Therapeutic Intervention in Multiple Sclerosis with Alpha B-Crystallin: A Randomized Controlled Phase IIa Trial

Johannes M van Noort, Malika Bsibsi, Peter J Nacken, Richard Verbeek, Edna H G Venneker, Johannes M van Noort, Malika Bsibsi, Peter J Nacken, Richard Verbeek, Edna H G Venneker

Abstract

As a molecular chaperone and activator of Toll-like receptor 2-mediated protective responses by microglia and macrophages, the small heat shock protein alpha B-crystallin (HspB5) exerts therapeutic effects in different animal models for neuroinflammation, including the model for multiple sclerosis (MS). Yet, HspB5 can also stimulate human antigen-specific memory T cells to release IFN-γ, a cytokine with well-documented detrimental effects during MS. In this study, we explored in a Phase IIa randomized clinical trial the therapeutic application of HspB5 in relapsing-remitting MS (RR-MS), using intravenous doses sufficient to support its protective effects, but too low to trigger pathogenic memory T-cell responses. These sub-immunogenic doses were selected based on in vitro analysis of the dose-response profile of human T cells and macrophages to HspB5, and on the immunological effects of HspB5 in healthy humans as established in a preparatory Phase I study. In a 48-week randomized, placebo-controlled, double-blind Phase IIa trial, three bimonthly intravenous injections of 7.5, 12.5 or 17.5 mg HspB5 were found to be safe and well tolerated in RR-MS patients. While predefined clinical endpoints did not differ significantly between the relatively small groups of MS patients treated with either HspB5 or placebo, repeated administration especially of the lower doses of HspB5 led to a progressive decline in MS lesion activity as monitored by magnetic resonance imaging (MRI), which was not seen in the placebo group. Exploratory linear regression analysis revealed this decline to be significant in the combined group receiving either of the two lower doses, and to result in a 76% reduction in both number and total volumes of active MRI lesions at 9 months into the study. These data provide the first indication for clinical benefit resulting from intervention in RR-MS with HspB5.

Trial registration: ClinicalTrials.gov Phase I: NCT02442557; Phase IIa: NCT02442570.

Conflict of interest statement

Competing Interests: JMvN, MB, PJN and RV are paid employees of Delta Crystallon BV; EHGV is a paid consultant of Delta Crystallon BV. JMvN holds equity in Delta Crystallon BV. Intervention in multiple sclerosis with HspB5 is the subject of international patent application PCT/NL2014/050383.

