1
|
Mahadevan A, Yazdanpanah O, Patel V, Benjamin DJ, Kalebasty AR. Ophthalmologic toxicities of antineoplastic agents in genitourinary cancers: Mechanisms, management, and clinical implications. Curr Probl Cancer 2025; 54:101171. [PMID: 39708456 DOI: 10.1016/j.currproblcancer.2024.101171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/19/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Abstract
Genitourinary cancers affect over 480,000 patients in the United States annually. While promising therapeutic modalities continue to emerge, notably immune checkpoint inhibitors, molecular targeted therapies, antibody-drug conjugates, and radioligand therapies, these treatments are associated with a spectrum of adverse side-effects, including ophthalmologic toxicities. In this review, we cover the most commonly used antineoplastic agents for the kidneys, bladder, urinary tracts, prostate, testis, and penis, detailing mechanism, indication, and recent trials supporting their use. For each category of antineoplastic therapy, we describe the epidemiology, management, and clinical presentation, of common ophthalmologic toxicities stemming from these agents. This review serves to augment awareness and recognition of possible ophthalmologic manifestations resulting from the use of antineoplastic agents in genitourinary malignancy. Early identification of these side effects can hasten ophthalmology referral and ultimately improve visual outcomes in patients experiencing medication-induced ocular toxicities.
Collapse
Affiliation(s)
- Aditya Mahadevan
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - Omid Yazdanpanah
- Division of Hematology/Oncology, University of California Irvine Health, Orange, CA, USA.
| | - Vivek Patel
- Department of Ophthalmology, University of California Irvine Health, Orange, CA, USA.
| | | | | |
Collapse
|
2
|
Lauricella E, Chaoul N, D'Angelo G, Giglio A, Cafiero C, Porta C, Palmirotta R. Neuroendocrine Tumors: Germline Genetics and Hereditary Syndromes. Curr Treat Options Oncol 2025:10.1007/s11864-024-01288-z. [PMID: 39821711 DOI: 10.1007/s11864-024-01288-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 01/19/2025]
Abstract
OPINION STATEMENT The vast majority of neuroendocrine 'neoplasms (NENs) are sporadic, although recent evidence has indicated that a subset of these cancers may also originate as a result of genetic germline mutations. To date, 10% of these cancers can be linked to an inherited genetic syndrome. Genetic diagnosis is crucial for patients with a suspected hereditary NEN syndrome, as it recognizes patients carrying germline mutations and allows for personalized clinical follow-up, considering the higher risk of developing other tumours. The potential for early genetic detection has significant implications for the treatment of patients with hereditary NEN syndrome, as it may facilitate the delivery of precision therapy that differs from that typically provided to other patients. Thus, the integration of genotypic and phenotypic diagnostic methods help clinicians to provide more informed treatment and to extend appropriate prevention to family members.
Collapse
Affiliation(s)
- Eleonora Lauricella
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Piazza G. Cesare 11, 70124, Bari, Italy
| | - Nada Chaoul
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Piazza G. Cesare 11, 70124, Bari, Italy
| | - Gabriella D'Angelo
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Piazza G. Cesare 11, 70124, Bari, Italy
| | - Andrea Giglio
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Piazza G. Cesare 11, 70124, Bari, Italy
| | - Concetta Cafiero
- Medical Oncology, SG Moscati Hospital, Via Per Martina Franca, 74010, Taranto, Italy
| | - Camillo Porta
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Piazza G. Cesare 11, 70124, Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico Di Bari, Piazza G. Cesare 11, 70124, Bari, Italy
| | - Raffaele Palmirotta
- Interdisciplinary Department of Medicine, Section of Sciences and Technologies of Laboratory Medicine, University of Bari "Aldo Moro", Piazza G. Cesare 11, 70124, Bari, Italy.
