMasoud Toloueas president of Quanterix and Diagnostics. He'll enter the role on June 9. He joins the team fromPerkinElmer, where he most recently served as senior vice president, diagnostics. He alsofounded and led Bioo Scientifics next generation sequencing business, which was acquired by PerkinElmer in 2016.Release

> Gene therapy firm AavantiBio has named Jessie Hanrahan, Ph.D., as Chief Regulatory Officer. She's the fifth senior executive to joins AavantiBio's leadership team in recent months and will be in charge ofglobal regulatory affairs for AavantiBios diversified pipeline of gene therapy programs targeting rare diseases with significant unmet medical need.Most recently, she was vice president of regulatory science at bluebird bio.Release

Go here to read the rest:
Chutes & LaddersAnother Novartis exec answers the biotech call, this time for gene therapy startup Tevard - FierceBiotech

Prevalence of Plasmodium falciparum isolates lacking the histidine rich protein 2 gene among symptomatic malaria patients in Kwilu Province of the…

Study design and setting

We used secondary data from a prospective health facility-based cross-sectional study conducted on individuals of all ages, seeking healthcare from October to December 2018 in 34 randomly selected health facilities of three health zones in the Kwilu Province (D.R. Congo), Fig.1.

The Kwilu Province is one of 26 provinces of D.R. Congo with an area of 79 906 km2. It is divided into five administrative territories: Bagata (including the city of Bandundu), Bulungu (including the city of Kikwit), Gungu, Idiofa, and Masimanimba [31].

The two selected cities (Bandundu and Kikwit) include three of the 24 health zones of the Kwilu Province [31]. They are the two main cities in the province and bear the highest burden of malaria. pfhrp2 gene deletions were previously reported in this region [14, 32].

Bandundu, the capital city of the Kwilu Province, is located 400km from Kinshasa, the capital of D.R. Congo [33]. Bandundu covers an area of 222 km2 with a population estimated at 950 683 as of 2015 [33]. It has a tropical wet and dry climate with two seasons. Heavy rainfalls and constant heat characterize the rainy season while fewer rainfalls are recorded during the dry season. The average annual temperature is 26.9C [33]. Bandundu City has one semi-urban health zone of the same name and 17 health areas, including 11 urban and six rural.

Kikwit is the second-largest city in Kwilu Province, located in the south-west of D.R. Congo, at 525km from Kinshasa and 400km from Bandundu. It is the main economic city of the province and a commercial hub that provides access to diamond-rich regions of Kasa Province and Angola. Kikwit covers an area of 92 km2 with an estimated population of 1 326 068 as of 2016 [34]. The city has a tropical wet and dry climate with a long rainy season from early September through to the end of May and a short dry season from early June to the end of August. Kikwit City has two urban health zones: Kikwit-Nord and Kikwit-Sud.

The study was approved by the Kwilu Province Division of Health (N ADM/707/01/DPS-KLU/CD/JPBK/632/2018), the Kinshasa School of Public Health Ethical Committee (ESP/CE/015/2019) and the School of Tropical Medicine and Global Health Ethical Review Committee.

The study was first explained to all participants, then written and verbal voluntary informed consent was obtained from all study participants including guardian/parents of non-adult participants.

The study population included individuals of all ages seeking health care in health facilities located in the three Health Zones of Bandundu (one) and Kikwit (two) cities. Health facilities included General Reference Hospitals, Reference Health Centres, and Health Centres. The smallest selection units were individuals attending these health facilities with symptoms suggestive of malaria. The study included all individuals seeking care in the selected health facilities with symptoms suggestive of malaria such as fever, headaches, malaise; during the study period for whom a laboratory test (PfHRP2-RDT and/or microscopic examination) was performed. Individuals who failed to meet the inclusion criteria or did not consent to participate in the study were excluded.

The minimum number of subjects required to enrol in this study was calculated based on a previously reported proportion of pfhrp2 gene deletion in the Kwilu Province (3%) and recommendations from WHO for studies on pfhrp2/3 deletion among symptomatic patients [14, 35]. According to the WHO protocol for estimating pfhrp2/3 deletion prevalence, for an expected prevalence of 3.2%, at least 370 individuals with P. falciparum infection are required per sampling domain [35]. In this study, the sampling domain was the Kwilu province, which included 34 health facilities. The study enrolled a total of 684 patients meeting the inclusion criteria of which 491 were positive for P. falciparum using PCR.

The primary study applied a two-stage random sampling to select health centres. At stage one, 27 health centres were randomly selected among the 62 health centres in the targeted areas. For neighbouring health centres, one health centre was randomly selected out of two. In order to increase the chance of catching individuals not respecting the referral system by directly seeking care in high-level health facilities, four reference health centres and three general reference hospitals from the three health zones were included, bringing the total number of selected health facilities to 34 (27 in Kikwit and seven in Bandundu).

At stage two, individuals attending the selected health facilities with symptoms indicative of malaria were recruited. The lead investigator weighed the number of individuals to recruit per health centre to the average rate of service utilization provided by the National Health Information System.

This study used four groups of variables: sociodemographic, malaria prevention, clinical and biological variables. Plasmodium falciparum HRP2 gene deletion (pfhrp2) was the main outcome variable. Explanatory variables were age, sex, health zones, household size, existence of mosquito breeding sites, LLIN (Long Lasting Insecticidal Net) ownership, use of LLIN, malaria drug intake, malaria clinical features, parasite density, and microscopy result.

Potential participants were introduced to the study by a research assistant. After securing consent/assent from the subjects or their guardians, socio-demographic, malaria prevention and treatment practices, and clinical variables were collected using a pre-tested structured questionnaire. Patients medical records were used to collect data from the physicians or health officers clinical examination.

Heel or finger-prick blood was collected from each individual. Samples for microscopy were prepared using two drops of blood. Then 50l of blood were applied on PfHRP2-RDT, and a few drops were spotted onto Whatman filter paper to prepare dried blood spots (DBS).

The membranes of spent PfHRP2-RDT cassettes and the DBSs were individually stored in plastic bags, sealed with a desiccant at room temperature before being shipped to the Institute of Tropical Medicine in Nagasaki (NEKKEN) where they were refrigerated at 4C.

The CareStart Malaria Pf (HRP2) Ag RDT (Access Bio, Inc., Somerset, New Jersey, USA) was used for the qualitative detection of malaria histidine-rich protein 2 in the whole blood according to the manufacturers instructions [ACCESSBIO, 2018, Somerset, New Jersey, USA]).

The test membrane strip is pre-coated with a P. falciparum HRP2 specific monoclonal antibody as a single line across the test strip. The reported panel detection score is 91.0% at 200 parasites/l with a false positive rate of 0.9% [36, 37]

A team of four medical technologists read the slides in the laboratories of health facilities where samples were collected. When a health facility did not have the necessary equipment to perform the examination, slides were read at the nearest laboratory possessing adequate equipment. For quality assurance, one expert microscopist randomly selected positive and negative slides to cross-check results. In the case results were not concordant, another reading was performed. About five percent of slides went through another quality control in the vector control laboratory of the Kinshasa School of Public Health.

Thick and thin smears were made on the same slide. The part of the slide containing the thin smear was fixed with methanol and dried. Then the whole slide was stained with 10% Giemsas solution for ten minutes and finally washed off with distilled water and air-dried. Stained smears were examined under a microscope for malaria parasite identification. For positive slides, malaria parasites were counted against 200 white blood cells (WBC), and parasite density was calculated based on a total of 8000 WBC/l using the following formula: (Number of Parasites counted8000)/Number of counted WBC.

Parasite density calculation was immediately performed when 100 parasites were counted against 200 WBC. However, in the case that fewer than 100 parasites were counted against 200 WBC, the count continued until 500 WBC.

Genomic DNA was extracted from membranes of spent PfHRP2-RDT cassettes and DBS using the QIAGEN QIAmpDNA extraction kit (company, city, country) according to the manufacturers instructions. We also adapted a previously described method to recover DNA from 197 spent RDTs membranes from Bandundu Health Zone [38].

To confirm P. falciparum infection, we designed specific primers targeting a 226 base pair region of the P. falciparum lactate dehydrogenase (pfldh) gene and performed a real-time PCR assay (Table 1). This assay was also used to ensure there was sufficient parasite DNA quantity in the samples to discriminate P. falciparum negative samples from samples with pfhrp2 gene deletion, as shown in Fig.2.

pfhrp2 gene deletion testing pipeline

Samples were duplicated and loaded in 96-wells plates along with serially diluted positive controls (gDNA from in vitro cultured P. falciparum strain 3D7) (1ng/l, 0.1ng/l, 0.01ng/l, 0.001ng/l), as well as negative controls containing DNA from blood spots prepared from known malaria negative individuals. We repeated the assay for all discordant duplicates and counted three consistent results as the final result.

For detection of the pfhrp2 gene, we performed a nested PCR assay using primers targeting a 228 base pair fragment spanning exon 1, the intron, and a portion of exon 2 of pfhrp2 as previously described (Table 1) [9]. We used a lower elongation temperature (68C) to improve PCR sensitivity, pfhrp2 being AT-rich, and increased the number of cycles to 40. We used genomic DNA from Dd2 (pfhrp2 negative) and 3D7 (pfhrp2 positive) as controls for all assays.

We repeated the nested PCR for all negative results. In the case of discordant results, we performed the amplification a third time and counted two consistent results as the final result.

Reaction components for both real-time and nested PCR are summarized in Table 1.

PCR amplicons were separated by electrophoresis on a 2% agarose gel stained with Gel Red Nucleic Acid Stain 10 000in water. A total of 12l of PCR amplicons (6l) and loading dye (6l) were loaded onto the gel, which was run for 35min at 100V and observed under UV light. A 500g/ml Gene Ruler 100bp DNA Ladder (BioLabsinc, Ipswich, USA) was loaded onto the same gel to determine the sizes of the resolved fragments.

Data were entered and analyzed using STATA15 (StataCorp LLC, Lakeway, Texas, USA). Tables have been used to describe categorical variables. Continuous variables have been summarized using median and interquartile ranges. Proportions have been used to summarize categorical variables.

Fischers exact test (for categorical variables) and the KruskalWallis test (for non-normally distributed continuous variables) were applied to look for associations between potential explanatory variables and the main outcome (pfhrp2 gene deletion). We computed the 95% Confidence Interval (95% CI) for the prevalence of pfhrp2 gene deletion. We considered a P-value less than 0.05 statistically significant.

Read more:
Prevalence of Plasmodium falciparum isolates lacking the histidine rich protein 2 gene among symptomatic malaria patients in Kwilu Province of the...

Companies Unlocking a New Class of Cancer Therapies with Sensitive METTL3 Tuning – BioSpace

University of Cambridge researchers published the first preclinical validation for a novel therapeutic strategy targeting epitranscriptomic modifiers of RNA, opening the door for spinout STORM Therapeutics acute myeloid leukemia therapy (AML)and potentially much more.

Tony Kouzarides, cofounder and director of Milner Therapeutics Institute at Cambridge and a Storm co-founder, led the team that published the paper in Nature in April, which was the first published data showing the efficacy of a catalytic METTL3 inhibitor in mouse models of AML.

Consistent with earlier findings on METTL3s role in regulating disease, Storms STM2457 decreased AML engraftment and increased survival. Last year the company nominated a related compound as its lead clinical candidate, and Kouzarides said Storm expects it will reach clinical testing early next year.

This is the first published in vivo data for a small molecule inhibiting an epitranscriptomic disease target, but the approach has been brewing for years. Just as epigenetic modifications to DNA can directly control gene expression, epitranscriptomic modifications control RNA gene expression indirectly via RNA translation, making them potential therapeutic targets. While the first epigenetic therapy was approved in 2004, the connection between epitranscriptomic modifiers and disease pathways have only begun to be characterized.

