Go to JCI Insight
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
  • Clinical Research and Public Health
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Gastroenterology
    • Immunology
    • Metabolism
    • Nephrology
    • Neuroscience
    • Oncology
    • Pulmonology
    • Vascular biology
    • All ...
  • Videos
    • Conversations with Giants in Medicine
    • Video Abstracts
  • Reviews
    • View all reviews ...
    • Complement Biology and Therapeutics (May 2025)
    • Evolving insights into MASLD and MASH pathogenesis and treatment (Apr 2025)
    • Microbiome in Health and Disease (Feb 2025)
    • Substance Use Disorders (Oct 2024)
    • Clonal Hematopoiesis (Oct 2024)
    • Sex Differences in Medicine (Sep 2024)
    • Vascular Malformations (Apr 2024)
    • View all review series ...
  • Viewpoint
  • Collections
    • In-Press Preview
    • Clinical Research and Public Health
    • Research Letters
    • Letters to the Editor
    • Editorials
    • Commentaries
    • Editor's notes
    • Reviews
    • Viewpoints
    • 100th anniversary
    • Top read articles

  • Current issue
  • Past issues
  • Specialties
  • Reviews
  • Review series
  • Conversations with Giants in Medicine
  • Video Abstracts
  • In-Press Preview
  • Clinical Research and Public Health
  • Research Letters
  • Letters to the Editor
  • Editorials
  • Commentaries
  • Editor's notes
  • Reviews
  • Viewpoints
  • 100th anniversary
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
Top
  • View PDF
  • Download citation information
  • Send a comment
  • Terms of use
  • Standard abbreviations
  • Need help? Email the journal
  • Top
  • Abstract
  • Genetic basis of HHT
  • Clinical manifestations of HHT
  • The second-hit hypothesis
  • Signaling pathways involved in HHT
  • Preclinical data, clinical trials, and perspectives in HHT
  • Future challenges in HHT
  • Summary and conclusions
  • Acknowledgments
  • Footnotes
  • References
  • Version history
Article has an altmetric score of 5

See more details

Posted by 7 X users
On 1 Facebook pages
44 readers on Mendeley
  • Article usage
  • Citations to this article (14)

Advertisement

Review Series Open Access | 10.1172/JCI176379

Hereditary hemorrhagic telangiectasia: from signaling insights to therapeutic advances

Tala Al Tabosh,1 Mohammad Al Tarrass,1 Laura Tourvieilhe,2 Alexandre Guilhem,2,3 Sophie Dupuis-Girod,1,2 and Sabine Bailly1

1Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, Grenoble, France.

2Hospices Civils de Lyon, National HHT Reference Center and Genetics Department, Femme-Mère-Enfants Hospital, Bron, France.

3TAI-IT Autoimmunité Unit RIGHT-UMR1098, Burgundy University, INSERM, EFS-BFC, Besancon, France.

Address correspondence to: Sabine Bailly, Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, F-38000 Grenoble, France. Email: sabine.bailly@cea.fr.

Authorship note: TAT, MAT, and LT are co–first authors. SDG and SB are co–last authors.

Find articles by Al Tabosh, T. in: JCI | PubMed | Google Scholar

1Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, Grenoble, France.

2Hospices Civils de Lyon, National HHT Reference Center and Genetics Department, Femme-Mère-Enfants Hospital, Bron, France.

3TAI-IT Autoimmunité Unit RIGHT-UMR1098, Burgundy University, INSERM, EFS-BFC, Besancon, France.

Address correspondence to: Sabine Bailly, Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, F-38000 Grenoble, France. Email: sabine.bailly@cea.fr.

Authorship note: TAT, MAT, and LT are co–first authors. SDG and SB are co–last authors.

Find articles by Al Tarrass, M. in: JCI | PubMed | Google Scholar

1Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, Grenoble, France.

2Hospices Civils de Lyon, National HHT Reference Center and Genetics Department, Femme-Mère-Enfants Hospital, Bron, France.

3TAI-IT Autoimmunité Unit RIGHT-UMR1098, Burgundy University, INSERM, EFS-BFC, Besancon, France.

Address correspondence to: Sabine Bailly, Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, F-38000 Grenoble, France. Email: sabine.bailly@cea.fr.

Authorship note: TAT, MAT, and LT are co–first authors. SDG and SB are co–last authors.

Find articles by Tourvieilhe, L. in: JCI | PubMed | Google Scholar

1Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, Grenoble, France.

2Hospices Civils de Lyon, National HHT Reference Center and Genetics Department, Femme-Mère-Enfants Hospital, Bron, France.

3TAI-IT Autoimmunité Unit RIGHT-UMR1098, Burgundy University, INSERM, EFS-BFC, Besancon, France.

Address correspondence to: Sabine Bailly, Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, F-38000 Grenoble, France. Email: sabine.bailly@cea.fr.

Authorship note: TAT, MAT, and LT are co–first authors. SDG and SB are co–last authors.

Find articles by Guilhem, A. in: JCI | PubMed | Google Scholar

1Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, Grenoble, France.

2Hospices Civils de Lyon, National HHT Reference Center and Genetics Department, Femme-Mère-Enfants Hospital, Bron, France.

3TAI-IT Autoimmunité Unit RIGHT-UMR1098, Burgundy University, INSERM, EFS-BFC, Besancon, France.

Address correspondence to: Sabine Bailly, Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, F-38000 Grenoble, France. Email: sabine.bailly@cea.fr.

Authorship note: TAT, MAT, and LT are co–first authors. SDG and SB are co–last authors.

Find articles by Dupuis-Girod, S. in: JCI | PubMed | Google Scholar

1Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, Grenoble, France.

2Hospices Civils de Lyon, National HHT Reference Center and Genetics Department, Femme-Mère-Enfants Hospital, Bron, France.

3TAI-IT Autoimmunité Unit RIGHT-UMR1098, Burgundy University, INSERM, EFS-BFC, Besancon, France.

Address correspondence to: Sabine Bailly, Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, F-38000 Grenoble, France. Email: sabine.bailly@cea.fr.

Authorship note: TAT, MAT, and LT are co–first authors. SDG and SB are co–last authors.

Find articles by Bailly, S. in: JCI | PubMed | Google Scholar

Authorship note: TAT, MAT, and LT are co–first authors. SDG and SB are co–last authors.

Published February 15, 2024 - More info

Published in Volume 134, Issue 4 on February 15, 2024
J Clin Invest. 2024;134(4):e176379. https://doi.org/10.1172/JCI176379.
© 2024 Tabosh et al. This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
Published February 15, 2024 - Version history
View PDF
Abstract

Hereditary hemorrhagic telangiectsia (HHT) is an inherited vascular disorder with highly variable expressivity, affecting up to 1 in 5,000 individuals. This disease is characterized by small arteriovenous malformations (AVMs) in mucocutaneous areas (telangiectases) and larger visceral AVMs in the lungs, liver, and brain. HHT is caused by loss-of-function mutations in the BMP9-10/ENG/ALK1/SMAD4 signaling pathway. This Review presents up-to-date insights on this mutated signaling pathway and its crosstalk with proangiogenic pathways, in particular the VEGF pathway, that has allowed the repurposing of new drugs for HHT treatment. However, despite the substantial benefits of these new treatments in terms of alleviating symptom severity, this not-so-uncommon bleeding disorder still currently lacks any FDA- or European Medicines Agency–approved (EMA-approved) therapies.

Genetic basis of HHT

Hereditary hemorrhagic telangiectasia (HHT) or Osler-Weber-Rendu syndrome is a hereditary disease that is transmitted in an autosomal dominant manner and is caused by loss-of-function (LOF) mutations in certain components of the predominantly endothelial BMP9-10/ENG/ALK1/SMAD4 signaling pathway, which is an important mediator of vascular quiescence (1). Specifically, mutations in ENG (chromosomal locus 9q34.11, encoding the coreceptor endoglin) and ACVRL1 (chromosomal locus 12q13.13, encoding the type I receptor ALK1) are nearly equally responsible for the majority of HHT cases and give rise to the two major forms of the disease, HHT1 (OMIM 187300) and HHT2 (OMIM 600376), respectively (2–4). Collectively, over 600 different HHT-associated pathogenic mutations in ENG and ACVRL1, spanning the entire coding sequences of both genes, have been documented in the ClinVar database (5). While reported ENG mutations were mostly nonsense or frameshift mutations leading to premature termination codons, a higher prevalence of missense mutations was observed for ACVRL1. Mutations in the SMAD4 gene (chromosomal locus 18q21.2) were also found in HHT patients, although more rarely than ENG and ACVRL1 mutations, resulting in a combined syndrome of HHT and juvenile polyposis (JP-HHT; OMIM 175050) (4, 6). Based on the SMAD4 mutation repository developed by Wooderchak et al. (7), at least 26 different mutations in SMAD4 have been described in patients with the combined JP-HHT syndrome, most of which are missense mutations and deletions that are mainly concentrated at the MH2 domain of SMAD4.

Together, mutations in ENG, ACVRL1, and SMAD4 are responsible for more than 90% of HHT cases, leaving a minority of clinically diagnosed individuals with an unknown genetic basis. In some cases, this can be attributed to limitations of current sequencing technologies in detecting certain variants in the previously identified loci or to the exclusion of testing noncoding regions (8); however, additional chromosomal loci could also be implicated in HHT development. As such, mutations in GDF2 (chromosomal locus 10q11.22, encoding BMP9) were described in few cases displaying an HHT-like syndrome (9–13). This rare form of HHT was annotated as HHT5 (OMIM #615506). In addition, mutations in RASA1, which are commonly associated with hereditary capillary malformations with or without arteriovenous malformations (AVMs) (14, 15), have been reported in few patients presenting HHT symptoms (16–18). More recently, LOF variants in EPHB4 (encoding ephrin receptor B4) were reported in a few individuals exhibiting atypical HHT symptoms and HHT-like hepatic abnormalities (19). Furthermore, one study described an overrepresentation of rare mutations in DROSHA, a key enzyme involved in miRNA processing, among clinically diagnosed HHT patients who did not carry any mutations in the typical HHT-associated genes (20). Interestingly, this study also demonstrated that zebrafish and mice with endothelial-specific DROSHA deficiency developed vascular defects resembling those observed in HHT patients (20).