| |
Collapse
|
3
|
Zhao W, Kim B, Coffey NJ, Bowers S, Jiang Y, Bowman CE, Noji M, Jang C, Simon MC, Arany Z, Kim B. HIF2α inhibits glutaminase clustering in mitochondria to sustain growth of clear cell Renal Cell Carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.04.592520. [PMID: 38746132 PMCID: PMC11092754 DOI: 10.1101/2024.05.04.592520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Clear cell renal cell carcinomas (ccRCC) are largely driven by HIF2α and are avid consumers of glutamine. However, inhibitors of glutaminase1 (GLS1), the first step in glutaminolysis, have not shown benefit in phase III trials, and HIF2α inhibition, recently FDA-approved for treatment of ccRCC, shows great but incomplete benefits, underscoring the need to better understand the roles of glutamine and HIF2α in ccRCC. Here, we report that glutamine deprivation rapidly redistributes GLS1 into isolated clusters within mitochondria across diverse cell types, but not in ccRCC. GLS1 clustering is rapid (1-3 hours) and reversible, is specifically driven by reduced intracellular glutamate, and is mediated by mitochondrial fission. Clustered GLS1 markedly enhances glutaminase activity and promotes cell death under glutamine-deprived conditions. HIF2α prevents GLS1 clustering, independently of its transcriptional activity, thereby protecting ccRCC cells from cell death induced by glutamine deprivation. Reversing this protection, by genetic expression of GLS1 mutants that constitutively cluster, enhances ccRCC cell death in culture and suppresses ccRCC growth in vivo . These findings provide multiple insights into cellular glutamine handling, including a novel metabolic pathway by which HIF2α promotes ccRCC, and reveals a potential therapeutic avenue to synergize with HIF2α inhibition in the treatment of ccRCC.
Collapse
|
4
|
Tsoli M, Panagaki M, Tasouli E, Kolomodi D, Kaltsas G. New Developments in VHL-Associated Neuroendocrine Neoplasms. Curr Oncol Rep 2025:10.1007/s11912-024-01631-5. [PMID: 39757325 DOI: 10.1007/s11912-024-01631-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE OF REVIEW The purpose of this review is to outline the current knowledge on epidemiology, diagnosis and management of neuroendocrine neoplasms (NENs) that develop in the context of Von Hippel-Lindau (VHL) syndrome. RECENT FINDINGS Pancreatic NENs develop in 8-17% of VHL patients (vPNENs) and are mostly multi-focal, cystic and non-functioning. Surgical resection is recommended for vPNENS > 3 cm that exhibit higher metastatic potential or in tumors with short doubling time while in the 20% of cases with metastatic disease the HIF-2 A inhibitor belzutifan is considered a promising option. Pheochromocytomas arising in VHL type 2 are often bilateral and have a noradrenergic phenotype while they are associated with increased risk of recurrence. High-specific activity [131I]-MIBG and sunitinib are the treatment options with the highest level of evidence whereas studies on belzutifan are evolving. Life-long surveillance and management in the context of a multidisciplinary team are suggested to achieve the best clinical outcome.
Collapse
Affiliation(s)
- Marina Tsoli
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Agiou Thoma 17, Athens, 11527, Greece.