Figures

Fig 1. Phase IIa study profile.
Fig 1. Phase IIa study profile.
Fig 2. Selection of therapeutic intravenous doses…
Fig 2. Selection of therapeutic intravenous doses of HspB5 in humans.
Therapeutic application of HspB5 in humans relies on activation of M2-like protective microglia and macrophage responses, while avoiding pathogenic IFN-γresponses by memory T cells. In Fig 2A, the existence of a substantial HspB5-reactive memory T-cell repertoire in humans is illustrated by a representative fresh PBMC sample from a healthy subject. Proliferative responses to HspB5 of CD45RO+ memory T cells in this sample are reflected by proliferation-induced dilution of the fluorescent tracer CFSE over 9 days in culture. With CFSEdim CD45RO+ T cells representing proliferated memory T cells, Fig 1A illustrates that a substantial part of all human memory T cells, an estimated one in about every 4,000 cells, do indeed respond to HspB5. In Fig 2B, it is illustrated, again with a representative example, that human T cells are triggered by HspB5 to release IFN-γ, but only when the HspB5 concentration exceeds a threshold of about 20 μg/mL. The protective macrophage response, on the other hand, is already activated by markedly lower μg/mL-concentrations of HspB5, as exemplified by IL-10 secretion as marker for protective responses. At these low sub-immunogenic concentrations, HspB5 can therefore be therapeutically exploited in humans. In Fig 3C, it is shown that single intravenous doses of up to 37.5 mg HspB5 leads to such sub-immunogenic peak serum concentrations in humans, remaining well below the 20-μg/mL threshold. Fig 3C shows the mean ± standard deviation peak serum concentrations of HspB5 found in groups of 8 healthy subjects 10–20 min after receiving varying amounts of intravenous HspB5 during the Phase I study.
Fig 3. Effects of intravenous HspB5 on…
Fig 3. Effects of intravenous HspB5 on peripheral T-cell reactivity to HspB5 itself, and tetanus toxoid.
Fig 3 illustrates the proliferative response in vitro of CD4+ (A) as well as CD45RO+ memory T-cells (B) to either 50 μg/mL HspB5 or 0.2 μg/mL tetanus toxoid as a control antigen, at various time points after a single intravenous dose of 12.5 mg HspB5 in 8 healthy subjects, as evaluated using a CFSE assay during Phase I. Percentages of proliferated CFSEdim cells are expressed relative to all other lymphocytes detected by flowcytometry in the sample after 9 days in culture, including CD4/CD45RO-negative cells. The results not only confirm that following an intravenous dose of 12.5 mg HspB5, its levels remain at sub-immunogenic levels, but even illustrate significant suppressive effect on in vitro T-cell reactivity that is markedly more lasting for HspB5-reactive T cells than for tetanus toxoid-reactive T cells. Background proliferation found in cultures without any antigen was well below 1% in all cases. *: p< 0.05; ***: p<0.005.
Fig 4. The impact of intravenous HspB5…
Fig 4. The impact of intravenous HspB5 on active MRI lesions in RR-MS patients.
RR-MS patients received three bimonthly intravenous administrations of either PBS as placebo, or HspB5 at the indicated doses, and mean numbers of Gd+ T1 lesions for each group were evaluated by MRI over 36 weeks. Fig 4 shows mean values ± standard deviation, as calculated for the full analysis set of patients, including readings for 7 to 8 subjects at different time points for each group except for the 7.5-mg group, which included readings for 4 to 8 subjects. Linear regression analysis was used to evaluate the trends of change in each group over 36 weeks; p values reflect the probability that the deviation of such trends from a horizontal line is a chance event. Arrows indicate times of administration of placebo or HspB5.
Fig 5. Decline in MRI lesions RR-MS…
Fig 5. Decline in MRI lesions RR-MS patients treated with the two lower doses of HspB5.
RR-MS patients received three bimonthly intravenous administrations of either 7.5 or 12.5 mg HspB5, and mean numbers as well as total volumes of Gd+ T1 lesions for the combined groups were evaluated by MRI over the course of 36 weeks. Data show mean values ± standard deviation, as calculated for the full analysis set of patients, including readings for 11 to 14 subjects at different time points. Linear regression analysis was used to evaluate the trends of change over 36 weeks; p values reflect the probability that the deviation of such trends from a horizontal line is a chance event. Arrows indicate times of administration of HspB5.

