Arteriovenous Malformations in Osler-Weber-Rendu Syndrome: Pathophysiology, Clinical Manifestations, and Advances in Management
Janeddie Rivera Lopez1, Kennett Collazo Aparicio1, Arashdeep Singh2, Linda Boahemaah-Kontoh3, Lilian R Diaz Rosero4, Rishita Dave1, Molly Sanjay Jain5, Kevin Isaac Villatoro Guzmán6, Jhoset Molinares Hernandez7, Reem Faiz Hussein Eisa8 and Maria Isabel Gomez-Coral9*
1University of Medicine and Health Sciences, St. Kitts
2Government Medical College, Amritsar, India
3Vinnitsa National Medical University, Ukraine
4Universidad Nacional de Colombia, Colombia
5Parkview Health, Indiana, USA
6Universidad de El Salvador, Facultad Multidisciplinaria Oriental, El Salvador
7Universidad Americana, Nicaragua
8Faculty of Medicine, Ain Shams University, Egypt
9Universidad del Valle, México
Submission: August 16, 2024; Published: August 23, 2024
*Corresponding author: Maria Isabel Gomez-Coral, Universidad del Valle, México, México
How to cite this article: Janeddie Rivera L, Kennett Collazo A, Arashdeep S, Linda Boahemaah-K, Lilian R Diaz R, et al. Arteriovenous Malformations in Osler-Weber-Rendu Syndrome: Pathophysiology, Clinical Manifestations, and Advances in Management. J Cardiol & Cardiovasc Ther. 2024; 19(4): 556017 DOI: 10.19080/JOCCT.2024.19.556017
Abstract
Osler-Weber-Rendu Syndrome, or Hereditary Hemorrhagic Telangiectasia (HHT), is a rare genetic disorder marked by the development of arteriovenous malformations (AVMs) throughout the body. These abnormal vascular connections bypass the capillary network, leading to significant clinical challenges. This article reviews the pathophysiology, clinical manifestations, and advances in the management of AVMs in HHT. The genetic basis of HHT involves mutations in the ENG, ACVRL1, and SMAD4 genes, which play critical roles in the TGF-β signaling pathway. These mutations result in endothelial dysfunction, contributing to the formation of AVMs. The clinical impact of AVMs is significant, particularly when they occur in the brain, lungs, liver, or gastrointestinal tract, where they can lead to life-threatening complications. This review also explores current diagnostic and management strategies, emphasizing the importance of a multidisciplinary approach. Interventional techniques such as embolization, stereotactic radiosurgery, and microsurgical resection are discussed, alongside emerging therapies like anti-angiogenic drugs and gene therapy. These advancements have led to more personalized and effective treatment plans for patients. As a result, the management of AVMs in HHT has seen significant progress, with a growing emphasis on individualized care. Continued research and the development of specialized treatment centers are crucial for further improving patient outcomes in this complex condition.
Keywords: Arteriovenous malformations (AVMs); Osler-Weber-Rendu Syndrome (OWR); Hereditary hemorrhagic telangiectasia (HHT); Vascular endothelial growth factor; Interventional techniques
Abbreviations: AVMs: Arteriovenous Malformations; OWR: Osler-Weber-Rendu Syndrome; HHT: Hereditary Hemorrhagic Telangiectasia; DSA: Digital Subtraction Angiography; MRA: Magnetic Resonance Angiography; SRS: Stereotactic Radiosurgery; VEGF: Vascular Endothelial Growth Factor; PDGF: Platelet-Derived Growth Factor; TKIs: Tyrosine Kinase Inhibitors; ENG: Endoglin; ACVRL1: Activin A Receptor Type II-Like 1; SMAD4: Mothers Against Decapentaplegic Homolog 4; MRI: Magnetic Resonance Imaging; 3D: Three-Dimensional; GWAS: Genome-Wide Association Studies; NGS: Next-Generation Sequencing; TGF-β: Transforming Growth Factor Beta; PAVMs: Pulmonary Arteriovenous Malformations; CRISPR-Cas9: Clustered Regularly Interspaced Short Palindromic Repeats-Associated Protein 9
Introduction
Osler-Weber-Rendu Syndrome, also known as Hereditary Hemorrhagic Telangiectasia (HHT), is a rare genetic disorder characterized by the formation of abnormal blood vessels throughout the body. This autosomal dominant condition arises from mutations in genes critical for vascular formation and repair, notably ENG (Endoglin), ACVRL1 (Activin A receptor-like kinase 1), and SMAD4. These genetic mutations drive the pathology of HHT, particularly through the development of arteriovenous malformations (AVMs), which are central to the disease’s clinical challenges [1]. The primary clinical features of HHT include telangiectasias-small, dilated blood vessels visible on the skin and mucous membranes-and recurrent nosebleeds. While these symptoms are troubling, the most severe complications arise from the formation of AVMs. These abnormal vascular connections between arteries and veins bypass the capillary network, leading to significant health issues depending on their location [2].
AVMs can develop in critical organs such as the brain, lungs, liver, and gastrointestinal tract. Although HHT affects approximately 1 in 5,000 individuals globally, its effects on patients can be profound, often leading to serious complications and impacting quality of life [3]. Understanding the pathophysiology of AVMs, their clinical manifestations, and recent advancements in their management is essential for enhancing patient care and outcomes. This paper aims to delve into the complex pathophysiology of arteriovenous malformations in the context of Osler-Weber-Rendu Syndrome. We will explore the genetic mechanisms underlying these malformations, their clinical implications, and recent progress in their management. By examining these aspects, we seek to provide a comprehensive overview of how HHT-related AVMs are identified, treated, and managed, ultimately contributing to better clinical care and patient support.