| | - Maria Panagaki
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Agiou Thoma 17, Athens, 11527, Greece
| | - Elisavet Tasouli
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Agiou Thoma 17, Athens, 11527, Greece
| | - Dionysia Kolomodi
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Agiou Thoma 17, Athens, 11527, Greece
| | - Gregory Kaltsas
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Agiou Thoma 17, Athens, 11527, Greece
| |
Collapse
|
5
|
Cotton CC, Chandrabhatla AS, Andrews PH, Purrow BW, Shildkrot Y(E. BELZUTIFAN FOR TREATMENT OF GIANT RETINAL HEMANGIOBLASTOMA WITH EXTRASCLERAL EXTENSION ASSOCIATED WITH VON HIPPEL-LINDAU SYNDROME. Retin Cases Brief Rep 2025; 19:66-69. [PMID: 37973032 PMCID: PMC11649176 DOI: 10.1097/icb.0000000000001516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 10/03/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE To describe the clinical response and safety profile of the novel HIF-2ɑ inhibitor belzutifan in treating a giant retinal hemangioblastoma with extrascleral extension associated with von Hippel-Lindau syndrome. METHODS A 71-year-old woman with Von Hippel-Lindau syndrome presented with a giant retinal hemangioblastoma with extrascleral extension in her only remaining eye. She had no light perception in the right eye and intraocular pressure was 48. She requested enucleation because of chronic pain, but because of concern for significant bleeding given the size of the neoplasm, a trial of belzutifan was initiated. RESULTS Within 3 months of treatment initiation, the patient reported an 80% reduction in pain. Magnetic resonance imaging showed 30% reduction in longest tumor diameter. Dose adjustments were guided by serum hemoglobin levels, allowing the patient to remain on the medication for over a year with continued tumor regression on MRI and avoid enucleation. CONCLUSION Retinal hemangioblastoma with extrascleral extension is exceedingly rare and its treatment is complex, often requiring enucleation or external beam radiotherapy. This report demonstrates the use of belzutifan to safely and successfully reduce ocular tumor burden of complicated retinal hemangioblastoma with extrascleral extension, ultimately decreasing the need for enucleation.
Collapse
Affiliation(s)
- Caroline C. Cotton
- Department of Ophthalmology, University of Virginia Health Sciences Center, Charlottesville, Virginia
| | - Anirudha S. Chandrabhatla
- Department of Ophthalmology, University of Virginia Health Sciences Center, Charlottesville, Virginia
| | - Patrick H. Andrews
- Department of Ophthalmology, University of Virginia Health Sciences Center, Charlottesville, Virginia
| | - Benjamin W. Purrow
- Department of Neurology, Division of Neuro-oncology, University of Virginia Health Sciences Center, Charlottesville, Virginia; and
| | | |
Collapse
|
6
|
Duan E, Robinson M, Davis C, Pruthi S, Shin C, Lewis M, Martinez-Agosto JA, Gorin MB, Shuch BM, Friedman DL, Chang VY. Pediatric patients with von Hippel-Lindau and hemangioblastomas treated successfully with belzutifan. Pediatr Blood Cancer 2025; 72:e31371. [PMID: 39415342 DOI: 10.1002/pbc.31371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/24/2024] [Accepted: 09/23/2024] [Indexed: 10/18/2024]
Abstract
Hemangioblastoma is the most common tumor associated with von Hippel-Lindau (VHL), and are a leading cause of mortality. We present five pediatric patients with VHL-associated hemangioblastomas treated with belzutifan, a hypoxia-inducible factor 2a (HIF2a) inhibitor. Three patients were started on belzutifan due to vision loss from progressive retinal hemangioblastomas. Within one year of treatment, all three patients had improvement in hemangioblastoma size and visual acuity. For patients with intracranial lesions, belzutifan resulted in an improvement in neurologic symptoms and hemangioblastoma size. Four patients experienced grade 1-2 anemia and two patients required a dose reduction. Our report suggests that belzutifan can be an effective therapy for pediatric, adolescent, and young adult patients with VHL-associated hemangioblastomas.
