odactra (2)

FDA expand drug label of ALK’s Odactra for the Treatment of House Dust Mite Allergy in Young Children

Written By Lavanya Chavhan B.Pharm

Reviewed and Fact Checked by Vikas Londhe, M.Pharm (Pharmacology)

odactra (2)

The U.S. Food and Drug Administration (FDA) has recently expanded label for ALK-Abelló’s Odactra, a sublingual immunotherapy tablet, to treat house dust mite (HDM) allergies in children as young as five years old. This landmark decision expands access to a groundbreaking therapy previously approved for adults, offering a safe and convenient option for young children grappling with this pervasive allergen. 

What is Odactra?

Odactra s the allergen extract of House Dust Mite (Dermatophagoides farinae and Dermatophagoides pteronyssinus). It is prescription medication designed to address house dust mite (HDM)-induced allergic rhinitis, with or without conjunctivitis. It is indicated in case of Confirmed positive in vitro testing for IgE antibodies to Dermatophagoides farinae or Dermatophagoides pteronyssinus house dust mites.  It is approved in the population from 5 years old to 65 years old age patient.

Administered as a once-daily tablet as sublingual that dissolves under the tongue, it works by exposing the immune system to small, controlled amounts of dust mite allergens. Over time, this exposure helps desensitize the body, reducing the severity of allergic reactions. Unlike traditional treatments such as antihistamines or nasal corticosteroids, which only alleviate symptoms, Odactra targets the root cause of allergies, offering potential long-term relief.

Significance of the Approval

House dust mites (HDMs) are microscopic arachnids (related to ticks and spiders) that thrive in warm, humid environments. They feed on dead human skin cells and are commonly found in mattresses, pillows, upholstered furniture, and carpets. The allergens are primarily from their faeces and body fragments. House dust mites are a leading trigger of perennial allergies, affecting millions of children worldwide.

For sensitized individuals, short-term exposure can cause immediate allergic reactions, including Allergic rhinitis, Asthma exacerbations, Allergic conjunctivitis, and Skin irritation. Prolonged exposure and chronic sensitization can lead to Persistent asthma, chronic rhino sinusitis, Atopic dermatitis, increased risk of developing new allergies.

Chronic symptoms like sneezing, nasal congestion, and itchy eyes can disrupt sleep, school performance, and overall quality of life. For young children, whose immune systems are still developing, uncontrolled allergies may also heighten the risk of developing asthma.

Global Prevalence of HDM allergy estimated to affect 65–130 million people globally, in some regions, up to 80% of asthmatic children are sensitized to HDM however in adults’ sensitization rates range from 20% to 30% in general populations.

A renowned pediatric allergist involved in Odactra’s clinical trials, emphasized the importance of this approval: “Immunotherapy at an early age can alter the course of allergic disease. Odactra’s sublingual form is particularly advantageous for children, avoiding the anxiety associated with allergy shots.” 

Clinical Trial Insights

The clinical trial (NCT04145219) performed in children was double-blind, placebo-controlled, randomized field efficacy study conducted in Europe, the United States and Canada for a duration of approximately 12 months comparing the efficacy of ODACTRA (N=693) to placebo (N=706) in the treatment of HDM allergic rhinitis/rhino conjunctivitis with or without asthma in children 5 through 11 years of age.

The FDA’s decision followed this phase III study which evaluated safety and efficacy of odactra in young children. Results demonstrated a significant reduction in allergy symptoms and medication use compared to placebo. Participants also reported improved sleep and daily functioning. The trial highlighted Odactra’s tolerability, with most side effects being mild, such as oral itching or throat irritation. Severe reactions were rare, aligning with the therapy’s established safety profile in adults. 

Safety and Accessibility

During the pediatric clinical trial, the most commonly reported adverse reactions included ear and mouth itching, followed by throat irritation, abdominal pain, altered taste, and lip swelling. However, since its initial approval, Odactra has carried a black box warning for anaphylaxis and is contraindicated in individuals with uncontrolled asthma.

Conclusion

The FDA’s expansion of Odactra’s approval marks a pivotal advancement in managing HDM allergies in children. By offering a convenient, home-based therapy, Odactra empowers families to address allergies proactively, potentially reducing long-term health burdens. Parents of affected children are encouraged to consult allergists to determine if Odactra is a suitable option. 

As research continues to underscore the benefits of early intervention, Odactra stands out as a beacon of innovation in the quest to improve pediatric health outcomes. 