Pathophysiology of AVMs in HHT
Genetic Mutations:
The majority of the cases (>90%) of Hereditary hemorrhagic telangiectasia (HHT) have been linked to heterozygous mutations in endothelial cell surface receptors, notably ENG (endoglin, responsible for HHT type 1), ACVRL1 (activin receptor like kinase 1 gene, notable for HHT type 2), whereas mutations in MADH4 (encoding SMAD4) causes a combined syndrome of juvenile polyposis and hereditary hemorrhagic telangiectasia in less than 5% of the cases [4-7]. Several mutations have been described including missense, nonsense, frameshifts, splicing errors, and gross deletions. HHT is inherited in an autosomal dominant fashion.
ENG (located on chromosome 9q), ACVRL1 (located on chromosome 12q), and MADH4 are involved in the Transforming growth factor-beta (TGF-β) signaling pathway, which is a superfamily of secreted factors comprising over 30 members and divided into two groups. The first includes activins, inhibins, and nodal growth Differentiation factors, while the second contains Bone Morphogenetic proteins (BMPs), most of the Growth Differentiation Factors, anti-Mullerian hormone, etc [8]. These proteins are expressed primarily on the surface of endothelial cells. They are also present in activated monocytes, syncytiotrophoblasts, and some leukemic cells [9]. In the pathogenesis of HHT, BMP 9 and 10 are the main ligands of the TGF-β pathway, which exert their actions through two distinct type 1 receptors, ALK1 and ALK5, which are known to mediate TGF-β signaling through a cascade of intracellular effectors which belong to the SMAD protein family. While ALK1 signals by SMAD1/SMAD5, ALK2 prefers SMAD2/SMAD3 [10-12].
There are two phases of angiogenesis. In the activation phase, endothelial cells degrade the perivascular basement membrane, invade and migrate into extracellular space, proliferate and form capillary lumen. In contrast, during the resolution phase, they stop these activities and rebuild the basement membrane. While activation of the ALK1 pathway favors the resolution phase, the ALK5 pathway prolongs the activation phase, leading to continuous proliferation. In HHT type 1, mutated endoglin disrupts both the ALK1 and ALK5 pathways, thereby disrupting the intricate balance between them. On the other hand, only the ALK1 pathway is disrupted in HHT type 2, leading to over proliferation of endothelial cells during the activation phase and development of aberrant communication between dilated arterioles and venules, resulting in AVMs [9].
Finally, some theories have been put forward to explain the pathogenesis of disease development in HHT. The most accepted theory is the haploinsufficiency model. It states that mutated ENG and ACVRL1 genes create altered proteins that are not expressed adequately on the cell membranes of endothelial cells [13]. Other less approved theories include dominant negative mutation and double-hit hypothesis. In dominant negative theory, McAllister et al. proposed that the mutated allele prevents the wild type allele from exerting its normal effects [5]. In two-hit hypothesis, Snellings et al. suggested that vascular malformations associated with HHT, specifically cutaneous telangiectasia, follow a genetic two-hit mechanism [14].
Vascular Abnormalities
AVMs are direct connections between an artery and a vein without an intervening capillary bed. The earliest aberration in the development of a telangiectasia is dilatation of a post-capillary venule. During the disease development, these venules further dilate and branch extensively to become convoluted, usually extending through the entire dermis. The walls of these post capillary venules have excessive layers of smooth muscle cells while lacking sufficient elastic fibers. Finally, these dilated post capillary venules connect directly to dilated arterioles during the fully developed stage, forming a small arteriovenous shunt surrounded by a perivascular mononuclear cell infiltrate [15,16].
Larger AVMs, which are usually found in lungs, brain, or the gastrointestinal tract, are a result of progressive vascular remodeling [15]. Since there is no capillary network to blunt the arterial pressure, veins must undergo certain adaptations to minimize wall stress, like increasing their thickness [17]. In HHT, however, this phenomenon is blunted, leading to the development of fragile vessels that are prone to rupture and can cause significant bleeding complications [18].
Clinical Manifestations of AVMs
Pulmonary Arteriovenous Malformations (PAVMs)
Most patients with Pulmonary Arterio-Venous Malformations (PAVMS)are asymptomatic but they may show symptoms depending on the complexity of the PAVMS. There are simple or complex PAVMS with large, complex or multiple PAVMs, shortness of breath may be present, especially with exertion [19]. There is dyspnea due to hypoxemia resulting from the arteriovenous shunting or limitations in the air and blood flow direction [19]. There is also platypnea orthodeoxia syndrome, this is when dyspnea and hypoxemia are triggered by standing and relieved with recumbency. This occurs due to the coexistence of diverse structural and physiological abnormalities in addition to the gravitational forces that induce blood shunting after standing. This flow of deoxygenated blood through the PAVM shunts occur mostly at the lung base. The increased blood flow through the basilar regions in standing position increases the shunts and generates this symptom [20,21]. Hemoptysis is another symptom of PAVM, the AVM may bleed due to rupture of endobrachial or intra-parenchyma PAVMS, causing coughing of blood or collections of blood around the lung [22,23]. Other symptoms are exercise intolerance, chest pain, cough, clubbing and cyanosis [22]. A well-known complication is paradoxical embolism, this occurs when a thrombus in the deep venous circulation embolizes through a PAVM into the systemic circulation. This is due to the permanent right to left shunt caused by the pathological connection between the pulmonary arteries to the pulmonary veins returning to the left atrium [24]. The above complication has been linked to cryptogenic strokes as clots will travel to big vessels feeding the brain [25,26]. A brain abscess can form due to walled off emboli caused from the paradoxical embolism [27].
Cerebral Arteriovenous Malformations (CAVMs)
Cerebral arteriovenous malformations (CAVMS) occur in 10-20% of patients. The risk of rupture (which causes hemorrhage and potential neurologic symptoms) is estimated to be less than 2.5% per year [28]. In CAVMS the main cause of symptoms are due to bleeding, this is seen more in young people ages 15-20 [29]. These hemorrhages are mostly spontaneous, usually with a parenchymal location but can also be seen in the subarachnoid region. Some symptoms seen with hemorrhages are loss of consciousness, headache (severe and sudden), nausea and vomiting. Due to the complication CAVM affecting the galen vein there is an increase in pressure of the brain, this then causes seizures and even failure to thrive in children [30]. The seizures gotten from CAVM complications are typically focal, and the location of the AVM determines the seizure type. These focal seizures often become tonic clonic and or even generalized. Occasionally, a cranial bruit can be detected via auscultation [31]. Another complication that can occur are memory deficits, this occurs due to decreased supply of oxygen and nutrients to parts of the brain. Due to the above deficit hemiparesis, loss of sensation of one side of body, vertigo, slurred speech, balanced disorders and other symptoms of stroke can occur [31-33].
