Double-blind, placebo-controlled study of HGF gene therapy in diabetic neuropathy

John A Kessler, A Gordon Smith, Bong-Soo Cha, Sung Hee Choi, James Wymer, Aziz Shaibani, Senda Ajroud-Driss, Aaron Vinik, VM202 DPN-II Study Group, John A Kessler, A Gordon Smith, Bong-Soo Cha, Sung Hee Choi, James Wymer, Aziz Shaibani, Senda Ajroud-Driss, Aaron Vinik, VM202 DPN-II Study Group

Abstract

Objective: To evaluate the safety and efficacy of a plasmid (VM202) containing two human hepatocyte growth factor isoforms given by intramuscular injections in patients with painful diabetic neuropathy.

Methods: In a double-blind, placebo-controlled study, patients were randomized to receive injections of 8 or 16 mg VM202 per leg or placebo. Divided doses were administered on Day 0 and Day 14. The prospective primary outcome was change in the mean pain score measured by a 7 day pain diary. Secondary outcomes included a responder analysis, quality of life and pain measures, and intraepidermal nerve fiber density.

Results: There were no significant adverse events attributable to VM202. Eighty-four patients completed the study. Patients receiving 8 mg VM202 per leg improved the most in all efficacy measures including a significant (P = 0.03) reduction at 3 months in the mean pain score and continued but not statistically significant reductions in pain at 6 and 9 months. Of these patients, 48.4% experienced a ≥50% reduction in pain compared to 17.6% of placebo patients. There were also significant improvements in the brief pain inventory for patients with diabetic peripheral neuropathy and the questionnaire portion of the Michigan Neuropathy Screening Instrument. Patients not on pregabalin or gabapentin had the largest reductions in pain.

Interpretation: VM202 was safe, well tolerated and effective indicating the feasibility of a nonviral gene therapy approach to painful diabetic neuropathy. Two days of treatment were sufficient to provide symptomatic relief with improvement in quality of life for 3 months. VM202 may be particularly beneficial for patients not taking gabapentin or pregabalin.

Figures

Figure 1
Figure 1
Analysis populations in the study. Intent to treat (ITT) population: All subjects enrolled in the study for whom there were data (n = 103). One subject randomized to the high dose (HD) group withdrew consent prior to any treatment. Safety population: All subjects enrolled in the study who received any study injections. The safety population is the same as the ITT population (n = 103). Efficacy population: All subjects in ITT/safety population receiving all study injections, except for those subjects excluded by protocol in a blinded review after a database lock (n = 84). Exclusion criteria were as follows: does not meet the eligibility criteria or does not have 6 month data. Use of protocol prohibited COX1/COX2 inhibitors. Use of opioid medications. Two patients in the HD group (one for shoulder pain, one for chronic cough), and one patient in the LD group (hernia repair) were excluded for this reason.
Figure 2
Figure 2
Mean pain scores on the daily pain diary expressed as change from baseline. (A) Scores for all patients in the efficacy population. The means, standard deviations, medians, minimum scores, and maximum scores are given in Table1. The low-dose (LD) group differed significantly from the P group at 3 months (*P = 0.04) by ANOVA with Dunnett's post hoc test. (B) Mean pain scores for patients not taking gabapentin or pregabalin. The LD group differed significantly from the P group both at 3 months (*P = 0.007) and at 6 months (**P = 0.005).
Figure 3
Figure 3
(A) BPI-DPN pain interference scores expressed as change from baseline. The means, standard deviations, medians, minimum scores, and maximum scores are given in Table2. The LD group differed significantly from the P group both at 3 months (*P = 0.046) and at 6 month (*P = 0.046) by ANOVA with Dunnett's post hoc test. (B) Patient's global impression in change (PGIC) expressed as the % of patients who were much or very much improved. The LD group differed significantly from the P group (*P = 0.008) by the Generalized Estimating Equation for repeated measurements. BPI-DPN, brief pain inventory for patients with diabetic peripheral neuropathy; LD, low dose.
Figure 4
Figure 4
Monofilament test administered as part of the MNSI expressed as the percent of patients improved or normal. Scores are given for (A) right leg, (B) left leg, and (C) both legs. A significant number of subjects in the LD group at 9 months was experienced improvement in sensory threshold of their left leg as compared with the P group (*P = 0.05 by Fischer exact test). MNSI, Michigan Neuropathy Screening Instrument; LD, low dose.