METTL3 is an epigenetic writer of N6-methyladenosine (m6A) modifications, the most prevalent reversable epitranscriptomic modification in human cells. METTL3 and other writers form m6A modifications via methylation of certain RNA sequences, which is key for numerous processes in normal cells. This would seem to make it an unlikely therapeutic target.

But in 2017, two key publications began to elucidate the role of m6A in AML and demonstrate the possibility of targeting the pathway without toxicity in healthy cells. Kouzarides and colleagues published findings showing the role of METTL3 in both establishing disease and in leukemia cell differentiation.

Separately, a team from Weill Cornell Medicine and Memorial Sloan Kettering Cancer Center showed in a Nature Medicine paper that leukemia cells were more dependent on METTL3 than normal hematopoietic cells. Samie Jaffrey, an author on the paper and a professor of pharmacology at Weill Cornell, said that the abnormal differentiation seen in leukemia cells was abrogated by only mild inhibition of METTL3important because a complete METTL3 inhibitor delivered systemically would be highly toxic.

The new paper from the Cambridge researchers was therefore a proof-of-concept for Jaffreys approach as well.

I think a lot of other people were cautious because they felt it was a fools errand to go after METTL3, he said. I think this makes the METTL3 target seem much more validated.

Researchers from both groups founded companies to exploit the therapeutic pathway in leukemia. Kouzarides and fellow Cambridge professor Eric Miska co-founded Storm Therapeutics in 2015 to develop first-in-class therapies against RNA epigenetic targets. Jaffrey co-founded Gotham Therapeutics in 2017. Storm has raised 42 million ($59.4 million) seed and Series A rounds, while Gotham raised $54 million in a Series A.

At least one other company appears to be chasing METTL3: Accent Therapeutics, which last year raised a $63 million B round and announced a co-development deal with AstraZeneca utilizing its RNA-modifying protein platform, has presented in vitro data in AML for its inhibitors of METTL3 and METTL14, another m6A writer.

Additional publications have shown more therapeutic possibilities for targeting the m6A pathway, suggesting it may be possible to increase sensitivity to existing therapies in pancreatic and ovarian cancer, for example. m6A methylation has also been linked to a suppressed immune response in infectious diseases like Zika and HIV, and in April a team led by University of California San Diego School of Medicine professor Tariq Rana published in vitro data showing METTL3 inhibition could suppress SARS-CoV-2 replication.

Ranas team has previously shown that the inhibition of ALKBH5, an m6A eraser, can improve the efficacy of cancer immunotherapies in mice, and Rana has spun out ViRx Pharmaceuticals with plans to license intellectual property from the university around RNA therapeutics for broad-spectrum antivirals.

But Storm, Accent and Gotham are starting in AML for a reason. Optimizing compounds for AML is an easier task because unlike other cancers, when you inhibit the METTL3 pathway, you can see the cells differentiating with your eyes, Jaffrey said. In most cancers, you can inhibit METTL3 and they will die but youre inhibiting it to such a degree that probably normal cells will die too. it all comes down to one thing: therapeutic index.

Gotham has identified several solid tumor types with comparable m6A addictions it is looking at closely, he added.

Several other companies are likely targeting different epitranscriptomic targets. Academic work by an Accent scientific cofounder, Chuan He, has focused on YTHDF1, an RNA reader that could function as an immune system control switch for modulating the response to cancer immunotherapy. He is also a chemistry professor at the University of Chicago. Accent is also pursuing ADAR1 inhibitors for cancer.

Silicon Therapeutics, which uses a physics-based approach to drug design for intractable protein targets, is developing an ADAR1 antagonist for cancer. The company said inhibiting ADAR1 can both activate innate antitumor immune cells and directly kill tumors.

Gotham is also looking into additional epitranscriptomic regulators that are much less prevalent than m6A, Jaffrey says, using its screening platform to identify and analyze RNA modifiers.

Kouzarides envisions a lot more opportunities ahead for the burgeoning space. These enzymes can potentially regulate many different processes in many different diseases, he said. He declined to specify which other epitranscriptomic targets Storm is chasing, but the company is currently raising a Series B round to support its anticipated clinical testing for a METTL3 inhibitor in 2022.

Link:
Companies Unlocking a New Class of Cancer Therapies with Sensitive METTL3 Tuning - BioSpace

ARUP Sequencing Specimens Positive for SARS-CoV-2 to Detect the UK Variant and Other Possible Strains of the Virus – PRNewswire

SALT LAKE CITY, Jan. 19, 2021 /PRNewswire/ --ARUP Laboratories, a major provider of COVID-19 molecular diagnostic testing in Utah and nationwide, today announced that it is sequencing variants of SARS-CoV-2 in specimens positive for COVID-19 to enable detection of the more contagious "United Kingdom (UK) variant" of the virus.

ARUP's announcement came after the Utah Department of Health (UDOH) on Jan. 15, 2021, said that a Salt Lake County man had been found to have the so-called "UK," or B.1.1.7 variant, which so far has been detected in 20 states. The first known case of the variant in Utah, it was identified by the UDOH Public Health Laboratory, which since early in the pandemic has been sequencing a percentage of specimens positive for COVID-19 as part of an effort in which ARUP is collaborating.

Independently, ARUP has begun sequencing all specimens identified as positive for SARS-CoV-2 through molecular testing on one of three platforms it uses to test for the virus. ARUP, which offers a full menu of COVID-19 testing for hospital and health system clients nationwide, does not release sequencing variant information for individual patients. Rather, sequencing is performed for epidemiologic purposes and to monitor and, if necessary, improve the performance of the tests used to diagnose COVID-19.

"Our sequencing research seeks to characterize not just the UK variant, but other circulating strains of SARS-CoV-2, so that we may continue to develop accurate tests for the virus," said Adam Barker, PhD, director of the ARUP Institute for Clinical and Experimental Pathology.

The UK variant is known to contain a mutation of the spike protein, or S gene, which one of ARUP's tests uses as a target for SARS-CoV-2 identification, along with two other genes. Read more about ARUP's tests and about how ARUP is sequencing variants here.

Genetic sequencing of SARS-CoV-2 variants for research and epidemiologic purposes is a good example of ARUP's unique ability to offer vital testing while also contributing to scientific advancement, thanks to its affiliation with the University of Utah (U of U). ARUP is a not-for-profit enterprise of the U of U School of Medicine and its Department of Pathology, and Barker and all ARUP medical directors are also U of U faculty members.

"Our work to help advance laboratory medicine and improve patient care is at the heart of what we do at ARUP," said CEO Sherrie L. Perkins, MD, PhD. "As this public health crisis persists, our commitment is stronger than ever."

More information about ARUP's COVID-19 test menu and about other COVID-19-related research in which ARUP is involved is available at aruplab.com.

About ARUP Laboratories

Founded in 1984, ARUP Laboratories is a leading national reference laboratory and a nonprofit enterprise of the University of Utah and its Department of Pathology. ARUP offers more than 3,000 tests and test combinations, ranging from routine screening tests to esoteric molecular and genetic assays. ARUP serves clients across the United States, including many of the nation's top university teaching hospitals and children's hospitals, as well as multihospital groups, major commercial laboratories, group purchasing organizations, military and other government facilities, and major clinics. In addition, ARUP is a worldwide leader in innovative laboratory research and development, led by the efforts of the ARUP Institute for Clinical and Experimental Pathology. ARUP is ISO 15189 CAP accredited.

Media ContactARUP: Lisa Carricaburu,[emailprotected],801-541-5041

SOURCE ARUP Laboratories

http://www.aruplab.com

Visit link:
ARUP Sequencing Specimens Positive for SARS-CoV-2 to Detect the UK Variant and Other Possible Strains of the Virus - PRNewswire

Cell and Gene Therapies Shatter Prior Records, with Continued Growth Expected – BioSpace

Cell and gene therapies achieved record growth in 2020, surpassing prior high points in terms of financings and approvals. Janet Lambert, CEO of the Alliance for Regenerative Medicine (ARM), painted a rosy picture despite a few challenges at the 2021 Cell & Gene State of the Industry Briefing during this years Biotech Showcase, held virtually January 11-15.

2020 was a record-shattering year for financing, with $19.9 billion raised in 2020, up from the $9.8 billion raised in 2019 and the $13.3 billion raised in 2018 the previous record, she said.

Follow-on financings, at $6.8 billion, accounted for the largest portion of funding, followed by venture capital at $5.6 billion and IPOs at $3.7 billion. Upfront payments from partnerships totaled $3 billion and private placements totaled $1.2 billion.

The number of large financings exceeding $200 million grew noticeably. Sana Bios $700 million private financing, announced last June, topped the field. Iovance and bluebird bio gained the most from follow-on financing, with fundings of $604 million and $575 million, respectively. Legend Bio led the list of IPOs, raising $487 million last May.

Large pharma continued to buy-in to biotechs for research, development, commercialization, and licensing agreements focuses largely on oncology and CNS disorders, Lambert said. She cited Sangamo, with two major collaborations with Biogen ($350 million) and Novartis ($75 million), though adding, Bayer is especially active.

In the stock market, Regenerative medicine companies outperformed the NASDAQ Biotech Index, she continued. Although stock prices for virtually everything plummeted in mid-March, biotech stocks rebounded. Cell based immuno-oncology (IO) share prices increased 80% from their January 1 levels. Gene therapy was close behind, with a 70% increase, and all publicly traded regenerative medicine companies saw a 50% increase.

Given the overall environment, it seems safe to predict that 2021 will be another good year for regenerative therapies.

Currently, there are approximately 1,100 gene, cell and tissue-based therapeutic developers throughout the world. Of those, the overwhelming majority are in North America, with 543. We saw a lot of growth in China, Japan, and Korea, she said, with 295 companies. Europe boasts 209.

Globally, 1,220 clinical trials are underway for regenerative medicine.

More than 100 clinical trials commended in the fourth quarter alone, Lambert said. Roughly, they are divided evenly among cell, gene, and IO. With 152 trials in phase III and nearly half of those in gene therapy, this offers strong support for predictions by both the FDA and EMA that there will be 10 to 20 advanced therapies approved each year through 2025.

What people often forget, Lambert said, quoting Amy Price, a mother of two gene therapy recipients, is that cell and gene therapies arent some fantastical futuristic thing. Two of the Price children received gene therapy in a clinical trial 10 years ago for metachromatic leukodystrophy (MLD), a historically fatal disease. That drug, Libmeldy, by Orchard Therapeutics, was approved in Europe in 2020, making it one of the most significant milestones of the year.

The benefits of cell and gene therapy have expanded beyond experimental treatments.

Patients are continuing to benefit from innovative therapies, Lambert said. We saw a significant number of gene therapy approvals in 2020. In addition to Libmeldy, she cited approvals of Zolgensma (by Novartis Gene Therapies) in Europe, Japan, and Canada; Tecartus (by Kite, a Gilead company) in the U.S.; and Luxturna (by Spark/Roche) in Canada.

Looking forward, oncology, and particularly IO, dominates the regenerative medicine landscape. Some 554 oncology trials are underway.

Investors have invested heavily in this space for some time, and IO comprises 50% of Phase I trials in cell and gene therapy, Lambert noted. Focus is increasing on allogeneic therapies as well as gene editing.

Central nervous system therapeutics are the second most popular therapeutic indication for regenerative therapies for the second year in a row, with 94 trials. 2020 saw promising data from the first-ever attempt iPSC therapy for Parkinsons disease.

Gene editing continues to advance in the clinic, she added. For the first time, a patient was treated with CRISPR therapy in vivo and, later, systemically with CRISPR. CRISPR Therapeutics and Vertex Pharmaceuticals shared data (during J.P. Morgan week) from a sickle cell trial of 20 patients.

Despite these scientific advances and investor enthusiasm, cell and gene therapies face challenges in terms of dosing and delivery, and chemistry, manufacturing and controls (CMC), Lambert admitted. Gene therapy represents almost half of the Phase III pipeline, so we expect to learn a lot quite soon.