Hepatic Arteriovenous Malformations (HAVMs)
There is a high possibility of Hepatic Arteriovenous malformations(HAVMS) forming in patients with Osler Weber Rendu syndrome. With a percentage being at approximately 70%. The symptoms that develop are usually due to the complexity (size, type) and mostly the location of the HAVM while many hepatic AVMs are asymptomatic, large AVMs can have varying symptoms from telengiectasis, to high-output cardiac failure due to vascular shunting [27]. HAVMs hunts complications are high output cardiac failure (HOCF), ischemic cholangitis, portal hypertension, gastrointestinal bleed and mesenteric ischemia [33]. The HAVMS cause chronic liver disease, and patients can present with symptoms of hepatic encephalopathy (ranging from grade 0 -minimal change in concentration to grade 4-coma) [34,35]. In patients with portal hypertension, showing symptoms such as melena, hematemesis, ascites and edema, with negative initial liver disease, HAVMS are a likely cause [35]. Melena and hematemesis are the symptoms that occur once the gastrointestinal tract is involved. HOCF, is seen mostly found in women. Large HAVMS shunts reduce systemic vascular resistance and increase cardiac output, these lead to a wide pulse pressure, systolic cardiac murmur, and liver bruit [36]. The Increased cardiac output cause increased ventricular volume, which causes a dilation and this affects the cardiac valves and further leading to cardiac failure. Patients with HOCF have what we call a “preserved” left ventricular ejection fraction despite chronic LV volume overload, because the low systemic vascular resistance associated with their liver AVMs reduces the LV afterload [36]. Symptoms of HOCF include fatigue, weakness, edema, orthopnea and paroxysmal nocturnal dyspnea. Some conditions like pregnancy cause an increase in shunting, this exacerbates existing symptoms.
Diagnosis of AVMs in HHT
Clinical Criteria
The diagnosis of Hereditary Hemorrhagic Telangiectasia (HHT) is based on the Curaçao Criteria, which include four key features: spontaneous, recurrent epistaxis, a positive family history, cutaneous or mucosal telangiectasias, and visceral lesions. A definitive diagnosis is confirmed if a patient meets three of these criteria, while a possible or suspected diagnosis is considered when two criteria are present. It is important to note that these criteria have a poor negative predictive value in children under the age of 16 [37,38].
Imaging Modalities
Pulmonary AVMs are often silent but can lead to strokes, massive hemoptysis, spontaneous hemothorax, transient ischemic attacks, and brain abscesses [39-42]. Sensitive screening tests to detect pulmonary AVMs include thoracic CT scan and transthoracic contrast/bubble echocardiography; both modalities can also detect pulmonary hypertension. Most screening protocols use contrast echocardiography as a first-line test, followed by a thoracic CT to determine the anatomic location and if embolization is a viable option. Chest x-rays, right-to-left shunt measurements, and blood oxygen measurements are less sensitive to identifying the presence of pulmonary AVMs. Multiple cerebral AVMs are predictive of HHT [43]. The role of screening remains controversial due to the overall low risk, albeit significant morbidity or mortality, of hemorrhage. Additionally, the risks associated with treating asymptomatic cerebral AVMs potentially outweigh the benefits. An MRI brain with and without contrast is initially recommended for patients with cerebral symptoms, who have a known unstable cerebral aneurysm, or a family member who has had a cerebral hemorrhage since familial aneurysms have a higher risk of hemorrhage [44]. The gold standard for diagnosing and treating cerebral AVMs is diagnostic angiography, which carries a 0.5% risk of stroke [45].
Screening for asymptomatic hepatic AVMs with doppler ultrasonography is recommended because it is non-invasive and can improve patient management and outcomes. While doppler ultrasonography is ideal due to its accuracy, cost, safety, and tolerability depending on resources available and operator expertise, patients can be screened by alternate means, such as multiphase contrast CT or MRI [46].
Genetic Testing
Genetic mutation testing should be done to confirm a diagnosis of HHT, including patients who meet 1-2 of the Curaçao criteria or young children with affected parents who are yet to develop the clinical manifestations [41]. Initial genetic testing should screen for the three most prevalent mutations, ENG, ACVRL1, and SMAD4. Testing should also be extended to family members. It is important to note that genetic mutations are not identified in up to 10 to 15% of HHT families, and a negative genetic test does not exclude the diagnosis of HHT. Once the diagnosis is confirmed, additional tests can be done to evaluate for other HHT manifestations. Screening should be done, regardless of a patient’s clinical symptoms, due to the danger of undiagnosed silent AVMs [40-45].
Management and Treatment of AVMs
Interventional Techniques
Interventional techniques have become a cornerstone in the management of arteriovenous malformations (AVMs) in patients with Osler-Weber-Rendu Syndrome (OWR). The primary goal of these techniques is to reduce the risk of hemorrhage and other complications associated with AVMs by selectively targeting abnormal vascular structures. One of the most widely used interventional procedures is embolization, which involves the injection of embolic agents, such as coils, liquid embolic materials, or particles, into the abnormal vessels to occlude them and prevent further blood flow. Embolization can be performed as a standalone treatment or as an adjunct to surgical resection, helping to minimize intraoperative blood loss and improve the safety of surgery [47,48].
Endovascular approaches, such as transcatheter embolization, have gained prominence due to their minimally invasive nature and ability to reach deep-seated AVMs that are challenging to access surgically. These procedures are often guided by imaging modalities like digital subtraction angiography (DSA) or magnetic resonance angiography (MRA), which provide detailed visualization of the AVMs and enable precise targeting of the embolic agents. However, the success of embolization is influenced by the complexity and size of the AVM, as well as the presence of high-flow shunts, which can complicate the procedure and increase the risk of complications [48].