Clinical manifestations of HHT

AVMs are the hallmark of HHT. According to the international classification of vascular anomalies, AVMs are characterized by malformed arteries, veins, and capillaries with direct arteriovenous communications resulting in arteriovenous shunting (21) (Figure 1). The stepwise development of AVMs in the context of HHT was histologically documented in cutaneous biopsies by Braverman et al. in 1990 (22). This process was found to commence with focal dilatations of the postcapillary venules that progressively encompass the normal capillaries, finally establishing a direct connection with dilated arterioles. The venous origin of AVMs was later supported by studies specifically deleting Alk1 or Eng from venous and capillary beds, which was sufficient to obtain AVMs in retinas (23, 24). In addition, the process seemed to involve an immune arm, as perivascular lymphocytic infiltrates were evident at the site of the AVM (22). It is noteworthy that all clinical signs of HHT result from AVMs of varying size and number affecting different organ systems.

AVMs: The hallmark of HHT and its clinical consequences.Figure 1

AVMs: The hallmark of HHT and its clinical consequences. In HHT, focal dilations of the postcapillary venules (indicated on images by V) progressively encompass the normal capillaries and establish a direct connection with dilated arterioles (indicated by A), leading to AVM formation, muscularization of large vessels, and dilation of capillary beds, which are often surrounded by inflammatory cells. At the microvascular level, AVMs appear as telangiectases on specific mucocutaneous areas (finger pads, lips, tongue, nasal and digestive mucosa). They are responsible for spontaneous and recurrent epistaxis/bleeding, leading to bleeding iron-deficiency anemia. At the macrovascular level, large AVMs mainly affect the liver, leading to high-output cardiac failure, and more rarely biliary ischemia and portal hypertension; lung AVMs can provoke ischemic stroke and cerebral abscess and, more rarely, hypoxemia and hemoptysis. Central nervous system AVM is rarely complicated by hemorrhage. Skin and liver sections were stained for H&E. Photo of the liver section was provided by J.Y. Scoazec (Institut Gustave Roussy, University Paris-Saclay, Paris, France).

At the mucocutaneous surfaces, HHT AVMs appear as telangiectases, after which the disease is named. HHT-associated telangiectases are typically small, red, flat, or slightly elevated spots that blanch under pressure. They are classically present on the palmar faces of finger pads and hands, the lips, the tongue, and mucosal areas inside of the mouth, the nasal cavity, and the gastrointestinal (GI) tract (25, 26) (Figure 1). On the nasal mucosa, telangiectases are responsible for the spontaneous and recurrent epistaxis leading to iron-deficiency anemia in about 50% of patients (27). Similarly, GI telangiectases, which can affect any part of the tract, occasionally result in occult bleeding.

In visceral organs, AVMs can reach a larger size in the lungs, the central nervous system, and the liver and result in life-threatening complications (Figure 1). Pulmonary AVMs can cause direct complications, such as hemorrhage/hemoptysis, due to rupture and hypoxemia due to shunting, but can also cause indirect complications by allowing systemic embolic events resulting in brain abscesses and embolic strokes (28). On the other hand, hepatic AVMs can lead to high-output cardiac failure, biliary ischemia, and portal hypertension (29), warranting liver transplantation. Interestingly, hepatic relapse of the vascular lesions has been described in some HHT patients receiving liver transplants (30), suggesting a potential role of circulating endothelial precursors, which bear the HHT-causal mutation, in populating the newly transplanted liver and driving the formation of new vascular lesions. Cerebral and spinal AVMs are less frequent and generally asymptomatic. Although they can cause serious complications, such as hemorrhagic stroke or epilepsy, the risks associated with treatment are currently higher than those of natural progression and unruptured cerebral AVMs are generally not treated. Widespread screening of asymptomatic HHT patients is therefore still controversial (31).

Cohort studies have demonstrated interorgan differences in the natural history of AVMs. Mucocutaneous telangiectases and liver AVMs appear progressively over the life span, with complications generally occurring in late adulthood. The majority of lung AVMs are probably present from childhood, but some of them become detectable only during adulthood. Most brain AVMs appear in utero or in early childhood and generally remain stable later in life (32). Although HHT1 and HHT2 patients are clinically indistinguishable, ACVRL1 mutations are associated with higher rates of liver AVMs and digestive telangiectases, while ENG mutations are more predominantly linked to pulmonary and cerebral AVMs (33).

In addition to the well-documented HHT symptoms, i.e., epistaxis, telangiectases, and AVMs, HHT patients exhibit a heightened infectious risk in soft tissues, bones, and joints involving Staphylococcus aureus as well as cerebral infections involving bacteria from the orodigestive flora (34). HHT is also associated with immunological abnormalities, mainly characterized by a T-helper lymphopenia, although these abnormalities lack clear correlation with the aforementioned infectious risks (35).

The second-hit hypothesis

The LOF nature of HHT causal mutations and the autosomal dominant inheritance of the disease led to the longstanding belief that HHT is caused by haploinsufficiency of the mutated gene product. This was corroborated by several reports demonstrating a reduction in endoglin levels in HHT1-derived cells (36–38) and in ALK1 levels in some HHT2-derived cells compared with control counterparts (37–39). In line with these studies, mice heterozygous for mutations in Eng or Acvrl1 (Eng+/– or Alk1+/–) display reduced expression levels of the affected gene and develop some HHT-like lesions at the adult stage, albeit with a low penetrance, including telangiectases, nosebleeds, and dilated vessels with reduced vascular smooth muscle cell (VSMC) coverage (40–42).

However, the haploinsufficiency model does not explain why HHT vascular lesions develop focally in characteristic vascular beds despite the presence of the germline HHT causal mutation in all endothelial cells (ECs) of the body (43). This model also fails to elucidate the differences in disease expressivity that are observed even between related patients carrying the same mutation (44). These disparities put forth the second-hit hypothesis in HHT, which is becoming more and more accepted by the HHT scientific community (45). This hypothesis implies that the germline mutation (first hit) predisposes the endothelium to vascular defects that strictly develop in the presence of additional, local factors (second hit) that could be either environmental or genetic. Along these lines, several preclinical studies have demonstrated the role of proangiogenic and proinflammatory triggers in driving HHT pathogenesis (46). For instance, Eng+/– or Alk1+/– mice acquired cerebrovascular dysplasia 8 to 10 weeks after intracranial administration of viral vectors encoding VEGF, as opposed to normal angiogenesis in WT mice receiving the same treatment (47, 48). In addition, unlike WT mice, inducible Eng- or Alk1-knockout mice readily and consistently developed AVMs in their brains only upon local VEGF overexpression (49–52) as well as dermal AVMs strictly after wounding or upon local VEGF or LPS treatment (53–56). Interestingly, blocking VEGF in inducible Alk1–/– mice partially reversed or blocked the development of AVMs induced by VEGF, LPS, or wounding (51, 55), in line with the well-documented effectiveness of the humanized anti-VEGF antibody bevacizumab in alleviating HHT symptoms (Table 1) (57, 58). In addition to angiogenic and inflammatory stimuli, several other environmental triggers have been proposed as potential second hits in HHT, including mechanical stress and sun exposure. A study on 103 HHT patients revealed a higher number of telangiectases on the dominant hand and on the lower lip, which are expected to be more frequently exposed to mechanical stimuli, than the other hand and the upper lip (59). The number of telangiectases on the lips was also found to be positively correlated with sunlight exposure (59).

Table 1

Therapeutic strategies targeting the VEGF pathway

Another study supporting the two-hit hypothesis in HHT patients was released in 2019, but this time involving a genetic second hit (60). Using next-generation sequencing, Snellings et al. detected low-frequency somatic mutations, mostly in trans configuration, leading to biallelic loss of ENG or ACVRL1 in cutaneous telangiectases on the hands of some HHT patients (60). Consequently, HHT vascular malformations were proposed to develop when specific endothelial clones acquire somatic mutations leading to loss of heterozygosity (60). This is a well-known pathogenic mechanism in cancer, known as the Knudsonian two-hit mechanism (61), that was also shown in some heritable vascular anomalies, including venous, glomuvenous, and cerebral cavernous malformations (62, 63). It remains to be validated whether this hypothesis holds true for HHT-related AVMs. As sunlight comprises mutagenic ultraviolet radiation, it is plausible that prolonged exposure to sunlight may trigger somatic mutations that potentially support the development of dermal telangiectases on the hands. This hypothesis might explain the late-onset development of some AVMs (skin, liver) (32), but cannot explain the ones that appear early in life (cerebral and pulmonary AVMs) (32, 64). Therefore, the somatic second-hit hypothesis could indeed be responsible for some HHT-related vascular defects, but might not represent a universal pathogenic mechanism in HHT.

Signaling pathways involved in HHT

BMP9-10/ENG/ALK1/SMAD4 pathway. Endoglin and ALK1 are two transmembrane receptors mainly expressed on ECs, explaining why mutations in this pathway result in vascular abnormalities. BMP9 and BMP10 are the high-affinity ligands of the receptors ALK1 and endoglin (65, 66). Endoglin is a coreceptor for BMP9 and BMP10 and serves as a reservoir of these ligands on the surface of ECs (67), enhancing ligand-induced responses (65). In line wit endoglin’s role as an upstream coreceptor of the ALK1 pathway, it was shown that AVMs induced by endothelial loss of Eng could be corrected by overexpression of ALK1, whereas endoglin overexpression could not compensate for the loss of Alk1 (68). BMP9 or BMP10 recruits a heterocomplex of two type II receptors (BMPRII or ActRIIA, which are the main type II receptors expressed on ECs) and two type I receptors (ALK1) (1, 69). Upon BMP9 or BMP10 binding, the serine/threonine kinase type II receptor phosphorylates the serine/threonine kinase type I receptor ALK1, leading to its activation. Subsequently, activated ALK1 phosphorylates the transcription factors SMAD1/5, allowing their binding to SMAD4, which is a common downstream signaling mediator shared with the TGF-β pathway (Figure 2). The trimeric SMAD complex then migrates to the nucleus, where it interacts with other transcription factors to regulate the transcription of many target genes (1, 69). Accordingly, endothelium-specific Smad1/5 or Smad4 deletions resulted in AVM formation in the retina (70–72). In addition, few studies show that BMP9 and BMP10 can activate noncanonical BMP signaling pathways, including P38, ERK, Wingless (Wnt), and NOTCH signaling (73). It is widely accepted that BMP9 and BMP10 lead to vascular maturation and quiescence (74). The current working model suggests that BMP9-10/ENG/ALK1/SMAD4 signaling maintains vascular homeostasis via attenuation of proangiogenic pathways and, in particular, the VEGF signaling pathway. However, the mechanisms underlying this attenuation are still not fully characterized.