While it goes without saying that 2020 was a challenging year, many of the repercussions of the COVID-19-related disruptions remain to be seen. They extend not only to operational and clinical disruptions but also to regulatory backlogs and the politicization of diagnostics and therapeutics, all set against the usual challenges of fast-moving science.

None-the-less, Lambert pointed out, There were some positive developments.

As she said, Medicare approved a new diagnosis related group (DRG) for CAR T cell therapy and promulgated a new rule for outcomes-based therapies, thus enabling a new payment model that ARM deems essential for cell and gene therapies. In Europe, the European Commissions pharmaceutical strategy now recognizes the importance of cell and gene therapy. We are encouraged that we can build on that starting point with the Commission, Lambert said. ARM also is expecting progress on n-of-one therapies for ultra-orphan indications this year.

Looking ahead to 2021, Lambert identified six regenerative therapies on the FDAs docket from Mallinckrodt, bluebird, BMS, PTC Bio, and Gensight Bio. All indications are that 2021 will be a fantastic year of scientific, technological, and clinical progress in this sector, Lambert predicted.

See the rest here:
Cell and Gene Therapies Shatter Prior Records, with Continued Growth Expected - BioSpace

NIH awards UT Southwestern researchers $4.4 million to study the genetic basis of vocal learning – UT Southwestern

DALLAS Oct. 5, 2021 A UT Southwestern research team has received the National Institutes of Healths prestigious Transformative Research Award to further their study of zebra finches to investigate the genetic basis of vocal imitation abilities.

Todd Roberts, Ph.D.

The award grants $4.4 million over five years to Todd Roberts, Ph.D., Associate Professor of Neuroscience, Joseph Takahashi, Ph.D., Professor and Chair of Neuroscience, and Kent Hamra, Ph.D., a Senior Research Associate in Obstetrics and Gynecology. Drs. Roberts and Takahashi are members of the Peter ODonnell Jr. Brain Institute.

TheTransformative Research Award is part of nearly$9 millionin prestigious NIH Directors Awards received by UT Southwestern researchers today from the NIH Common Funds High-Risk, High-Reward Program, which supports scientists pursuing highly innovative research with the potential to have a broad impact on biomedical, behavioral, or social sciences.

Joseph Takahashi, Ph.D.

Zebra finches are a vocal learning species that provide the only practical platform for systematically identifying the genes involved in this important social behavior. Like speech, zebra finch song is a culturally transmitted behavior learned via imitation, said Dr. Roberts, the principal investigator on this award. We think a forward genetic screen for mutations that affect vocal imitation, followed by the detailed genetic mapping and manipulations developed through this proposal, will identify genetic signatures for this polygenic trait.

The scientists are seeking to establish the first mutagenesis screen in a vocal learning species and the genetic tools to independently test the function of identified genes by developing novel transgenic zebra finches using germline gene targeting technologies. The research may shed new light on speech and language deficits associated with autism spectrum disorder.

Kent Hamra, Ph.D.

Previous research by Dr. Roberts, published in Science Advances, found that inactivating a gene closely associated with autism prevents songbirds from replicating their fathers songs.

UTSouthwestern ratesNo. 1 among global institutions in the health care sectorin the2021NatureIndexfor its published research, as well as among thetop 20 U.S. institutions overall for published research in life sciences journals.

Dr. Roberts is the Thomas O. Hicks Scholar in Medical Research. Dr. Takahashi holds the Loyd B. Sands Distinguished Chair in Neuroscience.

About UTSouthwestern Medical Center

UT Southwestern, one of the nations premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institutions faculty has received six Nobel Prizes, and includes 25 members of the National Academy of Sciences, 16 members of the National Academy of Medicine, and 14 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 2,800 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UTSouthwestern physicians provide care in about 80 specialties to more than 117,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 3 million outpatient visits a year.

Read the original here:
NIH awards UT Southwestern researchers $4.4 million to study the genetic basis of vocal learning - UT Southwestern

M6P Therapeutics to Host Key Opinion Leader Webinar on Lysosomal Storage Disorders – Business Wire

ST. LOUIS--(BUSINESS WIRE)--M6P Therapeutics (M6PT or the Company), a privately held life sciences company developing next-generation recombinant enzyme and gene therapies for lysosomal storage disorders (LSDs), today announced that it will host a key opinion leader (KOL) webinar on LSDs on Wednesday, July 28, 2021 at 10:00 a.m. ET.

The webinar will feature a fireside chat with KOLs Gregory Enns, M.D., Lucile Salter Packard Childrens Hospital Stanford School of Medicine, and Mark S. Sands, Ph.D., Departments of Medicine and Genetics at Washington University School of Medicine, who will discuss the current treatment landscape and unmet medical needs in LSDs, including Gaucher disease, Fabry disease, Pompe disease, mucopolysaccharidoses, and mucolipidoses. LSDs are a family of approximately 50 rare, genetic, and life-threatening diseases characterized by a deficiency in a specific lysosomal enzyme.

The event will also feature an update from the M6PT management team on its recombinant enzyme and gene therapy S1S3 bicistronic technology platform for the treatment of LSDs. The Company plans to initiate its first clinical program in 2022.

Dr. Enns, Dr. Sands, and M6PT management will also take questions from the audience.

To register for the webinar, please click here.

Dr. Enns is a Professor of Pediatrics and Genetics at the Lucile Salter Packard Childrens Hospital Stanford School of Medicine. He completed his medical education at the University of Glasgow (1990) in Scotland and completed his residency at the Children's Hospital Los Angeles Pediatric Residency in California. He then went on to complete his fellowship at the UCSF Medical Center in California. He is board certified in Clinical Genetics and Genomics. Dr. Enns research interests include novel means of diagnosing and treating mitochondrial disorders, with an emphasis on antioxidant therapy, lysosomal disorders, and newborn screening by tandem mass spectrometry. His current pursuits include the analysis of glutathione and antioxidant status in patients who have mitochondrial disorders and the development of new techniques for diagnosing and treating these conditions.

Dr. Sands is a Professor in the Departments of Medicine and Genetics at Washington University School of Medicine in St. Louis. Dr. Sands received his Ph.D. in Molecular Pharmacology from the State University of New York at Stony Brook. He was a postdoctoral fellow at The Jackson Laboratory (Bar Harbor, ME) and at the University of Pennsylvania School of Veterinary Medicine before joining the faculty at Washington University School of Medicine. The goals of Dr. Sands laboratory are to better understand the underlying pathogenesis and developing effective therapies for inherited childhood diseases, specifically LSDs. A major focus of his group is to determine the safety and efficacy of adeno-associated viral gene transfer vectors for the treatment of both the central nervous system (CNS) and systemic manifestations of these diseases. In addition, his group has developed lentiviral-mediated hematopoietic stem cell-directed gene therapy approaches, as well as small molecule drugs, and more recently rational combinations of these approaches. The primary diseases that Dr. Sands studies are mucopolysaccharidosis type VII (MPS VII), Krabbe disease, and Infantile Neuronal Ceroid Lipofuscinosis.

About M6P Therapeutics

M6P Therapeutics is a privately held, venture-backed biotechnology company developing the next-generation of targeted recombinant enzyme and gene therapies for lysosomal storage disorders (LSDs). M6P Therapeutics proprietary S1S3 bicistronic platform has the unique ability to enhance phosphorylation of lysosomal enzymes for both recombinant enzyme and gene therapies, leading to improved biodistribution and cellular uptake of recombinant proteins and efficient cross-correction of gene therapy product. This can potentially lead to more efficacious treatments with lower therapy burden, as well as new therapies for currently untreated diseases. M6P Therapeutics team, proven in rare diseases drug development and commercialization, is dedicated to fulfilling the promise of recombinant enzyme and gene therapies by harnessing the power of protein phosphorylation using its S1S3 bicistronic platform. M6P Therapeutics mission is to translate advanced science into best-in-class therapies that address unmet needs within the LSD community. For more information, please visit: http://www.m6ptherapeutics.com.

See the article here:
M6P Therapeutics to Host Key Opinion Leader Webinar on Lysosomal Storage Disorders - Business Wire

Cancer gene sequencing and an unexpected transmission – Health Report – ABC News

The New England Journal of Medicine has a potentially disturbing report from Japan of two little boys, one aged two, the other aged six, diagnosed with lung cancer (they are not related) whose mothers had cervical cancer. Genome sequencing found that the tumours in the kids, although they seemed very different from the mothers, were genetically related to their mum's tumour. The assumption was that cancer cells had spread to the boys in the physical process of being born.

Dr Alison Brand is director of gynaecological oncology at Westmead Hospital in Sydney. Welcome to the Health Report, Alison.

Alison Brand: Thanks for having me.

Norman Swan: So have you ever heard of this before?

Alison Brand: No, I had not heard of this before this and, I have to say, it's virtually unheard of, and the New England Journal of Medicine, which is the peak journal in medicine to publish a case report. So you know if they've published this case report they have really examined the data very closely to verify that it is true. And I think we have to do believe some of it because it has been published in the New England Journal of Medicine.

Norman Swan: Let me just divert from the core story here, which is about cervical cancer moving to the kids and getting into their lungs. You're a gynaecological surgeon, you do a lot of surgery, what's the story with gynaecological seeding and surgery? There has been a lot of discussion about that, whether in fact you can spread cancer with surgery itself.

Alison Brand: The old wives' tale was that once you open up the belly and let the air in, then the cancer just spreads terribly, and of course we know that's not true. That really came from the fact that when patients had operations many years ago, they found cancer but then they couldn't do much about it, there was no chemotherapy or there was no radiotherapy. So really this whole business of surgery spreading cancer is probably not true at all. We do know that cancer from the mum can sometimes cross the placenta and get into babies, but that's usually haematologic malignancies.

Norman Swan: So it's leukaemia.

Alison Brand: Leukaemia, that's right, and otherand basically the babies then have widespread disease because it got into their bloodstream and then went throughout the body. These particular two case reports are really unusual because it doesn't look like it came transplacentally or through the placenta, it looks like it came as the baby has passed through the birth canal, and landed in the lung, which is the kind of closest place that the babies could breathe in some of the cells that were in the vagina as they pass through the birth canal.

Norman Swan: And because of that you wouldn't think it was human papilloma virus related, which is the cause of cervical cancer, because it was the actual tumour itself that got transmitted.

Alison Brand: Yes, that's right, although we haven't often thought that cancer is catching. I think that's the one thing that we've said; you can't touch someone who has cancer and then catch it, and in many ways you catch lots of viruses, and women can pass their HPV infections in some rare cases to their babies. And so this is unusual in that the cancer has really been caught from the mother and that's highly unusual.

I guess when we look at this we have to look atas we examine any reports, we have to say is this biologically plausible, and I guess in rare cases it is biologically plausible, although you mostly expect that the tumour cells on the top of tumours are really those ones that are often non-viable or not living and therefore can't attach to something and grow there. But I think that the next generation sequencing that they have done here really suggests to us that maybe there is some truth to all of this, albeit rare, rare, rare.

Norman Swan: Is it routine to screen for cervical cancer in pregnancy?

Alison Brand: It is routine that patients should have had a recent screen prior to their pregnancy, and if they haven't, to have one done during pregnancy. What you have to remember though, Norman, is certainly the mother of the first patient had had a normal cervical screen seven months prior to delivering her baby, and it's important because she had a very rare neuroendocrine tumour, so a very rare type of cervical tumour that probably wouldn't have been picked up by screening anyway. But those are very rare tumours, and the vast majority of cervical cancers can be picked up by screening, and certainly are much better picked up by the new screening test that we have that looks at HPV presence.

Norman Swan: And before we go, just tell usbecause the screening program has changed, it now happens every five years if I remember rightly, and you are checking for HPV. So, just give us a very brief outline of the screening program now.

