Targeting TGF-β for treatment of osteogenesis imperfecta

I-Wen Song, Sandesh Cs Nagamani, Dianne Nguyen, Ingo Grafe, Vernon Reid Sutton, Francis H Gannon, Elda Munivez, Ming-Ming Jiang, Alyssa Tran, Maegen Wallace, Paul Esposito, Salma Musaad, Elizabeth Strudthoff, Sharon McGuire, Michele Thornton, Vinitha Shenava, Scott Rosenfeld, Shixia Huang, Roman Shypailo, Eric Orwoll, Brendan Lee, I-Wen Song, Sandesh Cs Nagamani, Dianne Nguyen, Ingo Grafe, Vernon Reid Sutton, Francis H Gannon, Elda Munivez, Ming-Ming Jiang, Alyssa Tran, Maegen Wallace, Paul Esposito, Salma Musaad, Elizabeth Strudthoff, Sharon McGuire, Michele Thornton, Vinitha Shenava, Scott Rosenfeld, Shixia Huang, Roman Shypailo, Eric Orwoll, Brendan Lee

Abstract

BACKGROUNDCurrently, there is no disease-specific therapy for osteogenesis imperfecta (OI). Preclinical studies demonstrate that excessive TGF-β signaling is a pathogenic mechanism in OI. Here, we evaluated TGF-β signaling in children with OI and conducted a phase I clinical trial of TGF-β inhibition in adults with OI.METHODSHistology and RNA-Seq were performed on bones obtained from children. Gene Ontology (GO) enrichment assay, gene set enrichment analysis (GSEA), and Ingenuity Pathway Analysis (IPA) were used to identify dysregulated pathways. Reverse-phase protein array, Western blot, and IHC were performed to evaluate protein expression. A phase I study of fresolimumab, a TGF-β neutralizing antibody, was conducted in 8 adults with OI. Safety and effects on bone remodeling markers and lumbar spine areal bone mineral density (LS aBMD) were assessed.RESULTSOI bone demonstrated woven structure, increased osteocytes, high turnover, and reduced maturation. SMAD phosphorylation was the most significantly upregulated GO molecular event. GSEA identified the TGF-β pathway as the top activated signaling pathway, and IPA showed that TGF-β1 was the most significant activated upstream regulator mediating the global changes identified in OI bone. Treatment with fresolimumab was well-tolerated and associated with increases in LS aBMD in participants with OI type IV, whereas participants with OI type III and VIII had unchanged or decreased LS aBMD.CONCLUSIONIncreased TGF-β signaling is a driver pathogenic mechanism in OI. Anti-TGF-β therapy could be a potential disease-specific therapy, with dose-dependent effects on bone mass and turnover.TRIAL REGISTRATIONClinicalTrials.gov NCT03064074.FUNDINGBrittle Bone Disorders Consortium (U54AR068069), Clinical Translational Core of Baylor College of Medicine Intellectual and Developmental Disabilities Research Center (P50HD103555) from National Institute of Child Health and Human Development, USDA/ARS (cooperative agreement 58-6250-6-001), and Sanofi Genzyme.

Keywords: Bone disease; Clinical Trials; Drug therapy; Therapeutics.