Another interventional technique that has shown promise in the management of AVMs is stereotactic radiosurgery (SRS). SRS delivers focused, high-dose radiation to the AVM, leading to gradual obliteration of the abnormal vessels over time. This technique is particularly useful for small to medium-sized AVMs that are located in eloquent areas of the brain or are otherwise inaccessible for conventional surgery [49,50]. Despite its effectiveness, SRS carries a risk of radiation-induced side effects, and its therapeutic effect may take months or even years to manifest, necessitating close long-term follow-up [49].
Surgical Approaches
Surgical resection remains a definitive treatment option for AVMs in patients with OWR, particularly for lesions that are symptomatic, at high risk of rupture, or located in areas where they pose a significant threat to vital structures. The choice of surgical approach depends on the size, location, and complexity of the AVM, as well as the overall health of the patient. In general, smaller AVMs with well-defined borders are more amenable to surgical removal, while larger, more diffuse AVMs may require a combination of surgical and interventional strategies [51].
Micrurgical resection is the most common surgical technique used to treat AVMs, especially those located in the brain. This approach involves the use of an operating microscope and microsurgical instruments to carefully excise the AVM while preserving surrounding healthy tissue. The success of microsurgical resection depends on the surgeon's ability to achieve complete removal of the AVM without causing significant neurological deficits. In some cases, preoperative embolization may be performed to reduce the size of the AVM and minimize intraoperative bleeding [52].
For AVMs located in the lungs, a condition frequently associated with OWR, pulmonary resection may be necessary. This involves the surgical removal of the affected lung tissue, either through lobectomy (removal of a lung lobe) or segmentectomy (removal of a lung segment), depending on the extent of the AVM. Pulmonary resection is typically reserved for patients with severe symptoms or those who have not responded to less invasive treatments, such as embolization [53]. The decision to proceed with surgery must be carefully weighed against the potential risks, including respiratory complications and the impact on lung function. The management and treatment of AVMs in OWR patients involve a multidisciplinary approach that includes both interventional techniques and surgical interventions. The choice of treatment depends on various factors, including the size, location, and complexity of the AVM, as well as the patient's overall condition. Advances in imaging and interventional techniques have improved the safety and efficacy of these treatments, offering new hope for patients with this challenging condition.
Pharmacological therapies
Angiogenesis, the formation of new blood vessels from pre-existing ones, is essential for both normal physiological functions and malignant states, such as cancer. The growth and spread of tumors rely significantly on angiogenesis, a process that provides the tumor with essential nutrients and oxygen. Antiangiogenic drugs are a specific group of medications developed to hinder the process of angiogenesis, which in turn limits the growth of tumors and enhances the overall results of medical treatment. Although not typically the initial treatment for AVMs, antiangiogenic therapy is being investigated as a possible therapeutic approach [59]. Antiangiogenic medications specifically focus on different aspects of the angiogenic process, such as inhibiting VEGF, inhibiting VEGFR, and targeting other pathways involved in angiogenesis [59,60]. Some examples of these drugs are bevacizumab, sunitinib, and sorafenib. TKIs such as sunitinib and sorafenib inhibit signaling pathways that stimulate the growth of endothelial cells [59]. Certain medications have a direct impact on endothelial cells, either by inhibiting their growth or inducing programmed cell death. Notable examples include thalidomide and its derivatives [59,60].
Bevacizumab is a monoclonal antibody that inhibits vascular endothelial growth factor-A (VEGF-A), thereby minimizing angiogenesis and abnormal vascular growth. It has been demonstrated in clinical trials that it can reduce the extent of AVMs and improve symptoms. Some studies have demonstrated an improvement in symptoms and a reduction in bleeding episodes in patients with AVMs [60-62]. Nevertheless, the efficacy of bevacizumab in AVMs can fluctuate, and it can result in hypertension, bleeding, and gastrointestinal perforations, which necessitate meticulous management [62].
Sunitinib is a tyrosine kinase inhibitor that targets multiple tyrosine kinases and inhibits angiogenesis and tumor growth by blocking VEGF, PDGF, and c-KIT receptors [63,64]. It has been investigated in the context of AVMs, primarily in research settings, with the objective of reducing symptoms and size. However, more studies are required to verify its efficacy. Limited data on its role in AVMs, as well as side effects such as hypertension, fatigue, and hepatic function impacts, are among the challenges [63,64]. Aflibercept is a fusion protein that functions as a decoy receptor for VEGF-A, VEGF-B, and PlGF, thereby preventing their activation on endothelial cells. It has been investigated for its potential to reduce abnormal blood vessel formation and complications, particularly in the context of AVMs. On the other hand, its efficacy is restricted by its experimental nature and potential adverse effects, such as hypertension, similar to other anti-VEGF therapies [65].
Bevacizumab, sunitinib, thalidomide, and aflibercept have all been investigated for their capacity to regulate the angiogenic processes associated with AVMs. Although there is evidence that these pharmaceuticals have the potential to reduce the size and symptoms of AVMs, their efficacy and safety profiles require further investigation through rigorous clinical trials. The necessity of a personalized treatment approach is emphasized by the variability in patient responses and potential adverse effects. Further research is necessary to gain a comprehensive understanding of the function of these agents in the management of AVMs and to refine therapeutic strategies to achieve the best possible patient outcomes.
Advances in Management
Multidisciplinary Approach
The management of arteriovenous malformations (AVMs) in patients with Osler-Weber-Rendu Syndrome (OWR) has seen significant advancements through the adoption of a multidisciplinary approach. This approach integrates the expertise of various specialists, including interventional radiologists, neurosurgeons, pulmonologists, geneticists, and otolaryngologists, to address the diverse manifestations of AVMs and their associated complications. The complexity of OWR, which can involve multiple organ systems, necessitates coordinated care that ensures comprehensive assessment, individualized treatment plans, and ongoing monitoring of the patient’s condition [48]. For instance, the decision to pursue endovascular interventions, surgical resection, or conservative management often requires input from a team of specialists to weigh the risks and benefits of each option. This collaborative effort has improved outcomes by allowing for tailored treatment strategies that take into account the unique clinical presentation and progression of AVMs in each patient.
