Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2023 Jun 15;21(1):387.
doi: 10.1186/s12967-023-04251-y.

Latozinemab, a novel progranulin-elevating therapy for frontotemporal dementia

Affiliations

Latozinemab, a novel progranulin-elevating therapy for frontotemporal dementia

Michael Kurnellas et al. J Transl Med. .

Abstract

Background: Heterozygous loss-of-function mutations in the progranulin (PGRN) gene (GRN) cause a reduction in PGRN and lead to the development of frontotemporal dementia (FTD-GRN). PGRN is a secreted lysosomal chaperone, immune regulator, and neuronal survival factor that is shuttled to the lysosome through multiple receptors, including sortilin. Here, we report the characterization of latozinemab, a human monoclonal antibody that decreases the levels of sortilin, which is expressed on myeloid and neuronal cells and shuttles PGRN to the lysosome for degradation, and blocks its interaction with PGRN.

Methods: In vitro characterization studies were first performed to assess the mechanism of action of latozinemab. After the in vitro studies, a series of in vivo studies were performed to assess the efficacy of a mouse-cross reactive anti-sortilin antibody and the pharmacokinetics, pharmacodynamics, and safety of latozinemab in nonhuman primates and humans.

Results: In a mouse model of FTD-GRN, the rodent cross-reactive anti-sortilin antibody, S15JG, decreased total sortilin levels in white blood cell (WBC) lysates, restored PGRN to normal levels in plasma, and rescued a behavioral deficit. In cynomolgus monkeys, latozinemab decreased sortilin levels in WBCs and concomitantly increased plasma and cerebrospinal fluid (CSF) PGRN by 2- to threefold. Finally, in a first-in-human phase 1 clinical trial, a single infusion of latozinemab caused a reduction in WBC sortilin, tripled plasma PGRN and doubled CSF PGRN in healthy volunteers, and restored PGRN to physiological levels in asymptomatic GRN mutation carriers.

Conclusions: These findings support the development of latozinemab for the treatment of FTD-GRN and other neurodegenerative diseases where elevation of PGRN may be beneficial. Trial registration ClinicalTrials.gov, NCT03636204. Registered on 17 August 2018, https://clinicaltrials.gov/ct2/show/NCT03636204 .

Keywords: Frontotemporal dementia; Latozinemab; Neuroprotection; Pharmacodynamics; Pharmacokinetics; Phase 1 clinical trial; Progranulin; Safety; Sortilin.

PubMed Disclaimer

Conflict of interest statement

A.M., H.L., and A.R. are employees of Alector LLC and may have an equity interest in Alector, Inc. M.K., T.S., R.P., M.W., and F.Y. were Alector employees at the time of manuscript conception and may have an equity interest in Alector, Inc. E.D.R. has served as a consultant to AGTC and as a DSMB member for Lilly.