Figures

Figure 1. Transcriptomic and bioinformatics analyses demonstrate…
Figure 1. Transcriptomic and bioinformatics analyses demonstrate activation of TGF-β signaling in OI type III bone.
(A) Principal component analysis (PCA) plot of transcriptomic data from non-OI and OI type III bones in 3 principal component dimensions. (B) Hierarchical clustering based on Euclidian distance using RPKM of all non-OI and OI type III bone data. Blue, downregulated; yellow, upregulated. (C) Gene set enrichment plot demonstrated activation of TGF-β signaling. C18, C14, and C15 represent 3 biologically distinct non-OI bone samples. OI85, OI33, and OI31 represent 3 biologically distinct OI type III bone samples. The expression pattern of genes involved in the TGF-β gene set in the analysis database is shown. NES, normalized enrichment score; FDR, false discovery rate. Blue, downregulated; red, upregulated.
Figure 2. Increased phosphorylated SMAD2 in OI…
Figure 2. Increased phosphorylated SMAD2 in OI type III bone.
(A) IHC staining of phosphorylated SMAD2 (pSMAD2) in non-OI and OI type III bone sections. Higher-magnification images are shown in black boxes on the bottom right. Increase in pSMAD2 signal was detected in all OI samples, especially in the osteocytes. Scale bar: 20 μm. (B) Western blot of p-SMAD2 and total SMAD2 (T-SMAD2) in protein extracted from non-OI and OI type III bone. A total of 50 μg protein was loaded. One OI bone sample (OI62) was treated with calf-intestinal alkaline phosphatase (CIP) to remove phosphorylation signal to serve as a negative control for accurate pSMAD2 signal (indicated by arrowhead). See complete unedited blots in the supplemental material. (C) Quantification of Western blot in B, showing the ratio of phosphorylated (phospho) versus total SMAD2. Data are shown as the mean ± SD. GAPDH was used as loading control. C, non-OI (n = 3); OI, OI type III (n = 5).
Figure 3. Effect of fresolimumab on bone…
Figure 3. Effect of fresolimumab on bone turnover markers and bone density.
The top row shows of serum levels of osteocalcin (Ocn), C-terminal telopeptide (CTX), and N-terminal propeptide of type 1 procollagen (P1NP) at each time point. The bottom row shows percentage changes in these markers of bone turnover as compared with baseline values. The solid lines with circles represent results for the 1 mg/kg dose cohort (n = 4), and the dotted lines with squares represent results for the 4 mg/kg dose cohort (n = 4). Data are shown as the mean ± SEM. *P < 0.05, GLM association.
Figure 4. Effect of fresolimumab on LS…
Figure 4. Effect of fresolimumab on LS aBMD.
The percentage change in LS aBMD in (A) the 1 mg/kg dose cohort (n = 4) and (B) the 4 mg/kg dose cohort (n = 4). (C) Average aBMD changes at each time point based on dose. In B, aBMD could not be assessed in FR012 at the 90-day time point; therefore, the result is shown as a dotted line. In C, the solid lines represent results for the 1 mg/kg dose cohort, and the dotted lines represent results for the 4 mg/kg dose cohort. Data are shown as the mean ± SEM.
Figure 5. CONSORT flow diagram depicting screening,…
Figure 5. CONSORT flow diagram depicting screening, enrollment, and follow up of participants in the trial.