Collapse
Affiliation(s)
- Emily Duan
- Department of Pediatrics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Michael Robinson
- Division of Pediatric Hematology/Oncology, Vanderbilt, Nashville, Tennessee, USA
| | - Charles Davis
- Department of Radiology, UCLA, Los Angeles, California, USA
| | - Sumit Pruthi
- Department of Neuroradiology, Vanderbilt, Nashville, Tennessee, USA
| | - Christina Shin
- Ronald Reagan UCLA Medical Center, UCLA, Los Angeles, California, USA
| | - Marisa Lewis
- Division of Pediatric Hematology/Oncology, UCLA, Los Angeles, California, USA
| | - Julian A Martinez-Agosto
- Division of Clinical Genetics, Department of Human Genetics, UCLA, Los Angeles, California, USA
- Department of Psychiatry, UCLA, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| | - Michael B Gorin
- Department of Ophthalmology, UCLA, Los Angeles, California, USA
| | - Brian M Shuch
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
- Department of Urology, UCLA, Los Angeles, California, USA
| | - Debra L Friedman
- Division of Pediatric Hematology/Oncology, Vanderbilt, Nashville, Tennessee, USA
| | - Vivian Y Chang
- Division of Pediatric Hematology/Oncology, UCLA, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
- Children's Discovery and Innovation Institute, UCLA, Los Angeles, California, USA
| |
Collapse
|
7
|
Zhang X, Zhang M, Sun H, Wang X, Wang X, Sheng W, Xu M. The role of transcription factors in the crosstalk between cancer-associated fibroblasts and tumor cells. J Adv Res 2025; 67:121-132. [PMID: 38309692 PMCID: PMC11725164 DOI: 10.1016/j.jare.2024.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Transcription factors (TFs) fulfill a critical role in the formation and maintenance of different cell types during the developmental process as well as disease. It is believed that cancer-associated fibroblasts (CAFs) are activation status of tissue-resident fibroblasts or derived from form other cell types via transdifferentiation or dedifferentiation. Despite a subgroup of CAFs exhibit anti-cancer effects, most of them are reported to exert effects on tumor progression, further indicating their heterogeneous origin. AIM OF REVIEW This review aimed to summarize and review the roles of TFs in the reciprocal crosstalk between CAFs and tumor cells, discuss the emerging mechanisms, and their roles in cell-fate decision, cellular reprogramming and advancing our understanding of the gene regulatory networks over the period of cancer initiation and progression. KEY SCIENTIFIC CONCEPTS OF REVIEW This manuscript delves into the key contributory factors of TFs that are involved in activating CAFs and maintaining their unique states. Additionally, it explores how TFs play a pivotal and multifaceted role in the reciprocal crosstalk between CAFs and tumor cells. This includes their involvement in processes such as epithelial-mesenchymal transition (EMT), proliferation, invasion, and metastasis, as well as metabolic reprogramming. TFs also have a role in constructing an immunosuppressive microenvironment, inducing resistance to radiation and chemotherapy, facilitating angiogenesis, and even 'educating' CAFs to support the malignancies of tumor cells. Furthermore, this manuscript delves into the current status of TF-targeted therapy and considers the future directions of TFs in conjunction with anti-CAFs therapies to address the challenges in clinical cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Meng Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Hui Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xu Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xin Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| |
Collapse
|
8
|