Figures

Fig. 1
Fig. 1
In vitro characterization of latozinemab. A Binding curve of sortilin-expressing HEK293T cells by latozinemab. n = 4 technical replicates for latozinemab, n = 5 technical replicates for isotype control. B Percentage of dose-dependent blocking of human PGRN binding to sortilin by latozinemab. n = 3 technical replicates for latozinemab, n = 2 technical replicates for isotype control. C Dose-dependent decrease in surface sortilin expression on U251 cells. n = 3 technical replicates for latozinemab, n = 2 technical replicates for isotype control. D Extracellular PGRN is increased relative to baseline (dashed line) in the presence of latozinemab. n = 3 technical replicates for latozinemab, n = 2 technical replicates for isotype control. All data represent the mean ± SD. 2–5 technical replicates shown for each experiment, each experiment was repeated N > 3, representative data are shown. PE, phycoerythrin; PGRN, progranulin
Fig. 2
Fig. 2
In vivo assessment of an anti-sortilin antibody in a mouse model of GRN haploinsufficiency. A Cell lysate sortilin levels and B plasma PGRN levels from WT and Grn+/− mice that received weekly i.p. injections of S15JG or isotype control for 4.5 weeks. C PGRN levels measured from ISF 33–48 h post injection demonstrated a significant main effect of antibody treatment (P = 0.0089) by Mann–Whitney U tests. (A-C) N = 4–6 per group. All data represent the mean ± SEM. P values are by planned post hoc Tukey’s tests and are adjusted for multiple comparisons. D Schematic of social dominance test. E–H Ratio of the number of matches won by each animal in a given matchup. Male and female pairs were tested at 19–21 months of age (n = 9–12 per group). P values are by Mann–Whitney U tests. All data represent the mean ± SEM. Anti-sort, anti-sortilin antibody S15JG; Ctl, control; ISF, interstitial fluid; PGRN, progranulin
Fig. 3
Fig. 3
Latozinemab decreases sortilin levels in WBCs and increases PGRN levels in plasma and CSF of cynomolgus monkeys. A–B Plasma A and CSF B concentrations of latozinemab antibody as a function of time. C Sortilin concentrations measured as a function of time in WBCs. D–E Plasma D and CSF E PGRN levels measured as a function of time. Male monkeys, 24–50 months of age, were used (n = 3 per group). All data represent the mean ± SEM. Dashed lines in C-E indicate baseline. CSF, cerebrospinal fluid; PGRN, progranulin
Fig. 4
Fig. 4
Pharmacokinetics of latozinemab in HVs and aFTD-GRN participants. A–B Mean ± SD serum concentrations of latozinemab plotted as a function of time for HVs in the SAD groups A and aFTD-GRN participants who received the 60-mg/kg dose B. For the HV SAD groups A, n = 4–13 participants/group/timepoint, and for the aFTD-GRN group B, n = 8 at all timepoints, except for days 15 (n = 7 at 4 h post dose), 85 (n = 4), and 141 (n = 3). C–D Mean ± SD CSF concentrations of latozinemab plotted as a function of time for HVs in the SAD groups C and aFTD-GRN participants D; note that CSF samples were not collected for HVs in the 2-mg/kg and 6-mg/kg dose groups. For the HV SAD groups C, n = 6 participants/group/timepoint, except for the 60-mg/kg dose group prior to dosing (n = 13) and on day 13 (n = 5). For the aFTD-GRN group D, n = 5 at all timepoints, except for day 25 (n = 1). Nominal time after dose is relative to the end of infusion. Placebo-treated subjects were excluded from the PK population. aFTD-GRN, asymptomatic carrier of GRN mutations causative of frontotemporal dementia; CSF, cerebrospinal fluid; HV, healthy volunteer
Fig. 5
Fig. 5
Latozinemab decreases sortilin in WBCs and increases PGRN levels in the plasma and CSF of HVs and aFTD-GRN participants. A–B Median percentage change from baseline in sortilin in WBCs from HVs who were administered a SAD of latozinemab A and from aFTD-GRN participants who received a single injection of 60-mg/kg latozinemab B. For the HV SAD groups A, n = 4–13 participants/group/timepoint and for the aFTD-GRN group B, n = 3–5 participants/timepont. C–D Median percentage change from baseline in PGRN levels in plasma from HVs C and from the aFTD-GRN group D. For the HV SAD groups C, n = 4–13 participants/group/timepoint and for the aFTD-GRN group D, n = 5 at all timepoints, except on days 2 (30 h post dose), 57, 85, and 113 (n = 3) E–F Median percent change from baseline in PGRN levels in CSF from HVs E and from the aFTD-GRN group F; note that CSF samples were not collected for HVs in the 2-mg/kg and 6-mg/kg dose groups. For the HV SAD groups E, n = 2–13 participants/group/timepoint and for the aFTD-GRN group F, n = 5 at baseline and day 13 and n = 1 at day 25. Nominal timepoints are relative to the end of infusion. The percent change from baseline was calculated for each individual based on their own predose value. Dashed lines indicate baseline. aFTD-GRN, asymptomatic carriers of GRN mutations causative of frontotemporal dementia; HV, healthy volunteer; PGRN, progranulin