Alison Brand: So it used to be that we looked at the cells on the cervix to check to see whether or not they had precancerous changes. That had up to a 30% false negative rate, and therefore we had to screen more often to make sure that we didn't miss anything. Now we check by looking at what we call high risk HPV virus, which is human papilloma virus, which is known to cause cervical cancer, and we check for that high risk HPV, and because the test is so sensitive, then if there is a negative test, we only need to do the test every five years. And I think the take-home message here for women who are pregnant is that we shouldn't worry so much about giving your baby cancer from you, what we should really worry about is making sure that we prevent cancer in the first place by having regular screening and, if eligible, making sure your boys and girls have vaccinations.

Norman Swan: Alison, thank you for joining us.

Alison Brand: Thank you.

Norman Swan: Dr Alison Brand is director of gynaecological oncology at Westmead Hospital in Sydney.

See original here:
Cancer gene sequencing and an unexpected transmission - Health Report - ABC News

Induction of muscle-regenerative multipotent stem cells from human adipocytes by PDGF-AB and 5-azacytidine – Science Advances

Abstract

Terminally differentiated murine osteocytes and adipocytes can be reprogrammed using platelet-derived growth factorAB and 5-azacytidine into multipotent stem cells with stromal cell characteristics. We have now optimized culture conditions to reprogram human adipocytes into induced multipotent stem (iMS) cells and characterized their molecular and functional properties. Although the basal transcriptomes of adipocyte-derived iMS cells and adipose tissuederived mesenchymal stem cells were similar, there were changes in histone modifications and CpG methylation at cis-regulatory regions consistent with an epigenetic landscape that was primed for tissue development and differentiation. In a non-specific tissue injury xenograft model, iMS cells contributed directly to muscle, bone, cartilage, and blood vessels, with no evidence of teratogenic potential. In a cardiotoxin muscle injury model, iMS cells contributed specifically to satellite cells and myofibers without ectopic tissue formation. Together, human adipocytederived iMS cells regenerate tissues in a context-dependent manner without ectopic or neoplastic growth.

The goal of regenerative medicine is to restore function by reconstituting dysfunctional tissues. Most tissues have a reservoir of tissue-resident stem cells with restricted cell fates suited to the regeneration of the tissue in which they reside (14). The innate regenerative capacity of a tissue is broadly related to the basal rate of tissue turnover, the health of resident stem cells, and the hostility of the local environment. Bone marrow transplants and tissue grafts are frequently used in clinical practice but for most tissues, harvesting and expanding stem and progenitor cells are currently not a viable option (5, 6). Given these constraints, research efforts have been focused on converting terminally differentiated cells into pluripotent or lineage-restricted stem cells (7, 8). However, tissues are often a complex mix of diverse cell types that are derived from distinct stem cells. Therefore, multipotent stem cells may have advantages over tissue-specific stem cells. To be of use in regenerative medicine, these cells would need to respond appropriately to regional cues and participate in context-dependent tissue regeneration without forming ectopic tissues or teratomas. Mesenchymal stem cells (MSCs) were thought to have some of these characteristics (911), but despite numerous ongoing clinical trials, evidence for their direct contribution to new tissue formation in humans is sparse, either due to the lack of sufficient means to trace cell fate in hosts in vivo or failure of these cells to regenerate tissues (12, 13).

We previously reported a method by which primary terminally differentiated somatic cells could be converted into multipotent stem cells, which we termed as induced multipotent stem (iMS) cells (14). These cells were generated by transiently culturing primary mouse osteocytes in medium supplemented with azacitidine (AZA; 2 days) and platelet-derived growth factorAB (PDGF-AB; 8 days). Although the precise mechanisms by which these agents promoted cell conversion was unclear, the net effect was reduced DNA methylation at the OCT4 promoter and reexpression of pluripotency factors (OCT4, KLF4, SOX2, c-MYC, SSEA-1, and NANOG) in 2 to 4% of treated osteocytes. iMS cells resembled MSCs with comparable morphology, cell surface phenotype, colony-forming unit fibroblast (CFU-F), long-term growth, clonogenicity, and multilineage in vitro differentiation potential. iMS cells also contributed directly to in vivo tissue regeneration and did so in a context-dependent manner without forming teratomas. In proof-of-principle experiments, we also showed that primary mouse and human adipocytes could be converted into long-term repopulating CFU-Fs by this method using a suitably modified protocol (14).

AZA, one of the agents used in this protocol, is a cytidine nucleoside analog and a DNA hypomethylating agent that is routinely used in clinical practice for patients with higher-risk myelodysplastic syndrome (MDS) and for elderly patients with acute myeloid leukemia (AML) who are intolerant to intensive chemotherapy (15, 16). AZA is incorporated primarily into RNA, disrupting transcription and protein synthesis. However, 10 to 35% of drug is incorporated into DNA resulting in the entrapment and depletion of DNA methyltransferases and suppression of DNA methylation (17). Although the relationship between DNA hypomethylation and therapeutic efficacy in MDS/AML is unclear, AZA is known to induce an interferon response and apoptosis in proliferating cells (1820). PDGF-AB, the other critical reprogramming agent, is one of five PDGF isoforms (PDGF-AA, PDGF-AB, PDGF-BB, PDGF-CC, and PDGF-DD), which bind to one of two PDGF receptors (PDGFR and PDGFR) (21). PDGF isoforms are potent mitogens for mesenchymal cells, and recombinant human (rh)PDGF-BB is used as an osteoinductive agent in the clinic (22). PDGF-AB binds preferentially to PDGFR and induces PDGFR- homodimers or PDGFR- heterodimers. These are activated by autophosphorylation to create docking sites for a variety of downstream signaling molecules (23). Although we have previously demonstrated induction of CFU-Fs from human adipocytes using PDGF-AB/AZA (14), the molecular changes, which underlie conversion, and the multilineage differentiation potential and in vivo regenerative capacity of the converted cells have not been determined.

Here, we report an optimized PDGF-AB/AZA treatment protocol that was used to convert primary human adipocytes, a tissue source that is easily accessible and requires minimal manipulation, from adult donors aged 27 to 66 years into iMS cells with long-term repopulating capacity and multilineage differentiation potential. We also report the molecular landscape of these human iMS cells along with that of MSCs derived from matched adipose tissues and the comparative in vivo regenerative and teratogenic potential of these cells in mouse xenograft models.

Primary mature human adipocytes were harvested from subcutaneous fat (Fig. 1A and table S1) and their purity confirmed by flow cytometry with specific attention to the absence of contaminating adipose-derived MSCs (AdMSCs) (fig. S1, A and B). As previously described (14), plastic adherent adipocytes were cultured in Alpha Minimum Essential Medium (MEM) containing rhPDGF-AB (200 ng/ml) and 20% autologous serum (AS) with and without 10 M AZA for 2 and 23 days, respectively (Fig. 1A). During daily observations, unilocular lipid globules were observed to fragment within adipocytes ~day 10 with progressive extrusion of fat into culture medium, coincident with changes in cell morphology (movie S1). Consistent with these observations, when fixed and stained with Oil Red O, adipocytes that were globular in shape at the start of culture resembled lipid laden stromal cells at day 12 and lipid-free stromal cells at day 25 (Fig. 1B).

(A) Generation and reprogramming of adipocytes. (B) Oil Red Ostained adipocytes (days 0, 12, and 25) during treatment with recombinant human platelet-derived growth factorAB (rhPDGF-AB) and AZA. (C) Flow cytometry plots of LipidTOX and PDGFR in adipocytes cultured as in (A). (D) CFU-F counts from treated and untreated adipocytes during conversion. (E) CFU-F counts from adipocytes treated (Rx) with indicated combinations of rhPDGF-AB, AZA, fetal calf serum (FCS), autologous serum (AS), or serum-free media (SFM). (F) CFU-F counts from adipocytes reprogrammed in the presence of 0, 1, or 10 M PDGFR/ inhibitor AG1296. (G) CFU-F counts per 400 reprogrammed adipocytes from three donor age groups (n = 3 for each) generated using indicated combinations of rhPDGF-AB and AZA. (H) Long-term growth of reprogrammed adipocytes from three donor age groups (n = 3 for each) generated using indicated combinations of rhPDGF-AB and AZA. (I) Long-term growth of iMS cells cultured in SFM or media supplemented with FCS, autologous, or allogeneic serum. Error bars indicate SD, n = 3; *P < 0.05, **P < 0.01, and ***P < 0.0001 calculated using either a Students t test (E and F) or a linear mixed model (H). Photo credit: Avani Yeola, UNSW Sydney.

To evaluate these changes in individual cells, we performed flow cytometry at multiple time points during treatment and probed for adipocyte (LipidTOX) (24) and stromal cell characteristics [PDGFR expression (25); Fig. 1C]. A subpopulation of adipocytes, when cultured in media supplemented with PDGF-AB/AZA and AS (Fig. 1C, top; treated), showed reduced LipidTOX staining intensity at day 10, with progressive reduction and complete absence in all cells by day 19. Adipocytes cultured in the absence of PDGF-AB/AZA retained LipidTOX staining, albeit with reduced intensity (Fig. 1C, bottom; untreated). Adipocytes expressed PDGFR [fig. S1C, (i) and (ii)] but not PDGFR (Fig. 1C) at day 0 but both the frequency and intensity of PDGFR staining increased from day 21. To record these changes in real time, we also continuously live-imaged treated adipocytes from days 15 to 25 and recorded the extrusion of fat globules, change in cell morphology from globular to stromal, and acquisition of cell motility and cell mitosis (movie S1 and fig. S1D). Intracellular fragmentation of fat globules was observed over time in untreated adipocytes (fig. S1E), consistent with variable LipidTOX staining intensity. CFU-F capacity was absent at day 10, present in day 15 cultures, and tripled by day 19 with no substantial increase at days 21, 23, and 25 (Fig. 1D). It is noteworthy that CFU-F potential was acquired before PDGFRA surface expression when adipocytes had started to display stromal cell morphology and had diminished fat content. There was also no CFU-F capacity in adipocytes cultured in MEM with fetal calf serum (FCS) or AS, unless supplemented with both PDGF-AB and AZA. CFU-F capacity was significantly higher with AS than with FCS and absent in serum-free media (SFM) (Fig. 1E and fig. S1F). As previously shown with reprogramming of murine osteocytes, there was dose-dependent inhibition of CFU-F capacity when AG1296, a potent nonselective PDGF receptor tyrosine kinase inhibitor (26), was added to the reprogramming media (Fig. 1F).

To evaluate the impact of patient age and concentrations of PDGF-AB and AZA on the efficiency of human adipocyte conversion, we harvested subcutaneous fat from donors aged 40 (n = 3), 41 to 60 (n = 3), and 61 (n = 3) years and subjected each to three different concentrations of PDGF-AB (100, 200, and 400 ng/ml) and three different concentrations of AZA (5, 10, and 20 M) (Fig. 1G). Although all combinations supported cell conversion in all donors across the three age groups, rhPDGF-AB (400 ng/ml) and 5 M AZA yielded the highest number of CFU-Fs (Fig. 1G). When these cultures were serially passaged in SFM (with no PDGF-AB/AZA supplementation, which was used for cell conversion only), adipocytes converted with reprogramming media containing rhPDGF-AB (400 ng/ml) and 5 M AZA were sustained the longest (Fig. 1H, fig. S2A, and table S2). The growth plateau that was observed even with these cultures [i.e., adipocytes converted with rhPDGF-AB (400 ng/ml) and 5 M AZA when expanded in SFM or FCS] was overcome when cells were expanded in either autologous or allogeneic human serum (Fig. 1I). The genetic stability of human iMS cells (RM0072 and RM0073) was also assessed using single-nucleotide polymorphism arrays and shown to have a normal copy number profile at a resolution of 250 kb (fig. S2B). Together, these data identify an optimized protocol for converting human primary adipocytes from donors across different age groups and show that these can be maintained long term in culture.