Choueiri TK, Merchan JR, Figlin R, McDermott DF, Arrowsmith E, Michaelson MD, Tykodi SS, Heath EI, Spigel DR, D'Souza A, Kassalow L, Perini RF, Vickery D, Bauer TM. Belzutifan plus cabozantinib as first-line treatment for patients with advanced clear-cell renal cell carcinoma (LITESPARK-003): an open-label, single-arm, phase 2 study. Lancet Oncol 2025; 26:64-73. [PMID: 39756444 DOI: 10.1016/s1470-2045(24)00649-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Belzutifan, a first-in-class HIF-2α inhibitor, has shown antitumour activity as monotherapy and in combination with cabozantinib in patients with previously treated advanced kidney cancer. The phase 2 LITESPARK-003 study was designed to determine the antitumour activity and safety of belzutifan in combination with cabozantinib in patients with advanced clear-cell renal cell carcinoma that was previously untreated (cohort 1) or previously treated with immunotherapy (cohort 2). Here, we report results from cohort 1 of this clinical trial. METHODS LITESPARK-003 is an open-label, single-arm, phase 2 study at ten hospitals and cancer centres in the USA. In cohort 1, eligible patients were at least 18 years of age, had an Eastern Cooperative Oncology Group performance status of 0 or 1, and had received no previous systemic therapy for locally advanced or metastatic renal cell carcinoma. Patients received belzutifan 120 mg orally once daily and cabozantinib 60 mg orally once daily until unacceptable adverse events, disease progression, or patient withdrawal. The primary endpoint was investigator-assessed confirmed objective response according to Response Evaluation Criteria in Solid Tumors version 1.1. Antitumour activity and safety were assessed in all patients who received at least one dose of study treatment. This trial is registered with ClinicalTrials.gov, NCT03634540, and is ongoing. FINDINGS Between Sept 27, 2018, and Jan 10, 2023, we screened 138 patients for eligibility, and 50 (36%) were enrolled and assigned to cohort 1. The median age was 64 years (IQR 57-72). 40 (80%) of 50 patients were male and ten (20%) were female. 48 (96%) patients were White, one (2%) patient was Black or African American, and one (2%) was of a race in the other category. As of the data cutoff (May 15, 2023), median follow-up was 24·3 months (IQR 13·9-32·0). 35 (70%, 95% CI 55-82) of 50 patients had a confirmed objective response, including four (8%) who had a complete response and 31 (62%) who had a partial response. The most frequent grade 3-4 treatment-related adverse events were hypertension (six [12%] patients), anaemia (five [10%] patients), and fatigue (four [8%] patients). Seven (14%) of 50 patients had serious treatment-related adverse events. No treatment-related deaths occurred. INTERPRETATION Belzutifan plus cabozantinib has promising antitumour activity in treatment-naive patients with advanced clear-cell renal cell carcinoma and further investigation of an HIF-2α inhibitor in combination with a multitargeted tyrosine kinase inhibitor as a treatment option in this population is warranted. FUNDING Merck Sharp & Dohme LLC, a subsidiary of Merck & Co, Inc, Rahway, NJ, USA, and the National Cancer Institute.
Collapse
Affiliation(s)
- Toni K Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| | | | | | | | | | | | - Scott S Tykodi
- University of Washington and Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | | | - Anishka D'Souza
- Genitourinary Medical Oncology, University of Southern California, Los Angeles, CA, USA
| | | | | | | | - Todd M Bauer
- Tennessee Oncology/Greco-Hainsworth Centers for Research, Nashville, TN, USA
| |
Collapse
|
9
|
Santamarina MG, Necochea Raffo JA, Lavagnino Contreras G, Recasens Thomas J, Volpacchio M. Predominantly multiple focal non-cystic renal lesions: an imaging approach. Abdom Radiol (NY) 2025; 50:224-260. [PMID: 38913137 DOI: 10.1007/s00261-024-04440-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/25/2024]
Abstract
Multiple non-cystic renal lesions are occasionally discovered during imaging for various reasons and poses a diagnostic challenge to the practicing radiologist. These lesions may appear as a primary or dominant imaging finding or may be an additional abnormality in the setting of multiorgan involvement. Awareness of the imaging appearance of the various entities presenting as renal lesions integrated with associated extrarenal imaging findings along with clinical information is crucial for a proper diagnostic approach and patient work-up. This review summarizes the most relevant causes of infectious, inflammatory, vascular, and neoplastic disorders presenting as predominantly multiple focal non-cystic lesions.
Collapse
Affiliation(s)
- Mario G Santamarina
- Radiology Department, Hospital Naval Almirante Nef, Subida Alesandri S/N., Viña del Mar, Provincia de Valparaíso, Chile.