BMP9-10/ENG/ALK1/SMAD4 signaling pathway maintains vascular quiescence by rFigure 2

BMP9-10/ENG/ALK1/SMAD4 signaling pathway maintains vascular quiescence by repressing angiogenic pathways. HHT occurs due to LOF mutations in ENG, ALK1, SMAD4, and, more rarely, BMP9 (respective proteins indicated with red asterisks), which are all in the same signaling pathway. On endothelial cells, BMP9 or BMP10 recruits a heterocomplex composed of two type II receptors (BMPRII or ActRIIA, which are the main type II receptors expressed on ECs, and two similar type I receptors (ALK1), and the coreceptor ENG (endoglin). Upon BMP binding, the type II receptor phosphorylates ALK1, which subsequently phosphorylates the transcription factors SMAD1/5. SMAD1/5 bind SMAD4, which is shared with the TGF-β pathway, to regulate transcription of many genes (in association with other transcription factors). BMP9 and BMP10 maintain vascular quiescence (middle panel) via repression of angiogenesis pathways (right panel). VEGF-A (red) binds to VEGFR2, which activates the ERK1/2 and P38 MAPK pathways and the PI3K/AKT/mTORC1 pathway. In turn, the PI3K/AKT/mTORC1 pathway activates the signaling cascade P70S6K/S6. VEGF can also activate the calcineurin phosphatase, which activates, via dephosphorylation, the NFAT transcription factor family. The PI3K/AKT/mTOR pathway is negatively regulated by the phosphatase PTEN, which is active when unphosphorylated. VEGF-A can also bind to VEGFR1, but this will not generate a signal. Two other members of the VEGF family, VEGF-B (yellow) and PlGF (blue), also bind to VEGFR1. BMP9 induces the expression of VEGFR1, thus inhibiting VEGF signaling. BMP9 also induces PTEN expression and phosphorylation, which inhibit AKT activity as well as the expression of SGK1 kinase, which can activate the mTORC1/P70S6K/S6 pathway. Moreover, BMP9 inhibits ERK activation and CDK4/6 kinases through not-yet-characterized mechanisms. Ang1 activates the TIE2 receptor to maintain vascular quiescence, and this pathway can be antagonized by Ang2.

The immunosuppressor tacrolimus (FK506) has been identified in two independent screens as a potent activator of SMAD1/5 signaling using a reporter assay based on the Id1 promoter (75, 76). However, the mechanism by which tacrolimus activates this pathway is not clearly understood. Tacrolimus is a macrolide with immunomodulatory and antiangiogenic properties commonly used in patients who have undergone organ transplantation (77). It inhibits the phosphatase calcineurin, which dephosphorylates NFAT proteins (Figure 3) (78, 79). Activation of calcineurin has also been shown to be downstream of VEGF (80) (Figure 2). Interestingly, we have previously shown that BMP9 regulates the calcineurin/NFAT pathway in lymphatic differentiation (81). Tacrolimus can also activate the BMP signaling pathway by blocking 12 kDa FK506 (FKBP12) (Figure 3) (82), which is known to bind and suppress ALK1 activation (76). In ECs, it was shown that tacrolimus activated SMAD1/5 signaling and inhibited AKT and P38 phosphorylation induced by VEGF (75). The same group showed that tacrolimus injection in BMP9/BMP10-immunodepleted postnatal retinas prevented hypervascularization (Table 2). However, the molecular mechanisms involved in this protective effect of tacrolimus are not yet fully elucidated.

Therapeutic targets of antiangiogenic drugs tested in preclinical models anFigure 3

Therapeutic targets of antiangiogenic drugs tested in preclinical models and in HHT patients. Figure shows the targets of drugs tested in completed clinical trials (red), drugs currently under testing or with a case report (orange), and drugs tested only in preclinical models (blue). For further details, see Table 1 and Table 2. Drugs have been developed to block VEGF-A signaling using neutralizing anti–VEGF-A mAbs (bevacizumab) or soluble trap/decoy receptors that bind VEGF-A (red), VEGF-B (yellow), and PlGF (blue) (aflibercept), or neutralizing anti-VEGFR2 antibodies (D5B1 and DC101). Drugs developed to block intracellular signaling, such as tyrosine kinase receptor inhibitors that block VEGFR2 activity but also other receptors, are currently undergoing testing: pazopanib (VEGFR, PDGFR, c-KIT, and FGFR) and nintedanib (VEGFR, PDGFR, and FGFR). Drugs have also been developed to block PI3K and AKT (VAD044), as well as mTORC1 (sirolimus) and calcineurin and FKBP12 (tacrolimus). Immunomodulatory imide drugs (IMIDS), such as thalidomide and pomalidomide, have been tested in HHT patients. Other drugs have been tested so far only in preclinical models, such as the neutralizing anti-Ang2 monoclonal antibodies (LC10) and inhibitors of CDK4/6 (palbociclib and ribociclib).

Table 2

Therapeutic strategies targeting intracellular pathways

VEGF/VEGFR2 pathway. VEGFs, which include VEGF-A, VEGF-B, and PlGF (placenta growth factor), are some of the most potent and extensively studied angiogenic factors. VEGF signaling occurs through its binding to the receptor tyrosine kinase VEGFR2, which activates several downstream pathways (Figure 2). These include the ERK1/2 pathway, the PI3K/AKT/mTOR pathway, the SRC and small GTPases pathways, and others that are poorly understood, including the p38 MAPK pathway (80, 83) (Figure 2). The PI3K/AKT/mTOR pathway is negatively regulated by the phosphatase and tensin homolog (PTEN), which is active when unphosphorylated. VEGF-A, VEGF-B, and PlGF can also bind VEGFR1 with higher affinity than VEGFR2, but the former exhibits low kinase activity, making VEGFR1 a decoy receptor (80). VEGF signaling is thus modulated by the different relative expression levels of VEGFR2 versus VEGFR1 in ECs.

Awareness of the beneficial effect of blocking VEGF signaling in HHT patients dates back to 2006, when an HHT patient suffering from a malignant mesothelioma unexpectedly showed amelioration of HHT symptoms following antiangiogenic cancer treatment with an anti-VEGF-A monoclonal antibody (bevacizumab) (84). The following year, BMP9 and BMP10 were identified as two high-affinity ligands for the receptors ALK1 and endoglin (65, 66). It was shown that these two ligands inhibited angiogenesis in vitro (EC proliferation and migration) and in ex vivo models (66, 85). A few years later, the first clinical trial using bevacizumab showed very positive results on 25 patients suffering from HHT (Table 1) (86). Since then, preclinical HHT models have been developed in order to test different therapeutic approaches. The main model used is the murine retina, a two-dimensional–like vascular structure that forms via angiogenesis during early postnatal days. It has been shown that the loss of Alk1, Eng, Smad1/5, Smad4, or Bmp9/Bmp10 led to spontaneous AVMs in the retina (46). Using these models, it was shown that blocking VEGF signaling reduced the development of vascular malformations (51, 55). These mouse models supported that AVM formation involved aberrant EC responses, including enhanced proliferation, impaired flow-migration coupling, and abnormal cellular behavior in response to angiogenic signals such as VEGF (46).

To date, there is a limited understanding of how BMP9 or BMP10 mitigates VEGF signaling. It has been found that BMP9 induces the expression of VEGFR1, thus restricting downstream VEGF signaling (87) (Figure 2). Conversely, inhibiting ALK1 signaling in ECs using ALK1 ligand trap (ALK1-Fc) promoted an elevation of several key proangiogenic regulators (DLL4, ANGPT2 [encoding angiopoietin 2], and KDR [encoding VEGFR2]) (75). Accordingly, it was shown that Alk1+/– mice presented a reduced level of VEGFR1 expression and that VEGFR1 levels were reduced in skin biopsies from HHT2 patients (88).

Concerning the molecular mechanisms involved downstream of the VEGF signaling pathway, studies in mice have shown that the loss of Alk1 or Smad4 resulted in the activation of the PI3K/AKT pathway (70, 89, 90). Similar results were obtained in vitro in HUVECs using siRNA against ALK1 (70, 90). Inversely, BMP9 treatment for 2 hours in HUVECs was found to increase PTEN expression and activity, leading to a decrease in AKT activity (Figure 2) (89, 90). Activated AKT subsequently activates the mTORC1 complex, which in turn activates the signaling cascade P70S6K/S6 (Figure 2). Interestingly, this pathway has been found activated in skin telangiectases from HHT patients (89, 91). In HUVECs, BMP9 was also found to induce the expression of SGK1 kinase, which can also activate the mTORC1/P70S6K/S6 pathway (92). In this work, it was proposed that activation of this pathway would play a role in regulating protein synthesis. Additionally, BMP9 was also shown to inhibit ERK activation, although the specific mechanism behind this inhibition remains unclear (Figure 2). In parallel, BMP9 and BMP10 have been shown to inhibit endothelial cell proliferation, but the underlying mechanism is not yet clearly characterized. One study showed that BMP9-induced inhibition of EC proliferation was SMAD1/5 dependent and required the expression of the CDK4/6 inhibitor P27KIP (93). In contrast, Smad4 loss enhanced flow-mediated KLF4/TIE2/PI3K/AKT signaling, leading to cell-cycle progression, which was inhibited using CDK4/6 inhibitors (94). Accordingly, the CDK4/6 inhibitors and ribociclib inhibited the formation of retinal AVMs and the knockdown of CDK6 prevented the development of retinal AVMs (Table 2) (94, 95).