Given the stromal characteristics observed in human adipocytes treated with PDGF-AB/AZA (Fig. 1), we performed flow cytometry to evaluate their expression of MSC markers CD73, CD90, CD105, and STRO1 (13) and noted expression levels comparable to AdMSCs extracted from the same subcutaneous fat harvest (Fig. 2A). Primary untreated adipocytes (day 25 in culture) did not express any of these MSC markers (fig. S3A). The global transcriptomes of iMS cells and matched AdMSCs were distinct from untreated control adipocytes but were broadly related to each other [Fig. 2B, (i) and (ii)]. Ingenuity pathway analysis (IPA) using genes that were differentially expressed between AdMSCs versus adipocytes [3307 UP/4351 DOWN in AdMSCs versus adipocytes; false discovery rate (FDR) 0.05] and iMS versus adipocytes (3311 UP/4400 DOWN in iMS versus adipocytes; FDR 0.05) showed changes associated with gene expression, posttranslational modification, and cell survival pathways and organismal survival and systems development [Fig. 2B(iii)]. The number of differentially expressed genes between iMS cells and AdMSCs was limited (2 UP/26 DOWN in iMS versus AdMSCs; FDR 0.05) and too few for confident IPA annotation. All differentially expressed genes and IPA annotations are shown in table S3 (A to E, respectively).

(A) Flow cytometry for stromal markers on AdMSCs (green) and iMS cells (purple) from matched donors. Gray, unstained controls. (B) (i) Principal components analysis (PCA) plot of adipocyte, AdMSC, and iMS transcriptomes. (ii) Hierarchical clustering of differentially expressed genes (DEGs, FDR 0.05). (iii) Ingenuity pathway analysis (IPA) of DEG between AdMSCs/adipocytes (top) or iMS cells/adipocytes (bottom). The most enriched annotated biological functions are shown. (C) (i) Chromatin immunoprecipitation sequencing (ChIP-seq) profiles in AdMSCs and iMS cells from matched donors at a representative locus. Gray bar indicates differential enrichment. (ii) Volcano plots of H3K4me3, H3K27Ac, and H3K27me3 enrichment peaks significantly UP (red) or DOWN (blue) in iMS cells versus AdMSCs. (iii) IPA of corresponding genes. log2FC, log2 fold change. (D) (i) DNA methylation at a representative locus in AdMSCs and iMS cells from matched donors. (ii) Volcano plot of regions with significantly higher (red) or lower (blue) DNA methylation in iMS cells versus AdMSCs. (iii) IPA using genes corresponding to differentially methylated regions (DMRs). (E) OCT4, NANOG, and SOX2 expression in iPS, AdMSCs, and iMS cells. Percentage of cells expressing each protein is indicated. DAPI, 4,6-diamidino-2-phenylindole. (F) AdMSCs and iMS cells differentiated in vitro. Bar graphs quantify staining frequencies, error bars show SD, n = 3. ***P < 0.001 (Students t test). Photo credit: Avani Yeola, UNSW Sydney.

In the absence of significant basal differences in the transcriptomes of AdMSCs and iMS cells, and the use of a hypomethylating agent to induce adipocyte conversion into iMS cells, we examined global enrichment profiles of histone marks associated with transcriptionally active (H3K4me3 and H3K27Ac) and inactive (H3K27me3) chromatin. There were differences in enrichment of specific histone marks in matched AdMSCs versus iMS cells at gene promoters and distal regulatory regions [Fig. 2C(i) and fig. S3, B to D]. H3K4me3, H3K27ac, and H3K27me3 enrichments were significantly higher at 255, 107, and 549 regions and significantly lower at 222, 78, and 98 regions in iMS cells versus AdMSCs [Fig. 2C(ii) and table S4, A to C] and were assigned to 237, 84, and 350 and 191, 58, and 67 genes, respectively. IPA was performed using these gene lists to identify biological functions that may be primed in iMS cells relative to AdMSCs [Fig. 2C(iii) and table S4, D to F]. Among these biological functions, annotations for molecular and cellular function (cellular movement, development, growth, and proliferation) and systems development (general; embryonic and tissue development and specific; cardiovascular, skeletal and muscular, and hematological) featured strongly and overlapped across the different epigenetic marks.

We extended these analyses to also assess global CpG methylation in matched AdMSCs and iMS cells using reduced representation bisulfite sequencing [RRBS; (27)]. Again, there were loci with differentially methylated regions (DMRs) in iMS cells versus AdMSCs [Fig. 2D(i)] with increased methylation at 158 and reduced methylation at 397 regions among all regions assessed [Fig. 2D(ii) and table S4G]. IPA of genes associated with these DMRs showed a notable overlap in annotated biological functions [Fig. 2D(iii) and table S4H] with those associated with differential H3K4me3, H3K27Ac, and H3K27me3 enrichment [Fig. 2C(iii) and table S4, E to G]. Together, these data imply that although basal transcriptomic differences between iMS cells and AdMSCs were limited, there were notable differences in epigenetic profiles at cis-regulatory regions of genes that were associated with cellular growth and systems development.

We next compared iMS cells to adipocytes from which they were derived. Expression of genes associated with adipogenesis was depleted in iMS cells (fig. S4A and table S4I). The promoter regions of these genes in iMS cells had broadly retained an active histone mark (H3K4me3), but, in contrast with adipocytes, many had acquired an inactive mark (H3K27me3) (fig. S4B and table S4J). However, there were examples where iMS cells had lost active histone marks (H3K4me3 and H3K27ac) at gene promoters and potential regulatory regions and gained repressive H3K27me3 [e.g., ADIPOQ; fig. S4C(i)]. In contrast, stromal genes had acquired active histone marks and lost repressive H3K27me3 [e.g. EPH2A; fig. S4C(ii)]. It is noteworthy that promoter regions of genes associated with muscle and pericytes (table S4K) were enriched for active histone marks in iMS cells compared with adipocytes [fig. S4D, (i) and (ii)]. We also compared demethylated CpGs in iMS cells and adipocytes (fig. S4E). There were 7366 sites in 2971 genes that were hypomethylated in iMS cells, of which 236 showed increased expression and were enriched for genes associated with tissue development and cellular growth and proliferation (fig. S4E).

PDGF-AB/AZAtreated murine osteocytes (murine iMS cells), but not bone-derived MSCs, expressed pluripotency associated genes, which were detectable by immunohistochemistry in 1 to 4% of cells (14). To evaluate expression in reprogrammed human cells, PDGF-AB/AZAtreated human adipocytes and matched AdMSCs were stained for OCT4, NANOG, and SOX2 with expression noted in 2, 0.5, and 3.5% of iMS cells respectively, but no expression was detected in AdMSCs (Fig. 2E). In addition to these transcription factors, we also evaluated surface expression of TRA-1-60 and SSEA4. Both proteins were uniformly expressed on iPSCs and absent in AdMSCs [fig. S4F(i)] and adipocytes [fig. S4F(ii)]. Although TRA-1-60 was absent in iMS cells, most (78%) expressed SSEA4 but rarely (<1%) coexpressed OCT4 and NANOG [fig. S4F(i)].

MSCs can be induced to differentiate in vitro into various cell lineages in response to specific cytokines and culture conditions. To evaluate the in vitro plasticity of human iMS cells, we induced their differentiation along with matched AdMSCs and primary adipocytes, into bone, fat, and cartilage, as well as into other mesodermal Matrigel tube-forming assays for endothelial cells (CD31) and pericytes (PDGFR) and muscle (MYH, myosin heavy chain; SMA, smooth muscle actin), endodermal (hepatocyte; HNF4, hepatocyte nuclear factor ), and neuroectodermal (TUJ1; neuron specific class III beta tubulin) lineages (Fig. 2F and fig. S4G). Whereas primary adipocytes remained as such and were resistant to transdifferentiation, iMS cells and AdMSCs showed comparable differentiation potential with the notable exception that only iMS cells generated pericyte-lined endothelial tubes in Matrigel. In keeping with these findings, relative to AdMSCs, iMS cells showed permissive epigenetic marks at pericyte genes [increased H3K4me3 and H3K27Ac; EPHA2 and MCAM; fig. S4H(i); and reduced CpG methylation; NOTCH1, SMAD7, TIMP2, AKT1, and VWF; fig. S4H(ii)]. Together with the notable differences in epigenetic profiles, these functional differences and low-level expression of pluripotency genes in iMS cell subsets suggested that these cells could be more amenable than matched AdMSCs to respond to developmental cues in vivo.

To evaluate spontaneous teratoma formation and in vivo plasticity of iMS cells, we tagged these cells and their matched AdMSCs with a dual lentiviral reporter, LeGO-iG2-Luc2 (28), that expresses both green fluorescent protein (GFP) and luciferase under the control of the cytomegalovirus promoter (Fig. 3A). To test teratoma-initiating capacity, we implanted tagged cells under the right kidney capsules of NOD Scid Gamma (NSG) mice (n = 3 per treatment group) after confirming luciferase/GFP expression in cells in culture (fig. S5, A and B). Weekly bioluminescence imaging (BLI) confirmed retention of cells in situ [Fig. 3B(i)] with progressive reduction in signal over time [Fig. 3B(ii)] and the absence of teratomas in kidneys injected with either AdMSCs or iMS cells [Fig. 3B(iii)]. Injection of equivalent numbers of iPS cells and iPS + iMS cell mixtures (1:49) to approximate iMS fraction expressing pluripotency markers led to spontaneous tumor formation in the same timeframe [Fig. 3B(iii)].

(A) Generation of luciferase/GFP-reporter AdMSCs and iMS cells, and assessment of their in vivo function. (B) Assessment of teratoma initiating capacity; (i) bioluminescence images at 0, 2, 6, and 8 weeks after implantation of 1 106 matched AdMSCs and iMS cells (P2; RM0057; n = 2 per group) under the right kidney capsules. (ii) Quantification of bioluminescence. (iii) Gross kidney morphology 8 weeks following subcapsular implantation of cells (R) or vehicle control (L). (C) Assessment of in vivo plasticity in a posterior-lateral intertransverse lumbar fusion model; (i) bioluminescence images following lumbar implantation of 1 106 matched AdMSCs or iMS cells (P2; RM0038; n = 3 per group) at 1 and 365 days after transplant. (ii) Quantification of bioluminescence. (iii) Tissues (bone, cartilage, muscle, and blood vessels) harvested at 6 months after implantation stained with (left) hematoxylin and eosin or (right) lineage-specific anti-human antibodies circles/arrows indicate regions covering GFP and lineage markerpositive cells. Corresponding graphs show donor cell (GFP+) contributions to bone, cartilage, muscle, and blood vessels as a fraction of total (DAPI+) cells in four to five serial tissue sections. Bars indicate confidence interval, n = 3. Photo Credit: Avani Yeola, UNSW Sydney.

To evaluate whether iMS cells survived and integrated with damaged tissues in vivo, we implanted transduced human iMS cells and matched AdMSCs controls into a posterior-lateral intertransverse lumbar fusion mouse model (Fig. 3A) (29). Cells were loaded into Helistat collagen sponges 24 hours before implantation into the posterior-lateral gutters adjacent to decorticated lumbar vertebrae of NSG mice (n = 9 iMS and n = 9 AdMSC). Cell retention in situ was confirmed by intraperitoneal injection of d-luciferin (150 mg/ml) followed by BLI 24 hours after cell implantation, then weekly for the first 6 weeks and monthly up to 12 months from implantation [Fig. 3C(i)]. The BLI signal gradually decreased with time but persisted at the site of implantation at 12 months, the final assessment time point [Fig. 3C(ii)]. Groups of mice (n = 3 iMS and n = 3 AdMSC) were euthanized at 3, 6, and 12 months and tissues harvested from sites of cell implantation for histology and immunohistochemistry [Fig. 3C(iii)]. Although implanted iMS cells and AdMSCs were present and viable at sites of implantation at 3 months, there was no evidence of lineage-specific gene expression in donor human cells (fig. S5C). By contrast, at 6 months after implantation, GFP+ donor iMS cells and AdMSCs were shown to contribute to new bone (BMP2), cartilage (SOX9), muscle (MYH), and endothelium (CD31) at these sites of tissue injury [Fig. 3C(iii)]. The proportion of donor cells expressing lineage-specific markers in a corresponding tissue section was significantly higher in iMS cells compared with matched AdMSCs at 6 months [Fig. 3C(iii) and table S2] as well as 12 months (fig. S5, E and D, and table S2). There was no evidence of malignant growth in any of the tissue sections or evidence of circulating implanted GFP+ iMS cells or AdMSCs (fig. S5E). Together, these data show that implanted iMS cells were not teratogenic, were retained long term at sites of implantation, and contributed to regenerating tissues in a context-dependent manner with greater efficiency than matched AdMSCs.