References

    1. Haider L, Fischer MT, Frischer JM, Bauer J, Höftberger R, Botond G, et al. Oxidative damage in multiple sclerosis lesions. Brain 2011; 134: 1914–1924. 10.1093/brain/awr128
    1. Goldbaum O, Richter-Landsberg C. Stress proteins in oligodendrocytes: differential effects of heat shock and oxidative stress. J Neurochem 2001; 78: 1233–12342.
    1. van Noort JM, van Sechel AC, Bajramović JJ, el Ouagmiri M, Polman CH, Lassmann H, et al. The small heat-shock protein alpha B-crystallin as candidate autoantigen in multiple sclerosis. Nature 1995; 375: 798–801.
    1. Bajramović JJ, Lassmann H, van Noort JM. Expression of alpha B-crystallin in glia cells during lesional development in multiple sclerosis. J Neuroimmunol 1997; 78: 143–151.
    1. Chabas D, Baranzini SE, Mitchell D, Bernard CC, Rittling SR, Denhardt DT, et al. The influence of the proinflammatory cytokine, osteopontin, on autoimmune demyelinating disease. Science 2001; 294: 1731–1735.
    1. Tajouri L, Mellick AS, Ashton KJ, Tannenberg AE, Nagra RM, Tourtellotte WW, et al. Quantitative and qualitative changes in gene expression patterns characterize the activity of plaques in multiple sclerosis. Brain Res Mol Brain Res 2003; 119: 170–183.
    1. Sinclair C, Mirakhur M, Kirk J, Farrell M, McQuaid S. Up-regulation of osteopontin and alpha B-crystallin in the normal-appearing white matter of multiple sclerosis: an immunohistochemical study utilizing tissue microarrays. Neuropathol Appl Neurobiol 2005; 31: 292–303.
    1. Kagitani-Shimono K, Mohri I, Oda H, Ozono K, Suzuki K, Urade Y, et al. Lipocalin-type prostaglandin D synthase (beta-trace) is upregulated in the alpha B-crystallin-positive oligodendrocytes and astrocytes in the chronic multiple sclerosis. Neuropathol Appl Neurobiol 2006; 32: 64–73.
    1. van Noort JM, Bsibsi M, Gerritsen WH, van der Valk P, Bajramović JJ, Steinman L, et al. Alpha B-crystallin is a target for adaptive immune responses and a trigger of innate responses in preactive multiple sclerosis lesions. J Neuropathol Exp Neurol 2010; 69: 694–703. 10.1097/NEN.0b013e3181e4939c
    1. Masilamoni JG, Jesudason EP, Baben B, Jebaraj CE, Dhandayuthapani S, Jayakumar R. Molecular chaperone alpha-crystallin prevents detrimental effects of neuroinflammation. Biochim Biophys Acta 2006; 1762: 284–293.
    1. Ousman SS, Tomooka BH, van Noort JM, Wawrousek EF, O'Connor KC, Hafler DA, et al. Protective and therapeutic role for alpha B-crystallin in autoimmune demyelination. Nature 2007; 448: 474–479.
    1. Arac SE, Brownell JB, Rothbard C, Chen RM, Ko MP, Pereira GW, et al. Systemic augmentation of alpha B-crystallin provides therapeutic benefit twelve hours post-stroke onset via immune modulation. Proc Natl Acad Sci USA 2011; 108: 13287–13292. 10.1073/pnas.1107368108
    1. Pangratz-Fuehrer S, Kaur K, Ousman SS, Steinman L, Liao YL. Functional rescue of experimental ischemic optic neuropathy with αB-crystallin. Eye 2011; 25: 809–817. 10.1038/eye.2011.42
    1. Klopstein E, Santos-Nogueira A, Francos-Quijorna I, Redensek A, David S, Navarro X, et al. Beneficial effects of αB-crystallin in spinal cord contusion injury. J Neurosci 2012; 32: 14478–14488. 10.1523/JNEUROSCI.0923-12.2012
    1. Ying X, Zhang J, Wang Y, Wu N, Wang Y, Yew DT. Alpha-crystallin protected axons from optic nerve degeneration after crushing in rats. J Mol Neurosci 2008; 35: 253–258. 10.1007/s12031-007-9010-1
    1. Wu N, Yu J, Chen S, Xu J, Ying X, Ye M, et al. α-Crystallin protects RGC survival and inhibits microglial activation after optic nerve crush. Life Sci 2014; 94: 17–23. 10.1016/j.lfs.2013.10.034
    1. Kurnellas MP, Brownell SE, Su L, Malkovskiy AV, Rajadas J, Dolganov G, et al. Chaperone activity of small heat shock proteins underlies therapeutic efficacy in experimental autoimmune encephalomyelitis. J Biol Chem 2012; 287: 36423–36434. 10.1074/jbc.M112.371229
    1. Wang YH, Wang DW, Wu N, Wang Y, Yin ZQ. Alpha-crystallin promotes rat retinal neurite growth on myelin substrates in vitro. Ophthalmic Res 2011; 45: 164–168. 10.1159/000319944