Moreover, multidisciplinary clinics dedicated to the management of hereditary hemorrhagic telangiectasia (HHT), the underlying genetic disorder in OWR, have emerged as centers of excellence. These clinics offer patients access to a range of diagnostic and therapeutic options, along with specialized expertise in managing the rare and complex aspects of the disease. The integration of genetic counseling within these multidisciplinary teams has also become a critical component, helping patients and their families understand the hereditary nature of the syndrome and guiding decisions related to screening and preventive measures for at-risk relatives [44].
Emerging Therapies
In addition to established interventional and surgical techniques, emerging therapies are playing an increasingly important role in the management of AVMs in OWR. Advances in molecular biology and genetics have led to the identification of key pathways involved in the pathogenesis of AVMs, opening the door to targeted therapies. For example, anti-angiogenic agents, such as bevacizumab, have shown promise in reducing the severity of bleeding in patients with HHT by inhibiting the vascular endothelial growth factor (VEGF) pathway [54]. Bevacizumab, initially developed for cancer treatment, has been repurposed for use in OWR, particularly in cases where conventional treatments have failed or are contraindicated. Its ability to reduce epistaxis and gastrointestinal bleeding, common and debilitating symptoms of OWR, represents a significant advancement in the symptomatic management of the disease.
Gene therapy is another emerging area of interest, with the potential to correct the underlying genetic mutations that lead to AVM formation in OWR. Although still in the experimental stage, preclinical studies have demonstrated the feasibility of using viral vectors to deliver normal copies of the mutated genes responsible for HHT. If successful, gene therapy could offer a curative approach by addressing the root cause of the disease, rather than just managing its symptoms [55]. Additionally, advancements in imaging technologies, such as functional MRI and 3D angiography, have improved the ability to precisely map AVMs, allowing for more accurate and less invasive treatment planning [56].
Patient-Centered Care
The shift towards patient-centered care in the management of AVMs in OWR has been another critical advancement. This approach emphasizes the importance of involving patients in their treatment decisions, respecting their preferences, and addressing their individual needs and concerns. In the context of a chronic and potentially life-threatening condition like OWR, patient-centered care involves not only managing the physical aspects of the disease but also addressing the psychological and social impacts. Comprehensive care plans now often include psychological support, social services, and patient education programs, which are essential for improving the overall quality of life for patients and their families [57].
Patient-centered care also involves the use of shared decision-making tools, which help patients understand the potential risks and benefits of different treatment options, enabling them to make informed choices in collaboration with their healthcare providers. This approach fosters a sense of empowerment and partnership between patients and their care teams, which is particularly important in managing a disease as complex and variable as OWR. The availability of patient registries and online communities has further enhanced the patient-centered approach, providing individuals with OWR access to resources, support networks, and up-to-date information about their condition [58].
Future Directions
Osler-Weber-Rendu syndrome, or Hereditary Hemorrhagic Telangiectasia (HHT), presents several critical knowledge gaps, particularly in understanding genetic diversity and the influence of modifier genes. Although mutations in ENG, ACVRL1, and SMAD4 are known to cause the condition, the varying severity of symptoms among patients indicates that other genetic factors may play a role in disease expression [66]. Future research should prioritize the identification of these modifier genes using advanced methods like genome-wide association studies (GWAS) and next-generation sequencing (NGS) [67]. Additionally, the mechanisms behind vascular malformations, especially arteriovenous malformations (AVMs), are not fully understood. While the dysregulation of the TGF-β signaling pathway is implicated, further investigation is needed to elucidate its effects on vascular endothelial cells, which could uncover new therapeutic targets [68].
The development of new treatments for HHT is another essential area for future research. Current therapies mainly address symptoms, with limited options targeting the underlying molecular defects of the disease. Bevacizumab, an anti-VEGF antibody, shows promise, but more research is necessary to evaluate its long-term effectiveness and safety [69]. Exploring small molecules, gene therapy, or RNA-based therapies that can normalize TGF-β signaling or inhibit abnormal angiogenesis could lead to significant advancements in HHT treatment.
Improvements in diagnostic techniques for HHT are also needed. Diagnosis currently relies on clinical criteria and genetic testing, which may not detect all causative mutations. Imaging methods to identify AVMs often involve high radiation exposure or invasive procedures. Future research should aim to develop non-invasive imaging techniques, such as high-resolution ultrasound, magnetic resonance angiography (MRA), and molecular imaging, to enhance early AVM detection [47]. Additionally, the development of blood-based biomarkers could assist in early diagnosis and monitoring disease progression. The management of pulmonary arteriovenous malformations (PAVMs) in HHT patients is another area needing attention. PAVMs carry significant risks, including stroke and brain abscess. While embolization is a standard treatment, there is a need for better preventive strategies and a deeper understanding of long-term outcomes. Research into pharmacological agents that can prevent PAVM formation or growth, along with longitudinal studies on stroke prevention strategies, could improve patient outcomes [47].
Lastly, the impact of HHT on quality of life and mental health remains underexplored. The chronic nature of the disease, with recurrent bleeding and the risk of severe complications, significantly affects patients' quality of life, yet there is limited research on the psychological and social aspects of living with HHT [70]. Future studies should examine the mental health burden, coping strategies, and social support systems for HHT patients, with the goal of incorporating psychosocial care into HHT management guidelines. Addressing these knowledge gaps and focusing on these areas for future research could lead to significant advancements in the diagnosis, treatment, and overall management of HHT.