Although appropriate to assess in vivo plasticity and teratogenicity of implanted cells, the posterior-lateral intertransverse lumber fusion mouse model is not suited to address the question of tissue-specific differentiation and repair in vivo. To this end, we used a muscle injury model (30) where necrosis was induced by injecting 10 M cardiotoxin (CTX) into the left tibialis anterior (TA) muscle of 3-month-old female severe combined immunodeficient (SCID)/Beige mice. CTX is a myonecrotic agent that spares muscle satellite cells and is amenable to the study of skeletal muscle regeneration. At 24 hours after injury, Matrigel mixed with either 1 106 iMS cells or matched AdMSCs (or no cells as a control) was injected into the damaged TA muscle. The left (injured) and right (uninjured control) TA muscles were harvested at 1, 2, or 4 weeks after injury to assess the ability of donor cells to survive and contribute to muscle regeneration without ectopic tissue formation (Fig. 4A; cohort A). Donor human iMS cells or AdMSCs compete with resident murine muscle satellite cells to regenerate muscle, and their regenerative capacity is expected to be handicapped not only by the species barrier but also by having to undergo muscle satellite cell commitment before productive myogenesis. Recognizing this, a cohort of mice was subject to a second CTX injection, 4 weeks from the first injury/cell implantation followed by TA muscle harvest 4 weeks later (Fig. 4A; cohort B).

(A) Generation of iMS and AdMSCs and their assessment in TA muscle injury model. (B) (i) Confocal images of TA muscle stained for human CD56+ satellite cells (red) and laminin basement membrane protein (green; mouse/human). Graph shows donor hCD56+ satellite cell fraction for each treatment group. (ii) Confocal images of TA muscle harvested at 4 weeks and stained for human spectrin (red) and laminin (green; mouse/human). For each treatment, the left panel shows a tile scan of the TA muscle and the right panel a high magnification confocal image. Graph shows contribution of mouse (M), human (H), or chimeric (C) myofibers in three to five serial TA muscle sections per mouse (n = 3 mice per treatment group). (C) Confocal images of TA muscle 4 weeks following re-injury with CTX, stained for human spectrin (red) and laminin (green; mouse/human). For each treatment, left panel shows a tile scan of the TA muscle, upper right panel a low-magnification image, and lower right panel a high magnification image of the area boxed above. Graph shows contribution of mouse (M), human (H), or chimeric (C) myofibers in three to five serial TA muscle sections per mouse (n = 3 mice per treatment group). Graph bars indicate confidence interval. *P < 0.05, **P < 0.01, and ***P < 0.001 (linear mixed model). Photo credit: Avani Yeola, UNSW Sydney.

In tissue sections harvested from cohort A, donor-derived muscle satellite cells (31) [hCD56 (Thermo Fisher Scientific, MA5-11563)+; red] were evident in muscles implanted with both iMS cells and AdMSCs at each time point but were most numerous at 2 weeks after implantation [Fig. 4B(i) and fig. S6A]. The frequency of hCD56+ cells relative to total satellite cells [sublaminar 4,6-diamidino-2-phenylindolepositive (DAPI+) cells] was quantified in three to five serial sections of TA muscles per mouse in each of three mice per treatment group and was noted to be higher following the implantation of iMS cells compared with AdMSCs at all time points [week 1, 5.6% versus 2.4%; week 2, 43.3% versus 18.2%; and week 4, 30.7% versus 14.6%; Fig. 4B(i), table S2, and fig. S6A]. Donor cell contribution to regenerating muscle fibers was also assessed by measuring human spectrin (32) costaining with mouse/human laminin [(33) at 4 weeks (Fig. 4B(ii)]. At least 1000 myofibers from three to five serial sections of TA muscles for each of three mice in each treatment group were scored for human [H; hSpectrin+ (full circumference); laminin+], murine (M; mouse; hSpectrin; laminin+), or mouse/human chimeric [C; hSpectrin+ (partial circumference); laminin+] myofibers. Although none of the myofibers seen in cross section appeared to be completely human (i.e., donor-derived), both iMS cells and AdMSCs contributed to chimeric myofibers [Fig. 4B(ii)]. iMS cell implants contributed to a substantially higher proportion of chimeric fibers than AdMSC implants (57.7% versus 30.7%; table S2). In cohort B, TA muscles were allowed to regenerate following the initial CTX injection/cell implantation, and re-injured 4 weeks later with a repeat CTX injection. In these mice, although total donor cell contributions to myofibers in TA muscles harvested 4 weeks after re-injury were comparable to that observed in cohort A, there were no myofibers that appeared to be completely human (Fig. 4C). There were substantially more human myofibers following iMS cell implants than with AdMSCs (9.7% versus 5.4%; table S2). There was no evidence of ectopic tissue formation in TA muscles following implantation of either iMS cells or AdMSCs in either cohort.

To assess the physiological properties of muscles regenerated with human myofibers, we performed tetanic force contractions in extensor digitorum longus (EDL) muscles following the schema shown in Fig. 4A. Tetanic forces evoked by electrical pulses of various stimulus frequencies were not significantly different between the experimental cohorts or between the experimental cohorts and control animals [fig. S6B, (i) to (iii)]. However, when challenged with a sustained train of electrical pulses [fig. S6C(i)], the iMS group demonstrated significantly greater absolute [fig. S6C(ii)] and specific [fig. S6C(iii)] forces over a 3- to 6-s period. Together, these data showed that iMS cells had the capacity to respond appropriately to the injured environment and contribute to tissue-specific regeneration without impeding function.

We have optimized a protocol, originally designed for mouse osteocytes, to convert human primary adipocytes into iMS cells. We show that these long-term repopulating cells regenerate tissues in vivo in a context-dependent manner without generating ectopic tissues or teratomas.

PDGF-AB, AZA, and serum are indispensable ingredients in reprograming media, but the underlying reasons for their cooperativity and the observed dose-response variability between patients are not known. PDGF-AB is reported to bind and signal via PDGFR- and PDGFR- but not PDGFR- subunits (21). Mouse osteocytes and human adipocytes lack PDGFR, although surface expression was detectable as cells transition during reprogramming [mouse; day 2 of 8 (14) and human day 21 of 25]. However, these cells express PDGFR (14). Given that PDGFR inhibition attenuates iMS cell production in both mice (14) and humans, a degree of facilitated binding of PDGF-AB to PDGF- subunits or signaling through a noncanonical receptor is likely to occur, at least at the start of reprogramming. PDGF-Bcontaining homo- and heterodimers are potent mitogens that increase the pool of undifferentiated fibroblasts and preosteoblasts with rhPDGF-BB used in the clinic to promote healing of chronic ulcers and bone regeneration (34). However, the unique characteristics of PDGF-AB but not PDGF-BB or PDGF-AA that facilitate reversal and plasticity of cell identity in combination with AZA and serum (14) remain unknown.

PDGF-AB was replenished in culture throughout the reprogramming period, but AZA treatment was limited to the first 2 days for both mouse osteocyte and human adipocyte cultures. DNA replication is required for incorporation of AZA into DNA (35) and hence DNA demethylation is unlikely to be an initiating event in the conversion of terminally differentiated nonproliferating cells such as osteocytes and mature adipocytes. However, the majority of intracellular AZA is incorporated into RNA, which could directly affect the cellular transcriptome and proteome as an early event (36, 37). It is feasible that subsequent redistribution of AZA from RNA to DNA occurs when cells replicate resulting in DNA hypomethylation as a later event (38).

In the absence of serum, we could neither convert primary human adipocytes into iMS cells nor perpetuate these cells long term in culture. The efficiency of conversion and expansion was significantly higher with human versus FCS and highest with AS. The precise serum factor(s) that are required for cell conversion in conjunction with PDGF-AB and AZA are not known. The volumes of blood (~50 ml 2) and subcutaneous fat (5 g) that we harvested from donors were not limiting to generate sufficient numbers of P2 iMS cells (~10 106) for in vivo implantation and are in the range of cell numbers used in prospective clinical trials using mesenchymal precursor cells for chronic discogenic lumbar back pain (NCT02412735; 6 106) and hypoplastic left heart syndrome (NCT03079401; 20 106).

Our motivation was to optimize a protocol that could be applied to primary uncultured and easily accessible cells for downstream therapeutic applications, and adipose tissue satisfied these criteria. We have not surveyed other human cell types for their suitability for cell conversion using this protocol. It would be particularly interesting to establish whether tissue-regenerative properties of allogeneic mesenchymal precursor populations that are currently in clinical trials could be boosted by exposure to PDGF-AB/AZA. However, given that iMS cells and MSCs share stromal cell characteristics, identifying a unique set of cell surface markers that can distinguish the former is a priority that would assist in future protocol development and functional assessment of iMS cells.

Producing clinical-grade autologous cells for cell therapy is expensive and challenging requiring suitable quality control measures and certification. However, the advent of chimeric antigen receptor T cell therapy into clinical practice (39) has shown that production of a commercially viable, engineered autologous cellular product is feasible where a need exists. Although there were no apparent genotoxic events in iMS cells at P2, ex vivo expansion of cells could risk accumulation of such events and long-term follow-up of ongoing and recently concluded clinical trials using allogeneic expanded mesenchymal progenitor cells will be instructive with regard to their teratogenic potential. The biological significance of the observed expression of pluripotency-associated transcription factors in 2 to 3% of murine and human iMS cells is unknown and requires further investigation. However, their presence did not confer teratogenic potential in teratoma assays or at 12-month follow-up despite persistence of cells at the site of implantation. However, this risk cannot be completely discounted, and the clinical indications for iMS or any cell therapy require careful evaluation of need.

In regenerating muscle fibers, it was noteworthy that iMS cells appeared to follow canonical developmental pathways in generating muscle satellite cells that were retained and primed to regenerate muscle following a second muscle-specific injury. Although iMS cells were generated from adipocytes, there was no evidence of any adipose tissue generation. This supports the notion that these cells have lost their native differentiation trajectory and adopted an epigenetic state that favored response to local differentiation cues. The superior in vivo differentiation potential of iMS cells vis--vis matched AdMSCs was consistent with our data showing that despite the relatively minor transcriptomic differences between these cell types, the epigenetic state of iMS cells was better primed for systems development. Another clear distinction between iMS cells and AdMSCs was the ability of the former to produce CD31+ endothelial tube-like structures that were enveloped by PDGFR+ pericytes. An obvious therapeutic application for iMS cells in this context is vascular regeneration in the setting of critical limb ischemia to restore tissue perfusion, an area of clear unmet need (40).

