<<

Research

ISSN: 0300-8207 (Print) 1607-8438 (Online) Journal homepage: https://www.tandfonline.com/loi/icts20

Advances in the pathogenesis and possible treatments for multiple hereditary exostoses from the 2016 international MHE conference

Anne Q. Phan, Maurizio Pacifici & Jeffrey D. Esko

To cite this article: Anne Q. Phan, Maurizio Pacifici & Jeffrey D. Esko (2018) Advances in the pathogenesis and possible treatments for multiple hereditary exostoses from the 2016 international MHE conference, Connective Tissue Research, 59:1, 85-98, DOI: 10.1080/03008207.2017.1394295 To link to this article: https://doi.org/10.1080/03008207.2017.1394295

Published online: 03 Nov 2017.

Submit your article to this journal

Article views: 323

View related articles

View Crossmark data

Citing articles: 1 View citing articles

Full Terms & Conditions of access and use can be found at https://www.tandfonline.com/action/journalInformation?journalCode=icts20 CONNECTIVE TISSUE RESEARCH 2018, VOL. 59, NO. 1, 85–98 https://doi.org/10.1080/03008207.2017.1394295

PROCEEDINGS Advances in the pathogenesis and possible treatments for multiple hereditary exostoses from the 2016 international MHE conference Anne Q. Phana, Maurizio Pacificib, and Jeffrey D. Eskoa aDepartment of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA; bTranslational Research Program in Pediatric Orthopaedics, Division of Orthopaedic , The Children’s Hospital of Philadelphia, Philadelphia, PA, USA

ABSTRACT KEYWORDS Multiple hereditary exostoses (MHE) is an autosomal dominant disorder that affects about 1 in 50,000 Multiple hereditary children worldwide. MHE, also known as hereditary multiple exostoses (HME) or multiple osteochon- exostoses; multiple dromas (MO), is characterized by -capped outgrowths called that develop osteochondromas; EXT1; adjacent to the growth plates of skeletal elements in young patients. These benign tumors can affect EXT2; heparan sulfate; growth plate; skeletal growth plate function, leading to skeletal growth retardation, or deformations, and can encroach on development nerves, , muscles, and other surrounding tissues and cause motion impairment, chronic pain, and early onset osteoarthritis. In about 2–5% of patients, the osteochondromas can become malig- nant and life threatening. Current treatments consist of surgical removal of the most symptomatic tumors and correction of the major skeletal defects, but physical difficulties and chronic pain usually continue and patients may undergo multiple throughout life. Thus, there is an urgent need to find new treatments to prevent or reverse formation. The 2016 International MHE Research Conference was convened to provide a forum for the presentation of the most up-to-date and advanced clinical and basic science data and insights in MHE and related fields; to stimulate the forging of new perspectives, collaborations, and venues of research; and to publicize key scientific findings within the biomedical research community and share insights and relevant information with MHE patients and their families. This report provides a description, review, and assessment of all the exciting and promising studies presented at the Conference and delineates a general roadmap for future MHE research targets and goals.

Introduction threatening, because of their common resistance to chemo- or radiation therapy (5,6). No new osteochondro- Multiple hereditary exostoses (MHE) is a rare, pediatric, mas form after the end of puberty when all the growth and autosomal dominant musculoskeletal disorder that plates close and the skeleton reaches maturity. affects about 1 in 50,000 children worldwide (1,2). Current treatments for MHE mainly consist of surgical MHE, also known as Hereditary Multiple Exostoses removal of the most symptomatic tumors and correction (HME) or Multiple Osteochondromas (MO), is amongst of major skeletal defects such as deformations, limb the most common disorders within the NIH Office of length discrepancies, and joint ankylosis, and patients Rare Diseases classification and one of the most frequent often undergo more than 40–50 surgeries by age 18 (7). causes of tumors. As its name implies, MHE is However, because the osteochondromas are so numerous characterized by osteochondromas, benign cartilage- and often difficult to reach and resect, many are left in capped outgrowths with a bony stem developing adjacent place; consequently, a considerable number of patients to the growth plates of long , vertebrae, ribs, and struggle with pain and physical difficulties through life pelvis in juvenile patients (3,4). The tumors can affect the (8,9). As recent studies indicate, patients can suffer from normal functioning of growth plates, leading to growth additional non-skeletal health problems that include retardation and skeletal deformations. They can also social and learning difficulties, sleep disorders, and neu- impinge on nerves, tendons, blood vessels, muscles, and ropathies (10–12). MHE may also involve alterations in other surrounding tissues, causing motion impairment, postprandial lipid clearance and pancreatic beta-cell chronic pain, and even early onset osteoarthritis. In reserve due to smaller pancreas volume (13,14), making about 2–5% of patients, the osteochondromas transform MHE a syndrome rather than a pure skeletal condition. into malignant that can be life

CONTACT Jeffrey D. Esko [email protected] Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093- 0687, USA. © 2017 Taylor & Francis 86 A. Q. PHAN ET AL.