Emerging technologies and treatments hold the potential to significantly transform the management landscape for arteriovenous malformations (AVMs) in hereditary hemorrhagic telangiectasia (HHT). Advances in genetic therapies, such as CRISPR-Cas9 gene editing, could offer targeted interventions to correct the underlying genetic mutations responsible for HHT, potentially preventing AVMs from forming [71,72]. Additionally, innovations in imaging technologies, including high-resolution 3D imaging and artificial intelligence-enhanced diagnostics, could enable earlier and more precise detection of AVMs, allowing for timely intervention and improved patient outcomes [73,74]. The development of novel pharmacological agents, such as angiogenesis inhibitors, may provide non-invasive alternatives to traditional surgical treatments, reducing the risks associated with current interventions [75,76]. Furthermore, personalized medicine approaches, driven by advancements in genomics and biomarker discovery, could lead to individualized treatment plans that are more effective and less invasive [77,78]. These emerging technologies and treatments collectively have the potential to shift the focus from reactive to proactive management of AVMs in HHT, improving both the quality of life and survival rates for affected individuals.
Conclusion
Osler-Weber-Rendu Syndrome (HHT) presents a complex and multifaceted challenge in the field of vascular medicine. The pathophysiology of AVMs in this genetic disorder underscores the delicate balance between angiogenic pathways, where mutations in key genes lead to significant clinical manifestations across multiple organ systems. From pulmonary and cerebral AVMs to hepatic complications, the spectrum of disease highlights the critical need for precise and multidisciplinary management strategies. The advancements in interventional techniques, surgical approaches, and pharmacological therapies, particularly the use of antiangiogenic drugs like bevacizumab, have significantly enhanced the ability to manage the symptoms and complications associated with AVMs. However, the importance of a tailored, patient-centered approach cannot be overstated. The emergence of specialized multidisciplinary clinics has proven vital in optimizing outcomes, and providing patients with comprehensive care that addresses both the immediate and long-term challenges of the disease.
Looking forward, the potential of emerging therapies, including gene therapy and targeted molecular interventions, offers hope for more effective treatments and even curative approaches. The ongoing integration of cutting-edge imaging technologies continues to refine the accuracy of diagnosis and treatment planning, reducing risks and improving patient quality of life. While significant progress has been made in understanding and managing AVMs in HHT, the pursuit of more effective and personalized treatment strategies remains a dynamic and essential focus. Continued collaboration across medical specialties, combined with advances in research and technology, will be key to improving outcomes for patients with this challenging syndrome.
References
- Gallagher JR, Wooderchak-Donahue W, McDonald JS (2015) Genetic Basis of Hereditary Hemorrhagic Telangiectasia: Implications for Diagnosis and Management. Curr Treat Opt Cardiovasc Med 17(8): 46.
- Plauchu H, Koussa S, Rameh C (2006) Clinical and Genetic Aspects of Hereditary Hemorrhagic Telangiectasia. Orphanet J Rare Dis 1(1): 40.
- Shovlin CL (2010) Hereditary Hemorrhagic Telangiectasia: Pathophysiology and Management. Hematology/Oncology Clin North Am 24(6): 1257-1278.
- Park H, Jessica F, Mathilde P, Minhwan C, Sanguk Y, et al. (2021) Defective Flow-Migration Coupling Causes Arteriovenous Malformations in Hereditary Hemorrhagic Telangiectasia. Circulation 144(10): 805-822.
- McAllister KA, Grogg KM, Johnson DW, Gallione CJ, Baldwin MA, et al. (1994) Endoglin, a TGF-β binding protein of endothelial cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat Genet 8(4): 345-351.
- Johnson DW, Berg JN, Baldwin MA, Gallione CJ, Marondel I, et al. (1996) Mutations in the activin receptor–like kinase 1 gene in hereditary haemorrhagic telangiectasia type 2. Nat Genet 13(2): 189-195.
- Gallione CJ, Gabriela MR, Eric L, Anil KR, Susan LS, et al. (2004) A combined syndrome of juvenile polyposis and hereditary haemorrhagic telangiectasia associated with mutations in MADH4 (SMAD4). The Lancet 363(9412): 852-859.
- Weiss A, Attisano L (2013) The TGFbeta Superfamily Signaling Pathway. WIREs DeV Biol 2(1): 47-63.
- Azuma H (2024) Genetic and molecular pathogenesis of hereditary hemorrhagic telangiectasia. J Med Invest 47(3-4): 81-90.
- Derynck R, Zhang YE (2024) mad-dependent and Smad-independent pathways in TGF-β family signalling. Nature 425(6958): 577-84.
- Fernández-L A, Sanz-Rodriguez F, Blanco FJ, Bernabéu C, Botella LM (2006) Hereditary Hemorrhagic Telangiectasia, a Vascular Dysplasia Affecting the TGF-β Signaling Pathway. Clin Med Res 4(1): 66-78.
- Viteri-Noël A, Andrés González-G, José LP, Martin F, Nuria BL, et al. (2022) Hereditary Hemorrhagic Telangiectasia: Genetics, Pathophysiology, Diagnosis, and Management. J Clin Med 11(17): 5245.
- Shovlin CL, Hughes JMB, Scott J, Seidman CE, Seidman JG (1997) Characterization of Endoglin and Identification of Novel Mutations in Hereditary Hemorrhagic Telangiectasia. Am J Hum Genet 61(1): 68-79.
- Snellings DA, Gallione CJ, Clark DS, Vozoris NT, Faughnan ME, et al. (2019) Somatic Mutations in Vascular Malformations of Hereditary Hemorrhagic Telangiectasia Result in Bi-allelic Loss of ENG or ACVRL1. Am J Hum Genet 105(5): 894-906.
- Braverman IM, Keh A, Jacobson BS (1990) Ultrastructure and three-dimensional organization of the telangiectases of hereditary hemorrhagic telangiectasia. J Invest Dermatol 95(4): 422-427.
- Guttmacher AE, Marchuk DA, White RI (1995) Hereditary Hemorrhagic Telangiectasia. N Engl J Med 333(14): 918-924.
- Owens CD (2010) Adaptive changes in autogenous vein grafts for arterial reconstruction: Clinical Implications. J Vasc Surg 51(3): 736-746.