Angiopoietin/Tie2 pathway. Genetic HHT mouse models have also shown an increase in Angiopoietin 2 (Ang2) levels (96, 97). The angiopoietin/Tie2 pathway is critical for maintaining vessel stability by regulating, as for VEGF, the PI3K/AKT pathway (Figure 2). Angiopoietin 1 (Ang1), produced by mural cells, activates the receptor tyrosine kinase TIE2 to maintain VSMC coverage. An increase in TIE2 antagonist Ang2 produced by ECs inhibits Ang1-mediated TIE2 activation, leading to destabilization of VSMC-EC interaction and a decrease in vascular quiescence (83, 98). Accordingly, it was recently shown that blocking Ang2 (using LC10 monoclonal antibody) can prevent or reduce AVMs and other HHT-associated abnormalities in mice (96, 97) (Figure 3). However, in contrast with these mouse models, Ang2 circulating levels in HHT patients tend to be reduced compared with those in healthy individuals (99, 100). Thus, although Ang2 seems to be a promising new therapeutic target, further work is needed before pursuing clinical trials (101).

Preclinical data, clinical trials, and perspectives in HHT

Since 2009, antiangiogenic drugs developed in oncology have been increasingly used in drug-repurposing strategies in patients with HHT (102, 103). Today, numerous preclinical (51, 55, 70, 75, 88, 90, 94–97, 104–112) and clinical studies (58, 86, 104, 113–136) targeting angiogenesis in the scope of HHT have been carried out and are summarized in Table 1 and Table 2. Drugs used in the aforementioned studies and their distinct sites of action are depicted in Figure 3.

Today, with our current knowledge of the pathophysiological mechanisms of HHT owing to preclinical models and clinical trials, the main therapeutic lines of action to be considered include the following: first, avoiding angiogenic and inflammatory stimuli by all means, in line with the second hit hypothesis. Whenever possible, “mechanical” prevention, such as sun protection, application of nasal ointments, and avoidance of nasal cauterization during childhood, is recommended to reduce the development of cutaneous and mucosal telangiectases. In addition, it is essential to prevent and treat anemia and iron deficiency, whose repercussions are probably not limited to hematology alone, but also include the risk of infection and stroke (43, 137).

Another important point is moving toward personalized medicine for each patient. While studies across 14 years that involved more than 400 patients ensured the safety of bevacizumab, they demonstrated that the response varied depending on its elimination rate (58, 86). Indeed, pharmacokinetic studies highlighted a correlation between a progressive decrease in response to the drug and low plasma bevacizumab concentration. Thus, as in cancer, there is a concentration threshold for efficacy. So taking into account the metabolisms of different patients and adapting the dosage accordingly would allow us to maintain its efficacy and optimize the risk-benefit balance. It is therefore essential to monitor clinical improvement as a function of bevacizumab levels during maintenance therapy in order to maintain bioactive levels within the therapeutic window, especially if efficacy appears to be declining (120). In case of treatment failure, it is mandatory to verify that bevacizumab residual levels are above 25 mg/L (120, 138).

Another aspect that should be considered more carefully is tailoring treatment to disease severity in order to limit drug toxicity. To reduce systemic adverse effects, antiangiogenic treatments applied locally have been studied. For example, 0.1% tacrolimus ointment applied twice daily has promising outcomes (139) and could be a good option for patients with moderate epistaxis, although this awaits further confirmation by larger studies. Moreover, continued use of tacrolimus in HHT patients could be called into question, as a few HHT patients using this regimen after liver transplantation developed new hepatic vascular lesions and needed a second transplantation (30). Other antiangiogenic drugs with local use are more debated. Two randomized trials showed that intranasal bevacizumab was not effective (140, 141). This result was confirmed in a systematic review (142) conducted on 13 studies. Some studies evaluated submucosal administration alone (143, 144), with electrocautery (145) or cyanoacrylate glue (146). In addition, nonselective β-adrenergic receptor blockers with antiangiogenic properties that reduce VEGF and matrix metalloproteinase-9 (MMP-9) tissue expression have been studied (147). For instance, four randomized studies using topical timolol in the form of a nasal spray or gel for epistaxis and an ophthalmic solution for cutaneous telangiectases reported discordant results in cohorts ranging from 6 to 58 patients (148–151). This is consistent with a systematic review from 2022 (152) on topical beta blockers suggesting that propranolol is more promising than timolol. Topical propranolol was reported to increase hemoglobin levels (153, 154), but its use warrants monitoring for bradycardia occurrence, even when administered locally.

Future challenges in HHT

Although great progress has been made in the treatment of HHT patients, there are still some remaining challenges. Perhaps the greatest challenge is determining whether large AVMs observed in HHT patients would respond to antiangiogenic therapies. Indeed, several antiangiogenic drugs, particularly those administered orally, including tyrosine kinase inhibitors, AKT inhibitors, pomalidomide, and mTOR inhibitors, are currently being tested on visceral AVMs, but no regression of visceral AVMs has been observed so far. Only skin or mucosal telangiectases have been found to be reduced in a few cases (125, 128). In the Metafore study (86), cardiac index and hepatic blood flow were decreased after bevacizumab treatment, but large liver AVMs on CT scan were unchanged, and despite a marked improvement in digestive bleeding, the number of GI telangiectases was not reduced. Although some preclinical studies reported that antiangiogenic treatments, such as PI3K inhibitors, could revert established AVMs in preclinical models (90), these AVMs were only seen in neonatal retinas of mice. Similarly, it was shown that in zebrafish Eng mutants, the HHT-like phenotype was abrogated by inhibiting VEGF signaling with drugs targeting VEGFR2, but only in embryos (112). In both models, the treatment was effective during active angiogenesis (embryonic or postnatal stages), which is not the case after remodeling of vascular structure in large AVMs. Thus there is a real need for testing the efficacy of drugs in preclinical models on large AVMs or AVMs at adult stages. Accordingly, recent preclinical HHT models that better mimic HHT symptoms have been developed in adult mice and zebrafish (55, 106, 107, 112). However, to maximize the potential of currently available models for monitoring the response of AVMs to tested drugs, advancements in visualization methodologies are required for the deeper investigation of AVM formation, progression, and regression. Such advancements might also allow the revelation of previously undetected visceral AVMs in HHT mouse models, necessitating further investigation of these models using advanced visualization tools in the future.

Another challenge is to identify the best target to inhibit angiogenesis in HHT patients. VEGF signaling is complex and drives numerous downstream pathways, so adverse complications can be caused by inhibiting the VEGF pathway as a whole (155). Thus, one could imagine that targeting specific downstream pathways of VEGF could reduce these complications. A recent study addressed this point by using an adult endoglin-mutant zebrafish model that developed several HHT symptoms (112). They found that inhibiting mTORC1 (using rapamycin, also known as sirolimus) or MEK pathways prevented vascular abnormalities (Table 2), whereas inhibiting P38 or nitric oxide synthase pathways had no effect. Combined subtherapeutic mTORC1 and MEK inhibitors demonstrated synergistic effects in treating HHT. However, it remains unclear at this stage whether it will be more beneficial in the future to only target particular downstream targets of VEGF (PI3K, MEK), which are activated by many signaling pathways, or to target a specific proangiogenic growth factor, such as VEGF.

In the present Review, we focus on therapeutic approaches that aim to block activated signaling pathways due to LOF of the BMP9-10/ALK1 signaling pathway, but another possibility would be to increase the deficient signaling pathway (more ligands, more receptors, activation of the downstream signaling pathway). However, the validity of such a therapeutic option fundamentally relies on whether HHT vascular lesions are driven by haploinsufficiency or biallelic loss of the affected gene.

Summary and conclusions

Ever since the discovery that HHT is due to mutations in a single signaling pathway nearly 30 years ago, considerable progress has been made in treating HHT symptoms by blocking the VEGF angiogenic pathway. However, the molecular mechanism underlying the interaction between BMP9/BMP10 and VEGF signaling is still not fully elucidated. In addition, as described in this Review, several points remain to be better characterized, such as the development of AVMs, preclinical adult models for AVMs, biomarkers for personalized antiangiogenic treatments, the choice of therapeutic target (growth factors such as VEGF or more focused downstream signaling [AKT, MTORC1, MEK]), the possibility and efficacy of localized treatment and the benefit of preventative antiangiogenic treatments. Larger scale and real-life data are particularly difficult to obtain, since these treatments are used off-label due to a lack of better options. This is partly due to the general abstention of pharmaceutical industries from repurposing drugs in rare diseases. In the meantime, antiangiogenic drugs hold a promising potential in HHT and represent avenues worth investigating before taking on the future challenges associated with gene therapy.

Acknowledgments

The team is supported by the National Institute for Health and Medical Research (INSERM), the University of Grenoble-Alpes, the Commissariat à l’Énergie Atomique et aux Énergies Alternatives DRF/IRIG/DS (CEA), the Hospices Civils de Lyon (HCL), the Fondation Pour la Recherche Médicale (EQU202003010188), the Association Maladie de Rendu-Osler (AMRO/HHT France), the Association FAVA-Multi, the H2020-MSCA-ITN-2018 (VA Cure-84316), the French National Agency for Research (ANR) grant no. ANR-20-CE14-0002 (SMAD4pathy), and GRAL, a program from the Chemistry Biology Health (CBH) Graduate School of University Grenoble Alpes (ANR-17-EURE-0003).

Address correspondence to: Sabine Bailly, Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, F-38000 Grenoble, France. Email: sabine.bailly@cea.fr.

Footnotes

Conflict of interest: The authors have declared that no conflict of interest exists.

Copyright: © 2024, Al Tabosh et al. This is an open access article published under the terms of the Creative Commons Attribution 4.0 International License.

Reference information: J Clin Invest. 2024;134(4):e176379. https://doi.org/10.1172/JCI176379.