MHE is linked to heterozygous loss-of-function muta- (35–37). Other studies showed that chon- tions in EXT1 (8q24.1) or EXT2 (11p11-12) (15–17)that drocytes in zebrafish mutants lacking dackel (correspond- encode Golgi-associated glycosyltransferases responsible ing to mammalian Ext2) grew aberrantly and lost polarity, for the biosynthesis of heparan sulfate (HS) (18). HS is a invading surrounding tissues, thus mimicking traits of complex polymerized from alternat- MHE (38). Human MHE growth plate ing N-acetylglucosamine and glucuronic acid residues, chondrocytes display disorganized primary cilia and loss and both EXT1 (the enzyme) and EXT2 (the chaperone- of cellular polarity, suggesting that the cells were unable to like and non-enzymatically active partner of EXT1) are perceive cilia-mediated signals and maintain cell align- needed for its synthesis (19,20). The HS chains are poly- ment and location, thus contributing to osteochondromas merized and covalently linked to small subset of HS-rich (39). Huegel et al. showed that Ext1 and HS regulate (HSPGs) and undergo extensive, concur- perichondrial phenotype and border function in develop- rent chain modifications including sulfation at various ing long bones and that the loss of Ext1 in perichondrial positions and glucuronic acid epimerization, resulting in progenitor cells led to osteochondroma formation (40). chains with a high degree of structural complexity and Boundary defects may be reinforced by changes in biological specificity (21,22). The HSPG family includes microRNAs demonstrated in MHE chondrocytes (41). cell surface proteoglycans (4 syndecans, 6 glypicans, Studies also indicated that the HS deficiency in MHE CD44V3, betaglycan, and CD47), pericellular/extracellu- cartilage and perichondrium could be exacerbated by lar matrix proteoglycans (perlecan, agrin, and Type XVIII abnormally high expression of heparanase (42,43). In collagen), and serglycin (21,23,24). The HSPGs are summary, there has been significant progress in uncover- expressed in a tissue-specific manner and have a number ing aspects of the genetics and cellular pathogenesis of of important developmental and physiologic functions MHE using both animal models and human MHE speci- (24). One of their main roles is to interact with growth mens. However, much remains to be understood factors, morphogens, and receptors, all characterized by mechanistically, and relatively little has been achieved so possessing a specific HS-binding domain (25,26). The far toward the development of treatments to prevent HSPGs regulate protein availability, distribution, turn- osteochondroma formation and/or growth (44). over, and signaling, thus affecting cell adhesion and As pointed out above, MHE is not simply a skeletal migration, cell–cell interactions and communication, disease, but encompasses non-skeletal pathologies (9). cell differentiation and morphogenesis, and lipoprotein Unlike skeletal abnormalities, these additional patholo- metabolism and proteases involved in hemostasis and gies occur with varying frequency and have been dis- development (21,23,24). The EXT deficiency in MHE counted or unappreciated in the past. However, recent results in partial truncation of the HS chains, which human and mouse studies have provided evidence that leads to lower levels of HS in multiple tissues and plasma HS deficiency can in fact cause physiological defects in (27–29), potentially explaining the impact of the muta- kidney organization and podocyte structure (13), neuro- tions on multiple organ systems. nal cell signaling and mouse behavior (45,46), lipid The haploinsufficiency in EXT1 or EXT2 in MHE metabolism (47,48), intestinal epithelial barrier function patients can in itself cause certain pathologies such as (49), pancreatic beta-cell reserve (50), and inflammation lipid metabolism defects and scar formation (14,30), but and infection (51,52). Thus, there is little doubt that HS it is not sufficient to cause osteochondroma formation has encompassing roles in many tissues and organs (21) (31,32). In line with Knudson’s hypothesis of tumorigen- and that the HS deficiency in MHE can provoke a esis (33), osteochondroma formation requires a “second variety of pathologies or deficiencies in patients. hit” such as loss of heterozygosity (LOH), aneuploidy or other genetic changes that would further lower the HS The 5th International Research Conference on levels in affected cells and tissues (34). Murine models for MHE MHE have provided strong and direct evidence for this thesis. Single heterozygous Ext1+/- or Ext2+/- mutant mice The 5th International MHE Research Conference was were found to be largely normal and less than 10% of organized to assess the status of most recent MHE them developed solitary osteochondromas only in ribs research and to provide a springboard for the future of and the penetrance varied with the genetic background the field. Its key and specific goals were to: (i) maintain of the mice (20,29). In sharp contrast, numerous osteo- research momentum; (ii) provide a forum for sharing the at typical anatomical locations developed in most recent and unpublished findings from major labora- double heterozygous Ext1+/-; Ext2+/- mice (29), and in tories working on MHE; (iii) include experts from related mice in which both Ext1 alleles were conditionally and fields to share in this knowledge, provide critical input stochastically ablated in growth plate chondrocytes and and fresh perspectives, and be inspired to work directly on TREATMENTS FOR MULTIPLE HEREDITARY EXOSTOSES 87 this debilitating pediatric disorder individually or in col- Heparan sulfate in MHE and related laboration; (iv) improve our understanding of the medical pathologies and clinical complexities of MHE; and (v) use all the Over 90% of MHE patients bear heterozygous muta- biological and biomedical insights gained in animal tions in EXT1 or EXT2 (53), and direct measurements model systems and from patients and donated specimens of blood and tissue HS levels have shown that the to begin to envision therapeutic approaches for treating patients have a systemic decrease in HS of about 50% MHE by pharmacological, cellular, or genetic means (27). As indicated above, such partial loss of HS may in together or without surgery. To accomplish these objec- itself be sufficient to cause physiologic abnormalities in tives, the Conference was organized around major themes tissues and organs such as the liver (14), but it is not including HS biochemistry and metabolism, skeletogen- sufficient to alter the behavior and functioning of esis and protein signaling, cancer biology, stem cell biol- growth plate and perichondrial cells and provoke osteo- ogy, and therapeutic approaches. It also included a clinical formation (34). The latter appears to session for patients and families. As in past MHE require a much greater loss of HS, and genetic studies Conferences, the 5th Conference provided many oppor- with human osteochondroma specimens and in mouse tunities for brainstorming and promoting collaborations models have solidified this tenet as pointed out above. and synergy amongst laboratories studying MHE and Genomic analyses have indicated that LOH or other related diseases. The program united an eclectic mixture chromosomal rearrangements are detectable in at least of investigators from several clinical and biological dis- a portion of human osteochondroma samples ciplines, enabling the participants to formulate compre- (31,32,54,55), thus likely leading to a steep decrease in hensive and multi-disciplinary concepts in MHE and to HS production within the tumors. In mice, compound envision strategies and goals. Expected outcomes heterozygous loss of Ext1 and Ext2 or conditional abla- included: a refined and more comprehensive understand- tion of both Ext1 alleles are needed to trigger a stereo- ing of MHE as a syndrome; dissemination of the most typic MHE phenotype in which the mice develop current data and insights; sharing of reagents, methods, multiple tumors at multiple and typical locations, and technologies across the research community; and including long bones, vertebrae, and ribs (29,35,36). identification of possible and plausible means for preven- The conclusions stemming from these studies are that tive or therapeutic interventions to counter osteochon- a partial loss of HS seems to be tolerated by growth droma formation and to rectify other MHE-associated plate cartilage and perichondrium and does not alter pathologies. their behaviors and function, but a steeper HS loss The Conference was held in May 2016 at St. Mary’s would render the cells tumorigenic and prone to pro- Medical Center and The Paley Advanced Limb duce osteochondromas. Lengthening Institute in West Palm Beach and consisted Matthew Hilton (Duke University) engineered two of six biomedical and clinical research sessions, followed new mouse models in which loss of both Ext1 alleles by a special session for patients and orthopedic specialists. could be induced by mating “floxed” Ext1 mice (Ext1f/f) Each session had five to six speakers and followed an with Matrilin1 Cre or Col2a1CreTM. The former model interactive workshop format. A total of 85 individuals— inactivated Ext1 in all chondrocytes, but the animals including 35 senior speakers, 13 trainees (PhD, MD, and did not develop osteochondromas; instead, they exhib- BS), representatives from the MHE research foundations, ited skeletal developmental defects similar to those in staff members from the Paley Institute and the University other models in which Ext1 is compromised systemi- of California, and 26 MHE family members—participated cally by reduced expression (e.g., Ext1Gt/Gt)(56). in the meeting. The format, roster, and size of the meeting The second genetic model—Col2a1CreTM;Ext1f/f results were designed to maximize interactions among partici- in a stochastic deletion of both Ext1 alleles within pants in an informal setting. In addition to the scientific chondrocytes and perichondrial cells (both targeted by sessions, a final session for families focused on direct Col2a1Cre)(57). The mutant mice contained islands of interactions with MHE medical specialists to discuss indi- chondrocytes lacking HS and exhibited a high fre- vidual MHE prognosis. Sarah Ziegler, Director of the quency of osteochondroma formation. One interpreta- MHE Research Foundation, chaired this session. tion of the data is that osteochondromas arise from Speakers and attendees hailed from 11 countries on 4 uneven expression of HS across cartilage. continents: the United States, Argentina, Canada, China, Interestingly, systemic transgenic overexpression of France, Germany, Israel, Japan, the Netherlands, Norway, heparanase, the endo- β-D-glucuronidase that cleaves and the United Kingdom. HS at specific subsites in the chain, was found to reduce Major themes and research areas covered at the HS across all tissues and to reduce osteochondroma conference included the following: 88 A. Q. PHAN ET AL. incidence in Col2a1CreTM;Ext1f/f mice, perhaps by per- time. Histological analyses revealed excessive accumu- vasive and pleiotropic effects and collapsing HS gradi- lation of hypertrophic chondrocytes possibly originat- ents. Though intriguing, the data would need to be ing from growth plates, but no mononuclear infiltrates reconciled with the strong endogenous heparanase indicative of inflammation. Osteochondroma-like expression observed in human osteochondromas from lesions developed faster and were more severe in MHE patients (43), possibly suggesting that gene DKOCD4 mice that lacked T cells, indicating that T dosage and/or restricted heparanase expression might cells play an important role in regulating cartilage for- be important. mation and homeostasis. Development of osteochon- Marion Kusche-Gullberg (University of Bergen, droma-like lesions was also influenced by changes in Norway) presented work examining the influence of microbiota and inflammatory stimuli, demonstrating HS chains on bi-directional communication and inter- previously unappreciated roles of the immune system actions that occur between stromal fibroblasts and in cartilage homeostasis (60). Joseph Yost (University tumor cells. An in vitro model was described based on of Utah) reported studies indicating that defects in mixed multicellular three-dimensional spheroids com- immunological cell lineages and wound responses posed of human tumor cells and wild type or Ext1- occurred in zebrafish with mutations in genes encoding deficient fibroblasts. Differential effects in expression Syndecan 2 core protein, Ext2 or HS chain modification profiles were noted in genes involved in tumor cell enzymes. A surprising number of certain homozygous proliferation and invasion, whereas several genes encod- recessive mutants appeared to have normal develop- ing extracellular matrix proteins or those regulating cell ment and morphology and were viable through adult- motility were suppressed (58). The data indicated how hood. Live imaging distinguishing specific immune HS can have non-cell autonomous effects, possibly rele- lineages revealed that the more penetrant mutants had vant to models of MHE in which only small cohorts of underlying defects in specification and propagation of cells undergo stochastic loss of Ext function or to certain immune cell lineages and substandard immune system anatomical sites such as tissue boundaries where the responses to wounding. These findings suggested that levels of HS can vary and be modulated. defects in the immune system and wound healing could Andrea Vortkamp (University of Duisburg, Germany) be possible cofactors in non-skeletal symptoms and presented studies on HS in osteoarthritis and articular osteochondroma formation in MHE patients. cartilage maintenance. Osteoarthritis (OA) is character- ized by progressive loss of articular cartilage due to slow Heparan sulfate, protein signaling, and and chronic degradation of cartilage matrix and loss of skeletogenesis phenotypic expression in chondrocytes (59). Clonal dele- tion of Ext1 in type II collagen-expressing chondrocytes The mechanisms by which osteochondromas exclusively in Col2rtTACre; Ext1e2fl/e2fl mice was found to induce for- form next to the growth plates remain unclear and some- mation of enlarged, a typical cells within articular carti- what controversial. Several studies have suggested that HS lage, as seen in previous studies (37). The abnormal cells loss in MHE causes aberrant distribution and action by were largely devoid of HS, produced an altered extracel- HS-binding signaling proteins and growth factors, includ- lular matrix, and appeared to separate themselves from ing members of the hedgehog, bone morphogenetic pro- surrounding wild type chondrocytes. Surprisingly, the tein (BMP)/transforming growth factor β (TGFβ), clonal loss of HS did not lead to OA during ageing or in fibroblast growth factor (FGF), and Wnt families a surgical OA model, but actually seemed to have a (29,40). Alterations in growth factor distribution and protective effect on cartilage maintenance. It remains action could potentially provoke abnormal behaviors in unclear whether this surprising effect is due to protective chondrocytes and/or neighboring perichondrial cells, release of beneficial growth factors by the HS-deficient leading to osteochondroma initiation and growth (34). cell clones, increased turnover and remodeling of the Hence, one of the most pressing issues in current MHE matrix, or other mechanisms operating within articular research is to uncover the mechanisms by which HS cartilage over time. regulates the normal function of signaling proteins and Two studies presented at the Conference suggested factors (25,26) and how alterations in such key homeo- that the immune system might affect osteochondroma static mechanisms affect skeletal cell behavior and induce formation. Maureen A. McGargill (St. Jude Children’s tumor formation. It is interesting to point out that the Research Hospital) showed that mice with a germline osteochondromas and their growth plate-like cartilage deletion of Erk1 and a conditional deletion of Erk2 caps are usually oriented perpendicularly to the axis of mediated by CD4Cre (DKOCD4 mice) spontaneously the endogenous growth plates, and it is wholly unknown developed multiple osteochondroma-like lesions over how this occurs and what it may mean pathologically. TREATMENTS FOR MULTIPLE HEREDITARY EXOSTOSES 89

