Semin Respir Crit Care Med 2019; 40(06): 762-774
DOI: 10.1055/s-0039-1696664
Review Article
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Treating the Underlying Cystic Fibrosis Transmembrane Conductance Regulator Defect in Patients with Cystic Fibrosis

Senne Cuyx
1   UZ Leuven, University Hospital of Leuven, Leuven, Belgium
,
Kris De Boeck
2   Division Pediatric Pulmonology, Department of Pediatrics, University Hospital of Leuven, Leuven, Belgium
› Author Affiliations
Further Information

Publication History

Publication Date:
28 October 2019 (online)

Abstract

Detailed knowledge of how mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene disturb the trafficking or function of the CFTR protein and the use of high-throughput drug screens have allowed novel therapeutic strategies for cystic fibrosis (CF). The main goal of treatment is slowly but surely shifting from symptomatic management to targeting the underlying CFTR defect to halt disease progression and even to prevent occurrence of CF complications. CFTR potentiators for patients with class III mutations, mutation R117H (and in United States also for patients with specific residual function mutations) and the combination of a CFTR modulator plus a potentiator for patients homozygous for F508del, are the two classes of modulators that are in use in the clinic. Approval of these therapeutics has progressively expanded to include both younger patients and a wider range of CFTR mutations. For a significant proportion of patients with CF, current treatment is however still insufficient or unavailable.

This review provides an overview of the clinical trial results and the real-life efficacy data of approved CFTR modulators. In addition, we discuss the entire pipeline of CFTR modulators: novel potentiators and correctors, amplifiers, stabilizers, and read-through agents. Furthermore, we discuss other strategies to improve CFTR function like nonsense-mediated decay inhibitors, modified transfer ribonucleic acids, antisense oligonucleotides, and genetic therapies.

CFTR modulators are already changing the face of CF and the pipeline of new therapies continues to be exciting.

 
  • References

  • 1 Riordan JR, Rommens JM, Kerem B. , et al. Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA. Science 1989; 245 (4922): 1066-1073
  • 2 Spielberg DR, Clancy JP. Cystic fibrosis and its management through established and emerging therapies. Annu Rev Genomics Hum Genet 2016; 17 (01) 155-175
  • 3 Mehta G, Macek Jr M, Mehta A. ; European Registry Working Group. Cystic fibrosis across Europe: EuroCareCF analysis of demographic data from 35 countries. J Cyst Fibros 2010; 9 (Suppl. 02) S5-S21
  • 4 Cystic Fibrosis Mutation Database. Available at: http://www.genet.sickkids.on.ca/ . Accessed May 30, 2019
  • 5 CFTR2. Available at: https://www.cftr2.org/ . Accessed May 30, 2019
  • 6 Reddy MM, Quinton PM. Selective activation of cystic fibrosis transmembrane conductance regulator Cl- and HCO3- conductances. JOP 2001; 2 (4, Suppl): 212-218
  • 7 Pezzulo AA, Tang XX, Hoegger MJ. , et al. Reduced airway surface pH impairs bacterial killing in the porcine cystic fibrosis lung. Nature 2012; 487 (7405): 109-113
  • 8 Vankeerberghen A, Cuppens H, Cassiman J-J. The cystic fibrosis transmembrane conductance regulator: an intriguing protein with pleiotropic functions. J Cyst Fibros 2002; 1 (01) 13-29
  • 9 Castellani C, Duff AJA, Bell SC. , et al. ECFS best practice guidelines: the 2018 revision. J Cyst Fibros 2018; 17 (02) 153-178