- Radiology Department, Hospital Dr. Eduardo Pereira, Valparaiso, Chile.
| | - Javier A Necochea Raffo
- Radiology Department, Hospital Naval Almirante Nef, Subida Alesandri S/N., Viña del Mar, Provincia de Valparaíso, Chile
| | | | - Jaime Recasens Thomas
- Departamento de Radiología, Escuela de Medicina, Universidad de Valparaíso, Valparaiso, Chile
| | - Mariano Volpacchio
- Radiology Department, Centro de Diagnóstico Dr. Enrique Rossi, Buenos Aires, Argentina
| |
Collapse
|
10
|
Wang Y, Song J, Zheng S, Wang S. Advancements in understanding the molecular mechanisms and clinical implications of Von Hippel-Lindau syndrome: A comprehensive review. Transl Oncol 2025; 51:102193. [PMID: 39571489 PMCID: PMC11617254 DOI: 10.1016/j.tranon.2024.102193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/06/2024] [Accepted: 10/30/2024] [Indexed: 12/08/2024] Open
Abstract
Von Hippel-Lindau Syndrome (VHL) is a rare genetic disorder characterized by tumors in multiple organs, including the kidneys, pancreas, and central nervous system. This comprehensive review discusses the genetic basis and clinical manifestations of VHL, as well as recent advancements in understanding the molecular mechanisms that lead to tumor formation. The authors highlight the role of hypoxia-inducible factors and the ubiquitin-proteasome system in VHL-associated cancer development .The review also discusses the potential clinical implications of these findings, such as the development of targeted therapies for VHL-associated cancers. However, the authors note the challenges associated with developing effective treatments for this complex disease, including limited patient availability for clinical trials due to its rarity .Overall, this review provides valuable insights into our current understanding of VHL and offers important avenues for future research aimed at improving the diagnosis, treatment, and management of VHL patients. By illuminating the molecular underpinnings of VHL-associated cancers, this work may ultimately help to develop more effective treatments and improve outcomes for patients with this challenging disease.
Collapse
Affiliation(s)
- Yaochun Wang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Jingzhuo Song
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Shuxing Zheng
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Shuhong Wang
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China.
| |
Collapse
|
11
|
Guo X, Song J, Liu M, Ou X, Guo Y. The interplay between the tumor microenvironment and tumor-derived small extracellular vesicles in cancer development and therapeutic response. Cancer Biol Ther 2024; 25:2356831. [PMID: 38767879 PMCID: PMC11110713 DOI: 10.1080/15384047.2024.2356831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 05/14/2024] [Indexed: 05/22/2024] Open
Abstract
The tumor microenvironment (TME) plays an essential role in tumor cell survival by profoundly influencing their proliferation, metastasis, immune evasion, and resistance to treatment. Extracellular vesicles (EVs) are small particles released by all cell types and often reflect the state of their parental cells and modulate other cells' functions through the various cargo they transport. Tumor-derived small EVs (TDSEVs) can transport specific proteins, nucleic acids and lipids tailored to propagate tumor signals and establish a favorable TME. Thus, the TME's biological characteristics can affect TDSEV heterogeneity, and this interplay can amplify tumor growth, dissemination, and resistance to therapy. This review discusses the interplay between TME and TDSEVs based on their biological characteristics and summarizes strategies for targeting cancer cells. Additionally, it reviews the current issues and challenges in this field to offer fresh insights into comprehending tumor development mechanisms and exploring innovative clinical applications.