References

    1. Jensen MP, Chodroff MJ, Dworkin RH. The impact of neuropathic pain on health-related quality of life: review and implications. Neurology. 2007;68:1178–1182.
    1. Vinik AI, Nevoret ML, Casellini C, Parson H. Diabetic neuropathy. Endocrinol Metab Clin North Am. 2013;42:747–787.
    1. Davies M, Brophy S, Williams R, Taylor A. The prevalence, severity, and impact of painful diabetic peripheral neuropathy in type 2 diabetes. Diabetes Care. 2006;29:1518–1522.
    1. O'Connor AB. Neuropathic pain: quality-of-life impact, costs and cost effectiveness of therapy. Pharmacoeconomics. 2009;27:95–112.
    1. Callaghan BC, Cheng HT, Stables CL, et al. Diabetic neuropathy: clinical manifestations and current treatments. Lancet Neurol. 2012;11:521–534.
    1. Veves A, Backonja M, Malik RA. Painful diabetic neuropathy: epidemiology, natural history, early diagnosis, and treatment options. Pain Med. 2008;9:660–674.
    1. Funakoshi H, Nakamura T. Identification of HGF-like protein as a novel neurotrophic factor for avian dorsal root ganglion sensory neurons. Biochem Biophys Res Commun. 2001;283:606–612.
    1. Maina F, Hilton MC, Ponzetto C, et al. Met receptor signaling is required for sensory nerve development and HGF promotes axonal growth and survival of sensory neurons. Genes Dev. 1997;11:3341–3350.
    1. Gascon E, Gaillard S, Malapert P, et al. Hepatocyte growth factor-Met signaling is required for Runx1 extinction and peptidergic differentiation in primary nociceptive neurons. J Neurosci. 2010;30:12414–12423.
    1. Hashimoto N, Yamanaka H, Fukuoka T, et al. Expression of HGF and cMet in the peripheral nervous system of adult rats following sciatic nerve injury. Neuroreport. 2001;12:1403–1407.
    1. Cheng C, Guo GF, Martinez JA, et al. Dynamic plasticity of axons within a cutaneous milieu. J Neurosci. 2010;30:14735–14744.
    1. Kato N, Nemoto K, Nakanishi K, et al. Nonviral HVJ (hemagglutinating virus of Japan) liposome-mediated retrograde gene transfer of human hepatocyte growth factor into rat nervous system promotes functional and histological recovery of the crushed nerve. Neurosci Res. 2005;52:299–310.
    1. Ebens A, Brose K, Leonardo ED, et al. Hepatocyte growth factor scatter factor is an axonal chemoattractant and a neurotrophic factor for spinal motor neurons. Neuron. 1996;17:1157–1172.
    1. Wong V, Glass DJ, Arriaga R, et al. Hepatocyte growth factor promotes motor neuron survival and synergizes with ciliary neurotrophic factor. J Biol Chem. 1997;272:5187–5191.
    1. Thompson J, Dolcet X, Hilton M, et al. HGF promotes survival and growth of maturing sympathetic neurons by PI-3 kinase- and MAP kinase-dependent mechanisms. Mol Cell Neurosci. 2004;27:441–452.
    1. Yang XM, Toma JG, Bamji SX, et al. Autocrine hepatocyte growth factor provides a local mechanism for promoting axonal growth. J Neurosci. 1998;18:8369–8381.
    1. Bottaro DP, Rubin JS, Faletto DL, et al. Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science. 1991;251:802–804.
    1. Zheng LF, Wang R, Yu QP, et al. Expression of HGF/c-Met is dynamically regulated in the dorsal root ganglions and spinal cord of adult rats following sciatic nerve ligation. Neurosignals. 2010;18:49–56.