  • 10 European Cystic Fibrosis Society Annual Patient Registry. Data Report 2016. 2016 . Available at: https://www.ecfs.eu/sites/default/files/general-content-images/working-groups/ecfs-patient-registry/ECFSPR_Report2016_06062018.pdf . Accessed May 30, 2019
  • 11 De Boeck K, Amaral MD. Progress in therapies for cystic fibrosis. Lancet Respir Med 2016; 4 (08) 662-674
  • 12 Dewulf J, Vermeulen F, Wanyama S. , et al. Treatment burden in patients with at least one class IV or V CFTR mutation. Pediatr Pulmonol 2015; 50 (12) 1230-1236
  • 13 Veit G, Avramescu RG, Chiang AN. , et al. From CFTR biology toward combinatorial pharmacotherapy: expanded classification of cystic fibrosis mutations. Mol Biol Cell 2016; 27 (03) 424-433
  • 14 Ramsey BW, Davies J, McElvaney NG. , et al; VX08-770-102 Study Group. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med 2011; 365 (18) 1663-1672
  • 15 Davies JC, Wainwright CE, Canny GJ. , et al; VX08-770-103 (ENVISION) Study Group. Efficacy and safety of ivacaftor in patients aged 6 to 11 years with cystic fibrosis with a G551D mutation. Am J Respir Crit Care Med 2013; 187 (11) 1219-1225
  • 16 Davies J, Sheridan H, Bell N. , et al. Assessment of clinical response to ivacaftor with lung clearance index in cystic fibrosis patients with a G551D-CFTR mutation and preserved spirometry: a randomised controlled trial. Lancet Respir Med 2013; 1 (08) 630-638
  • 17 De Boeck K, Munck A, Walker S. , et al. Efficacy and safety of ivacaftor in patients with cystic fibrosis and a non-G551D gating mutation. J Cyst Fibros 2014; 13 (06) 674-680
  • 18 Davies JC, Cunningham S, Harris WT. , et al; KIWI Study Group. Safety, pharmacokinetics, and pharmacodynamics of ivacaftor in patients aged 2–5 years with cystic fibrosis and a CFTR gating mutation (KIWI): an open-label, single-arm study. Lancet Respir Med 2016; 4 (02) 107-115
  • 19 Rosenfeld M, Cunningham S, Harris WT. , et al. An open-label extension study of ivacaftor in children with CF and a CFTR gating mutation initiating treatment at age 2–5 years (KLIMB). J Cyst Fibros 2019 (e-pub ahead of print). Doi: 10.1016/j.jcf.2019.03.00
  • 20 Rosenfeld M, Wainwright CE, Higgins M. , et al; ARRIVAL study group. Ivacaftor treatment of cystic fibrosis in children aged 12 to <24 months and with a CFTR gating mutation (ARRIVAL): a phase 3 single-arm study. Lancet Respir Med 2018; 6 (07) 545-553
  • 21 Kalydeco: summary of product characteristics. Available at: https://www.ema.europa.eu/en/documents/product-information/kalydeco-epar-product-information_en.pdf . Accessed May 30, 2019
  • 22 Kalydeco: highlights of prescribing information. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/203188lbl.pdf . Accessed May 30, 2019
  • 23 Moss RB, Flume PA, Elborn JS. , et al; VX11-770-110 (KONDUCT) Study Group. Efficacy and safety of ivacaftor in patients with cystic fibrosis who have an Arg117His-CFTR mutation: a double-blind, randomised controlled trial. Lancet Respir Med 2015; 3 (07) 524-533
  • 24 Van Goor F, Yu H, Burton B, Hoffman BJ. Effect of ivacaftor on CFTR forms with missense mutations associated with defects in protein processing or function. J Cyst Fibros 2014; 13 (01) 29-36
  • 25 McGarry ME, Illek B, Ly NP. , et al. In vivo and in vitro ivacaftor response in cystic fibrosis patients with residual CFTR function: N-of-1 studies. Pediatr Pulmonol 2017; 52 (04) 472-479
  • 26 Barry PJ, Plant BJ, Nair A. , et al. Effects of ivacaftor in patients with cystic fibrosis who carry the G551D mutation and have severe lung disease. Chest 2014; 146 (01) 152-158
  • 27 Sawicki GS, McKone EF, Pasta DJ. , et al. Sustained benefit from ivacaftor demonstrated by combining clinical trial and cystic fibrosis patient registry data. Am J Respir Crit Care Med 2015; 192 (07) 836-842