Yingzi Yang (Harvard School of Dental Medicine) between osteogenesis and and overall reported that FGF and Wnt5a signaling coordinates skeletal growth through FGF receptors 1 and 2 (Fgfr1 directional proximo-distal elongation and patterning and Fgfr2), both of which are expressed in the osteo- in developing limbs through regulation of planar cell progenitor lineage. Conditional knockout mice lacking polarity (PCP), a mechanism by which cells within both receptors were created using an osteoprogenitor- developing fields such as the limb bud become polar- specific Osx Cre driver. The mutants showed a ~50% ized in a specific direction (61). Genetic evidence was reduction in body weight and bone mass, and impaired provided suggesting that altering the Wnt5a gradient longitudinal skeletal growth due to cell autonomous direction affected the spatial orientation of PCP as loss of FGF signaling. The double knockout mice also revealed by Vangl2 phosphorylation. The data raise showed growth plate defects that appeared to be due to the possibility that the directionality of osteochon- a non-cell autonomous feedback pathway regulating droma formation may reflect orthogonal re- Fgfr3 in growth plate chondrocytes, leading to suppres- orientation of such mechanisms and growth factors/ sion of proliferation (64). Wentian Yang morphogen gradients with respect to endogenous (Brown University) and collaborators conditionally growth plate and perichondrium. deleted Ptpn11 in mouse limb and head mesenchyme Elazar Zelzer (Weizmann Institute of Science, Israel) that encodes the protein tyrosine phosphatase Shp2. discussed proprioceptive mechanosensors and regula- They found that the Shp2-deficient mice exhibited tion of spinal alignment. A series of genetic studies in increased cartilage mass and deficient intramembra- mice in which Runx3 was inactivated selectively in nous and endochondral , suggesting that peripheral sensory neurons induced peripubertal sco- Shp2-deficient OCPs become more frequently chondro- liosis without skeletal dysplasia, which led to the con- cytes than . Interestingly, mosaic Shp2 dele- clusion that these sensors coordinate the alignment of tion at E13.5 in Prrx1CreERt2-expressing OCPs led to the vertebral column. These findings uncover a central osteochondromas and . Mechanistic stu- role for the mechanosensory system in maintaining dies suggested that Shp2 regulates fate determination of spinal alignment and provide a mechanistic explanation OCPs via post-translational phosphorylation and for adolescent idiopathic scoliosis. The spine is not SUMOylation of Sox9, at least in part via the protein usually included in general physical assessment of kinase A signaling pathway. MHE patients (62), but a recent study from Japan Condyles are present at the epiphyseal ends of long strongly indicates that this may need to change (63). bones where they participate in articulation between The authors clinically assessed 50 MHE patients (aver- skeletal elements. Karen Lyons (University of age age 28), used a disease severity classification system California, Los Angeles) reported that mice lacking that considered skeletal deformities and osteochon- the type I TGFβ receptor Alk5 in the growth plate droma number, and closely inspected the spine of (ALK5CKO) exhibited loss of specific condyles and each patient. They found that mild-to-moderate scolio- the development/appearance of cartilaginous protru- sis was actually common amongst the patients. Scoliosis sions in their limbs and also a severe and lethal chon- spanned King type I to type IV in severity, but not type drodysplasia. Preliminary data indicated that the loss of V. Though the data need to be verified in other and Alk5 prevented isotropic expansion of epiphyseal/rest- larger patient cohorts, they point to the possibility that ing zone cartilage and instead promoted growth plate defects in skeletal development and/or mechanosensory column formation. Surprisingly, Alk5 ablation led to mechanisms may affect aspects of spine physiology only a modest reduction in Smad2/3 activation, which during the course and progression of MHE. If con- is downstream from Alk5; instead, there was a massive firmed and further assessed, the data could possibly up-regulation of BMP Smad1/5/8 activation. The data, lead to changes in standard of care of MHE patients along with the considerably milder and viable pheno- in the future. type of mice lacking Smad2/3, suggested the very inter- Chondrocytes and osteoblasts originate from a com- esting conclusion that a major role for Alk5 in cartilage mon mesenchymal precursor, the osteochondropro- might not be to activate the TGFβ pathway, but rather genitor (OCP), and help build the vertebrate skeleton. to prevent excessive BMP signaling. As described The signaling pathways that control lineage decisions in below, Yu Yamaguchi and one of us (Maurizio OCPs are incompletely understood. Two studies Pacifici) have provided evidence that inhibitors of demonstrated the importance of FGF signaling in fate BMP signaling can reduce osteochondroma formation, decision and differentiation in the chondrogenic and signifying that this signaling pathway and/or modula- osteogenic lineages. David M. Ornitz (Washington tion of TGFβ signaling may serve as novel therapeutic University) showed that FGFs regulate the balance strategies for MHE. 90 A. Q. PHAN ET AL.