    1. Hashimoto N, Yamanaka H, Fukuoka T, et al. Expression of hepatocyte growth factor in primary sensory neurons of adult rats. Brain Res Mol Brain Res. 2001;97:83–88.
    1. Kato N, Nemoto K, Nakanishi K, et al. Nonviral gene transfer of human hepatocyte growth factor improves streptozotocin-induced diabetic neuropathy in rats. Diabetes. 2005;54:846–854.
    1. Tsuchihara T, Ogata S, Nemoto K, et al. Nonviral retrograde gene transfer of human hepatocyte growth factor improves neuropathic pain-related phenomena in rats. Mol Ther. 2009;17:42–50.
    1. Bussolino F, Di Renzo MF, Ziche M, et al. Hepatocyte growth factor is a potent angiogenic factor which stimulates endothelial cell motility and growth. J Cell Biol. 1992;119:629–641.
    1. Matsumoto K, Nakamura T. Emerging multipotent aspects of hepatocyte growth factor. J Biochem. 1996;119:591–600.
    1. Taniyama Y, Morishita R, Hiraoka K, et al. Therapeutic angiogenesis induced by human hepatocyte growth factor gene in rat diabetic hind limb ischemia model: molecular mechanisms of delayed angiogenesis in diabetes. Circulation. 2001;104:2344–2350.
    1. Taniyama Y, Morishita R, Aoki M, et al. Therapeutic angiogenesis induced by human hepatocyte growth factor gene in rat and rabbit hindlimb ischemia models: preclinical study for treatment of peripheral arterial disease. Gene Ther. 2001;8:181–189.
    1. Lokker NA, Mark MR, Luis EA, et al. Structure- function analysis of hepatocyte growth factor: identification of variants that lack mitogenic activity yet retain high affinity receptor binding. EMBO J. 1992;11:2503–2510.
    1. Shima N, Tsuda E, Goto M, et al. Hepatocyte growth factor and its variant with a deletion of five amino acids are distinguishable in their biological activity and tertiary structure. Biochem Biophys Res Commun. 1994;200:808–815.
    1. Liu ML, Mars WM, Zarnegar R, Michalopoulos GK. Uptake and distribution of hepatocyte growth factor in normal and regenerating adult rat liver. Am J Pathol. 1994;144:129–140.
    1. Pyun WB, Hahn W, Kim DS, et al. Naked DNA expressing two isoforms of hepatocyte growth factor induces collateral artery augmentation in a rabbit model of limb ischemia. Gene Ther. 2010;17:1442–1452.
    1. Hahn W, Pyun WB, Kim DS, et al. Enhanced cardioprotective effects by coexpression of two isoforms of hepatocyte growth factor from naked plasmid DNA in a rat ischemic heart disease model. J Gene Med. 2011;13:549–555.
    1. Ajroud-Driss S, Christiansen M, Allen JA, Kessler JA. Phase 1/2 open-label dose-escalation study of plasmid DNA expressing two isoforms of hepatocyte growth factor in patients with painful diabetic peripheral neuropathy. Mol Ther. 2013;21:1279–1286.
    1. Henry TD, Hirsch AT, Goldman J, et al. Safety of a non-viral plasmid-encoding dual isoforms of hepatocyte growth factor in critical limb ischemia patients: a phase I study. Gene Ther. 2011;18:788–794.
    1. Cleeland CS, Ryan KM. Pain assessment: global use of the Brief Pain Inventory. Ann Acad Med Singapore. 1994;23:129–138.
    1. Zelman DC, Gore M, Dukes E, et al. Validation of a modified version of the brief pain inventory for painful diabetic peripheral neuropathy. J Pain Symptom Manage. 2005;29:401–410.