References

  1. Package Insert, Highlights of Prescribing Information, Odactra, ALK-Abelló A/S available from https://www.fda.gov/media/103380/download

2. Odactra, US Food and Drug Administration, 17 March 2025 available from https://www.fda.gov/vaccines-blood-biologics/allergenics/odactra

3. Bracken SJ, Adami AJ, Szczepanek SM, et al, Long-Term Exposure to House Dust Mite Leads to the Suppression of Allergic Airway Disease Despite Persistent Lung Inflammation. Int Arch Allergy Immunol. 2015;166(4):243-58. doi: 10.1159/000381058. Epub 2015 Apr 28. PMID: 25924733; PMCID: PMC4485530.

4. Dust Mites, American Lung Association, 05 Nov 2024, available from https://www.lung.org/clean-air/indoor-air/indoor-air-pollutants/dust-mites

5. Schuster, Antje et al., Efficacy and safety of SQ house dust mite sublingual immunotherapy-tablet (12 SQ-HDM) in children with allergic rhinitis/rhino conjunctivitis with or without asthma (MT-12): a randomised, double-blind, placebo-controlled, phase III trial, The Lancet Regional Health – Europe, Volume 48, 101136

6. Solarz, K., Obuchowicz, A., Asman, M. et al. Abundance of domestic mites in dwellings of children and adolescents with asthma in relation to environmental factors and allergy symptoms. Sci Rep 11, 18453 (2021). https://doi.org/10.1038/s41598-021-97936-7.

Qfitlia_optimized_2000

FDA Approves Qfitlia: First siRNA Therapy for Haemophilia Prophylaxis

Written by Aishwarya Shinde (B.Pharm)

Reviewed and Fact Checked by Vikas Londhe M.Pharm (Pharmacology)

Qfitlia_optimized_2000

The U.S. Food and Drug Administration (FDA) has granted approval for Qfitlia (fitusiran), a groundbreaking therapy designed for the routine prophylaxis of bleeding episodes in individuals with haemophilia A or B. This approval, announced on March 28, 2025, marks a significant advancement in the treatment of these rare genetic bleeding disorders. Qfitlia is authorized for use in adults and pediatric patients aged 12 years and older, regardless of the presence of factor VIII or IX inhibitors.

Haemophilia A and B

Haemophilia is a rare, inherited bleeding disorder caused by deficiencies in clotting factors, leading to prolonged bleeding episodes. The two main types are Haemophilia A (deficiency of Factor VIII) and Haemophilia B (deficiency of Factor IX), both inherited as X-linked recessive disorders. Since the defective gene is located on the X chromosome, males (XY) are typically affected, while females (XX) are usually carriers. However, rare cases of female haemophilia can occur due to lyonization, where the healthy X chromosome is inactivated.

Conventional treatments for haemophilia focus on managing bleeding episodes and preventing complications. Replacement therapy includes recombinant or plasma-derived Factor VIII concentrates for Haemophilia A and Factor IX concentrates for Haemophilia B. Prophylactic treatment involves regular infusions to prevent bleeding, particularly in severe cases. Additional therapies include Desmopressin (DDAVP) for mild Haemophilia A, which stimulates Factor VIII release, and antifibrinolytics like tranexamic acid to stabilize clots, especially in mucosal bleeding.

Recent advances in treatment include gene therapy, such as etranacogene dezaparvovec for Haemophilia B, offering potential long-term solutions. Haemophilia A is more prevalent, affecting approximately 1 in 5,000 male births, while Haemophilia B occurs in 1 in 30,000 male births. Globally, an estimated 400,000 people are affected by the disorder. Ongoing research and novel therapies aim to improve quality of life and reduce the burden of this chronic condition.

Need of an Advance Therapy

However the advanced therapies are needed for haemophilia A and B to address the limitations of conventional treatments and improve patient outcomes. While traditional factor replacement therapies are effective, they require frequent intravenous infusions, which can be burdensome for patients and may lead to complications like inhibitor development (antibodies against clotting factors). Additionally, some patients experience breakthrough bleeding despite prophylaxis, highlighting the need for more durable and convenient solutions.

Qfitlia: A Novel Approach to Haemophilia Management

Developed by Sanofi, Qfitlia (Fitusiran) is an RNA interference (RNAi) therapeutic developed for the treatment of haemophilia A and B, as well as other bleeding disorders. It is designed to reduce bleeding episodes by silencing the production of antithrombin (AT), a natural anticoagulant protein, through RNA interference rather than replacing the missing clotting factors (as in conventional therapies).