MHE and related cancers The data raise the interesting possibility that “oncometa- bolites” could contribute to formation (67). The hallmark of MHE is the formation of benign Ernest (Chappie) Conrad (Seattle Children’sHospital) osteochondromas next to the growth plates. EXT muta- discussed the clinical difficulties in diagnosing patients tions occur in other types of cancer as well, and EXT1 with malignant chondrosarcoma and the need for bio- and EXT2 are classified as tumor suppressors (18,65). markers. Equally perplexing is the difference in malignant In about 2–5% of MHE patients, osteochondromas transformation in MHE patients (2–5%) versus patients progress to malignant , but the with /Multiple Endochondromas (10–46%) mechanisms underlying such transformation are (68). Although association of risk varies with the size of unknown (54). Thus, one of the sessions was devoted the primary tumors, other contributing factors remain to discussion of bone and cartilage tumors and the roles unknown. Judith V.M.G. Bovée (Leiden University of HS function and metabolism in tumor formation. Medical Center, Netherlands) discussed other potential Matthew L. Warman (Children’s Hospital, Boston) molecular culprits in cartilage tumors. Secondary muta- gave a keynote presentation entitled “Why don’t all tions causing malignant transformation of osteochondro- cancer causing mutations cause cancer?” A high per- mas or enchondromas may include alterations in the pRb centage of mutations in oncogenes or tumor suppres- and p53 pathways, Hedgehog signaling, metabolic path- sors often cause tissue malformation, but may not ways (e.g. mTOR), apoptosis and survival mechanisms result in cancer. Explanations for this observation (e.g. Bcl family members, survivin), and several tyrosine include phenotypic suppression by surrounding wild kinases (e.g. Src) (69,70). These genes represent potential type cells or the need for mutations or epigenetic targets for chemotherapy that would be especially relevant changes in modifier genes. Some tumors also undergo and beneficial to patients with un-resectable “reversibility”, i.e. the initial tumor growth is subse- chondrosarcomas. quently suppressed, leading to spontaneous regression, Veronique M. Lefebvre (Cleveland Clinic) presented possibly due to secondary mutations or epigenetic work on cell fate specification in skeletogenesis and alterations. Another complicating factor is that cells/ chondrosarcoma. CHIP-seq was used to study enhancer mutations responsible for development of the tumor elements in cartilage specific genes. “Super-enhancers,” may be gone by the time the tumor is biopsied. To genomic regions composed of multiple enhancers bound gain insight into mosaicism and genetic alterations to multiple transcription factors, were found in several taking place in tumor cells, modern techniques for genes involved in cartilage differentiation, including sequencing DNA and RNA from single cells from Sox6 and Sox9 and genes involved in pro- tumors using Nanopore technology are being devel- duction and glycosaminoglycan modification (Acan, oped. Their application to osteochondromas might pro- Chst11, Syndecan 4,andNdst1). Interestingly, Sox 4,11, vide clues about the variable penetrance of and 12 (termed SoxC genes) are expressed in the peri- osteochondromas and the overall MHE severity pheno- chondrium. Inactivation of any of these genes by Prrx1- type in patients bearing identical mutations, including Cre (that targets the early limb bud mesenchymal popu- family members (66). lation) resulted in defects in the growth plate, lack of Isocitrate dehydrogenases are soluble enzymes synovial joints, poor definition of the perichondrium- encoded by IDH1 and IDH2. Somatic mutations in either growth plate boundary, and, interestingly, formation of gene are present in the majority of enchondromas, which cartilaginous outgrowths. Clearly, the SoxC genes are are benign cartilage tumors forming inside (but not next needed to maintain the normal phenotype of perichon- to) the bone and can be precursors to malignant chon- drium and this may be in association with strong Wnt/ drosarcomas. Benjamin A. Alman (Duke University) β-catenin signaling. Decreased SoxC expression would showed that mice expressing the Idh1-R132Q mutation lead to decreased Wnt/β-catenin signaling, which is a in one allele in chondrocytes exhibited disordered growth well-known anti-chondrogenic mechanism (71), and plates, with persistence and scattered distribution of type ectopic chondrogenesis and cartilage formation in peri- X-collagen expressing hypertrophic chondrocytes. chondrium. The data point to the interesting possibility Homozygous mutants did not survive after neonatal that SoxC genes may be part of the mechanisms derailed stages, but induction by tamoxifen of Col2a1 CreERT2; by Ext and HS deficiencies. Idh1-R132 mutant conditional knock-in mice developed Joanna Phillips (University of California, San multiple -like lesions. Interestingly, this Francisco) presented studies on HS modifications and mutation alters enzymatic activity of IDH from conver- the impact on platelet-derived growth factor (PDGF) sion of isocitrate to α-ketoglutarate to production of signaling in glioblastoma (GBM) (72,73). Of particular D-2-hydroxyglutarate, which can alter DNA methylation. interest was the observation that expression of Sulf2,an TREATMENTS FOR MULTIPLE HEREDITARY EXOSTOSES 91 endosulfatase that modifies the level and pattern of Conceptually, MHE might also share elements with 6-O-sulfation of HS chains at the cell surface, was regenerative processes in which a stem cell undergoes elevated in GBM subtypes that exhibit amplification activation. HS regulates many of the cell signaling pathways of PDGF receptor expression. Sulfs have not yet been important for wound repair and regeneration (21). To analyzed in osteochondromas, but as indicated above, examine the impact of Ext1 on wound repair and regen- other studies suggested higher heparanase expression eration, one of us (Anne Phan, University of California, that could amplify the impact of HS reduction due to San Diego) examined the response to cutaneous skin Ext deficiency (42,43). Overall, the studies raise the lesions and digit amputations in Ext deficient mice. possibility that extracellular alterations in HS structure Surprisingly, double Ext1+/-; Ext2+/- heterozygous mice and levels could contribute to tumorigenicity. Ralph showed faster repair of full-thickness dorsal skin excisional Sanderson (University of Alabama at Birmingham) dis- wounds. This phenotype was recapitulated in wild type cussed how trimming of HS chains by heparanase animals treated with glycosides that modulate glycosami- could impact cell signaling and promote tissue remo- noglycan formation, suggesting a pharmacological deling, driving tumor growth, invasion, metastasis, approach to stimulate wound repair. Additionally, digit osteolysis, and angiogenesis in . His regeneration studies showed that wild type mice, like chil- group discovered that treatment of myeloma cells with dren (78), can regenerate the digit tip if amputations occur anti-myeloma drugs such as bortezomib and melphalan in the 3rd phalangeal element above the nail root and the significantly up-regulated the expression and secretion wound is allowed to close and form a blastema. More of heparanase and resulted in chemoresistance (74,75). proximal amputations below the nail root and in the 2nd These findings indicate that heparanase is a viable phalangeal element do not. No effect on 3rd digit regen- target for anti-cancer therapy. Roneparstat (previously eration was noted in Ext1+/- ; Ext2+/- mice, but amazingly, known as SST0001) is a rationally developed hepara- some mutant mice regenerated digit-tips after an amputa- nase inhibitor produced by modification of porcine tion through the more proximal 2nd phalangeal element. mucosal heparin to eliminate its anticoagulant activity. We note however, digit regeneration was highly variable Preclinical studies of Roneparstat in animal models of and poorly penetrant amongst mice. These findings raise multiple myeloma indicated anti-tumor efficacy and the possibility that the formation of osteochondromas may pharmacodynamic effects consistent with its anti- involve recruitment of processes and progenitor cells nor- heparanase activity in vivo (76). A recently completed mally involved in regenerative and repair responses. Phase I study in advanced refractory multiple myeloma Yang Chai (University of Southern California) dis- patients revealed excellent tolerability of this drug. cussed craniofacial mesenchymal stem cells in bone These exciting findings might be adapted for the treat- tissue homeostasis and repair. Craniofacial bones are ment of MHE. Roneparstat was shown to inhibit chon- derivatives of the neural crest and thus have a different drogenesis in vitro; since this step is the first to occur in origin as compared to those producing much of the osteochondroma development, Roneparstat may indeed skeleton. Gli1+ progenitors within the suture mesench- be a possible therapeutic for MHE as well (42). yme were found to serve as the major mesenchymal cell population for craniofacial bones. Ablation of Gli1+ cells led to craniosynostosis and arrest of skull growth. Interestingly, the calvarial sutures possessed much Stem cell biology stronger regeneration capacity than non-suture areas Original studies of specimens from MHE patients sug- during experimental calvarial bone repair, and the heal- gested the presence of early microchondromas within ing rate of calvarial bone was inversely related to the perichondrium along the physis (growth plate) and suture-injury distance. Some of these very exciting the groove of Ranvier, suggesting that precursor cells in data have been published (79). They strongly suggest the perichondrium give rise to neo-chondrocytes that that the potential for is not evenly dis- clonally expand and develop into osteochondromas tributed within the calvaria bones but is greatly (32). Conditional Ext1 ablation in the perichondrium enriched within the sutures and their mesenchymal flanking the epiphyseal long bone region, which stem cells and niche. This is analogous to the prefer- encompasses the groove of Ranvier, does indeed cause ential distribution of progenitors in perichondrium and ectopic cartilage formation in mice (40). Like the peri- groove of Ranvier and their likely roles in osteochon- chondrium, the groove of Ranvier is thought to house droma formation (40), a process invoking skeletal progenitor cells (77), suggesting the possibility that repair processes as postulated above osteochondroma formation in MHE involves defects T. Michael Underhill (University of British Columbia, in resident stem cell behavior and niches. Canada) also discussed the role of mesenchymal 92 A. Q. PHAN ET AL. progenitors in tissue renewal and regeneration. Sulf1 has roles in progenitor cell function and osteo- Mesenchymal progenitors, one type of adult progenitor chondroma formation. cells, can remain in a quiescent state, but become acti- Noriaki Ono (University of Michigan) presented data vated in response to various signals. Mouse models were suggesting that a novel type of skeletal stem cells are presented that involve knock-in of a reporter into the located in the resting zone of postnatal growth plate. Hic1 gene (hypermethylated in cancer), which is largely These cells display distinct characteristics compared to restricted to quiescent mesenchymal progenitor cells. adult bone marrow mesenchymal stromal stem cells. To Lineage tracing and marker analysis showed that the study if resting growth plate cells are important in bone Hic1+ cells generally resided in a perivascular location growth as well, his group utilized a genetic label- and significantly contributed to multiple mesenchymal retention strategy based on a Tet-Off system regulating linages, including beige and white , osteocytes, expression of a histone-linked GFP. Exploiting the fact marrow stromal cells, and myofibroblasts. RNA-seq ana- that the resting cells express PTHrP (82), the cells were lyses of isolated mesenchymal progenitors revealed that irreversibly labeled using PTHrPCreER BAC transgenic “activated” cells had multiple roles in regeneration, in mice, and they and their progenies were traced over particular in muscle regeneration. Mouse models of this time. The cells subsequently formed columnar chondro- type might prove useful for tracing the origin of cells in cytes, hypertrophic chondrocytes, and subchondral bone osteochondromas. in the primary spongiosa. Reporter-positive cells also April Craft (Boston Children’s Hospital) discussed formed large colonies in vitro, indicating their replicative how pluripotent stem cells (PSCs), including both capacity. Thus, the data suggested that cells in the resting human and mouse embryonic stem cells (ESCs) and zone of growth plate represent a novel type of skeletal induced pluripotent stem cells (iPSCs), can be used to stem cells. Whether these cells contribute to bony reproducibly and efficiently generate articular chondro- tumors, such as osteochondromas or endochondromas, cytes and growth plate-like chondrocytes (80). Human remains unknown. PSCs expressed lineage specific mRNAs in appropriate sequence during in vitro differentiation, and retained Therapeutic approaches their lineage-specific phenotypes when implanted in vivo. Cartilage tissues generated from PSCs resisted Surgery is currently the major treatment option for ossification when implanted in vivo, whereas hyper- MHE patients, through which the most symptomatic trophic chondrocytes derived from PSCs underwent osteochondromas are resected and major skeletal defects endochondral ossification, akin to endogenous growth can be corrected. However, as pointed out above, plate maturation. Additional studies compared the patients often have numerous osteochondromas that articular and growth plate cartilage structures created cannot be resected or would require multiple surgeries. in vitro and in vivo from PSCs to respective endogen- These lingering tumors can cause life-long problems in ous tissues. These techniques might prove useful for addition to increasing the probability of malignant trans- studying diseased tissues from MHE patients and could formation. Currently, there are no pharmacological elicit expandable and renewable populations of mutant treatments for MHE other than palliative care for pain. cells to study their basic pathogenic mechanisms. Thus, there is an urgent need to identify potential drug- Hiroshi Nakato (University of Minnesota) reported gable targets and drug-like agents that might prove of studies on the control of stem cell activity by HS in therapeutic value. One of the major goals of the Drosophila. One area of research was to understand Conference was the identification of potential pharma- how changes in HS sulfation regulate intestinal stem cological approaches for treating the disease, taking cell division during normal midgut homeostasis and advantage of various model organisms, high- regeneration. Deficiency in Sulf1, the extracellular throughput screening technologies, and studies of other endosulfatase that reduces 6-O-sulfation of HS, resulted bone and cartilage disorders where drug development in increased intestinal stem cell division. Using a regen- efforts have succeeded or have made progress. eration model, his group found that Sulf1 mutant One of us (Jeffrey Esko, University of California, San intestinal stem cells failed to properly halt division at Diego) discussed several novel approaches to develop the termination stage. Conversely, overexpression of drugs for treating MHE. Given that the primary defect in Sulf1 during early regeneration suppressed intestinal MHE is in HS biosynthesis, methods to restore HS were stem cell division. Taken together, these findings indi- discussed. Ideas included use of subclinical doses of low cate that Sulf1 is required for terminating intestinal molecular weight heparin as a method of “substrate repla- stem cell division at the end of regeneration (81). As cement therapy.” Initial studies in murine model of MHE pointed out above, it remains to be explored whether did not reveal significant reduction in the number, size, or