    1. Moghtaderi A, Bakhshipour A, Rashidi H. Validation of Michigan neuropathy screening instrument for diabetic peripheral neuropathy. Clin Neurol Neurosurg. 2006;108:477–481.
    1. Snedecor SJ, Sudharshan L, Cappelleri JC, et al. Systematic review and meta-analysis of pharmacological therapies for painful diabetic peripheral neuropathy. Pain Pract. 2014;14:167–184.
    1. Christ GJ, Andersson K-E, editors. Regenerative pharmacology. Cambridge, UK: Cambridge University Press; 2013. p. 55.
    1. Calabrese EJ, Baldwin LA. Hormesis: the dose-response revolution. Annu Rev Pharmacol Toxicol. 2003;43:175–197.
    1. Calabrese EJ. Enhancing and regulating neurite outgrowth. Crit Rev Toxicol. 2008;38:391–418.
    1. Konstorum A, Sprowl SA, Waterman ML, et al. Predicting mechanism of biphasic growth factor action on tumor growth using a multi-species model with feedback control. J Coupled Syst Multiscale Dyn. 2013;1:459–467.
    1. Micheva KD, Taylor CP, Smith SJ. Pregabalin reduces the release of synaptic vesicles from cultured hippocampal neurons. Mol Pharmacol. 2006;70:467–476.
    1. Taylor CP, Angelotti T, Fauman E. Pharmacology and mechanism of action of pregabalin: the calcium channel alpha2-delta (alpha2-delta) subunit as a target for antiepileptic drug discovery. Epilepsy Res. 2007;73:137–150.
    1. Bissonette GB, Bae MH, Suresh T, et al. Prefrontal cognitive deficits in mice with altered cerebral cortical GABAergic interneurons. Behav Brain Res. 2014;259:143–151.
    1. Russo AJ, Krigsman A, Jepson B, Wakefield A. Decreased serum hepatocyte growth factor (HGF) in autistic children with severe gastrointestinal disease. Biomark Insights. 2009;4:181–190.
    1. Russo AJ, Pietsch SC. Decreased hepatocyte growth factor (HGF) and gamma aminobutyric acid (GABA) in individuals with obsessive-compulsive disorder (OCD) Biomark Insights. 2013;8:107–114.
    1. Sharma S. Hepatocyte growth factor in synaptic plasticity and Alzheimer's disease. ScientificWorldJournal. 2010;10:457–461.
    1. McDowell I. Alzheimer's disease: insights from epidemiology. Aging (Milano) 2001;13:143–162.
    1. Hebert LE, Weuve J, Scherr PA, Evans DA. Alzheimer disease in the United States (2010–2050) estimated using the 2010 census. Neurology. 2013;80:1778–1783.
    1. Koike H, Ishida A, Shimamura M, et al. Prevention of onset of Parkinson's disease by in vivo gene transfer of human hepatocyte growth factor in rodent model: a model of gene therapy for Parkinson's disease. Gene Ther. 2006;13:1639–1644.
    1. Elbaz A, Bower JH, Maraganore DM, et al. Risk tables for parkinsonism and Parkinson's disease. J Clin Epidemiol. 2002;55:25–31.
    1. Nomura M, Oketa Y, Yasui K, et al. Expression of hepatocyte growth factor in the skin of amyotrophic lateral sclerosis. Acta Neurol Scand. 2012;125:389–397.
    1. Akita H, Takagi N, Ishihara N, et al. Hepatocyte growth factor improves synaptic localization of the NMDA receptor and intracellular signaling after excitotoxic injury in cultured hippocampal neurons. Exp Neurol. 2008;210:83–94.
    1. Keizer D, Fael D, Wierda JMKH, van Wijhe M. Quantitative sensory testing with Von Frey monofilaments in patients with allodynia: what are we quantifying? Clin J Pain. 2008;24:463–466.

Source: PubMed

3
Suscribir