- Shovlin CL (2010) Hereditary haemorrhagic telangiectasia: Pathophysiology, diagnosis and treatment. Blood Rev 24(6): 203-219.
- National Center for Biotechnology Information (2024) Pulmonary Arteriovenous Malformation - StatPearls - NCBI Bookshelf.
- Pascual J (2019) Mechanisms of platypnea-orthodeoxia syndrome. Arch Cardiol Mex 92(2): 274-282.
- Mahjoub F, Majidi S (2017) Platypnea-Orthodeoxia Syndrome: A Rare and Treatable Cause of Positional Dyspnea. Cureus 12(7): e9052.
- EMCrit Project (2024) Pulmonary arteriovenous malformations (PAVMs) & other rare pulmonary vascular disorders.
- Eryk Hakman N, Kathleen Cowling M (2023) Paradoxical Embolism - StatPearls - NCBI Bookshelf. National Center for Biotechnology Information.
- American Heart Association Journals (n.d.) Acute Ischemic Stroke in Patients With Pulmonary Arteriovenous Malformations: Paradoxical Embolism or Epiphenomenon? Stroke: Vascular and Interventional Neurology.
- Dubey A, Williams M (2019) Pulmonary arteriovenous malformations: diagnosis. Cardiovasc Diagn Ther 8(3): 325-337.
- American Society of Hematology (2017) Not So BenignHereditary Hemorrhagic Telangiectasia: More Than Just Recurrent Nosebleeds. The Hematologist.
- Mount Sinai Health System (n.d.) Cerebral arteriovenous malformation Information.
- Mayo Clinic Staff (2021) Brain AVM (arteriovenous malformation) - Symptoms and causes. Mayo Clinic.
- National Center for Biotechnology Information (2023) Arteriovenous Malformation of the Brain - StatPearls - NCBI Bookshelf.
- Barrow Neurological Institute (2023) Brain Arteriovenous Malformation (AVM) - Diagnosis and Treatment.
- Merck Manual Professional Edition (2023) Cerebral Arteriovenous Malformations (AVMs) - Neurologic Disorders.
- Luca Ielasi, Matteo Tonnini, Fabio Piscaglia, Ilaria Serio (2023) Current guidelines for diagnosis and management of hepatic involvement in hereditary hemorrhagic teleangiectasia. World J Hepatol 15(5): 675-687.
- Cleveland Clinic (2023) Hepatic Encephalopathy: Symptoms, Causes, Grading & Treatment.
- Inês Nunes da S, Clara M, Fábio C, Sofia C, Maria João G (2020) Osler-Weber-Rendu Syndrome with Severe Hepatic Manifestations: A Rare Clinical Case. Eur J Rep Intern Med 7(11): 001831.
- Binet Q, Annet L, Danse E, Goffette P, Lanthier N (2022) Hepatobiliary and Pancreatic: An uncommon cause of portal hypertension. J Gastroenterol Hepatol 37(10): 1843.
- Agnes SK, Katharine JH, Sumeet P, Min Jung K, Shahnaz P, et al. (2020) Subaortic Membranes in Patients With Hereditary Hemorrhagic Telangiectasia and Liver Vascular Malformations. J Am Heart Assoc 9(20): e016197.
- Shovlin CL, Guttmacher AE, Buscarini E, Faughnan ME, Hyland RH, et al. (2000) Diagnostic criteria for hereditary hemorrhagic telangiectasia (Rendu-Osler-Weber syndrome). Am J Med Genet 91(1): 66-67.
- Pahl KS, Choudhury A, Wusik K, Hammill A, White A, et al. (2018) Applicability of the Curaçao Criteria for the Diagnosis of Hereditary Hemorrhagic Telangiectasia in the Pediatric Population. J Pediatr 197: 207-213.
- Plauchu H, de Chadarévian JP, Bideau A, Robert JM (1989) Age-related clinical profile of hereditary hemorrhagic telangiectasia in an epidemiologically recruited population. Am J Med Genet 32(3): 291-297.
- Shovlin CL, Hughes JM, Tuddenham EG, Temperley I, Perembelon YF, et al. (1994) A gene for hereditary haemorrhagic telangiectasia maps to chromosome 9q3. Nature Genet 6(2): 205-209.
- Grigg C, Anderson D, Earnshaw J (2017) Diagnosis and Treatment of Hereditary Hemorrhagic Telangiectasia. Ochsner J 17(2): 157-161.
- Faughnan ME, Mager JJ, Hetts SW, Palda VA, Lang-Robertson K, et al. (2020) Second International Guidelines for the Diagnosis and Management of Hereditary Hemorrhagic Telangiectasia. Ann Internal Med 173(12): 989-1001.
- Bharatha A, Faughnan ME, Kim H, Pourmohamad T, Krings T, et al. (2012) Brain arteriovenous malformation multiplicity predicts the diagnosis of hereditary hemorrhagic telangiectasia: Quantitative assessment. Stroke 43(1): 72-78.
- Shovlin CL, Sodhi V, McCarthy A, Lasjaunias P, Jackson JE, et al. (2008) Estimates of maternal risks of pregnancy for women with hereditary haemorrhagic telangiectasia (Osler-Weber-Rendu syndrome): Suggested approach for obstetric services. BJOG: An Int J Obstetr Gynaecol 115(9): 1108-1115.
- Garg N, Khunger M, Gupta A, Kumar N (2014) Optimal management of hereditary hemorrhagic telangiectasia. J Blood Med 5: 191-206.
- Grigg C, Anderson D, Earnshaw J (2017) Diagnosis and Treatment of Hereditary Hemorrhagic Telangiectasia. Ochsner J 17(2): 157-161.
- Faughnan ME, Palda VA, Garcia-Tsao G, Geisthoff UW, McDonald J, et al. (2011) International guidelines for the diagnosis and management of hereditary hemorrhagic telangiectasia. J Med Genet 48(2): 73-87.