References
  1. Desroches-Castan A, et al. BMP9 and BMP10: Two close vascular quiescence partners that stand out. Dev Dyn. 2022;251(1):178–197.
    View this article via: CrossRef PubMed Google Scholar
  2. McAllister KA, et al. Endoglin, a TGF-P binding protein of endothelial cells, is the gene for hereditary haemorrhagic telangiectasia type. Nat Genet. 1994;8(4):345–351.
    View this article via: CrossRef PubMed Google Scholar
  3. Johnson DW, et al. Mutations in the activin receptor-like kinase 1 gene in hereditary haemorrhagic telangiectasia type 2. Nat Genet. 1996;13(2):189–195.
    View this article via: CrossRef PubMed Google Scholar
  4. McDonald J, et al. Curaçao diagnostic criteria for hereditary hemorrhagic telangiectasia is highly predictive of a pathogenic variant in ENG or ACVRL1 (HHT1 and HHT2). Genet Med. 2020;22(7):1201–1205.
    View this article via: CrossRef PubMed Google Scholar
  5. National Library of Medicine. ClinVar. https://www.ncbi.nlm.nih.gov/clinvar/ Accessed November 9, 2023.
  6. Gallione CJ, et al. SMAD4 mutations found in unselected HHT patients. J Med Genet. 2006;43(10):793–797.
    View this article via: CrossRef PubMed Google Scholar
  7. Wooderchak WL, et al. Repository of SMAD4 mutations: reference for genotype/ phenotype correlation. J Data Mining in Genom Proteomics. 2010;1(1):1000101.
    View this article via: CrossRef Google Scholar
  8. Wooderchak-Donahue WL, et al. Genome sequencing reveals a deep intronic splicing ACVRL1 mutation hotspot in Hereditary Haemorrhagic Telangiectasia. J Med Genet. 2018;55(12):824–830.
    View this article via: CrossRef PubMed Google Scholar
  9. Wooderchak-Donahue WL, et al. BMP9 mutations cause a vascular-anomaly syndrome with phenotypic overlap with hereditary hemorrhagic telangiectasia. Am J Hum Genet. 2013;93(3):530–537.
    View this article via: CrossRef PubMed Google Scholar
  10. Balachandar S, et al. Identification and validation of a novel pathogenic variant in GDF2 (BMP9) responsible for hereditary hemorrhagic telangiectasia and pulmonary arteriovenous malformations. Am J Med Genet A. 2022;188(3):959–964.
    View this article via: CrossRef PubMed Google Scholar
  11. Liu J, et al. Homozygous GDF2-related hereditary hemorrhagic telangiectasia in a Chinese family. Pediatrics. 2020;146(2):e20191970.
    View this article via: CrossRef PubMed Google Scholar
  12. Hodgson J, et al. Homozygous GDF2 nonsense mutations result in a loss of circulating BMP9 and BMP10 and are associated with either PAH or an “HHT-like” syndrome in children. Molec Gen Gen Med. 2021;9(12):e1685.
    View this article via: CrossRef PubMed Google Scholar
  13. Farhan A, et al. Clinical manifestations of patients with GDF2 mutations associated with hereditary hemorrhagic telangiectasia type 5. Am J Med Genet A. 2022;188(1):199–209.
    View this article via: CrossRef PubMed Google Scholar
  14. Revencu N, et al. RASA1 mutations and associated phenotypes in 68 families with capillary malformation-arteriovenous malformation. Hum Mutat. 2013;34(12):1632–1641.
    View this article via: CrossRef PubMed Google Scholar
  15. Orme CM, et al. Capillary malformation--arteriovenous malformation syndrome: review of the literature, proposed diagnostic criteria, and recommendations for management. Pediatr Dermatol. 2013;30(4):409–415.
    View this article via: CrossRef PubMed Google Scholar
  16. El Hajjam M, et al. RASA1 phenotype overlaps with hereditary haemorrhagic telangiectasia: two case reports. J Med Genet. 2021;58(9):645–647.
    View this article via: CrossRef PubMed Google Scholar
  17. Hernandez F, et al. Mutations in RASA1 and GDF2 identified in patients with clinical features of hereditary hemorrhagic telangiectasia. Hum Genome Var. 2015;2:15040.
    View this article via: CrossRef PubMed Google Scholar
  18. Wooderchak-Donahue WL, et al. Expanding the clinical and molecular findings in RASA1 capillary malformation-arteriovenous malformation. Eur J Hum Genet. 2018;26(10):1521–1536.
    View this article via: CrossRef PubMed Google Scholar
  19. Guilhem A, et al. Seven cases of hereditary haemorrhagic telangiectasia-like hepatic vascular abnormalities associated with EPHB4 pathogenic variants. J Med Genet. 2023;60(9):905–909.
    View this article via: CrossRef PubMed Google Scholar
  20. Jiang X, et al. Inactivating mutations in Drosha mediate vascular abnormalities similar to hereditary hemorrhagic telangiectasia. Sci Signal. 2018;11(513):eaan6831.
    View this article via: CrossRef PubMed Google Scholar
  21. Wassef M, et al. Vascular anomalies classification: recommendations from the international society for the study of vascular anomalies. Pediatrics. 2015;136(1):e203–e214.
    View this article via: CrossRef PubMed Google Scholar
  22. Braverman IM, et al. Ultrastructure and three-dimensional organization of the telangiectases of hereditary hemorrhagic telangiectasia. J Invest Dermatol. 1990;95(4):422–427.
    View this article via: CrossRef PubMed Google Scholar
  23. Singh E, et al. Arterial endoglin does not protect against arteriovenous malformations. Angiogenesis. 2020;23(4):559–566.
    View this article via: CrossRef PubMed Google Scholar
  24. Park H, et al. Defective flow-migration coupling causes arteriovenous malformations in hereditary hemorrhagic telangiectasia. Circulation. 2021;144(10):805–822.
    View this article via: CrossRef PubMed Google Scholar
  25. McDonald J, et al. Frequency of epistaxis and telangiectasia in patients with hereditary hemorrhagic telangiectasia (HHT) in comparison with the general population: Curaçao diagnostic criteria revisited. Genet Med. 2023;25(8):100865.
    View this article via: CrossRef PubMed Google Scholar
  26. Hyldahl SJ, et al. Skin and mucosal telangiectatic lesions in hereditary hemorrhagic telangiectasia patients. Int J Dermatol. 2022;61(12):1497–1505.
    View this article via: CrossRef PubMed Google Scholar
  27. Kasthuri RS, et al. Prevalence and predictors of anemia in hereditary hemorrhagic telangiectasia. Am J Hematol. 2017;92(10):591–593.
    View this article via: CrossRef PubMed Google Scholar
  28. Dupuis-Girod S, et al. The lung in hereditary hemorrhagic telangiectasia. Respiration. 2017;94(4):315–330.
    View this article via: CrossRef PubMed Google Scholar
  29. Buscarini E, et al. Liver involvement in hereditary hemorrhagic telangiectasia. Abdom Radiol (NY). 2018;43(8):1920–1930.
    View this article via: CrossRef PubMed Google Scholar
  30. Dumortier J, et al. Recurrence of hereditary hemorrhagic telangiectasia after liver transplantation: clinical implications and physiopathological insights. Hepatology. 2019;69(5):2232–2240.
    View this article via: CrossRef PubMed Google Scholar
  31. Eker OF, et al. European Reference Network for Rare Vascular Diseases (VASCERN) position statement on cerebral screening in adults and children with hereditary haemorrhagic telangiectasia (HHT). Orphanet J Rare Dis. 2020;15(1):165.
    View this article via: CrossRef PubMed Google Scholar
  32. Plauchu H, et al. Age-related clinical profile of hereditary hemorrhagic telangiectasia in an epidemiologically recruited population. Am J Med Genet. 1989;32(3):291–297.
    View this article via: CrossRef PubMed Google Scholar
  33. Lesca G, et al. Genotype-phenotype correlations in hereditary hemorrhagic telangiectasia: data from the French-Italian HHT network. Genet Med. 2007;9(1):14–22.
    View this article via: CrossRef PubMed Google Scholar
  34. Dupuis-Girod S, et al. Hemorrhagic hereditary telangiectasia (Rendu-Osler disease) and infectious diseases: an underestimated association. Clin Infect Dis. 2007;44(6):841–845.
    View this article via: CrossRef PubMed Google Scholar
  35. Guilhem A, et al. Immunological abnormalities associated with hereditary haemorrhagic telangiectasia. J Intern Med. 2013;274(4):351–362.
    View this article via: CrossRef PubMed Google Scholar
  36. Cymerman U, et al. Identification of hereditary hemorrhagic telangiectasia type 1 in newborns by protein expression and mutation analysis of endoglin. Pediatr Res. 2000;47(1):24–35.
    View this article via: CrossRef PubMed Google Scholar
  37. Fernandez-L A, et al. Blood outgrowth endothelial cells from Hereditary Haemorrhagic Telangiectasia patients reveal abnormalities compatible with vascular lesions. Cardiovasc Res. 2005;68(2):235–248.
    View this article via: CrossRef PubMed Google Scholar
  38. Fernandez-L A, et al. Mutation study of Spanish patients with hereditary hemorrhagic telangiectasia and expression analysis of Endoglin and ALK1. Hum Mutat. 2006;27(3):295.
    View this article via: CrossRef PubMed Google Scholar
  39. Abdalla SA. Analysis of ALK-1 and endoglin in newborns from families with hereditary hemorrhagic telangiectasia type 2. Hum Mol Genet. 2000;9(8):1227–1237.
    View this article via: CrossRef PubMed Google Scholar
  40. Bourdeau A, et al. A murine model of hereditary hemorrhagic telangiectasia. J Clin Invest. 1999;104(10):1343–1351.
    View this article via: JCI CrossRef PubMed Google Scholar
  41. Srinivasan S. A mouse model for hereditary hemorrhagic telangiectasia (HHT) type 2. Hum Mol Genet. 2003;12(5):473–482.
    View this article via: CrossRef PubMed Google Scholar
  42. Arthur HM, et al. Endoglin, an ancillary TGFbeta receptor, is required for extraembryonic angiogenesis and plays a key role in heart development. Dev Biol. 2000;217(1):42–53.
    View this article via: CrossRef PubMed Google Scholar
  43. Faughnan ME, et al. Second international guidelines for the diagnosis and management of hereditary hemorrhagic telangiectasia. Ann Intern Med. 2020;173(12):989–1001.
    View this article via: CrossRef PubMed Google Scholar
  44. Kritharis A, et al. Hereditary hemorrhagic telangiectasia: diagnosis and management from the hematologist’s perspective. Haematologica. 2018;103(9):1433–1443.
    View this article via: CrossRef PubMed Google Scholar
  45. Bernabeu C, et al. Potential second-hits in hereditary hemorrhagic telangiectasia. J Clin Med. 2020;9(11):3571.
    View this article via: CrossRef PubMed Google Scholar
  46. Arthur HM, Roman BL. An update on preclinical models of hereditary haemorrhagic telangiectasia: Insights into disease mechanisms. Front Med (Lausanne). 2022;9:973964.
    View this article via: CrossRef PubMed Google Scholar
  47. Xu B, et al. Vascular endothelial growth factor induces abnormal microvasculature in the endoglin heterozygous mouse brain. J Cereb Blood Flow Metab. 2004;24(2):237–244.
    View this article via: CrossRef PubMed Google Scholar
  48. Hao Q, et al. VEGF induces more severe cerebrovascular dysplasia in endoglin than in Alk1 mice. Transl Stroke Res. 2010;1(3):197–201.
    View this article via: CrossRef PubMed Google Scholar
  49. Choi E-J, et al. Novel brain arteriovenous malformation mouse models for type 1 hereditary hemorrhagic telangiectasia. PLoS One. 2014;9(2):e88511.
    View this article via: CrossRef PubMed Google Scholar
  50. Choi E-J, et al. Minimal homozygous endothelial deletion of Eng with VEGF stimulation is sufficient to cause cerebrovascular dysplasia in the adult mouse. Cerebrovasc Dis. 2012;33(6):540–547.
    View this article via: CrossRef PubMed Google Scholar
  51. Walker EJ, et al. Bevacizumab attenuates VEGF-induced angiogenesis and vascular malformations in the adult mouse brain. Stroke. 2012;43(7):1925–1930.
    View this article via: CrossRef PubMed Google Scholar
  52. Walker EJ, et al. Arteriovenous malformation in the adult mouse brain resembling the human disease. Ann Neurol. 2011;69(6):954–962.
    View this article via: CrossRef PubMed Google Scholar
  53. Park SO, et al. Real-time imaging of de novo arteriovenous malformation in a mouse model of hereditary hemorrhagic telangiectasia. J Clin Invest. 2009;119(11):3487–3496.
    View this article via: JCI PubMed Google Scholar