    1. Wang YH, Wang DW, Wu N, Wang Y, Yin ZQ. α-Crystallin promotes rat axonal regeneration through regulation of RhoA/rock/cofilin/MLC signaling pathways. J Mol Neurosci 2012; 46: 138–144. 10.1007/s12031-011-9537-z
    1. Bsibsi M, Holtman IR, Gerritsen WH, Eggen BJ, Boddeke E, van der Valk P, et al. Alpha-B-crystallin induces an immune-regulatory and antiviral microglial response in preactive multiple sclerosis lesions. J Neuropathol Exp Neurol 2013; 72: 970–979. 10.1097/NEN.0b013e3182a776bf
    1. van Noort JM, Bsibsi M, Nacken PJ, Gerritsen WH, Amor S, Holtman IR, et al. Activation of an immune-regulatory macrophage response and inhibition of lung inflammation in a mouse model of COPD using heat-shock protein alpha B-crystallin-loaded PLGA microparticles. Biomaterials 2013; 34: 831–840. 10.1016/j.biomaterials.2012.10.028
    1. Bajramović JJ, Plomp AC, van der Goes A, Koevoets C, Newcombe J, Cuzner ML, et al. Presentation of alpha B-crystallin to T cells in active multiple sclerosis lesions: an early event following inflammatory demyelination. J Immunol 2000; 164: 4359–4366.
    1. Bsibsi M, Peferoen LA, Holtman IR, Nacken PJ, Gerritsen WH, Witte ME, et al. Demyelination during multiple sclerosis is associated with combined activation of microglia/macrophages by IFN-γ and alpha B-crystallin. Acta Neuropathol 2014; 128: 215–229. 10.1007/s00401-014-1317-8
    1. Ransohoff RM, Engelhardt B. The anatomical and cellular basis of immune surveillance in the central nervous system. Nat Rev Immunol 2012; 12: 623–635. 10.1038/nri3265
    1. Panitch HS, Hirsch RL, Haley AS, Johnson KP. Exacerbations of multiple sclerosis in patients treated with gamma interferon. Lancet 1987; 1: 893–895.
    1. Polman CH, Reingold SC, Banwell B, Clanet M, Cohen JA, Filippi M, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol 2011: 69: 292–302. 10.1002/ana.22366
    1. van Noort JM, Verbeek R, Meilof JF, Polman CH, Amor S. Autoantibodies against alpha B-crystallin, a candidate autoantigen in multiple sclerosis, are part of a normal human immune repertoire. Mult Scler 2006; 12: 287–293.
    1. Critchfield JM, Racke MK, Zúñiga-Pflücker JC, Cannella B, Raine CS, Goverman J, et al. T cell deletion in high antigen dose therapy of autoimmune encephalomyelitis. Science 1994; 263: 1139–1143.
    1. Racke MK, Critchfield JM, Quigley L, Cannella B, Raine CS, McFarland HF, et al. Intravenous antigen administration as a therapy for autoimmune demyelinating disease. Ann Neurol 1996; 39: 46–56.
    1. Daniel C, von Boehmer H. Extrathymic generation of regulatory T cells—chances and challenges for prevention of autoimmune disease. Adv Immunol 2011; 112: 177–213. 10.1016/B978-0-12-387827-4.00005-X
    1. Hilliard BA, Kamoun M, Ventura E, Rostami A, Mechanisms of suppression of experimental autoimmune encephalomyelitis by intravenous administration of myelin basic protein: role of regulatory spleen cells. Exp Mol Pathol 2000; 68: 29–37.
    1. David A, Crawford F, Garside P, Kappler JW, Marrack P, MacLeod M. Tolerance induction in memory CD4 T cells requires two rounds of antigen-specific activation. Proc Natl Acad Sci USA 2014; 111: 7735–7740. 10.1073/pnas.1406218111
    1. Schwartz M, Kipnis J, Rivest S, Prat A. How do immune cells support and shape the brain in health, disease, and aging? J Neurosci 2013; 33: 17587–17596. 10.1523/JNEUROSCI.3241-13.2013
    1. van Noort JM, Baker D, Amor S. Mechanisms in the development of multiple sclerosis lesions: reconciling autoimmune and neurodegenerative factors. CNS Neurol Disord Drug Targets 2012; 11: 556–569.
    1. van Sechel AC, Bajramović JJ, van Stipdonk JJ, Persoon-Deen C, Geutskens SB, van Noort JM. EBV-induced expression and HLA-DR-restricted presentation by human B cells of alpha B-crystallin, a candidate autoantigen in multiple sclerosis. J Immunol 1999; 162: 129–135.
    1. Akbar AN, Vukmanovic-Stejic M, Taams LS, Macallan DC. The dynamic co-evolution of memory and regulatory CD4+ T cells in the periphery. Nat Rev Immunol. 2007; 7: 231–237.

Source: PubMed

Подписаться