An alternative to ex vivo iMS cell production and expansion is the prospect of in situ reprogramming by local subcutaneous administration of the relevant factors to directly convert subcutaneous adipocytes into iMS cells, thereby eliminating the need for ex vivo cell production. AZA is used in clinical practice and administered as a daily subcutaneous injection for up to 7 days in a 28-day cycle, with responders occasionally remaining on treatment for decades (41). Having determined the optimal dose of AZA required to convert human adipocytes into iMS cells in vitro (2 days, 5 M), the bridge to ascertaining the comparable in vivo dose would be to first measure levels of AZA incorporation in RNA/DNA following in vitro administration and match the dose of AZA to achieve comparable tissue levels in vivo. A mass spectrometrybased assay was developed to measure in vivo incorporation of AZA metabolites (AZA-MS) in RNA/DNA and is ideally suited to this application (38). The duration of AZA administration for adipocyte conversion was relatively short (i.e., 2 days), but PDGF-AB levels were maintained for 25 days. One mechanism of potentially maintaining local tissue concentrations would be to engineer growth factors to bind extra cellular matrices and be retained at the site of injection. Vascular endothelial growth factor A (VEGF-A) and PDGF-BB have recently been engineered with enhanced syndecan binding and shown to promote tissue healing (42). A comparable approach could help retain PDGF-AB at the site of injection and maintain local concentrations at the required dose. While our current data show that human adipocytederived iMS cells regenerate tissues in a context-dependent manner without ectopic or neoplastic growth, these approaches are worth considering as an alternative to an ex vivo expanded cell source in the future.

Extended methods for cell growth and differentiation assays and animal models are available in the Supplementary Materials, and antibodies used are detailed in the relevant sections.

The primary objective of this study was to optimize conditions that were free of animal products for the generation of human iMS cells from primary adipocytes and to characterize their molecular landscape and function. To this end, we harvested subcutaneous fat from donors across a broad age spectrum and used multiple dose combinations of a recombinant human growth factors and a hypomethylating agent used in the clinic and various serum types. We were particularly keen to demonstrate cell conversion and did so by live imaging and periodic flow cytometry for single-cell quantification of lipid loss and gain of stromal markers. Using our previous report generating mouse iMS cells from osteocytes and adipocytes as a reference, we first characterized the in vitro properties of human iMS cells including (i) long-term growth, (ii) colony-forming potential, (iii) in vitro differentiation, and (iv) molecular landscape. Consistent with their comparative morphology, cell surface markers, and behavioral properties, the transcriptomes (RNA sequencing) were broadly comparable between iMS cells and matched AdMSCs, leading to investigation of epigenetic differences [Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) histone chromatin immunoprecipitation sequencing (ChIP-seq), and RRBS for DNA methylation differences] that might explain properties that were unique to iMS cells (expression of pluripotency factors, generation of endothelial tubes in vitro with pericyte envelopes, and in vivo regenerative potential). Context-dependent in vivo plasticity was assessed using a tissue injury model that was designed to promote bone/cartilage/muscle/blood vessel contributions from donor cells and simultaneously assess the absence of ectopic/malignant tissue formation by these cells (labeled and tracked in vivo using a bioluminescence/fluorescence marker). Tissue-specific regeneration and the deployment of canonical developmental pathways were assessed using a specific muscle injury model, and donor cell contributions in all injury models were performed on multiple serial tissue sections in multiple mice with robust statistical analyses (see below). Power calculations were not used, samples were not excluded, and investigators were not blinded. Experiments were repeated multiple times or assessments were performed at multiple time points. Cytogenetic and Copy Number Variation (CNV) analyses were performed on iMS and AdMSCs pretransplant, and their teratogenic potential was assessed both by specific teratoma assays and long-term implantation studies.

Subcutaneous fat and blood were harvested from patients undergoing surgery at the Prince of Wales Hospital, Sydney. Patient tissue was collected in accordance with National Health and Medical Research Council (NHMRC) National Statement on Ethical Conduct in Human Research (2007) and with approval from the South Eastern Sydney Local Health District Human Research Ethics Committee (HREC 14/119). Adipocytes were harvested as described (43). Briefly, adipose tissue was minced and digested with 0.2% collagenase type 1 (Sigma-Aldrich) at 37C for 40 min and the homogenized suspension passed through a 70-m filter, inactivated with AS, and centrifuged. Primary adipocytes from the uppermost fatty layer were cultured using the ceiling culture method (44) for 8 to 10 days. AdMSCs from the stromal vascular pellet were cultured in MEM + 20% AS + penicillin (100 g/ml) and streptomycin (250 ng/ml), and 200 mM l-glutamine (complete medium).

Adherent mature adipocytes were cultured in complete medium supplemented with AZA (R&D systems; 5, 10, and 20 M; 2 days) and rhPDGF-AB (Miltenyi Biotec; 100, 200, and 400 ng/ml; 25 days) with medium changes every 3 to 4 days. For inhibitor experiments, AG1296 was added for the duration of the culture. Live imaging was performed using an IncuCyte S3 [10 0.25numerical aperture (NA) objective] or a Nikon Eclipse Ti-E (20 0.45-NA objective). Images were captured every 30min for a period of 8 days starting from day 15. Twelve-bit images were acquired with a 1280 1024 pixel array and analyzed using ImageJ software. In vitro plasticity was determined by inducing the cells to undergo differentiation into various cell types using differentiation protocols adapted from a previous report (45).

Animals were housed and bred with approval from the Animal Care and Ethics Committee, University of New South Wales (UNSW; 17/30B, 18/122B, and 18/134B). NSG (NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ) and SCID/Beige (C.B-Igh-1b/GbmsTac-Prkdcscid-Lystbg N, sourced from Charles River) strains were used as indicated. The IVIS Spectrum CT (Perkin Elmer) was used to capture bioluminescence. Briefly, 15 min after intraperitoneal injection of d-luciferin (150 mg/kg), images were acquired for 5 min and radiance (photon s1 cm2 sr1) was used for subsequent data analysis. The scanned images were analyzed using the Living Image 5.0 software (Perkin Elmer).

Teratoma assays (46) were performed on 3- to 4-month-old female NSG mice. Lentiviral-tagged cells (5 105) in 20 l of phosphate-buffered saline containing 80% Matrigel were injected under the right kidney capsule using a fine needle (26 gauges) and followed weekly by BLI until sacrifice at week 8. Both kidneys were collected, fixed in 4% paraformaldehyde (PFA) for 48 hours, embedded in optimal cutting temperature compound (OCT), cryosectioned, and imaged for GFP.

Posterior-lateral intervertebral disc injury model (29). Lentiviral-tagged (28) AdMSCs (1 106) or iMS cells were loaded onto Helistat collagen sponges and implanted into the postero-lateral gutters in the L4/5 lumbar spine region of anesthetized NSG mice following decortication of the transverse processes. Animals were imaged periodically for bioluminescence to track the presence of transplanted cells. At 3, 6, or 12 months, mice were euthanized, and spines from the thoracic to caudal vertebral region, including the pelvis, were removed whole. The specimens were fixed in 4% PFA for 48 hours, decalcified in 14% (w/v) EDTA, and embedded in OCT.

Muscle injury model (47). The left TA and EDL muscles of 3- to 4-month-old female SCID/Beige mice were injured by injection with 15 l of 10 M CTX (Latoxan). Confocal images of three to four serial sections (TA) per mouse were captured by Zen core/AxioVision (Carl Zeiss) and visualized by ImageJ with the colocalization and cell counter plugins [National Institutes of Health; (48)]. Tetanic force contractions were performed on EDL muscles (49).

Total RNA was extracted using the miRNeasy Mini Kit (Qiagen) according to manufacturers instructions, and 200 ng of total RNA was used for Illumina TruSeq library construction. Library construction and sequencing was performed by Novogene (HK) Co. Ltd. Raw paired-end reads were aligned to the reference genome (hg19) using STAR (https://github.com/alexdobin/STAR), and HTSeq (50) was used to quantify the transcriptomes using the reference refFlat database from the UCSC Table Browser (51). The resulting gene expression matrix was normalized and subjected to differential gene expression using DeSeq2 (52). Normalized gene expression was used to compute and plot two-dimensional principal components analysis, using the Python modules sklearn (v0.19.1; https://scikit-learn.org/stable/) and Matplotlib (v2.2.2; https://matplotlib.org/), respectively. Differentially expressed genes (log2 fold change |1|, adjusted P < 0.05) were the input to produce an unsupervised hierarchical clustering heat map in Partek Genomics Suite software (version 7.0) (Partek Inc., St. Louis, MO, USA). Raw data are available using accession GSE150720.

ChIP was performed as previously described (53) using antibodies against H3K27Ac (5 g per IP; Abcam, ab4729), H3K4Me3 (5 g per IP; Abcam ab8580), and H3K27Me3 (5 g per IP; Diagenode, C15410195). Library construction and sequencing were performed by Novogene (HK) Co. Ltd. Paired-end reads were aligned to the hg38 genome build using Burrows Wheeler Aligner (BWA) (54) duplicate reads removed using Picard (http://broadinstitute.github.io/picard/), and tracks were generated using DeepTools bamCoverage (https://deeptools.readthedocs.io/en/develop/). Peaks were called using MACS2 (55) with the parameter (P = 1 109). Differentially bound regions between the AdMSC and iMS were calculated using DiffBind (http://bioconductor.org/packages/release/bioc/vignettes/DiffBind/inst/doc/DiffBind.pdf) and regions annotated using ChIPseeker (56). Raw data are available using accession GSE151527. Adipocyte ChIP data were downloaded from Gene Expression Omnibus (GEO); accession numbers are as follows for the three histone marks: GSM916066, GSM670041, and GSM772771.

Total genomic DNA was extracted using the DNA MiniPrep Kit (Qiagen), and RRBS library construction and sequencing were performed by Novogene (HK) Co. Ltd. Raw RRBS data in fastq format were quality and adapter trimmed using trim_galore (0.6.4) with rrbs parameter (www.bioinformatics.babraham.ac.uk/projects/trim_galore). The trimmed fastq files were then aligned to a bisulfite-converted genome (Ensembl GRCh38) using Bismark (2.3.5), and methylation status at each CpG loci was extracted (57). The cytosine coverage files were converted to BigWig format for visualization. Differentially methylated cytosines (DMCs) and DMRs were identified using methylKit (1.10) and edmr (0.6.4.1) packages in R (3.6.1) (58, 59). DMCs and DMRs were annotated using ChIPseeker (56), and pathway enrichment was performed as detailed below. Raw data are available using accession number GSE151527. Adipocyte RRBS data were downloaded from GEO: GSM2342293 and GSM2342392.

IPA (Qiagen) was used to investigate enrichment in molecular and cellular functions, systems development and function, and canonical pathways.

Statistical analysis was performed in SAS. For the dose-optimization experiments (Fig. 1), a linear mixed model with participant-level random effects was used to estimate maximum time by dose level and age group. A linear mixed model with participant-level random effects was used to analyze statistical differences in lineage contribution outcomes between treatment groups (Fig. 3) and at different time points posttransplant, to estimate the percentage of cells by treatment and lineage. For the in vivo regeneration experiment (Fig. 4), a linear model was used to model the percent of cells over time for each group. Quadratic time terms were added to account for the observed increase from 1 to 2 weeks and decrease from 2 to 4 weeks. In the muscle regeneration experiment, a linear model was applied to cohort A and cohort B, to estimate and compare percent cells by treatment and source. Statistical modeling data are included in table S2.