References

    1. Marini JC, et al. Osteogenesis imperfecta. Nat Rev Dis Primers. 2017;3:17052. doi: 10.1038/nrdp.2017.52.
    1. Marom R, et al. Pharmacological and biological therapeutic strategies for osteogenesis imperfecta. Am J Med Genet C Semin Med Genet. 2016;172(4):367–383. doi: 10.1002/ajmg.c.31532.
    1. Patel RM, et al. A cross-sectional multicenter study of osteogenesis imperfecta in North America - results from the linked clinical research centers. Clin Genet. 2015;87(2):133–140. doi: 10.1111/cge.12409.
    1. Rossi V, et al. Osteogenesis imperfecta: advancements in genetics and treatment. Curr Opin Pediatr. 2019;31(6):708–715. doi: 10.1097/MOP.0000000000000813.
    1. Tam A, et al. A multicenter study to evaluate pulmonary function in osteogenesis imperfecta. Clin Genet. 2018;94(6):502–511. doi: 10.1111/cge.13440.
    1. Adami S, et al. Intravenous neridronate in adults with osteogenesis imperfecta. J Bone Miner Res. 2003;18(1):126–130. doi: 10.1359/jbmr.2003.18.1.126.
    1. Bishop N. Characterising and treating osteogenesis imperfecta. Early Hum Dev. 2010;86(11):743–746. doi: 10.1016/j.earlhumdev.2010.08.002.
    1. Bishop N, et al. Risedronate in children with osteogenesis imperfecta: a randomised, double-blind, placebo-controlled trial. Lancet. 2013;382(9902):1424–1432. doi: 10.1016/S0140-6736(13)61091-0.
    1. Chevrel G, et al. Effects of oral alendronate on BMD in adult patients with osteogenesis imperfecta: a 3-year randomized placebo-controlled trial. J Bone Miner Res. 2006;21(2):300–306.
    1. DiMeglio LA, Peacock M. Two-year clinical trial of oral alendronate versus intravenous pamidronate in children with osteogenesis imperfecta. J Bone Miner Res. 2006;21(1):132–140.
    1. Gatti D, et al. Intravenous neridronate in children with osteogenesis imperfecta: a randomized controlled study. J Bone Miner Res. 2005;20(5):758–763.
    1. Gatti D, et al. Teriparatide treatment in adult patients with osteogenesis imperfecta type I. Calcif Tissue Int. 2013;93(5):448–452. doi: 10.1007/s00223-013-9770-2.
    1. Glorieux FH, et al. Cyclic administration of pamidronate in children with severe osteogenesis imperfecta. N Engl J Med. 1998;339(14):947–952. doi: 10.1056/NEJM199810013391402.
    1. Rauch F, et al. Risedronate in the treatment of mild pediatric osteogenesis imperfecta: a randomized placebo-controlled study. J Bone Miner Res. 2009;24(7):1282–1289. doi: 10.1359/jbmr.090213.
    1. Rauch F, et al. Bone mass, size, and density in children and adolescents with osteogenesis imperfecta: effect of intravenous pamidronate therapy. J Bone Miner Res. 2003;18(4):610–614. doi: 10.1359/jbmr.2003.18.4.610.
    1. Orwoll ES, et al. Evaluation of teriparatide treatment in adults with osteogenesis imperfecta. J Clin Invest. 2014;124(2):491–498. doi: 10.1172/JCI71101.
    1. Hoyer-Kuhn H, et al. Safety and efficacy of denosumab in children with osteogenesis imperfecta — a first prospective trial. J Musculoskelet Neuronal Interact. 2016;16(1):24–32.
    1. Bains JS, et al. A multicenter observational cohort study to evaluate the effects of bisphosphonate exposure on bone mineral density and other health outcomes in osteogenesis imperfecta. JBMR Plus. 2019;3(5):e10118. doi: 10.1002/jbm4.10118.
    1. Anissipour AK, et al. Behavior of scoliosis during growth in children with osteogenesis imperfecta. J Bone Joint Surg Am. 2014;96(3):237–243. doi: 10.2106/JBJS.L.01596.
    1. Rauch F, et al. Intracortical remodeling during human bone development — a histomorphometric study. Bone. 2007;40(2):274–280. doi: 10.1016/j.bone.2006.09.012.
    1. Dwan K, et al. Bisphosphonate therapy for osteogenesis imperfecta. Cochrane Database Syst Rev. 2014;10(10):CD005088.
    1. Dwan K, et al. Bisphosphonate therapy for osteogenesis imperfecta. Cochrane Database Syst Rev. 2016;10(7):CD005088.
    1. Shi CG, et al. Efficacy of bisphosphonates on bone mineral density and fracture rate in patients with osteogenesis imperfecta: a systematic review and meta-analysis. Am J Ther. 2016;23(3):e894–e904. doi: 10.1097/MJT.0000000000000236.