  54. Garrido-Martin EM, et al. Common and distinctive pathogenetic features of arteriovenous malformations in hereditary hemorrhagic telangiectasia 1 and hereditary hemorrhagic telangiectasia 2 animal models--brief report. Arterioscler Thromb Vasc Biol. 2014;34(10):2232–2236.
    View this article via: CrossRef PubMed Google Scholar
  55. Han C, et al. VEGF neutralization can prevent and normalize arteriovenous malformations in an animal model for hereditary hemorrhagic telangiectasia 2. Angiogenesis. 2014;17(4):823–830.
    View this article via: CrossRef PubMed Google Scholar
  56. Mahmoud M, et al. Pathogenesis of arteriovenous malformations in the absence of endoglin. Circ Res. 2010;106(8):1425–1433.
    View this article via: CrossRef PubMed Google Scholar
  57. Dupuis-Girod S, et al. European Reference Network for Rare Vascular Diseases (VASCERN): When and how to use intravenous bevacizumab in hereditary haemorrhagic telangiectasia (HHT)? Eur J Med Genet. 2022;65(10):104575.
    View this article via: CrossRef PubMed Google Scholar
  58. Al-Samkari H, et al. An international, multicenter study of intravenous bevacizumab for bleeding in hereditary hemorrhagic telangiectasia: the InHIBIT-Bleed study. Haematologica. 2021;106(8):2161–2169.
    View this article via: CrossRef PubMed Google Scholar
  59. Geisthoff U, et al. Trauma can induce telangiectases in hereditary hemorrhagic telangiectasia. J Clin Med. 2020;9(5):1507.
    View this article via: CrossRef PubMed Google Scholar
  60. Snellings DA, et al. Somatic mutations in vascular malformations of hereditary hemorrhagic telangiectasia result in bi-allelic loss of ENG or ACVRL1. Am J Hum Genet. 2019;105(5):894–906.
    View this article via: CrossRef PubMed Google Scholar
  61. Maris JM, Knudson AG. Revisiting tissue specificity of germline cancer predisposing mutations. Nat Rev Cancer. 2015;15(2):65–66.
    View this article via: CrossRef PubMed Google Scholar
  62. Brouillard P, Vikkula M. Genetic causes of vascular malformations. Hum Mol Genet. 2007;16 Spec No. 2(r2):R140–R149.
    View this article via: CrossRef PubMed Google Scholar
  63. Snellings DA, et al. Developmental venous anomalies are a genetic primer for cerebral cavernous malformations. Nat Cardiovasc Res. 2022;1(3):246–252.
    View this article via: CrossRef PubMed Google Scholar
  64. Brinjikji W, et al. Prevalence and characteristics of brain arteriovenous malformations in hereditary hemorrhagic telangiectasia: a systematic review and meta-analysis. J Neurosurg. 2017;127(2):302–310.
    View this article via: CrossRef PubMed Google Scholar
  65. David L, et al. Identification of BMP9 and BMP10 as functional activators of the orphan activin receptor-like kinase 1 (ALK1) in endothelial cells. Blood. 2007;109(5):1953–1961.
    View this article via: CrossRef PubMed Google Scholar
  66. Scharpfenecker M, et al. BMP-9 signals via ALK1 and inhibits bFGF-induced endothelial cell proliferation and VEGF-stimulated angiogenesis. J Cell Sci. 2007;120(pt 6):964–972.
    View this article via: CrossRef PubMed Google Scholar
  67. Lawera A, et al. Role of soluble endoglin in BMP9 signaling. Proc Natl Acad Sci U S A. 2019;116(36):17800–17808.
    View this article via: CrossRef PubMed Google Scholar
  68. Hwan Kim Y, et al. Overexpression of activin receptor-like kinase 1 in endothelial cells suppresses development of arteriovenous malformations in mouse models of hereditary hemorrhagic telangiectasia. Circ Res. 2020;127(9):1122–1137.
    View this article via: CrossRef PubMed Google Scholar
  69. Roman BL, Hinck AP. ALK1 signaling in development and disease: new paradigms. Cell Mol Life Sci. 2017;74(24):4539–4560.
    View this article via: CrossRef PubMed Google Scholar
  70. Ola R, et al. SMAD4 prevents flow induced arteriovenous malformations by inhibiting casein kinase 2. Circulation. 2018;138(21):2379–2394.
    View this article via: CrossRef PubMed Google Scholar
  71. Crist AM, et al. Vascular deficiency of Smad4 causes arteriovenous malformations: a mouse model of hereditary hemorrhagic telangiectasia. Angiogenesis. 2018;21(2):363–380.
    View this article via: CrossRef PubMed Google Scholar
  72. Benn A, et al. BMP-SMAD1/5 signaling regulates retinal vascular development. Biomolecules. 2020;10(3):488.
    View this article via: CrossRef PubMed Google Scholar
  73. García de Vinuesa A, et al. BMP signaling in vascular biology and dysfunction. Cytokine Growth Factor Rev. 2016;27:65–79.
    View this article via: CrossRef PubMed Google Scholar
  74. Ricard N, et al. The quiescent endothelium: signalling pathways regulating organ-specific endothelial normalcy. Nat Rev Cardiol. 2021;18(8):565–580.
    View this article via: CrossRef PubMed Google Scholar
  75. Ruiz S, et al. Tacrolimus rescues the signaling and gene expression signature of endothelial ALK1 loss-of-function and improves HHT vascular pathology. Hum Mol Genet. 2017;26(24):4786–4798.
    View this article via: CrossRef PubMed Google Scholar
  76. Spiekerkoetter E, et al. FK506 activates BMPR2, rescues endothelial dysfunction, and reverses pulmonary hypertension. J Clin Invest. 2013;123(8):3600–3613.
    View this article via: JCI CrossRef PubMed Google Scholar
  77. Posadas Salas MA, Srinivas TR. Update on the clinical utility of once-daily tacrolimus in the management of transplantation. Drug Des Devel Ther. 2014;8:1183–1194.
    View this article via: CrossRef PubMed Google Scholar
  78. Horsley V, Pavlath GK. NFAT: ubiquitous regulator of cell differentiation and adaptation. J Cell Biol. 2002;156(5):771–774.
    View this article via: CrossRef PubMed Google Scholar
  79. Tocci MJ, Sigal NH. Recent advances in the mechanism of action of cyclosporine and FK506. Curr Opin Nephrol Hypertens. 1992;1(2):236–242.
    View this article via: CrossRef PubMed Google Scholar
  80. Simons M, et al. Mechanisms and regulation of endothelial VEGF receptor signalling. Nat Rev Mol Cell Biol. 2016;17(10):611–625.
    View this article via: CrossRef PubMed Google Scholar
  81. Subileau M, et al. Bone Morphogenetic protein 9 regulates early lymphatic-specified endothelial cell expansion during mouse embryonic stem cell differentiation. Stem Cell Reports. 2019;12(1):98–111.
    View this article via: CrossRef PubMed Google Scholar
  82. Chen YG, et al. Mechanism of TGFbeta receptor inhibition by FKBP12. EMBO J. 1997;16(13):3866–3876.
    View this article via: CrossRef PubMed Google Scholar
  83. Graupera M, Potente M. Regulation of angiogenesis by PI3K signaling networks. Exp Cell Res. 2013;319(9):1348–1355.
    View this article via: CrossRef PubMed Google Scholar
  84. Flieger D, et al. Dramatic improvement in hereditary hemorrhagic telangiectasia after treatment with the vascular endothelial growth factor (VEGF) antagonist bevacizumab. Ann Hematol. 2006;85(9):631–632.
    View this article via: CrossRef PubMed Google Scholar
  85. David L, et al. Bone morphogenetic protein-9 is a circulating vascular quiescence factor. Circ Res. 2008;102(8):914–922.
    View this article via: CrossRef PubMed Google Scholar
  86. Dupuis-Girod S, et al. Bevacizumab in patients with hereditary hemorrhagic telangiectasia and severe hepatic vascular malformations and high cardiac output. JAMA. 2012;307(9):948–955.
    View this article via: CrossRef PubMed Google Scholar
  87. Larrivée B, et al. ALK1 signaling inhibits angiogenesis by cooperating with the Notch pathway. Dev Cell. 2012;22(3):489–500.
    View this article via: CrossRef PubMed Google Scholar
  88. Thalgott JH, et al. Decreased expression of vascular endothelial growth factor receptor 1 contributes to the pathogenesis of hereditary hemorrhagic telangiectasia type 2. Circulation. 2018;138(23):2698–2712.
    View this article via: CrossRef PubMed Google Scholar
  89. Alsina-Sanchís E, et al. ALK1 loss results in vascular hyperplasia in mice and humans through PI3K activation. Arterioscler Thromb Vasc Biol. 2018;38(5):1216–1229.
    View this article via: CrossRef PubMed Google Scholar
  90. Ola R, et al. PI3 kinase inhibition improves vascular malformations in mouse models of hereditary haemorrhagic telangiectasia. Nat Commun. 2016;7(1):13650.
    View this article via: CrossRef PubMed Google Scholar
  91. Iriarte A, et al. PI3K (Phosphatidylinositol 3-Kinase) activation and endothelial cell proliferation in patients with hemorrhagic hereditary telangiectasia type 1. Cells. 2019;8(9):971.
    View this article via: CrossRef PubMed Google Scholar
  92. Medina-Jover F, et al. SGK1 is a signalling hub that controls protein synthesis and proliferation in endothelial cells. FEBS Lett. 2020;594(19):3200–3215.
    View this article via: CrossRef PubMed Google Scholar
  93. Rostama B, et al. DLL4/Notch1 and BMP9 interdependent signaling induces human endothelial cell quiescence via P27KIP1 and thrombospondin-1. Arterioscler Thromb Vasc Biol. 2015;35(12):2626–2637.
    View this article via: CrossRef PubMed Google Scholar
  94. Banerjee K, et al. SMAD4 maintains the fluid shear stress set point to protect against arterial-venous malformations. J Clin Invest. 2023;133(18):e168352.
    View this article via: JCI CrossRef PubMed Google Scholar
  95. Dinakaran S, et al. CDK6-mediated endothelial cell cycle acceleration drives arteriovenous malformations in hereditary hemorrhagic telangiectasia [preprint]. Posted on bioRxiv September 16, 2023. https://doi.org/10.1101/2023.09.15.554413.
  96. Zhou X, et al. ANG2 blockade diminishes proangiogenic cerebrovascular defects associated with models of hereditary hemorrhagic telangiectasia. Arterioscler Thromb Vasc Biol. 2023;43(8):1384–1403.
    View this article via: CrossRef PubMed Google Scholar
  97. Crist AM, et al. Angiopoietin-2 inhibition rescues arteriovenous malformation in a smad4 hereditary hemorrhagic telangiectasia mouse model. Circulation. 2019;139(17):2049–2063.
    View this article via: CrossRef PubMed Google Scholar
  98. Kim M, et al. Opposing actions of angiopoietin-2 on Tie2 signaling and FOXO1 activation. J Clin Invest. 2016;126(9):3511–3525.
    View this article via: JCI CrossRef PubMed Google Scholar
  99. Ojeda-Fernandez L, et al. Reduced plasma levels of Ang-2 and sEng as novel biomarkers in hereditary hemorrhagic telangiectasia (HHT). Clin Chim Acta. 2010;411(7–8):494–499.
    View this article via: CrossRef PubMed Google Scholar
  100. Fernandez-L A, et al. Gene expression fingerprinting for human hereditary hemorrhagic telangiectasia. Hum Mol Genet. 2007;16(13):1515–1533.
    View this article via: CrossRef PubMed Google Scholar
  101. Bernabeu C. Therapeutic targeting of the Ang2/tie pathway in endothelial cells as a potential treatment of hereditary hemorrhagic telangiectasia. Arterioscler Thromb Vasc Biol. 2023;43(8):1404–1408.
    View this article via: CrossRef PubMed Google Scholar
  102. Ardelean DS, Letarte M. Anti-angiogenic therapeutic strategies in hereditary hemorrhagic telangiectasia. Front Genet. 2015;6:35.
    View this article via: CrossRef PubMed Google Scholar
  103. Snodgrass RO, et al. Hereditary haemorrhagic telangiectasia, an inherited vascular disorder in need of improved evidence-based pharmaceutical interventions. Genes (Basel). 2021;12(2):174.
    View this article via: CrossRef PubMed Google Scholar
  104. Lebrin F, et al. Thalidomide stimulates vessel maturation and reduces epistaxis in individuals with hereditary hemorrhagic telangiectasia. Nat Med. 2010;16(4):420–428.
    View this article via: CrossRef PubMed Google Scholar
  105. Ardelean DS, et al. Endoglin and activin receptor-like kinase 1 heterozygous mice have a distinct pulmonary and hepatic angiogenic profile and response to anti-VEGF treatment. Angiogenesis. 2014;17(1):129–146.
    View this article via: CrossRef PubMed Google Scholar
  106. Tual-Chalot S, et al. Loss of endothelial endoglin promotes high-output heart failure through peripheral arteriovenous shunting driven by VEGF signaling. Circ Res. 2020;126(2):243–257.