  • 28 Bessonova L, Volkova N, Higgins M. , et al. Data from the US and UK cystic fibrosis registries support disease modification by CFTR modulation with ivacaftor. Thorax 2018; 73 (08) 731-740
  • 29 Wainwright CE, Elborn JS, Ramsey BW. , et al; TRAFFIC Study Group; TRANSPORT Study Group. Lumacaftor-ivacaftor in patients with cystic fibrosis homozygous for Phe508del CFTR. N Engl J Med 2015; 373 (03) 220-231
  • 30 Taylor-Cousar JL, Jain M, Barto TL. , et al; VX14-809-106 Investigator Group. Lumacaftor/ivacaftor in patients with cystic fibrosis and advanced lung disease homozygous for F508del-CFTR. J Cyst Fibros 2018; 17 (02) 228-235
  • 31 Ratjen F, Hug C, Marigowda G. , et al; VX14-809-109 investigator group. Efficacy and safety of lumacaftor and ivacaftor in patients aged 6-11 years with cystic fibrosis homozygous for F508del-CFTR: a randomised, placebo-controlled phase 3 trial. Lancet Respir Med 2017; 5 (07) 557-567
  • 32 McNamara JJ, McColley SA, Marigowda G. , et al. Safety, pharmacokinetics, and pharmacodynamics of lumacaftor and ivacaftor combination therapy in children aged 2-5 years with cystic fibrosis homozygous for F508del-CFTR: an open-label phase 3 study. Lancet Respir Med 2019; 7 (04) 325-335
  • 33 Taylor-Cousar JL, Munck A, McKone EF. , et al. Tezacaftor-ivacaftor in patients with cystic fibrosis homozygous for Phe508del. N Engl J Med 2017; 377 (21) 2013-2023
  • 34 Donaldson SH, Pilewski JM, Griese M. , et al; VX11-661-101 Study Group. Tezacaftor/ivacaftor in subjects with cystic fibrosis and F508del/F508del-CFTR or F508del/G551D-CFTR. Am J Respir Crit Care Med 2018; 197 (02) 214-224
  • 35 Vertex. Press release: vertex reports third-quarter 2017 financial results. Available at: https://investors.vrtx.com/news-releases/news-release-details/vertex-reports-third-quarter-2017-financial-results . Accessed May 30, 2019
  • 36 Rowe SM, Daines C, Ringshausen FC. , et al. Tezacaftor-ivacaftor in residual-function heterozygotes with cystic fibrosis. N Engl J Med 2017; 377 (21) 2024-2035
  • 37 Vertex. Press release: positive phase 3 study for tezacaftor/ivacaftor combination in children aged 6–11 years with cystic fibrosis supports European Medicines Agency submission. Available at: https://investors.vrtx.com/news-releases/news-release-details/positive-phase-3-study-tezacaftorivacaftor-combination-children . Accessed May 30, 2019
  • 38 Davies JC, Moskowitz SM, Brown C. , et al; VX16-659-101 Study Group. VX-659-tezacaftor-ivacaftor in patients with cystic fibrosis and one or two Phe508del alleles. N Engl J Med 2018; 379 (17) 1599-1611
  • 39 Keating D, Marigowda G, Burr L. , et al; VX16-445-001 Study Group. VX-445-tezacaftor-ivacaftor in patients with cystic fibrosis and one or two Phe508del alleles. N Engl J Med 2018; 379 (17) 1612-1620
  • 40 Vertex. Press release: two phase 3 studies of the triple combination of VX-659, tezacaftor and ivacaftor met primary endpoint of improvement in lung function (ppFEV1) in people with cystic fibrosis. Available from: https://investors.vrtx.com/news-releases/news-release-details/two-phase-3-studies-triple-combination-vx-659-tezacaftor-and . Accessed May 30, 2019
  • 41 Vertex. Press release: two phase 3 studies of the triple combination of VX-445, tezacaftor and ivacaftor met primary endpoint of improvement in lung function (ppFEV1) in people with cystic fibrosis. Available at: https://investors.vrtx.com/news-releases/news-release-details/correcting-and-replacing-two-phase-3-studies-triple-combination . Accessed May 30, 2019