Similar articles

  • Phase 1 study of latozinemab in progranulin-associated frontotemporal dementia.
    Ward M, Carter LP, Huang JY, Maslyar D, Budda B, Paul R, Rosenthal A. Ward M, et al. Alzheimers Dement (N Y). 2024 Jan 31;10(1):e12452. doi: 10.1002/trc2.12452. eCollection 2024 Jan-Mar. Alzheimers Dement (N Y). 2024. PMID: 38356474 Free PMC article.
  • An anti-sortilin affibody-peptide fusion inhibits sortilin-mediated progranulin degradation.
    Ek M, Nilvebrant J, Nygren PÅ, Ståhl S, Lindberg H, Löfblom J. Ek M, et al. Front Immunol. 2024 Aug 8;15:1437886. doi: 10.3389/fimmu.2024.1437886. eCollection 2024. Front Immunol. 2024. PMID: 39185427 Free PMC article.
  • Development of AL101 (GSK4527226), a progranulin-elevating monoclonal antibody, as a potential treatment for Alzheimer's disease.
    Budda B, Mitra A, Park L, Long H, Kurnellas M, Bien-Ly N, Estacio W, Burgess B, Chao G, Schwabe T, Paul R, Kenkare-Mitra S, Rosenthal A. Budda B, et al. Alzheimers Res Ther. 2025 Jul 25;17(1):174. doi: 10.1186/s13195-025-01817-4. Alzheimers Res Ther. 2025. PMID: 40713730 Free PMC article. Clinical Trial.
  • A systematic review of progranulin concentrations in biofluids in over 7,000 people-assessing the pathogenicity of GRN mutations and other influencing factors.
    Swift IJ, Rademakers R, Finch N, Baker M, Ghidoni R, Benussi L, Binetti G, Rossi G, Synofzik M, Wilke C, Mengel D, Graff C, Takada LT, Sánchez-Valle R, Antonell A, Galimberti D, Fenoglio C, Serpente M, Arcaro M, Schreiber S, Vielhaber S, Arndt P, Santana I, Almeida MR, Moreno F, Barandiaran M, Gabilondo A, Stubert J, Gómez-Tortosa E, Agüero P, Sainz MJ, Gohda T, Murakoshi M, Kamei N, Kittel-Schneider S, Reif A, Weigl J, Jian J, Liu C, Serrero G, Greither T, Theil G, Lohmann E, Gazzina S, Bagnoli S, Coppola G, Bruni A, Quante M, Kiess W, Hiemisch A, Jurkutat A, Block MS, Carlson AM, Bråthen G, Sando SB, Grøntvedt GR, Lauridsen C, Heslegrave A, Heller C, Abel E, Gómez-Núñez A, Puey R, Arighi A, Rotondo E, Jiskoot LC, Meeter LHH, Durães J, Lima M, Tábuas-Pereira M, Lemos J, Boeve B, Petersen RC, Dickson DW, Graff-Radford NR, LeBer I, Sellami L, Lamari F, Clot F, Borroni B, Cantoni V, Rivolta J, Lleó A, Fortea J, Alcolea D, Illán-Gala I, Andres-Cerezo L, Van Damme P, Clarimon J, Steinacker P, Feneberg E, Otto M, van der Ende EL, van Swieten JC, Seelaar H, Zetterberg H, Sogorb-Esteve A, Rohrer JD. Swift IJ, et al. Alzheimers Res Ther. 2024 Mar 28;16(1):66. doi: 10.1186/s13195-024-01420-z. Alzheimers Res Ther. 2024. PMID: 38539243 Free PMC article.
  • Progranulin as a therapeutic target for dementia.
    Galimberti D, Fenoglio C, Scarpini E. Galimberti D, et al. Expert Opin Ther Targets. 2018 Jul;22(7):579-585. doi: 10.1080/14728222.2018.1487951. Epub 2018 Jun 22. Expert Opin Ther Targets. 2018. PMID: 29889573 Review.

Cited by

References

    1. Knopman DS, Roberts RO. Estimating the number of persons with frontotemporal lobar degeneration in the US population. J Mol Neurosci. 2011;45:330–335. doi: 10.1007/s12031-011-9538-y. - DOI - PMC - PubMed
    1. Onyike CU, Diehl-Schmid J. The epidemiology of frontotemporal dementia. Int Rev Psychiatry. 2013;25:130–137. doi: 10.3109/09540261.2013.776523. - DOI - PMC - PubMed
    1. Warren JD, Rohrer JD, Rossor MN. Clinical review. Frontotemporal dementia. BMJ. 2013;347:f4827. doi: 10.1136/bmj.f4827. - DOI - PMC - PubMed
    1. Ljubenkov PA, Miller BL. A clinical guide to frontotemporal dementias. Focus (Am Psychiatr Publ) 2016;14:448–464. - PMC - PubMed
    1. Mann DMA, Snowden JS. Frontotemporal lobar degeneration: pathogenesis, pathology and pathways to phenotype. Brain Pathol. 2017;27:723–736. doi: 10.1111/bpa.12486. - DOI - PMC - PubMed

Publication types

Substances

Associated data

LinkOut - more resources