Fitusiran is a synthetic siRNA molecule encapsulated in a lipid nanoparticle for targeted delivery to hepatocytes (liver cells). Once inside the liver, it binds to the messenger RNA (mRNA) encoding antithrombin (SERPINC1 gene).

The siRNA triggers the degradation of antithrombin mRNA, reducing antithrombin production. Lower antithrombin levels shift the hemostatic balance toward a pro-coagulant state, promoting thrombin generation and improving clot formation.

In haemophilia, deficient Factor VIII (Haemophilia A) or Factor IX (Haemophilia B) leads to impaired thrombin burst, by reducing antithrombin, fitusiran bypasses the need for exogenous clotting factors, allowing even low levels of endogenous Factors VIII/IX to work more effectively.

The therapy is administered via subcutaneous injections as few as six times per year, offering a significant reduction in treatment burden compared to existing options. It is available in a convenient prefilled pen or vial-and-syringe format, making it easier for patients and caregivers to manage.

Clinical Efficacy

The FDA’s approval was based on data from Sanofi’s ATLAS clinical trial program, which included phase 3 studies such as ATLAS-A/B and ATLAS-INH.

ATLAS-A/B study was a Phase 3, multicenter, open-label, randomized trial evaluating the efficacy and safety of fitusiran prophylaxis in males aged 12 years and older with severe haemophilia A or B without inhibitors. The study enrolled 120 participants across 45 sites in 17 countries.Participants were randomized in a 2:1 ratio to receive either once-monthly 80 mg subcutaneous fitusiran prophylaxis or continue with on-demand clotting factor concentrates for duration of 9 months.

Key Findings includes, Annualized Bleeding Rate (ABR): The median ABR was 0.0 (interquartile range [IQR] 0.0–3.4) in the fitusiran group, compared to 21.8 (IQR 8.4-41.0) in the on-demand group. The estimated mean ABR was significantly lower in the fitusiran group (3.1) than in the on-demand group (31.0), representing an approximate 90% reduction. Bleed-Free Participants: Approximately 51% of participants in the fitusiran group experienced no treated bleeds during the study period, compared to 5% in the on-demand group.

ATLAS-INH study, was a Phase 3, open-label, randomized trial evaluating the efficacy and safety of fitusiran in individuals aged 12 years and older with severe haemophilia A or B who have inhibitors to factor VIII or IX. Participants were randomly assigned in a 2:1 ratio to receive once-monthly 80 mg subcutaneous fitusiran prophylaxis or to continue with on-demand treatment using bypassing agents (BPAs).

Fitusiran prophylaxis led to a significant reduction in the annualized bleeding rate (ABR), with a 90.8% decrease compared to the on-demand BPA group. Approximately 66% of participants receiving fitusiran experienced zero treated bleeds during the study period, compared to 5% in the BPA group.

Based on the results from the ATLAS-INH and other related trials, the U.S. Food and Drug Administration (FDA) approved fitusiran, marketed as Qfitlia, for routine prophylaxis to prevent or reduce the frequency of bleeding episodes in adults and pediatric patients aged 12 years and older with haemophilia A or B, with or without inhibitors.

Safety Profile

The safety profile of Qfitlia includes warning for serious thrombotic event and acute and recurrent gall bladder disease. The most common treatment-emergent adverse event in the fitusiran group was increased alanine aminotransferase levels, observed in 32% of participants.

Benefits for Patients

Qfitlia represents a paradigm shift in haemophilia care by combining effective bleed protection with infrequent dosing and simplified administration. “This approval highlights our commitment to advancing innovation and improving care for the rare blood disorders community,” said Brian Foard, executive vice president at Sanofi Tanya Wroblewski, MD, deputy director at the FDA’s Center for Drug Evaluation and Research, emphasized that this therapy “can be administered less frequently than other existing options,” improving quality of life for patients.

Conclusion

The FDA’s approval of Qfitlia marks a transformative moment for individuals living with haemophilia A or B. By offering effective bleed prevention with minimal treatment burden, this innovative therapy has the potential to significantly improve patient outcomes and redefine standards of care worldwide.