  • 42 Davies J, Bell SC, Van Koningsbruggen-Rietschel S. , et al. GLPG1837 in subjects with cystic fibrosis (CF) and the G551D mutation: results from a phase II study (SAPHIRA1). J Cyst Fibros 2017; 16 (Suppl. 01) S24-S25
  • 43 Zemanick ET, Daines CL, Dellon EP. , et al. Highlights from the 2016 North American Cystic Fibrosis Conference. Pediatr Pulmonol 2017; 52 (08) 1103-1110
  • 44 Rowe SM, Haque N, Gleason S. , et al. A randomized, placebo controlled 4-week study in COPD of QBW251, a potentiator of the cystic fibrosis transmembrane conductance regulator (CFTR) protein. Eur Respir J 2018; 52 (Suppl. 62) PA617
  • 45 Kazani S, Alcantara J, Debonnett L. , et al. QBW251 is a safe and efficacious CFTR potentiator for patients with cystic fibrosis. Am J Respir Crit Care Med 2016; 193: A7789
  • 46 Bell SC, Barry PJ, De Boeck K. , et al. CFTR activity is enhanced by the novel corrector GLPG2222, given with and without ivacaftor in two randomized trials. J Cyst Fibros 2019 (e-pub ahead of print). Doi: 10.1016/j.jcf.2019.04.014
  • 47 Martiniano SL, Toprak D, Ong T. , et al. Highlights from the 2017 North American Cystic Fibrosis Conference. Pediatr Pulmonol 2018; 53 (07) 979-986
  • 48 In vitro efficacy of combination FDL169/FDL176 is greater than tezacaftor/ivacaftor. 2018 . Available at: http://www.flatleydiscoverylab.com/wp-content/uploads/2017/11/169-176-combination-final.pdf . Accessed May 30, 2019
  • 49 Miller J, Drew L, Green O. , et al. CFTR amplifiers: a new class of CFTR modulator that complements the substrate limitations of other CF therapeutic modalities. Am J Respir Crit Care Med 2016; 193: A5574
  • 50 Molinski SV, Ahmadi S, Ip W. , et al. Orkambi® and amplifier co-therapy improves function from a rare CFTR mutation in gene-edited cells and patient tissue. EMBO Mol Med 2017; 9 (09) 1224-1243
  • 51 Miller J, Drew L, Bastos C. , et al. Novel CFTR modulator combination of amplifier, corrector and potentiator provides advantages over two corrector-based combinations. Pediatr Pulmonol 2016; 51 (S45): S194-S485
  • 52 Flume P, Sawicki G, Pressler T. , et al. Phase 2 initial results evaluating PTI-428, a novel CFTR amplifier, in patients with cystic fibrosis. J Cyst Fibros 2018; 17: S1-S2
  • 53 Press release: proteostasis therapeutics announces broad new dataset from proprietary combination and add-on CFTR modulator studies in cystic fibrosis patients. 2019 . Available at: http://ir.proteostasis.com/news-releases/news-release-details/proteostasis-therapeutics-announces-broad-new-dataset . Accessed May 30, 2019
  • 54 Fukuda R, Okiyoneda T. Peripheral protein quality control as a novel drug target for CFTR stabilizer. Front Pharmacol 2018; 9: 1100
  • 55 Donaldson SH, Solomon GM, Zeitlin PL. , et al. Pharmacokinetics and safety of cavosonstat (N91115) in healthy and cystic fibrosis adults homozygous for F508DEL-CFTR. J Cyst Fibros 2017; 16 (03) 371-379
  • 56 Press release: nivalis therapeutics announces results from phase 2 clinical trial of cavosonstat added to ivacaftor for treatment of cystic fibrosis. 2017 . Available at: https://www.globenewswire.com/news-release/2017/02/23/927232/0/en/Nivalis-Therapeutics-Announces-Results-from-Phase-2-Clinical-Trial-of-Cavosonstat-Added-to-Ivacaftor-for-Treatment-of-Cystic-Fibrosis.html . Accessed May 30, 2019
  • 57 De Boeck K, Zolin A, Cuppens H, Olesen HV, Viviani L. The relative frequency of CFTR mutation classes in European patients with cystic fibrosis. J Cyst Fibros 2014; 13 (04) 403-409
  • 58 Wilschanski M, Famini C, Blau H. , et al. A pilot study of the effect of gentamicin on nasal potential difference measurements in cystic fibrosis patients carrying stop mutations. Am J Respir Crit Care Med 2000; 161 (3, Pt 1): 860-865