TREATMENTS FOR MULTIPLE HEREDITARY EXOSTOSES 93 time of onset of disease, but additional studies were sug- pathology associated with chondrodysplasia and thus gested using non-anticoagulant heparin (e.g. offer a novel model to develop therapeutic agents. Roneparastat). Jian Liu (University of North Carolina) Catherine Merry (University of Nottingham, United described current methods for chemoenzymatic synthesis Kingdom) developed a similar approach to derive of HS and heparin. Synthesis of heparin and HS oligosac- chondrocytes from hiPSCs obtained from MHE charides using a purely chemical approach remains chal- patients. She reported the identification of a plasma- lenging. However, a chemoenzymatic method was derived protein (inter-α-inhibitor) as a culture additive developed to prepare synthetic heparin and HS, using 15 that negates the traditional requirement for surface different enzymes including sulfotransferases, an epimer- coating of tissue culture plastic with extracellular ase, and glycosyltransferases (83). The availability of HS matrix for culturing human pluripotent stem cells. oligosaccharide libraries offers a unique opportunity to Her group also developed 3D culture environments to study the function and activity relationship of HS in bio- better mimic in vivo conditions, using hydrogel-based logical systems. Applications include studies of HS interac- or fibrous scaffold-based culture conditions. tions with signaling proteins, competition experiments José Luis Millán (Sanford Burnham Prebys Medical performed in cultured cells and substrate replacement Discovery Institute) pioneered methods for treating therapy. Jian Liu’s methods could yield large quantities of caused by mutations in tissue- defined, non-anticoagulant heparinoids for treating MHE. nonspecific alkaline phosphatase. These methods involve Esko also described a high-throughput screening assay enzyme replacement targeted to bone (84). Skeletal to identify compounds that augment HS on the plasma mineralization involves matrix vesicles that initiate and membrane. One compound was identified and is cur- propagate mineralization from intralumenal to extravesi- rently under development. In addition, genome wide cular spaces, followed by deposition of mineral onto the screens using CRISPR/Cas9 identified unexpected genes collagenous matrix (85). The phosphatases are involved in that modulate HS formation. These genes represent other mineralization in various ways, including the hydrolysis potential targets for drug development efforts. of pyrophosphate (a strong anti-mineralization agent) Laurence Legeai-Mallet (Paris Descartes University, and local release of phosphate. Thus, enzyme replacement France) discussed novel strategies to correct abnormal- therapy compensated for low alkaline phosphatase activ- ities in skeletal development. Innovative therapies ity, reversed pyrophosphate accumulation and allowed include replacement of defective/missing proteins or resumption of mineralization. Targeting these mechan- genes or normalization of aberrant signaling pathways. isms could be useful to prevent and treat diverse forms of Potential therapeutic strategies emerged from studies on ectopic calcification, including arterial calcification. , the most frequent form of . Whether these methods might prove useful for treating Several preclinical studies are being carried out that MHE remains speculative. include treatment with: C-type natriuretic peptide analog One of us (Maurizio Pacifici, Children’sHospitalof (BMN111), intermittent PTH injections, soluble FGFR3, Philadelphia) in collaboration with Kevin Jones at the Meclozine, Statin and a pan-FGFR tyrosine kinase inhi- University of Utah, re-examined craniofacial X-ray scans bitor (NVP-BGJ398). The success of any therapies tar- from MHE patients and over half of them exhibited geting pediatric disorders, such as MHE, relies on early moderate defects or osteochondroma-like outgrowths in diagnosis and effective targeted correction of the affected the endochondral cranial base, specifically in the clivus. pathway, while maintaining normal development and Similar phenotypes were observed in the cranial base of homeostasis of the remainder of the skeleton. mutant Col2CreER; Ext1f/f or AggrecanCreER;Ext1f/f mouse Chondrodysplasias and other genetic diseases that models of MHE (86). These findings indicate that not affect bone and cartilage formation are often studied only the cervical vertebrae (3) but also the cranial base in animal models, but sometimes there are differences may be affected by MHE, expanding the potential anato- in animal models versus humans. Because human tis- mical sites and structures that a pharmacologic therapy sues are difficult to obtain on a routine basis, the would need to reach in patients. advent of induced pluripotent cells (iPSCs) is making Yu Yamaguchi (Sanford Burnham Prebys Medical it possible to create human chondrocytes and cartilage Discovery Institute) presented data that targeted inac- from skin fibroblasts from patients with chondrodys- tivation of Ext1 to perichondral progenitor cells plasias. Noriyuki Tsumaki (Kyoto University, Japan) (Fsp1Cre; Ext1f/f mice) leads to osteochondroma forma- described the application of iPSC technology for mod- tion (87). This confirms data first reported by one of us eling chondrodysplasias. His group found that cartila- (Maurizio Pacifici) and collaborators using mouse ginous tissues derived from iPSCs generated from models of MHE (40). Osteochondromas were found patients with FGFR3 mutations reproduced the to develop from mesenchymal stem cell-like progenitor 94 A. Q. PHAN ET AL. cells residing along the boundary between growth plate osteochondroma-like lesions, respectively, making and perichondrium. Both groups showed that Ext1- MHE more pervasive than previously realized. In deficient progenitor cells display enhanced BMP signal- terms of cellular and molecular pathogenesis subtend- ing and chondrogenic differentiation in vitro. Most ing osteochondroma development, strong evidence was importantly, Yamaguchi reported that systemic admin- provided that mesenchymal progenitors residing in istration of the BMP inhibitor LDN-193189 effectively perichondrium and groove of Ranvier may be major reduced osteochondroma growth in Col2CreER;Ext1f/f culprits; the cells shift from a mesenchymal to a chon- and Fsp1Cre;Ext1f/f mice (87). A similar osteochon- drogenic lineage, undergo chondrogenesis and initiate droma-suppressive effect of LDN-193189 was published tumor formation (40). Transgenic mouse studies have by Pacifici’s group using AggrecanCreER;Ext1f/f mouse established that compound heterozygous loss of Ext1 models of MHE (86). In vitro studies with mouse and Ext2 or conditional loss of both Ext1 alleles are embryo chondrogenic cells showed that treatment necessary and sufficient to cause osteochondroma for- with LDN-193189 resulted in decreased canonical mation. In this regard, it is interesting to note that an BMP signaling through pSMAD1/5/8. These studies appreciable number of MHE patients have been found provide a novel therapeutic approach for treating MHE. to bear compound heterozygous mutations in both EXT1 and EXT2 (66,88), possibly indicating that their disease course may not require an additional “second Clinical care of MHE patients hit” such as LOH and could be more severe given that The MHE Research Foundation organized a final ses- the double mutation is in every cell. Presented work sion dedicated to patients. Patient advocacy by the indicates that osteochondroma formation may directly MHE Research Foundation has been a driving force or indirectly involve additional genes, including Erk1/2, in directing research interest and attention to this ske- Ptpn11 and SoxC, and may also rely on and exploit letal disorder. The session was attended by MHE mechanisms normally involved in tissue repair and patients and their families. The purpose was to provide regeneration and inflammation. Arguably, a most pro- a layperson summary of major scientific findings; offer mising outcome of the Conference was the identifica- an opportunity for researchers, patients and families to tion of potential therapeutic means by which mingle and interact; point out issues arising in care and osteochondroma formation could be countered. High treatment; and facilitate contacts between physicians throughput drug screens conducted by one of us and MHE patients. One of us (Jeffrey Esko) provided (Jeffrey Esko) have identified a promising small mole- an overview of the Conference. A MHE Live Patient cule compound that is undergoing active laboratory Clinic Demonstration, led by Drs. Dror Paley, David testing at the moment. Compounds that interfere with Friedman, and Craig Robbins, followed his presenta- canonical BMP signaling have demonstrated effective tion. Subsequent presentations focused on Best inhibitory activity against osteochondroma formation Treatment Strategies in Children and Adults for in mouse models, in line with the fact that BMP signal- Upper Extremity Deformities of MHE, Straight Talk ing is required for chondrogenesis. These and other on Crooked Legs, and MHE of the Hip and How to proof-of-principle studies have thus provided a strong Restore Hip Motion and MHE of the Spine. basis and much impetus to move ahead as readily as possible toward the testing of a therapeutic strategy in a MHE clinical trial. Conclusions and perspectives The Conference was thus a critical and thought- The above synopsis of data and insights by participat- provoking forum for assessing the most recent basic ing research groups and from follow-up studies demon- and translational research and for creating a platform strates that much has been learned over the past and springboard for future research. Future efforts 2–3 years about the clinical and biological complexities should focus on: (1) further studies into the enzymol- and the pathogenic mechanisms in MHE. It is clear that ogy and regulation of HS synthesis, in particular the MHE is more than a pure musculoskeletal disorder, possibility of modulating HS assembly through alter- and can affect patients in additional and insidious nate pathways; (2) analysis of how the immune system manners, including their overall well-being and self- and physical forces might alter the progression of regard, occupational possibilities, social interactions, MHE; (3) identification of the signaling and molecular and independence. Anatomical sites that were regarded mechanisms that go awry in MHE due to HS defi- to be less affected, such as the thoracic and lumbar ciency; (4) further development of current and new spine and the cranial base, have turned out to be therapeutic approaches, such as supplementation ther- affected by mild to moderate scoliosis and apy, gene therapy, and pharmacological agents to TREATMENTS FOR MULTIPLE HEREDITARY EXOSTOSES 95 restore normal levels of signaling; and (5) improvement 7. Dormans JP. Pediatric orthopaedics: core knowledge in of diagnostic and prognostic tools to assess patients’ orthopaedics. Philadelphia: Elsevier Mosby; 2005. overall well being and response to treatment. The latter 8. Darilek S, Wicklund CL, Novy D, Scott A, Gambello M, Johnston D, Hecht JT. Hereditary multiple exosto- was particularly resonant with the patients and their sis and pain. J Pediatr Orthop. 2005;25:369–376. families, and a multidisciplinary approach may even- 9. Goud AL, De Lange J, Scholtes VA, Bulstra SK, Ham tually be needed to fulfill that goal. SJ. Pain, physical and social functioning, and quality of life in individuals with multiple hereditary exostoses in The Netherlands: a national cohort study. J Bone Joint Acknowledgments Surg Am. 2012;94:1013–1020. 10. Bolton P, Powell J, Rutter M, Buckle V, Yates Jr, Ishikawa- We would like to thank all of the speakers who devoted BrushY,MonacoAP.Autism,mentalretardation,multi- their time and resources to attend the Conference and ple exostoses and short stature in a female with 46,X,t(X;8) present lectures about ongoing and largely unpublished (P22.13;q22.1). Psychiatric Genet. 1995;5:51–56. work in their labs. We also thank the Conference advisory 11.DeStefanoN,DottiMT,MalandriniA,FedericoA. committee that included Drs. Hank Kronenberg, Karen Association of myopathy with multiple exostoses and men- Lyons, Ben Neel, Dave Ornitz, Andrea Vortkamp, and talretardation:acasereport.Brain&Dev. Matthew Warman for many helpful suggestions. Special 1994;16:136–138. thanks and much gratitude go out to Sarah Ziegler and 12. Roberts IS, Gleadle JM. Familial nephropathy and mul- Craig Eaton (MHE Research Foundation Director and tiple exostoses with Exostosin-1 (EXT1) gene mutation. President, respectively) for their continuous generous sup- J Am Soc Nephrol. 2008;19:450–453. port and leadership; Casey Gerdes and Gaylene Eisenach of 13. Chen S, Wassenhove-McCarthy DJ, Yamaguchi Y, the Glycobiology Research and Training Center at UC San Holzman B, Th VK, Jenniskens G, Tj W, Ac W, Diego for their outstanding organizational skills; Dr. Dror McCarthy KJ. Loss of heparan sulfate glycosaminogly- Paley and the staff of the Paley Institute for providing the can assembly in podocytes does not lead to proteinuria. venue and local support for patients; Wendy Lubkin and Kidney Int. 2008;74:289–299. the Lubkin Fund of The Philadelphia Foundation and 14. Mooij HL, BernelotMoens SJ, Gordts PL, Stanford KI, Glycan Therapeutics, LLC for support. We express much Foley EM, Van Den Boogert MA, Witjes JJ, Hassig gratitude to all the MHE patients and their families for HC,TanckMW,VanDeSandeMA,LevelsJH, their fortitude and dedication. Kstelein JJ, Stroes ES, Dallinga-Thie GM, Esko JD, Ext1 NM. heterozygosity causes a modest effect on postprandial lipid clearance in humans. J Lipid Res. – Funding 2015;56:665 673. 15.AhnJ,LudeckeHJ,LindowS,HortonWA,LeeB, We gratefully acknowledge the financial support from grant WagnerMJ,HorsthemkeB,WellsDE.Cloningof R13 AR069987 received from the National Institute of theputativetumoursuppressorgeneforhereditary Arthritis, Musculoskeletal and Skin Diseases (NIAMS). multiple exostoses (EXT1). Nat Genet. 1995;11:137–143. 16. Hecht JT, Hogue D, Strong LC, Hansen MF, Blanton References SH, Wagner H. Hereditary multiple and chondrosarcoma: linkage to chromosome 11 and loss 1. Luckert Wicklund CL, Pauli RM, Johnson DR, Hecht of heterozygosity for EXT-linked markers on chromo- JT. Natural history of hereditary multiple exostoses. some 11 and 8. Am J Hum Genet. 1995;56:1125–1131. Am J Med Genet. 1995;55:43–46. 17. Stickens D, Clines G, Burbee D, Ramos P, Thomas S, 2. Stieber JR, Dormans JP. Manifestations of hereditary Houge D, Hecht JT, Lovett M, Evans GA. The EXT2 multiple exostoses. J Am Acad Orthop Surg. multiple exostoses gene defines a family of putative – 2005;13:110–120. tumor suppressor genes. Nat Genet. 1996;14:25 32. 3. Jones KB. Glycobiology and the growth plate: current 18. McCormick C, Duncan G, Goutsos KT, Tufaro F. The concepts in multiple hereditary exostoses. J Pediatr putative tumor suppressors EXT1 and EXT2 form a Orthop. 2011;31:577–586. stable complex that accumulates in the Golgi complex 4. Porter DE, Simpson AHRW. The neoplastic pathogen- and catalyzes the synthesis of heparan sulfate. Proc – esis of solitary and multiple osteochondromas. J Pathol. Natl Acad Sci USA. 2000;97:668 673. 1999;188:119–125. 19. Lin X, Wei G, Shi Z, Dryer L, Esko JD, Wells DE, Matzuk 5. De Andrea CE, Reijnders CM, Kroon HM, De Jong D, MM. Disruption of gastrulation and heparan sulfate bio- Hogendoorn PC, Szuhai K, Bovee JV. Secondary per- synthesis in EXT1-deficient mice. Dev Biol. – ipheral chondrosarcoma evolving from osteochon- 2000;224:299 311. droma as a result of outgrowth of cells with 20. Stickens D, Zak BM, Rougier N, Esko JD, Werb Z. functional EXT. Oncogene. 2012;9:1095–1104. Mice deficient in Ext2 lack heparan sulfate and develop – 6. Porter De, Lonie L, Fraser M, Dobson-Stone C, Porter exostoses. Development. 2005;132:5055 5068. Jr, Monaco Ap, Simpson AH. Severity of disease and 21. Bishop JR, Schuksz M, Esko JD. Heparan sulphate risk in malignant change in hereditary multiple proteoglycans fine-tune mammalian physiology. – exostoses. J Bone Joint Surg Br. 2004;86:1041–1046. Nature. 2007;446:1030 1037. 96 A. Q. PHAN ET AL.