    1. Lim J, et al. Genetic causes and mechanisms of Osteogenesis Imperfecta. Bone. 2017;102:40–49. doi: 10.1016/j.bone.2017.02.004.
    1. Grafe I, et al. Excessive transforming growth factor-β signaling is a common mechanism in osteogenesis imperfecta. Nat Med. 2014;20(6):670–675. doi: 10.1038/nm.3544.
    1. Nijhuis WH, et al. Current concepts in osteogenesis imperfecta: bone structure, biomechanics and medical management. J Child Orthop. 2019;13(1):1–11. doi: 10.1302/1863-2548.13.180190.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–15550. doi: 10.1073/pnas.0506580102.
    1. Hannan FM, et al. Genetic approaches to metabolic bone diseases. Br J Clin Pharmacol. 2019;85(6):1147–1160. doi: 10.1111/bcp.13803.
    1. Rachner TD, et al. Osteoporosis: now and the future. Lancet. 2011;377(9773):1276–1287. doi: 10.1016/S0140-6736(10)62349-5.
    1. Tauer JT, et al. Effect of anti-TGF-β treatment in a mouse model of severe osteogenesis imperfecta. J Bone Miner Res. 2019;34(2):207–214. doi: 10.1002/jbmr.3617.
    1. Greene B, et al. Inhibition of TGF-β increases bone volume and strengthin a mouse model of osteogenesis imperfecta. JBMR Plus. 2021;5(9):e10530.
    1. Kaupp S, et al. Combination therapy in the Col1a2(G610C) mouse model of Osteogenesis Imperfecta reveals an additive effect of enhancing LRP5 signaling and inhibiting TGFbeta signaling on trabecular bone but not on cortical bone. Bone. 2020;131:115084. doi: 10.1016/j.bone.2019.115084.
    1. Langdahl B, et al. Bone modeling and remodeling: potential as therapeutic targets for the treatment of osteoporosis. Ther Adv Musculoskelet Dis. 2016;8(6):225–235. doi: 10.1177/1759720X16670154.
    1. Morris JC, et al. Phase I study of GC1008 (fresolimumab): a human anti-transforming growth factor-beta (TGFbeta) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS One. 2014;9(3):e90353. doi: 10.1371/journal.pone.0090353.
    1. Vincenti F, et al. A phase 2, double-blind, placebo-controlled, randomized study of fresolimumab in patients with steroid-resistant primary focal segmental glomerulosclerosis. Kidney Int Rep. 2017;2(5):800–810. doi: 10.1016/j.ekir.2017.03.011.
    1. Rice LM, et al. Fresolimumab treatment decreases biomarkers and improves clinical symptoms in systemic sclerosis patients. J Clin Invest. 2015;125(7):2795–2807. doi: 10.1172/JCI77958.
    1. Formenti SC, et al. Focal irradiation and systemic TGFβ blockade in metastatic breast cancer. Clin Cancer Res. 2018;24(11):2493–2504. doi: 10.1158/1078-0432.CCR-17-3322.
    1. Strauss J, et al. Bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1, in patients with human papillomavirus-associated malignancies. J Immunother Cancer. 2020;8(2):e001395. doi: 10.1136/jitc-2020-001395.
    1. Tao JJ, et al. First-in-human phase I study of the activin A inhibitor, STM 434, in patients with granulosa cell ovarian cancer and other advanced solid tumors. Clin Cancer Res. 2019;25(18):5458–5465. doi: 10.1158/1078-0432.CCR-19-1065.
    1. Glorieux FH, et al. BPS804 anti-sclerostin antibody in adults with moderate osteogenesis imperfecta: results of a randomized phase 2a trial. J Bone Miner Res. 2017;32(7):1496–1504. doi: 10.1002/jbmr.3143.
    1. Eric T, et al. Proceedings of the 2020 Rare Bone Disease Working Group. JBMR Plus. 2021;5(s1):e10455
    1. Martinez-Hackert E, et al. Receptor binding competition: A paradigm for regulating TGF-β family action. Cytokine Growth Factor Rev. 2021;57:39–54. doi: 10.1016/j.cytogfr.2020.09.003.
    1. Chou CH, et al. Direct assessment of articular cartilage and underlying subchondral bone reveals a progressive gene expression change in human osteoarthritic knees. Osteoarthritis Cartilage. 2013;21(3):450–461. doi: 10.1016/j.joca.2012.11.016.
    1. Bu W, et al. Mammary precancerous stem and non-stem cells evolve into cancers of distinct subtypes. Cancer Res. 2019;79(1):61–71. doi: 10.1158/0008-5472.CAN-18-1087.

Source: PubMed

3
Abonner