References

1. FDA Approves Novel Treatment for Haemophilia A or B, with or without Factor Inhibitors, US Food and Drug Administration, 28 March 2025, available form https://www.fda.gov/news-events/press-announcements/fda-approves-novel-treatment-Haemophilia-or-b-or-without-factor-inhibitors

2. Press Release: Qfitlia approved as the first therapy in the US to treat Haemophilia A or B with or without inhibitors, Sanofi, 28 March 2025, available from https://www.sanofi.com/en/media-room/press-releases/2025/2025-03-28-20-07-38-3051637

3. Castaman G, Matino D. Haemophilia A and B: molecular and clinical similarities and differences. Haematologica. 2019 Sep;104(9):1702-1709. doi: 10.3324/haematol.2019.221093. Epub 2019 Aug 8. PMID: 31399527; PMCID: PMC6717582.

4. Mannucci PM, Franchini M. Is haemophilia B less severe than haemophilia A? Haemophilia. 2013 Jul;19(4):499-502. Doi: 10.1111/hae.12133. Epub 2013 Mar 21. PMID: 23517072.

5. Meeks SL, Batsuli G. Haemophilia and inhibitors: current treatment options and potential new therapeutic approaches. Haematology Am Soc Hematol Educ Program. 2016 Dec 2; 2016(1):657-662. Doi: 10.1182/asheducation-2016.1.657. PMID: 27913543; PMCID: PMC6142469.

6. Treatment of Haemophilia, US Centre for disease control and prevention, 13 Nov 2024, available from https://www.cdc.gov/Haemophilia/treatment/index.html

7. Miesbach W, Schwäble J, Müller MM, Seifried E. Treatment Options in Haemophilia. Dtsch Arztebl Int. 2019 Nov 22; 116(47):791-798. Doi: 10.3238/arztebl.2019.0791. PMID: 31847949; PMCID: PMC6937545.

8. Von Drygalski A., Giermasz A., Castaman G., Key N.S., Lattimore S., Leebeek F.W.G., Miesbach W., Recht M., Long A., Gut R., et al. Etranacogene Dezaparvovec (AMT-061 Phase 2b): Normal/near Normal FIX Activity and Bleed Cessation in Haemophilia B. Blood Adv. 2019;3:3241–3247. Doi: 10.1182/bloodadvances.2019000811.

9. Mehta P, Reddivari AKR. Haemophilia. [Updated 2023 Jun 5]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK551607/

10. Qfitlia (fitusiran) injection, Highlights Of Prescribing Information, Genzyme Corporation, A Sanofi Company, available from https://products.sanofi.us/qfitlia/qfitlia.pdf

11. Young G, Srivastava A, Kavakli K, et al, Rangarajan S. Efficacy and safety of fitusiran prophylaxis in people with haemophilia A or haemophilia B with inhibitors (ATLAS-INH): a multicentre, open-label, randomised phase 3 trial. Lancet. 2023 Apr 29;401(10386):1427-1437. Doi: 10.1016/S0140-6736(23)00284-2. Epub 2023 Mar 29. PMID: 37003287.

12. Srivastava A, Rangarajan S, Kavakli K, et al, Fitusiran prophylaxis in people with severe haemophilia A or haemophilia B without inhibitors (ATLAS-A/B): a multicentre, open-label, randomised, phase 3 trial. Lancet Haematol. 2023 May;10(5):e322-e332. Doi: 10.1016/S2352-3026(23)00037-6. Epub 2023 Mar 29. PMID: 37003278.

 

VHL Welireg

EMA Approves Welireg (Belzutifan): A Breakthrough for VHL-Associated Tumors and Advanced Kidney Cancer

Written By Yogita Bhadane, B.Pharm

 Reviewed and Fact Checked by Vikas Londhe M.Pharm (Pharmacology)

VHL Welireg

Introduction
Welireg (belzutifan), a significant advancement, has been approved by the European Medicines Agency (EMA) as the first clinically approved pharmaceutical therapy for tumors associated with von Hippel-Lindau (VHL) disease and for adults with advanced renal cell carcinoma (RCC) who have already received treatment. As a new targeted therapy for unmet medical needs, this approval holds promise for clinicians treating these difficult conditions.

Understanding VHL Disease and RCC

Von Hippel-Lindau disease (VHL) is a rare genetic disorder that predisposes people to develop tumors and cysts in the kidneys, pancreas, and central nervous system, with kidney cancer being the most common type. Renal cell carcinoma occurs in an abnormal 70% of individuals with VHL syndrome. These tumors are caused by mutations in the VHL gene, which affects how the gene regulates the breakdown of proteins called hypoxia-inducible factor (HIF), which are sensitive to tissue oxygen levels. Tumor formation results from aberrant HIF activation brought on by VHL mutation. Conventional management techniques involved frequent surgical procedures and constant monitoring, which increased risks and lowered patient living standards. Immunocheckpoint inhibitors and anti-angiogenic therapies are used in the treatment of advanced renal cell carcinoma; however, post-progression care options are still scarce.