Acknowledgments: We are indebted to the patients who donated tissue to this project. We thank E. Cook (Prince of Wales Private Hospital), B. Lee (Mark Wainwright Analytical Centre, UNSW Sydney), and technicians at the UNSW BRC Facility for assistance with sample and data collection and animal care; Y. Huang for technical assistance; and A. Unnikrishnan and C. Jolly for helpful discussions and critical reading of the manuscript. We acknowledge the facilities and scientific and technical assistance of the National Imaging Facility, a National Collaborative Research Infrastructure Strategy (NCRIS) capability, at the BRIL (UNSW). The STRO-1 antibody was a gift from S. Gronthos, University of Adelaide, Australia. Funding: We acknowledge the following funding support: A.Y. was supported by an Endeavour International Postgraduate Research scholarship from the Australian Government. S.S. is supported by an International Postgraduate Student scholarship from UNSW and the Prince of Wales Clinical School. P.S. is supported by an International Postgraduate Student scholarship from UNSW. M.L.T. and D.D.M. acknowledge funding from St. Vincents Clinic Foundation and Arrow BMT Foundation. K.A.K. acknowledges funding from Australian Research Council (FT180100417). J.M. is supported, in part, by the Olivia Lambert Foundation. M.K. is supported by a NHMRC Program Grant (APP1091261) and NHMRC Principal Research Fellowship (APP1119152). L.B.H. acknowledges funding from MTPConnect MedTech and Pharma Growth Centre (PRJ2017-55 and BMTH06) as part of the Australian Governmentfunded Industry Growth Centres Initiative Programme and The Kinghorn Foundation. D.B. is supported by a Peter Doherty Fellowship from the National Health and Medical Research Council of Australia, a Cancer Institute NSW Early Career Fellowship, the Anthony Rothe Memorial Trust, and Gilead Sciences. R.M. acknowledges funding from Jasper Medical Innovations (Sydney, Australia). J.E.P., V.C., and E.C.H. acknowledge funding from the National Health and Medical Research Council of Australia (APP1139811). Author contributions: The project was conceived by V.C. and J.E.P., and the study design and experiments were planned by A.Y., V.C., and J.E.P. Most of the experiments and data analyses were performed by A.Y., guided and supervised by V.C. and J.E.P. S.S., R.A.O., C.A.L., D.C., F.Y., M.L.T., P.S., T.H., J.R.P., P.H., W.R.W., and V.C. performed additional experiments and data analyses, with further supervision from R.M., C.P., J.A.I.T., D.C., J.W.H.W., L.B.H., D.B., and E.C.H. Statistical analyses were performed by J.O. R.M., D.D.M., J.M., K.A.K., and M.K. provided critical reagents. The manuscript was written by A.Y., J.A.I.T., V.C., and J.E.P., and reviewed and agreed to by all coauthors. Competing interests: V.C. and J.E.P. are named inventors on a patent A method of generating cells with multi-lineage potential (US 9982232, AUS 2013362880). All other authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors.

See the original post here:
Induction of muscle-regenerative multipotent stem cells from human adipocytes by PDGF-AB and 5-azacytidine - Science Advances

Study On Rabbit Brain Reveals Genetic Markers Of Domestication – Texas A&M University Today

Tolerance of humans and tameness are noted traits differentiating domestic and wild rabbits.

Getty Images

An international team of researchers led by Leif Andersson, a professor at the Texas A&M University College of Veterinary Medicine & Biomedical Sciences (CVMBS), has found changes in gene expression patterns across the brain between wild and domestic rabbits, which likely contributed to the evolution of tameness during domestication.

By demonstrating that domestic animals acquired tolerance toward humans through regulatory changes of certain genes, researchers are able to better understand the link between genetic changes and the mechanisms of evolution in rabbits and, at a larger scale, of plants and animals in general. The research builds off Anderssons previous work demonstrating a link between rabbit domestication and brain structure.

The European rabbit is one of the most recently domesticated animals, exhibiting distinct morphological, physiological and behavioral differences from their wild counterparts. Tolerance of humans and tameness are noted traits differentiating the domestic and wild rabbits.

Domestication is often associated with the rapid development of such traits in a rather short evolutionary time period. As such, domestication has long been a major topic in evolutionary biology, as it allows researchers to study the mechanisms of evolution on a more manageable time scale.

In the study, published in Genome Biology and Evolution, the researchers compared gene expression patterns in four brain regions between newborns of wild and domestic rabbits. They detected hundreds of genes differentially expressed between the two.

It is important to notice that the drastic changes in behavior between domestic animals and their wild ancestors are associated with changes in how genes are expressed during the development of the brain, Andersson said. The domestication of plants and animals is one of the most important developments during human history. In this study, we shed light on how this process has altered brain function in domestic animals and made them tamer.

One significant finding was that genes involved in dopamine signaling, a chemical involved in fear responses, were expressed at higher rates in the amygdala, or area of the brain associated with memory, decision-making, and emotional responses, of the domestic rabbit. Notably, the amygdala is known as the part of the brain driving the fight or flight response, which would play a part in determining whether a wild animal flees in the company of humans or a tame animal tolerates human presence.

Researchers also found that genes associated with ciliary function were consistently downregulated in the hippocampus, an area of the brain associated with learning and memory, of domestic rabbits. Cilia cells are tiny hair-like structures that aid in the circulation of cerebrospinal fluid.

The flow of this fluid contributes to the transmission of molecular signals across the brain. Since these ciliary genes are important for brain development, the researchers hypothesized that these changes contribute to the evolution of tameness.

We were really surprised by the consistent changes we observed that several genes involved in dopamine signaling were consistently unregulated in the domestic brain, and that many genes with associated with ciliary function was consistently downregulated, Andersson said.

Read the original post:
Study On Rabbit Brain Reveals Genetic Markers Of Domestication - Texas A&M University Today

Kite Taps Industry Vet to Lead Cell Therapy R&D – BioSpace

AaronP/Bauer-Griffin/GC Images

Kite Pharma has tapped immunotherapy expert Francesco Marincola, a former National Institutes of Health investigator, to helm the companys cell therapy research.

Marincola will be in charge of Kites research organization across hematological malignancies and solid tumors. Kite, a Gilead Sciences company, said Marincola will assume his new responsibilities on Feb. 1.

Marincola joins Kite from his most recent role as president and chief scientific officer of California-based Refuge Biotechnologies, where he led research into that companys cell therapy programs developed with Refuge's receptor-dCas platform. Refuges pipeline is led by RB-1916, a CAR-T cell therapy designed to inhibit the expression of the PD-1 gene, with a potential initial application in diffused large B-cell lymphoma.

Kite is certainly no stranger to CAR-T. Kite is focused on chimeric antigen receptor and T cell receptor engineered cell therapies. In 2017, the U.S. Food and Drug Administration approvedYescarta, the second CAR-T treatment in the United States. The approval came two months after Kite wasacquired by Gilead for $12 billion. Following the approval of Yescarta, Kite dove into cell therapy research.

In 2018, Kite forged a cell therapy-focused partnership with the National Cancer Institute to develop adoptive cell therapies targeting patient-specific tumor neoantigens, which are mutations found on the surface of cancer cells that are unique to each person and tumor. To support its continued research into cell therapies, Kite also scaled up its manufacturing efforts to support CAR-T development in the U.S. and Europe.

Earlier this month, Kite and Oxford BioTherapeutics entered into a research collaboration to evaluate five novel targets for a number of hematologic and solid tumor indications.

Kite Chief Executive Officer Christi Shaw said Marincolas expertise in cell therapy will enable the company to discover and advance new life-saving therapies for patients in need. CAR-T therapy has proven to be successful in some hematological cancers and now researchers are scrambling to aim the potent anti-cancer therapy at solid tumors. Challenges in this area remain, and Marincola is expected to use his own expertise to push this research forward.

Francos proven track record in oncology and robust research experience that spans the NIH to industry will be critical to the targeting and acceleration of our research efforts in CAR T and beyond, Shaw said in a statement.

It is an honor to bring my experience to an industry-leading team that is working to make cell therapies with curative intent available to patients, Marincola said. I look forward to helping build and diversify our research efforts in support of this mission.

In addition to his experience at Refuge, Marincola spent 23 years at the NIH, where he was tenured senior investigator in cancer immunotherapy and biomarker research. During his time at NIH, he spent 15 years as the Chief of the Infectious Disease and Immunogenetics Section at the NIH Clinical Center. Over the course of his career, Marincola also served as a distinguished research fellow in immune oncology discovery at AbbVie and as Chief Research Officer at Sidra Research in Doha, Qatar.

Marincola also served as president of the Society for Immunotherapy of Cancer and currently serves as Editor-in-Chief for several prominent peer-reviewed publications, includingJournal of Translational Medicine,Translational Medicine CommunicationsandImmunotherapy.

Most Read Today

Continue reading here:
Kite Taps Industry Vet to Lead Cell Therapy R&D - BioSpace

Repurposing a proven gene therapy approach to treat, prevent COVID-19 – Penn Today

When did you first start using AAV vectors in your gene therapy work?

It came about 10 years ago when I was helping the Gates Foundation develop an approach for preventing HIV. Any attempt to use a traditional vaccine, where you inject a component of the virus to activate the immune system to develop proteins such as antibodies, had been challenging for HIV. Regardless of what you used to immunizebecause the virus changed so muchmost of it would escape. Once the field realized that, we started to look at other approaches, and it turns out its possible in the lab to engineer an antibody that could be effective against many types of HIV.

HIV represents a different type of pandemic than COVID-19. When did you turn to AAV vectors as a potential approach for other kinds of pandemics?

About eight years ago I started thinking about this as a countermeasure for a pandemic. The pandemics that we worry about are primarily transmitted through a respiratory route. If it were direct contact like Ebola virus, its not as dangerous because you can avoid touching one another. But if you cant even be in the same room, thats a problem.

Respiratory viruses enter our body through the nose and throat. Thats how we get infected. We proposed delivering the vector through a nasal mist or spray to engineer the cells that line the nose and throat to express the antibody. If you can localize this at that site to prevent the virus from going farther, then you dont need the whole body to express the antibodies.

The antibodies youre using, called casirivimab and imdevimab, are monoclonal antibodies, meaning they were created in a lab. Can you describe how they work?

Regeneron developed these. Theyre highly active and potent against SARS-CoV-2. For treatment, antibodies can be useful. If youre starting to get sick, you get an infusion or two of the antibodies and then you dont get sicker. But what do you do with 99% of the population who isnt sick and never gets sick? Our idea was to use an AAV vector expressing the antibodies to engineer someones cells to produce the antibodies. If we do this right, the expression could go on for a long period of time. Its a one-time vector infusion.

We were able to show in animal models that an AAV sprayed into the nose that expresses an antibody is effective against flu virus that causes respiratory diseases and has the potential to cause a pandemic. The treated animals were completely protected when exposed to flu virus. Its all about having the right antibody and then engineering a delivery system to have this blockage. We call it a bioshield. It could be a way to stop COVID-19 in its tracks.

Would this approach replace COVID-19 vaccines or be used in conjunction with them?

Theoretically, it could be used in place of a vaccine, but I suspect that traditional vaccines are going to succeed for a lot of people. We see our approach being deployed in individuals for which traditional vaccines may not work as well, patients with diseases that compromise their immune system such as cancer, patients who are on immune-modulating drugs, or even the elderly.

Early data seem to suggest that the elderly have some level of response to the active COVID-19 vaccine, but, like with many other vaccines, older people dont mount the same immune response as those who are younger. That said, I dont see any reason why receiving a traditional vaccine would preclude one from using our nasal spray because they do two different things.

The other possibility is that the COVID-19 vaccines we have become less effective because the virus changes. I dont think this will happen, and I hope it doesnt, but, if it does, the question becomes, Would the antibodies that Regeneron created become a backup? When we roll out an active vaccine based on a single spike protein into large populations, it creates pressure on the SARS-CoV-2 to change and potentially become resistant. I hope a variant doesnt emerge, but I do think it behooves us to have some redundancy in place to squelch a potential second wave due to resistant coronaviruses.

What is your projected timeline?

We are conducting one final experiment over the holiday break and in early January before we submit our request to the FDA for clinical trials. Weve had discussions with the FDA and have already done some of the initial testing, including safety testing in nonhuman primates, as well as preparing to manufacture the product. Conducting this in the Gene Therapy Program is beneficial since we are comfortable with AAV vectors and moving them into clinical trials. We support up to eight traditional AAV gene therapy programs a year, and we have the staff and technology to move pretty quickly.

If we get the go in January, I think our technology could contribute to the global response in eliminating COVID-19. And you have to understand, until we eliminate it globally, we havent actually eliminated it.

See the original post:
Repurposing a proven gene therapy approach to treat, prevent COVID-19 - Penn Today