22. Sarrazin S, Lamanna WC, Esko JD. Heparan sulfate multiple hereditary exostoses. Proc Natl Acad Sci USA. proteoglycans. Cold Spring Harb Prospect Biol. 2010;107:10932–10937. 2011;3:a004952. 37. Sgariglia F, Candela ME, Huegel J, Jacenko O, Koyama 23. Bulow HE, Hobert O. The molecular diversity of gly- E, Yamaguchi Y, Pacifici M, Enomoto-Iwamoto M. cosaminoglycans shapes animal development. Annu Epiphyseal abnormalities, trabecular bone loss and Rev Cell Dev Biol. 2006;22:375–407. articular chondrocyte hypertrophy develop in the long 24. Lin X. Functions of heparan sulfate proteoglycans in bones of postnatal Ext1-deficient mice. Bone. cell signaling during development. Development. 2013;57:220–231. 2004;131:6009–6021. 38. Clement A, Wiweger M, Von Der Hardt S, Rusch Ma, 25. Billings PC, Pacifici M. Interactions of signaling pro- Selleck S, Chien C-B, Roehl HH. Regulation of zebra- teins, growth factors and other proteins with heparan fish skeletogenesis by /dackel and papst1/pinscher. sulfate: mechanisms and mysteries. Connect Tissue PLoS Genet. 2008;4(7):e1000136. Res. 2015;56:272–280. 39. De Andrea CE, Wiweger M, Prins F, Bovee JVMG, 26. Xu D, Esko JD. Demystifying heparan sulfate-protein Romeo S, Hogendoorn PC. Primary cilia organization interactions. Annu Rev Biochem. 2014;83:129–157. reflects polarity in the growth plate and implies loss of 27. Anower-E-Khuda MF, Matsumoto K, Habuchi H, polarity and mosaicism in osteochondroma. Lab Invest. Morita H, Yokochi T, Shimizu K, Kimata K. 2010;90:1091–1101. in the blood of hereditary multi- 40. Huegel J, Mundy C, Sgariglia F, Nygren P, Billings PC, ple exostoses patients: half reduction of heparan sulfate Yamaguchi Y, Koyama E, Pacifici M. Perichondrium to chondroitin sulfate ratio and the possible diagnostic phenotype and border function are regulated by Ext1 application. Glycobiology. 2013;23:865–876. and heparan sulfate in developing long bones: A 28. Cheung PK, McCormick C, Crawford BE, Esko JD, mechanism likely deranged in hereditary multiple Tufaro F, Duncan G. Etiological point mutations in exostoses. Dev Biol. 2013;377:100–112. the hereditary multiple exostoses gene EXT1: a func- 41. Zuntini M, Salvatore M, Pedrini E, Parra A, Sgariglia F, tional analysis of heparan sulfate polymerase activity. Magrelli A, Taruscio D, Sangiorgi L. MicroRNA profil- Am J Hum Genet. 2001;69:55–66. ing of multiple osteochondromas: identification of 29. Zak BM, Schuksz M, Koyama E, Mundy C, Wells DE, disease-specific and normal cartilage signatures. Clin Yamaguchi Y, Pacifici M, Esko JD. Compound hetero- Genet. 2010;78:507–516. zygous loss of Ext1 and Ext2 is sufficient for formation 42. Huegel J, Enomoto-Iwamoto M, Sgariglia F, Koyama E, of multiple exostoses in mouse ribs and long bones. Pacifici M. Heparanase stimulates chondrogenesis and Bone. 2011;48:979–987. is up-regulated in human ectopic cartilage. A mechan- 30. Hosalkar H, Greenberg J, Gaugler RL, Garg S, ism possibly involved in hereditary multiple exostoses. Dormans JP. Abnormal scarring with keloid formation Am J Path. 2015;185:1676–1685. after osteochondroma excision in children with multi- 43. Trebicz-Geffen M, Robinson D, Evron Z, Glaser T, ple hereditary exostoses. J Pediatr Orthop. Fridkin M, Kollander Y, Vlodavsky I, Ilan N, Law KF, 2007;27:333–337. Cheah KSE, Chan D, Werner H, Nevo Z. The molecu- 31. Bovee JV, Cleton-Jansen A-M, Wuyts W, Caethoven G, lar and cellular basis of exostosis formation in heredi- Taminiau AH, Bakker E, Hul WV, Cornelisse CJ. tary multiple exostoses. Int J Exp Path. Hogendoorn PC. EXT-mutation analysis and loss of 2008;89:321–331. heterozygosity in sporadic and hereditary osteochon- 44. Pacifici M. Hereditary mutliple exostoses: new insights dromas and secondary chondrosarcoma. Am J Hum into pathogenesis, clinical complications, and potential Genet. 1999;65:689–698. treatments. Curr Osteoporos Rep. 2017;15:142–152. 32. Hecht JT, Hayes E, Haynes R, Cole GC, Long RJ, 45. Inatani M, Irie F, Plump AS, Tessier-Lavigne M, Farach-Carson MC, Carson DD. Differentiation- Yamaguchi Y. Mammalian brain morphogenesis and induced loss of heparan sulfate in human exostosis midline axon guidance require heparan sulfate. derived chondrocytes. Differentiation. 2005;73:212–221. Science. 2003;302:1044–1046. 33. Knudson A. Mutation and cancer: statistical study of 46. Yamaguchi Y, Inatani M, Matsumoto Y, Ogawa J, Irie retinoblastoma. Proc Natl Acad Sci USA. F. Roles of heparan sulfate in mammalian brain devel- 1971;68:820–823. opment: current views based on the findings from Ext1 34. Huegel J, Sgariglia F, Enomoto-Iwamoto M, Koyama E, conditional knockout studies. Prog Mol Biol Transl Sci. Dormans JP, Pacifici M. Heparan sulfate in skeletal 2010;93:133–152. development, growth, and pathology: the case of heredi- 47. MacArthur JM, Bishop JR, Stanford KI, Wang LC, tary multiple exostoses. Dev Dyn. 2013;242:1021–1032. Bensadoun A, Wiltztum JL, Esko JD. Liver heparan 35. Jones KB, Piombo V, Searby C, Kurriger G, Yang B, sulfate proteoglycans mediate clearance of Grabellus F, Roughley PJ, Morcuende JA, Buckwalter triglyceride-rich lipoproteins independently of LDL JA, Capechhi MR, Vortkamp A, Sheffield VC. A mouse receptor family members. J Clin Invest. model of osteochondromagenesis from clonal inactiva- 2007;117:153–164. tion of Ext1 in chondrocytes. Proc Natl Acad Sci USA. 48. Stanford KI, Wang L, Castagnola J, Song D, Bishop JR, 2010;107:2054–2059. Lawrence R, Bai XM, Habuchi H, Tanaka M, Cardoso 36. Matsumoto K, Irie F, Mackem S, Yamaguchi Y. A WV, Kimata K, Esko JD. Heparan sulfate mouse model of chondrocyte-specific somatic muta- 2-O-sulfotransferase is required for tryglyceride-ricj tion reveals a role for Ext1 loss of heterozygosity in lipoprotein clearance. J Biol Chem. 2010;285:286–294. TREATMENTS FOR MULTIPLE HEREDITARY EXOSTOSES 97