Hemangioblastomas

These non-cancerous growths mostly appear in the retina, spinal cord, and brain tissue. Brain hemangioblastoma patients are more likely to experience symptoms like headaches, vision issues, and neurological impairments.

Renal Cell Carcinomas (RCC)

The risk of kidney cancer, primarily clear cell renal carcinoma, is increased in people with VHL disease. In addition to renal impairment, these disorders often manifest bilaterally and cause hematuria and dysuria. Some examples show that (heterogeneous) petrocalcic deposits give rise to saddle-shaped inclusions.

 Pheochromocytomas:

High blood pressure, headaches, sweating, and palpitations are some of the symptoms caused by these tumors growing on the adrenal gland, which strictly adhere to EPI while binding to norepinephrine. Although phenochromocytomas usually appear as benign tumors, if left untreated, they have the potential to become fatal.

 Pancreatic Cysts and Tumors:

Patients with VHL may develop pancreatic cysts. Although the majority of them are benign, tiny molecules have the potential to develop into pancreatic neuroendocrine tumors, which may release hormones that result in various symptoms.

 Endolymphatic Sac Tumors (ELSTs):

Almost all of these are tumors that are located in the patient’s ear near the inner ear. Tinnitus, instability, and hearing loss are possible symptoms for those who have this condition. Chemotherapy or even stereotactic radiation may be used to treat general growth and low-grade ELSTs.

 Retinal Angiomas:

Hemangioblastomas, also known as retinal angiomas, are tumors of the blood vessels that frequently develop in the membrane of the eye and can cause partial blindness. Hemangioblastomas appear when they cause bleeding or retinal detachment, or when they are discovered during an eye exam.

Welireg’s Mechanism:

Angiogenesis (the formation of blood vessels), erythropoiesis (the production of red blood cells), and tumor growth are all regulated by HIF-2α.
The Von Hippel-Lindau (VHL) protein breaks down HIF-2α under normal oxygen conditions.
HIF-2α builds up in tumors that lack VHL, such as clear cell renal cell carcinoma and von Hippel-Lindau disease-associated tumors, which causes the tumor to grow out of control.

 Blocking HIF-2α Dimerization with HIF-1β:

By binding specifically to HIF-2α, belzutifan stops it from interacting with HIF-1β, which is a prerequisite for HIF-2α to activate target genes.
VEGF (vascular endothelial growth factor), PDGF (platelet-derived growth factor), and EPO (erythropoietin) are among the genes involved in tumor survival whose transcription is inhibited by this.

 Anti-Tumor Effects:

Welireg is effective in cancers caused by HIF-2α overactivity, especially VHL-associated renal cell carcinoma (RCC), central nervous system hemangioblastomas, and pancreatic neuroendocrine tumors, because it inhibits HIF-2α signaling, which in turn reduces tumor growth, angiogenesis, and erythropoiesis.

An important tumor growth pathway is disrupted by the oral hypoxia-inducible factor 2 alpha (HIF-2α) inhibitor belzutifan. HIF-2α builds up in low oxygen environments, activating genes that support angiogenesis and cell division. Welireg deprives tumors of these signals by blocking HIF-2α, which slows growth and causes shrinkage. This mechanism is revolutionary because it provides a precision medicine approach that is specific to the molecular drivers of RCC and VHL.

Clinical Trial Efficacy

VHL-Associated Tumors: Belzutifan is being evaluated in patients with von Hippel-Lindau (VHL) disease-associated renal cell carcinoma (RCC) in the Phase 2 open-label clinical trial NCT03401788 (Study 004). 61 adults with at least one detectable RCC tumor and a germline VHL alteration were enrolled. Additionally, patients may have pancreatic neuroendocrine tumors (pNETs) or CNS hemangioblastomas. Prior use of HIF-2α inhibitors, systemic therapy, surgical necessity, and metastatic disease were excluded. Until toxicity or progression, participants were given 120 mg of belzutifan every day. Overall response rate (ORR), the main outcome, was 49% in RCC, 63% in CNS hemangioblastomas, and 83% in pNETs. With more than half of respondents continuing to respond for more than 12 months, the median response duration was not reached.