  • 59 Clancy JP, Bebök Z, Ruiz F. , et al. Evidence that systemic gentamicin suppresses premature stop mutations in patients with cystic fibrosis. Am J Respir Crit Care Med 2001; 163 (07) 1683-1692
  • 60 Wilschanski M, Yahav Y, Yaacov Y. , et al. Gentamicin-induced correction of CFTR function in patients with cystic fibrosis and CFTR stop mutations. N Engl J Med 2003; 349 (15) 1433-1441
  • 61 Sermet-Gaudelus I, Renouil M, Fajac A. , et al. In vitro prediction of stop-codon suppression by intravenous gentamicin in patients with cystic fibrosis: a pilot study. BMC Med 2007; 5 (01) 5
  • 62 Kerem E, Hirawat S, Armoni S. , et al. Effectiveness of PTC124 treatment of cystic fibrosis caused by nonsense mutations: a prospective phase II trial. Lancet 2008; 372 (9640): 719-727
  • 63 Sermet-Gaudelus I, Boeck KD, Casimir GJ. , et al. Ataluren (PTC124) induces cystic fibrosis transmembrane conductance regulator protein expression and activity in children with nonsense mutation cystic fibrosis. Am J Respir Crit Care Med 2010; 182 (10) 1262-1272
  • 64 Wilschanski M, Miller LL, Shoseyov D. , et al. Chronic ataluren (PTC124) treatment of nonsense mutation cystic fibrosis. Eur Respir J 2011; 38 (01) 59-69
  • 65 Kerem E, Konstan MW, De Boeck K. , et al; Cystic Fibrosis Ataluren Study Group. Ataluren for the treatment of nonsense-mutation cystic fibrosis: a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Respir Med 2014; 2 (07) 539-547
  • 66 Konstan MW, DiMango E, Kerem E. , et al. Efficacy and safety of ataluren in patients with nonsense-mutation cystic fibrosis not receiving chronic inhaled aminoglycosides: the international, randomized, double-blind, placebo-controlled Ataluren Confirmatory Trial in Cystic Fibrosis (ACT CF). J Cyst Fibros 2017; 16: S23-S24
  • 67 Press release: PTC therapeutics announces results from pivotal phase 3 clinical trial of ataluren in patients living with nonsense mutation cystic fibrosis. Available at: https://www.prnewswire.com/news-releases/ptc-therapeutics-announces-results-from-pivotal-phase-3-clinical-trial-of-ataluren-in-patients-living-with-nonsense-mutation-cystic-fibrosis-300416860.html . Accessed May 30, 2019
  • 68 Zainal Abidin N, Haq IJ, Gardner AI, Brodlie M. Ataluren in cystic fibrosis: development, clinical studies and where are we now?. Expert Opin Pharmacother 2017; 18 (13) 1363-1371
  • 69 Xue X, Mutyam V, Tang L. , et al. Synthetic aminoglycosides efficiently suppress cystic fibrosis transmembrane conductance regulator nonsense mutations and are enhanced by ivacaftor. Am J Respir Cell Mol Biol 2014; 50 (04) 805-816
  • 70 Leubitz A, Frydman-Marom A, Sharpe N, van Duzer J, Campbell KCM, Vanhoutte F. Safety, tolerability, and pharmacokinetics of single ascending doses of ELX-02, a potential treatment for genetic disorders caused by nonsense mutations, in healthy volunteers. Clin Pharmacol Drug Dev 2019 (e-pub ahead of print). Doi: 10.1002/cpdd.647
  • 71 Lueck JD, Yoon JS, Perales-Puchalt A. , et al. Engineered transfer RNAs for suppression of premature termination codons. Nat Commun 2019; 10 (01) 822
  • 72 Linde L, Boelz S, Nissim-Rafinia M. , et al. Nonsense-mediated mRNA decay affects nonsense transcript levels and governs response of cystic fibrosis patients to gentamicin. J Clin Invest 2007; 117 (03) 683-692
  • 73 Sharma N, Evans TA, Pellicore MJ. , et al. Capitalizing on the heterogeneous effects of CFTR nonsense and frameshift variants to inform therapeutic strategy for cystic fibrosis. PLoS Genet 2018; 14 (11) e1007723