49. Bode L, Salvestrini C, Park PW, Li JP, Esko JD, establish planar cell polarity by inducing Vangl2 phos- Yamaguchi Y, Murch S, Freeze HH. Heparan sulfate phorylation through Ror2. Dev Cell. 2011;20:163–176. and syndecan-1 are essential in maintaining murine 62. Oestreich AT, Huslig EL. Hereditary multiple exosto- and human epithelial intestinal barrier function. J sis: another etiology of short leg and scoliosis. J Clin Invest. 2008;118:229–238. Manipulative Physiol Ther. 1985;8:267–269. 50. Bernelot Moens SJ, Mooij HL, Hassing HC, Kruit JK, 63. Matsumoto Y, Matsumoto K, Harimaya K, Okada S, Witjes JJ, Van De Sande MA, Nederveen AJ, Xu D, Doi T, Iwamoto Y. Scoliosis in patients with multiple Dallinga-Thie GM, Esko JD, Stroes ES, Nieuwdorp hereditary exostoses. Eur Spine J. 2015;24:1568–1573. M. Carriers of loss-of-function mutations in EXT 64. Karuppaiah K, Yu K, Lim J, Chen JD, Smith CL, Long display impaired pancreatic beta-cell reserve due to F, Ornitz DM. FGF signaling in the osteoprogenitor smaller pancreas volume. PLoS ONE. 2014;9: lineage non-autonomously regulates postnatal chon- e115662. drocyte proliferation and skeletal growth. 51. Wang L, Fuster MM, Sriramarao P, Esko JD. Development. 2016;143:1811–1822. Endothelial heparan sulfate deficiency impairs 65. McCormick C, Leduc Y, Martindale D, Mattison K, L-selectin- and chemokine-mediated neurophil traf- Esford L, Dyer A, Tufaro F. The putative tumor sup- ficking during inflammatory responses. Nat Immunol. pressor EXT1 alters the expression of cell-surface 2005;6:902–910. heparan sulfate. Nat Genet. 1998;19:158–161. 52. Tsuboi K, Hitakawa J, Seki E, Imai Y, Yamaguchi Y, 66. Pedrini E, Jennes I, Tremosini M, Milanesi A, KFuluka M, Kawashima H. Role of high endothelial Mordenti M, Parra A, Sgariglia F, Zuntini M, venule-expressed herapan sulfate in chemokine presen- Campanacci L, Fabbri N, Pignotti E, Wuyts W, tation and lymphocyte homing. J Immunol. Sangiorgi L. Genotype-phenotype correlation study in 2013;191:448–455. 529 patients with hereditary multiple exostoses: identi- 53. Jennes I, Pedrini E, Zuntini M, Mordenti M, Balkassmi fication of “protective” and “risk” factors. J Bone Joint S, Asteggiano CG, Casey B, Bakker S, Sangiorgi L, Surg. 2011;93:2294–2302. Wuyts W. Multiple osteochondromas: mutation update 67. Hirata M, Sasaki M, Cairns RA, Inoue S, Puviindran V, and description of the multiple osteochondromas Li WY, Snow BE, Jones LD, Wei QX, Sato S, Tang YJ, mutation database (MOdb). Hum Mutat. Nadesan P, Rockel JS, Whetstone H, Poon R, Weng A, 2009;30:1620–1627. Gross S, Straley K, Gliser C, Xu Y, Wunder JS, Mak 54. Bernard MA, Hall CE, Hogue DA, Cole WG, Scott A, TW, Alman BA. Mutant IDH is sufficient to initiate Snuggs MB, Clines GA, Ludecke HJ, Lovett M, Van in mice. Proc Natl Acad Sci USA. Winkle WB, Hecht JT. Diminshed levels of the puttaive 2015;112:2829–2834. tumor suppressor proteins EXT1 and EXT2 in exosto- 68. Verdegaal SH, Bovee JV, Pansuriya TC, Grimer RJ, sis chondrocytes. Cell Motiil Cytoskeleton. Ozger H, Jutte PC, Julian MS, Biau DJ, Van Der 2001;48:149–162. Geest IC, Leitner A, Streitburger A, Klenke FM, 55. Hameetman L, Szuhai K, Yavas A, Knijnenburg J, Van Gouim FG, Campanacci DA, Marec-Berard P, Duin M, Van Dekken H, Timiniau AH, Cleton-Jansen Hogendoorn PC, Brand R, Taminiau AH. Incidence, A-M, Bovee JV, Hogendoorn PC. The role of EXT1 in predictive factors, and prognosis of chondrosarcoma in nonhereditary osteochondroma: identification of patients with Ollier disease and : an homozygous deletions. J Natl Cancer Inst. international multicenter study of 161 patients. 2007;99:396–406. Oncologist. 2011;16:1771–1779. 56. Koziel L, Kunath M, Kelly OG, Vortkamp A. Ext1- 69. De Jong Y, Jg VO, Ab K, Briare-De BIH, Agrogiannis G, dependent heparan sulfate regulates the range of Ihh Baranski Z, Ah C, Am C-J, Van De Water B, Danen E, signaling during endochondral ossification. Dev Cell. Bovee JV. Targeting survivin as a potential new treatment 2004;6:801–813. for chondrosarcoma of bone. Oncogenesis. 2016;5:e222. 57. Ono N, Ono W, Nagasawa T, Kronenberg HM. A 70. Van Oosterwijk JG, Van Ruler MA, Briare-De Bruijn subset of chondrogenic cells provides early mesenchy- IH, Herpers B, Gelderblom H, Van De Water B, Bovee mal progenitors in growing bones. Nat Cell Biol. JV. Src kinases in chondrosarcoma chemoresistance 2014;16:1157–1167. and migration: dasatinib sensitises to doxorubicin in 58. Osterholm C, Lu N, Liden A, Kerlsen TV, Gullberg D, TP53 mutant cells. Br J Cancer. 2013;109:1214–1222. Reed RK, Kusche-Gullberg M. Fibroblast EXT1-levels 71. Day TF, Guo X, Garrett-Beal L, Yang Y. Wnt/β-catenin influence tumor cell proliferation and migration in signaling in mesenchymal progenitors controls osteo- composite spheroids. PLoS ONE. 2012;7:e41334. blast and chondrocyte differentiation during vertebrate 59. Hamerman D. The biology of osteoarthritis. New Engl skeletogenesis. Dev Cell. 2005;8:739–750. J Med. 1989;320:1322–1330. 72. Phillips JJ, Huillard E, Robinson AE, Ward A, Lum DH, 60. Wehenkel M, Corr M, Guy CS, Edwards BA, Castellaw Polley MY, Rosen SD, Rowitch DH, Werb Z. Heparan AH, Calabrese C, Pages G, Pouyssegur J, Vogel P, sulfate sulfatase SULF2 regulates PDGFRalpha signaling McGargill MA. Extracellular signal-regulated kinase and growth in human and mouse malignant glioma. J signaling in CD4-expressing cells inhibits Clin Invest. 2012;122(3):911–922. osteochondromas. Front Immunol. 2017;8:1–11. 73. Tran VM, Wade A, McKinney A, Chen K, Lindberg 61. Gao B, Song H, Bishop K, Elliot G, Garrett L, English OR, Engler JR, Persson AI, Phillips JJ. Heparan sulfate MA, Andre P, Robinson J, Sood R, Minami Y, glycosaminoglycans in glioblastoma promote tumor Economides AN, Yang Y. Wnt signaling gradients invasion. Mol Cancer Res. In press. 98 A. Q. PHAN ET AL.