    View this article via: CrossRef PubMed Google Scholar
  107. Kim YH, et al. Selective effects of oral antiangiogenic tyrosine kinase inhibitors on an animal model of hereditary hemorrhagic telangiectasia. J Thromb Haemost. 2017;15(6):1095–1102.
    View this article via: CrossRef PubMed Google Scholar
  108. Jin Y, et al. Endoglin prevents vascular malformation by regulating flow-induced cell migration and specification through VEGFR2 signalling. Nat Cell Biol. 2017;19(6):639–652.
    View this article via: CrossRef PubMed Google Scholar
  109. Ruiz S, et al. Correcting Smad1/5/8, mTOR, and VEGFR2 treats pathology in hereditary hemorrhagic telangiectasia models. J Clin Invest. 2020;130(2):942–957.
    View this article via: JCI CrossRef PubMed Google Scholar
  110. Peng H-L, et al. Thalidomide effects in patients with hereditary hemorrhagic telangiectasia during therapeutic treatment and in fli-EGFP transgenic zebrafish model. Chin Med J (Engl). 2015;128(22):3050–3054.
    View this article via: CrossRef PubMed Google Scholar
  111. Zhu W, et al. Thalidomide reduces hemorrhage of brain arteriovenous malformations in a mouse model. Stroke. 2018;49(5):1232–1240.
    View this article via: CrossRef PubMed Google Scholar
  112. Snodgrass RO, et al. Therapeutic targeting of vascular malformation in a zebrafish model of hereditary haemorrhagic telangiectasia. Dis Model Mech. 2023;16(4):dmm049567.
    View this article via: CrossRef PubMed Google Scholar
  113. Chavan A, et al. Systemic therapy with bevacizumab in patients with hereditary hemorrhagic telangiectasia (HHT). Vasa. 2013;42(2):106–110.
    View this article via: CrossRef PubMed Google Scholar
  114. Thompson AB, et al. Very low dose bevacizumab for the treatment of epistaxis in patients with hereditary hemorrhagic telangiectasia. Allergy Rhinol (Providence). 2014;5(2):91–95.
    View this article via: CrossRef PubMed Google Scholar
  115. Guilhem A, et al. Intra-venous bevacizumab in hereditary hemorrhagic telangiectasia (HHT): A retrospective study of 46 patients. PLoS One. 2017;12(11):e0188943.
    View this article via: CrossRef PubMed Google Scholar
  116. Chavan A, et al. Emerging role of bevacizumab in management of patients with symptomatic hepatic involvement in Hereditary Hemorrhagic Telangiectasia. Am J Hematol. 2017;92(11):E641–E644.
    View this article via: CrossRef PubMed Google Scholar
  117. Iyer VN, et al. Intravenous bevacizumab for refractory hereditary hemorrhagic telangiectasia-related epistaxis and gastrointestinal bleeding. Mayo Clin Proc. 2018;93(2):155–166.
    View this article via: CrossRef PubMed Google Scholar
  118. Al-Samkari H, et al. An international survey to evaluate systemic bevacizumab for chronic bleeding in hereditary haemorrhagic telangiectasia. Haemophilia. 2020;26(6):1038–1045.
    View this article via: CrossRef PubMed Google Scholar
  119. Vázquez C, et al. Bevacizumab for treating Hereditary Hemorrhagic Telangiectasia patients with severe hepatic involvement or refractory anemia. PLoS One. 2020;15(2):e0228486.
    View this article via: CrossRef PubMed Google Scholar
  120. Dupuis-Girod S, et al. Efficacy and safety of intravenous bevacizumab on severe bleeding associated with hemorrhagic hereditary telangiectasia: A national, randomized multicenter trial. J Intern Med. 2023;294(6):761–774.
    View this article via: CrossRef PubMed Google Scholar
  121. Villanueva B, et al. Aflibercept for gastrointestinal bleeding in hereditary hemorrhagic telangiectasia: a case report. Medicina (Kaunas). 2023;59(9):1533.
    View this article via: CrossRef PubMed Google Scholar
  122. Parambil JG, et al. Pazopanib effective for bevacizumab-unresponsive epistaxis in hereditary hemorrhagic telangiectasia. Laryngoscope. 2018;128(10):2234–2236.
    View this article via: CrossRef PubMed Google Scholar
  123. Faughnan ME, et al. Pazopanib may reduce bleeding in hereditary hemorrhagic telangiectasia. Angiogenesis. 2019;22(1):145–155.
    View this article via: CrossRef PubMed Google Scholar
  124. Parambil JG, et al. Pazopanib for severe bleeding and transfusion-dependent anemia in hereditary hemorrhagic telangiectasia. Angiogenesis. 2022;25(1):87–97.
    View this article via: CrossRef PubMed Google Scholar
  125. Moon JY, et al. Improvement of cutaneous hereditary hemorrhagic telangiectasia with pazopanib-A multikinase inhibitor. JAMA Dermatol. 2022;158(2):214–216.
    View this article via: CrossRef PubMed Google Scholar
  126. Kovacs-Sipos E, et al. Nintedanib as a novel treatment option in hereditary haemorrhagic telangiectasia. BMJ Case Rep. 2017;2017:bcr2017219393.
    View this article via: CrossRef PubMed Google Scholar
  127. Geisthoff UW, et al. Improvement in hereditary hemorrhagic telangiectasia after treatment with the phosphoinositide 3-kinase inhibitor BKM120. Ann Hematol. 2014;93(4):703–704.
    View this article via: CrossRef PubMed Google Scholar
  128. Skaro AI, et al. Regression of cutaneous and gastrointestinal telangiectasia with sirolimus and aspirin in a patient with hereditary hemorrhagic telangiectasia. Ann Intern Med. 2006;144(3):226–227.
    View this article via: CrossRef PubMed Google Scholar
  129. Pruijsen JM, et al. Tacrolimus in gastrointestinal bleeding in a young boy with hereditary hemorrhagic telangiectasia. JPGN Rep. 2021;2(4):e133.
    View this article via: CrossRef PubMed Google Scholar
  130. Hessels J, et al. Efficacy and safety of tacrolimus as treatment for bleeding caused by hereditary hemorrhagic telangiectasia: an open-label, pilot study. J Clin Med. 2022;11(18):5280.
    View this article via: CrossRef PubMed Google Scholar
  131. Invernizzi R, et al. Efficacy and safety of thalidomide for the treatment of severe recurrent epistaxis in hereditary haemorrhagic telangiectasia: results of a non-randomised, single-centre, phase 2 study. Lancet Haematol. 2015;2(11):e465–e473.
    View this article via: CrossRef PubMed Google Scholar
  132. Fang J, et al. Thalidomide for epistaxis in patients with hereditary hemorrhagic telangiectasia: a preliminary study. Otolaryngol Head Neck Surg. 2017;157(2):217–221.
    View this article via: CrossRef PubMed Google Scholar
  133. Baysal M, et al. Thalidomide for the management of bleeding episodes in patients with hereditary hemorrhagic telangiectasia: effects on epistaxis severity score and quality of life. Turk J Haematol. 2019;36(1):43–47.
    View this article via: CrossRef PubMed Google Scholar
  134. Samour M, et al. Pomalidomide in hereditary hemorrhagic telangiectasia: interim results of a phase I study. Blood. 2016;128(22):210.
    View this article via: CrossRef Google Scholar
  135. Sommer N, et al. Treatment with low-dose tacrolimus inhibits bleeding complications in a patient with hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension. Pulm Circ. 2019;9(2):2045894018805406.
    View this article via: CrossRef PubMed Google Scholar
  136. Shovlin CL, et al. MEK 1 inhibition and bleeding in hereditary haemorrhagic telangiectasia. [published online October 23, 2023]. Br J Haematol. https://doi.org/10.1111/bjh.19167.
    View this article via: PubMed Google Scholar
  137. Shovlin CL. Pulmonary arteriovenous malformations. Am J Respir Crit Care Med. 2014;190(11):1217–1228.
    View this article via: CrossRef PubMed Google Scholar
  138. Azzopardi N, et al. Dose - response relationship of bevacizumab in hereditary hemorrhagic telangiectasia. MAbs. 2015;7(3):630–637.
    View this article via: CrossRef PubMed Google Scholar
  139. Dupuis-Girod S, et al. ELLIPSE Study: a Phase 1 study evaluating the tolerance of bevacizumab nasal spray in the treatment of epistaxis in hereditary hemorrhagic telangiectasia. MAbs. 2014;6(3):794–799.
    View this article via: CrossRef PubMed Google Scholar
  140. Dupuis-Girod S, et al. Effect of bevacizumab nasal spray on epistaxis duration in hereditary hemorrhagic telangectasia: A Randomized Clinical Trial. JAMA. 2016;316(9):934–942.
    View this article via: CrossRef PubMed Google Scholar
  141. Riss D, et al. Intranasal submucosal bevacizumab for epistaxis in hereditary hemorrhagic telangiectasia: a double-blind, randomized, placebo-controlled trial. Head Neck. 2015;37(6):783–787.
    View this article via: CrossRef PubMed Google Scholar
  142. Stokes P, Rimmer J. Intranasal bevacizumab in the treatment of HHT -related epistaxis: a systematic review. Rhinology. 2018;56(1):3–10.
    View this article via: CrossRef PubMed Google Scholar
  143. Dheyauldeen S, et al. Bevacizumab in hereditary hemorrhagic telangiectasia-associated epistaxis: effectiveness of an injection protocol based on the vascular anatomy of the nose. Laryngoscope. 2012;122(6):1210–1214.
    View this article via: CrossRef PubMed Google Scholar
  144. Steineger J, et al. Long-term experience with intranasal bevacizumab therapy. Laryngoscope. 2018;128(10):2237–2244.
    View this article via: CrossRef PubMed Google Scholar
  145. Khanwalkar AR, et al. Randomized, controlled, double-blinded clinical trial of effect of bevacizumab injection in management of epistaxis in hereditary hemorrhagic telangiectasia patients undergoing surgical cauterization. Int Forum Allergy Rhinol. 2022;12(8):1034–1042.
    View this article via: CrossRef PubMed Google Scholar
  146. Khoueir N, et al. Injection of bevacizumab and cyanoacrylate glue for hereditary hemorrhagic telangiectasia. Laryngoscope. 2019;129(10):2210–2215.
    View this article via: CrossRef PubMed Google Scholar
  147. Storch CH, Hoeger PH. Propranolol for infantile haemangiomas: insights into the molecular mechanisms of action. Br J Dermatol. 2010;163(2):269–274.
    View this article via: CrossRef PubMed Google Scholar
  148. Jeon H, Cohen B. Lack of efficacy of topical timolol for cutaneous telangiectasias in patients with hereditary hemorrhagic telangiectasia: Results of a pilot study. J Am Acad Dermatol. 2017;76(5):997–999.
    View this article via: CrossRef PubMed Google Scholar
  149. Peterson AM, et al. Efficacy of timolol in a novel intranasal thermosensitive gel for hereditary hemorrhagic telangiectasia-associated epistaxis: A Randomized Clinical Trial. JAMA Otolaryngol Head Neck Surg. 2020;146(11):1006–1014.
    View this article via: CrossRef PubMed Google Scholar
  150. Dupuis-Girod S, et al. Efficacy of TIMOLOL nasal spray as a treatment for epistaxis in hereditary hemorrhagic telangiectasia. A double-blind, randomized, placebo-controlled trial. Sci Rep. 2019;9(1):11986.
    View this article via: CrossRef PubMed Google Scholar
  151. Andorfer KEC, et al. TIMolol nasal spray as a treatment for epistaxis in hereditary hemorrhagic telangiectasia (TIM-HHT)-A prospective, randomized, double-blind, controlled, cross-over trial. Pharmaceutics. 2022;14(11):2335.
    View this article via: CrossRef PubMed Google Scholar
  152. Albarki H, Rimmer J. The use of beta-blockers in hereditary hemorrhagic telangiectasia-related epistaxis: a systematic review. Am J Rhinol Allergy. 2022;36(6):890–896.
    View this article via: CrossRef PubMed Google Scholar
  153. Mei-Zahav M, et al. Topical propranolol improves epistaxis in patients with hereditary hemorrhagic telangiectasia — a preliminary report. J Otolaryngology Head Neck Surg. 2017;46(1):58.
    View this article via: CrossRef Google Scholar
  154. Mei-Zahav M, et al. Topical propranolol improves epistaxis control in hereditary hemorrhagic telangiectasia (HHT): a randomized double-blind placebo-controlled trial. J Clin Med. 2020;9(10):3130.
    View this article via: CrossRef PubMed Google Scholar
  155. Totzeck M, et al. Cardiovascular adverse events in patients with cancer treated with bevacizumab: a meta-analysis of more than 20,000 patients. J Am Heart Assoc. 2017;6(8):e006278.
    View this article via: CrossRef PubMed Google Scholar
Version history
  • Version 1 (February 15, 2024): Electronic publication