- Faughnan ME, Mager JJ, Hetts SW, Palda VA, Lang-Robertson K, et al. (2020) Second international guidelines for the diagnosis and management of hereditary hemorrhagic telangiectasia. Ann Internal Med 173(12): 989-1001.
- Brown RD, Wiebers DO, Forbes GS, O'Fallon WM (1988) The natural history of unruptured intracranial arteriovenous malformations. J Neurosurg 68(3): 352-357.
- Friedman WA, Bova FJ (1995) Linear accelerator radiosurgery for arteriovenous malformations: The relationship of size to outcome. J Neurosurg 82(2): 180-189.
- Awad IA, Polster SP (2010) Cavernous malformations: Deconstructing a neurosurgical disease. J Neurosurg 113(4): 641-654.
- Pollock BE, Flickinger JC (2002) A proposed radiosurgery-based grading system for arteriovenous malformations (AVMs). J Neurosurg 96(1): 79-85.
- Ference BA, Shannon TM, White RI, Zawin M (1994) Life-threatening pulmonary hemorrhage: A complication of large pulmonary arteriovenous malformations in hereditary hemorrhagic telangiectasia. Chest 106(5): 1387-1390.
- Dupuis-Girod S, Bailly S, Plauchu H (2010) Hereditary hemorrhagic telangiectasia: From molecular biology to patient care. J Thrombosis Haemost 8(7): 1447-56.
- McDonald J, Wooderchak-Donahue W, VanSant Webb C, Whitehead K, Stevenson DA, et al. (2015) Hereditary hemorrhagic telangiectasia: Genetics and molecular diagnostics in a new era. Front Genet 6: 1.
- Meyer D, Wagner M, Ziyeh S, Tonn JC (2018) 3D angiography in the treatment of cerebral arteriovenous malformations. Neurosurg Rev 41(3): 789-797.
- Duke SM, Shenoy MP, Fritz KA (2020) Patient-centered care in the management of hereditary hemorrhagic telangiectasia. Journal of Vascular Surgery: Venous and Lymphatic Disord 8(6): 1043-1050.
- Shovlin CL, Buscarini E, Kjeldsen AD, Mager JJ, Sabba C, et al. (2020) The European rare disease network for hereditary hemorrhagic telangiectasia. Orphanet J Rare Dis 15(1): 1-13.
- Lopes-Coelho F, Martins F, Pereira SA, Serpa J (2021) Anti-Angiogenic Therapy: Current Challenges and Future Perspectives. Int J Mol Sci 22(7): 3765.
- Pan P, Weinsheimer S, Cooke D, Ethan Winkler, Adib Abla, et al. (2021) Review of treatment and therapeutic targets in brain arteriovenous malformation. J Cereb Blood Flow Metab 41(12): 3141-3156.
- Seebauer CT, Wiens B, Hintschich CA, Natascha PBS, Katja E, et al. (2024) Targeting the microenvironment in the treatment of arteriovenous malformations. Angiogenesis 27(1): 91-103.
- Williams BJ, Park DM, Sheehan JP (2012) Bevacizumab used for the treatment of severe, refractory perilesional edema due to an arteriovenous malformation treated with stereotactic radiosurgery. J Neurosurg 116(5): 972-977.
- Tessier S, Lipton BA, Ido F, Longo S, Nanda S (2021) Pathogenesis and therapy of arteriovenous malformations: A case report and narrative review. Int J Crit Illn Inj Sci 11(3): 167-176.
- Zhu X, Stergiopoulos K, Wu S (2009) Risk of hypertension and renal dysfunction with an angiogenesis inhibitor sunitinib: Systematic review and meta-analysis. Acta Oncologica 48(1): 9-17.
- Anguita R, Tasiopoulou A, Shahid S, Roth J, Sim SY, et al. (2021) A Review of Aflibercept Treatment for Macular Disease Ophthalmol Ther 10(3): 413-428.
- Ola R, Hessels J, Hammill A, Cassi F, Marianne C, et al. (2023) Executive summary of the 14th HHT international scientific conference. Angiogenesis 26(Suppl 1): 27-37.
- Shovlin CL, Simeoni I, Downes K, Zoe CF, Karyn M, et al. (2020) Mutational and phenotypic characterization of hereditary hemorrhagic telangiectasia. Blood 136(17): 1907-1918.
- Rossi E, Bernabeu C, Smadja DM (2019) Endoglin as an Adhesion Molecule in Mature and Progenitor Endothelial Cells: A Function Beyond TGF-β. Front Med (Lausanne) 6: 10.
- Dupuis-Girod S, Ginon I, Saurin JC, Denis M, Els G, et al. (2012) Bevacizumab in patients with hereditary hemorrhagic telangiectasia and severe hepatic vascular malformations and high cardiac output. JAMA 307(9): 948-955.
- Kjeldsen AD, Vase P, Green A (1999) Hereditary hemorrhagic telangiectasia: a population-based study of prevalence and mortality in Danish patients. J Intern Med 245(1): 31-39.
- Doudna JA, Charpentier E (2014) Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346(6213): 1258096.
- Yin H, Kauffman KJ, Anderson DG (2017) Delivery technologies for genome editing. Nat Rev Drug Discov 16(6): 387-399.
- Gupta A, Maclean DA (2016) 3D imaging and advanced visualization in radiology. J Digit Imaging 29(2): 129-139.
- Wang S, Summers RM, Liu J (2020) AI-enhanced medical imaging: insights into image acquisition, processing, and interpretation. J Clin Invest 130(5): 2240-2250.
- Simioni P, Rondon F (2015) Emerging angiogenesis inhibitors for the treatment of solid tumors. J Angiogenesis Res 7(1): 1.
- Ricciuti B, Genova C (2021) Current landscape and future directions of angiogenesis inhibition in non-small cell lung cancer. J Thorac Dis 13(5): 2981-2993.
- Collins FS, Varmus H (2015) A new initiative on precision medicine. N Engl J Med 372(9): 793-795.
- Ashley EA (2015) The precision medicine initiative: A new national effort. JAMA 313(21): 2119-21120.