74. Ramani VC, Zhan F, He J, Barbieri P, Noseda A, Tricot 82. Chen X, Macica CM, Nasiri A, Broadus AE. Regulation G, Sanderson RD. Targeting heparanase overcomes of articular chondrocyte proliferation and differentia- chemoresistance and diminishes relapse in myeloma. tion by Indian hedgehog and parathyroid Oncotarget. 2016;7:1598–1607. hormone-related proteins in mice. Arthr Rheum. 75. Ramani VC, Vlodavsky I, Ng M, Zhang Y, Barbiieri P, 2008;58:3788–3797. Noseda A, Sanderson RD. Chemotherapy induces 83. Wang Z, Hsieh PH, Xu Y, Thieker D, Chai EJ, Xie S, expression and release of heparanase leading to Cooley B, Woods RJ. L. C. Liu J. Synthesis of changes associated with an aggressive tumor 3-O-Sulphated Oligosaccharides to Understand the phenotype. Matrix Biol. 2016;55:22–34. Relationship between Structures and Functions of 76. Ritchie JP, Ramani VC, Ren Y, Naggi A, Torri G, Casu Heparan Sulfate. J Am Chem Soc. 2017;139:5249–5256. B, Penco S, Pisano C, Camminati P, Tortoreto M, 84. Millan JL, Whyte MP. Alkaline phosphatase and Zunino F, Vlodavsky I. SST0001, a chemically modified hyposphosphatasia. Calcif Tissue Int. 2016;98:398–416. heparin, inhibits myeloma growth and angiogenesis via 85. Bonucci E. The locus of initial calcification in car- disruption of the heparanase/syndecan-1 shedding: a tilage and bone. Clin Orth Relat Res. novel mechanism for stimulation of tumor growth and 1971;78:108–139. metastasis. Clin Cancer Res. 2011;17:1382–1393. 86. Sihna R, Mundy C, Bechtold T, Sgariglia F, Ibrahim 77. Karlsson C, Thornemo M, Barreto Henriksson H, Lindahl MM, Billings PC, Carroll K, Koyama E, Jones KB, A. Identification of a stem cell niche in the zone of Ranvier Pacifici M. Unsuspected osteochondroma-like out- within the knee joint. J Anat. 2009;215:355–363. growths in the cranial base of Hereditary Multiple 78. Shieh S-J, Cheng T-C. Regeneration and repair of Exostoses patients and modeling and treatment with a human digits and limbs: fact and fiction. BMP antagonist in mice. PLoS Genetics. 2017;13: Regeneration. 2015;2:149–168. e1006742. 79. Zhao H, Ho T-V, Grimes W, Urata M, Chai Y. The 87. Inubushi T, Nazawa S, Matsumoto K, Irie F, suture provides a niche for mesenchymal stem cells of Yamaguchi Y. Aberrant perichondrial BMP signaling craniofacial bones. Nat Cell Biol. 2015;17:386–396. mediates multiple osteochondromagenesis in mice. J 80. Craft AM, Rockel JS, Nartiss Y, Kandel RA, Alman BA, Clin Inv Insight. 2017;2:e90049. Keller GM. Generation of articular chondrocytes from 88. Sarrion P, Sangorrin A, Urreizti R, Delgado A, Artuch human pluripotent stem cells. Nat Biotechnol. R, Mantorell L, Armstrong J, Anton J, Torner F, 2015;33:638–645. Vilaseca MA, Nevado J, Lapunzina P, Asteggiano CG, 81. Takemura M, Nakato H. Drosophila sulf1 is required Balcells S, Grinberg D. Mutations in the EXT1 and for the termination of intestinal stem cell division EXT2 genes in Spanish patients with multiple during regeneration. J Cell Sci. 2017;130:332–343. osteochondromas. Scientic Reports. 2013;3:1346.