Article tools

  • View PDF
  • Download citation information
  • Send a comment
  • Terms of use
  • Standard abbreviations
  • Need help? Email the journal

Review Series

Vascular Malformations

  • Infantile hemangioma: the common and enigmatic vascular tumor
    Annegret Holm et al.
  • Central conducting lymphatic anomaly: from bench to bedside
    Luciana Daniela Garlisi Torales et al.
  • Pathophysiology of cerebral small vessel disease: a journey through recent discoveries
    Nicolas Dupré et al.
  • Capillary malformations
    Adrienne M. Hammill et al.
  • Shear stress and pathophysiological PI3K involvement in vascular malformations
    Salim Abdelilah-Seyfried et al.
  • Lymphatic malformations: mechanistic insights and evolving therapeutic frontiers
    Milena Petkova et al.
  • Hereditary hemorrhagic telangiectasia: from signaling insights to therapeutic advances
    Tala Al Tabosh et al.

Metrics

Article has an altmetric score of 5
  • Article usage
  • Citations to this article (14)

Go to

  • Top
  • Abstract
  • Genetic basis of HHT
  • Clinical manifestations of HHT
  • The second-hit hypothesis
  • Signaling pathways involved in HHT
  • Preclinical data, clinical trials, and perspectives in HHT
  • Future challenges in HHT
  • Summary and conclusions
  • Acknowledgments
  • Footnotes
  • References
  • Version history
Advertisement
Advertisement

Copyright © 2025 American Society for Clinical Investigation
ISSN: 0021-9738 (print), 1558-8238 (online)

Sign up for email alerts

Posted by 7 X users
On 1 Facebook pages
44 readers on Mendeley
See more details