  • 74 Aksit MA, Bowling AD, Evans TA. , et al. Decreased mRNA and protein stability of W1282X limits response to modulator therapy. J Cyst Fibros 2019 (e-pub ahead of print). Doi: 10.1016/j.jcf.2019.02.009
  • 75 Keenan MM, Huang L, Jordan NJ. , et al. Nonsense mediated RNA decay pathway inhibition restores expression and function of W1282X CFTR. Am J Respir Cell Mol Biol 2019 (e-pub ahead of print). Doi: 10.1165/rcmb.2018-0316OC
  • 76 Alroy I, Shohat M, Eshkar-Oren I, Huertas P. Translational read-through of CTNS nonsense mutations and attenuation of CTNS nonsense-mediated mRNA decay by ELX-02 treatment. Mol Genet Metab 2018; 123 (02) S18
  • 77 Sermet-Gaudelus I, Clancy JP, Nichols DP. , et al. Antisense oligonucleotide eluforsen improves CFTR function in F508del cystic fibrosis. J Cyst Fibros 2018; 18 (04) 536-542
  • 78 Irony-Tur Sinai M, Wilton S, Kerem B. Restoration of the CFTR function by antisense oligonucleotide splicing modulation. J Cyst Fibros 2014; 13: S2
  • 79 Friedman KJ, Kole J, Cohn JA, Knowles MR, Silverman LM, Kole R. Correction of aberrant splicing of the cystic fibrosis transmembrane conductance regulator (CFTR) gene by antisense oligonucleotides. J Biol Chem 1999; 274 (51) 36193-36199
  • 80 Igreja S, Clarke LA, Botelho HM, Marques L, Amaral MD. Correction of a cystic fibrosis splicing mutation by antisense oligonucleotides. Hum Mutat 2016; 37 (02) 209-215
  • 81 Viart V, Bergougnoux A, Bonini J. , et al. Transcription factors and miRNAs that regulate fetal to adult CFTR expression change are new targets for cystic fibrosis. Eur Respir J 2015; 45 (01) 116-128
  • 82 Bardin P, Sonneville F, Corvol H, Tabary O. Emerging microRNA therapeutic approaches for cystic fibrosis. Front Pharmacol 2018; 9: 1113
  • 83 Alton EWFW, Boyd AC, Davies JC. , et al. Genetic medicines for CF: hype versus reality. Pediatr Pulmonol 2016; 51 (S44): S5-S17
  • 84 Griesenbach U, Pytel KM, Alton EWFW. Cystic fibrosis gene therapy in the UK and elsewhere. Hum Gene Ther 2015; 26 (05) 266-275
  • 85 Alton EWFW, Boyd AC, Porteous DJ. , et al; UK Cystic Fibrosis Gene Therapy Consortium *. A phase I/IIa safety and efficacy study of nebulized liposome-mediated gene therapy for cystic fibrosis supports a multidose trial. Am J Respir Crit Care Med 2015; 192 (11) 1389-1392
  • 86 Alton EWFW, Armstrong DK, Ashby D. , et al; UK Cystic Fibrosis Gene Therapy Consortium. Repeated nebulisation of non-viral CFTR gene therapy in patients with cystic fibrosis: a randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Respir Med 2015; 3 (09) 684-691
  • 87 Alton EWFW, Beekman JM, Boyd AC. , et al. Preparation for a first-in-man lentivirus trial in patients with cystic fibrosis. Thorax 2017; 72 (02) 137-147
  • 88 Schwank G, Koo B-K, Sasselli V. , et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell 2013; 13 (06) 653-658
  • 89 Firth AL, Menon T, Parker GS. , et al. Functional gene correction for cystic fibrosis in lung epithelial cells generated from patient iPSCs. Cell Reports 2015; 12 (09) 1385-1390
  • 90 Ruan J, Hirai H, Yang D. , et al. Efficient gene editing at major CFTR mutation loci. Mol Ther Nucleic Acids 2019; 16: 73-81
  • 91 Sanz DJ, Hollywood JA, Scallan MF, Harrison PT. Cas9/gRNA targeted excision of cystic fibrosis-causing deep-intronic splicing mutations restores normal splicing of CFTR mRNA. PLoS One 2017; 12 (09) e0184009
  • 92 Zhou ZP, Yang LL, Cao H. , et al. In vitro validation of a CRISPR-mediated CFTR correction strategy for preclinical translation in pigs. Hum Gene Ther 2019 (e-pub ahead of print). Doi: 10.1089/hum.2019.074