Advanced RCC: Belzutifan, a hypoxia-inducible factor 2α (HIF-2α) inhibitor, is being evaluated in patients with advanced solid tumors as part of the LITESPARK-001 trial (NCT02974738), a phase 1 clinical study. 55 patients with advanced ccRCC who had previously received treatment were given 120 mg of belzutifan orally once daily as part of this cohort. The objective response rate (ORR), which included one complete response and thirteen partial responses, was 25% following a median follow-up of more than three years. The median progression-free survival (PFS) was 14.5 months, and the disease control rate (DCR) was 80%. Anaemia (24% grade 3) and hypoxia (13% grade 3) were the most common treatment-related adverse events; no grade 4 or grade 5 treatment-related adverse events were noted. These findings show that belzutifan has a manageable safety profile and long-lasting antitumor activity in patients with ccRCC who have received extensive pre-treatment.

Glioblastoma (GBM) Cohort: A cohort of 25 patients with recurrent IDH wild-type glioblastoma after radiation therapy and temozolomide was also included in the trial. Belzutifan 120 mg was given orally to the patients twice a day. There were no objective responses after a median follow-up of 1.9 months; the median PFS was 1.4 months, and the clinical benefit rate was 8%. Every patient had at least one adverse event, and 60% of them had grade 3-5 events. Anaemia (64%), exhaustion (52%), headache (32%), and muscle weakness (32%), were the most frequent adverse events. There were no deaths brought on by the treatment. These results imply that in this GBM cohort, belzutifan did not exhibit antitumor activity.
Belzutifan’s potential for treating advanced ccRCC is generally highlighted by the LITESPARK-001 trial, but its effectiveness in treating GBM has not yet been established.

Safety and Tolerability

Administration of Welireg during pregnancy can cause embryo-fetal harm. It was highly recommended to confirm the pregnancy status before initiating Welireg.

Anaemia (71%), Hypoxia (48%), and nausea (31%), which are usually mild to moderate, are common side effects. It is advised to regularly check oxygen and hemoglobin levels. When compared to intravenous treatments, the ease of oral administration (120 mg once daily) improves patient adherence.

EMA Approval and Patient Impact

With the EMA’s support, access is made possible throughout the EU, changing the way that care is provided. Welireg preserves organ function and quality of life for VHL patients by lowering the need for invasive surgeries. It closes a crucial gap left by post-standard therapies for advanced RCC. “Belzutifan represents a paradigm shift, offering a non-invasive option with meaningful clinical benefits,” says oncologist Dr. Maria Ruiz.

 Future Directions

Belzutifan is being studied for combination treatments and other cancers driven by HIF-2α. Its success highlights how hypoxia pathways can be targeted in oncology.

Conclusion
The approval of Welireg, which addresses long-standing issues in the management of VHL and RCC, is a victory for precision medicine. Belzutifan sets a new standard by focusing on innovation in rare and complex cancers and coordinating treatment with disease biology. This milestone opens the door for future advancements in targeted therapies in addition to providing immediate clinical benefits.

References:

1. First medicine to treat rare genetic disorder causing cysts and tumours, European Medicine Agency, 13 December 2024

2. WELIREG® (belzutifan) Receives First European Commission Approval for Two Indications, Merck, 18 February 2025

3. Kim E, Zschiedrich S. Renal Cell Carcinoma in von Hippel-Lindau Disease-From Tumor Genetics to Novel Therapeutic Strategies. Front Pediatr. 2018 Feb 9;6:16. doi: 10.3389/fped.2018.00016. PMID: 29479523; PMCID: PMC5811471.

4. Ashouri K, Mohseni S, Tourtelot J, Sharma P, Spiess PE. Implications of Von Hippel-Lindau Syndrome and Renal Cell Carcinoma. J Kidney Cancer VHL. 2015 Sep 25; 2(4):163-173. Doi: 10.15586/jkcvhl.2015.41. PMID: 28326271; PMCID: PMC5345519.

5. Eric Jonasch, Frede Donskov, Othon Iliopoulos, Belzutifan for Renal Cell Carcinoma in von Hippel–Lindau Disease, n engl j med 385;22, November 25, 2021.

6. Hu, J., Tan, P., Ishihara, M. et al.Tumor heterogeneity in VHL drives metastasis in clear cell renal cell carcinoma. Sig Transduct Target Ther8, 155 (2023). https://doi.org/10.1038/s41392-023-01362-2

7. Roy E. Strowd et al., Phase 1 LITESPARK-001 study of belzutifan in advanced solid tumors: Results of the glioblastoma cohort. JCO 42, 2054-2054(2024). DOI:10.1200/JCO.2024.42.16_suppl.2054