Collapse
Affiliation(s)
- Xuanyu Guo
- The Affiliated Hospital, Southwest Medical University, Luzhou, PR China
| | - Jiajun Song
- Department of Clinical Laboratory Medicine, the Affiliated Hospital, Southwest Medical University, Luzhou, PR China
| | - Miao Liu
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, PR China
| | - Xinyi Ou
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, PR China
| | - Yongcan Guo
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, PR China
| |
Collapse
|
12
|
Song G, Xue S, Zhu Y, Wu C, Ji X. The efficacy and safety of belzutifan inhibitor in patients with advanced or metastatic clear cell renal cell carcinoma: a meta-analysis. BMC Pharmacol Toxicol 2024; 25:100. [PMID: 39707485 DOI: 10.1186/s40360-024-00828-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND The belzutifan is a hypoxia inducible factor-2 alpha (HIF-2α) inhibitor for the treatment of advanced or metastatic clear cell renal cell carcinoma (mccRCC) and has exhibited good safety and efficacy in clinical trials. We conducted a meta-analysis of relevant studies to further clarify the efficacy and safety of belzutifan for the treatment of mccRCC. METHODS Multiple databases and abstracts from major scientific meetings were systematically reviewed for eligible articles published before June 1, 2024. The following outcomes were analyzed: objective response rate (ORR), disease control rate (DCR), median duration of response (mDOR), median progression-free survival (mPFS), median overall survival (mOS), and treatment-related adverse events (TRAes). 426 records were reviewed, and data were extracted by at least two individuals. RESULTS Seven studies involving 715 patients were included in this meta-analysis. The pooled ORR was 34% (95% confidence interval [CI]: 23-46%), the DCR was 79% (95% CI: 66-90%), the mDOR was 21.8 months (95% CI: 14.82-28.78), and the mPFS time was 8.8 months (95% CI: 6.15-11.44). The pooled incidence of grade 3-5 TRAes was 46%, and the most common TRAe was anemia. Further subgroup analysis revealed that, compared with belzutifan monotherapy, the combination of belzutifan with tyrosine kinase inhibitors (TKIs) as second- or later-line therapy was associated with a statistically significant increase in the ORR. Toxicity was also greater with combined inhibition therapy. CONCLUSIONS Our meta-analysis revealed moderate antitumor activity and a manageable safety profile of the inhibitor belzutifan in patients with mccRCC.
Collapse
Affiliation(s)
- Ge Song
- Department of Critical Care Medicine, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, 250014, China
| | - Song Xue
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yingming Zhu
- Department of Radiation Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunling Wu
- Nephrology Blood Purification Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China.
| | - Xiaowei Ji
- Department of Critical Care Medicine, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, 250014, China.
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
13
|
Valdés A, Pizarro G, González-Montero J, Rojas C, Burotto M. Targeting HIF-2α: the role of belzutifan in clear cell renal carcinoma management. Expert Rev Clin Pharmacol 2024:1-11. [PMID: 39670660 DOI: 10.1080/17512433.2024.2436433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION Belzutifan is a first-in-class hypoxia-inducible factor-2 alpha (HIF-2α) inhibitor. It targets the von Hippel-Lindau protein (pVHL)-HIF-vascular endothelial growth factor (VEGF) pathway, which is crucial in cellular responses to hypoxia. By inhibiting HIF-2α, belzutifan disrupts the transcription of genes involved in tumor growth and angiogenesis. AREAS COVERED In this review, we describe the pVHL-HIF-VEGF pathway and how it led to the development of HIF inhibitors, including belzutifan. A search was conducted for trials involving Belzutifan, including phase I-III trials. We describe the relevant toxicity, with emphasis on hypoxia and anemia. EXPERT OPINION Belzutifan is a relatively safe drug, with manageable adverse events, including anemia and hypoxia as on-target toxicity. Ongoing trials are studying its benefit in overall survival for RCC in first-line treatment and its potential in other malignancies. The LITESPARK-005 trial reported the benefit of belzutifan in progression-free survival (PFS) compared to everolimus in later lines of treatment, with improvement in quality-of-life outcomes. Given its different mechanism of action to currently available treatments, belzutifan is expected to play a prominent role in the treatment of clear cell renal carcinoma and other cancers.
Collapse
Affiliation(s)
- Alejandro Valdés
- Department of Medical Oncology, Bradford Hill Clinical Research Center, Santiago, Chile
- Department of Medical Oncology, Instituto Nacional del Cáncer, Santiago, Chile
| | - Gonzalo Pizarro
- Department of Medical Oncology, Bradford Hill Clinical Research Center, Santiago, Chile
- Department of Medical Oncology, Hospital Sótero del Río, Santiago, Chile
| | | | - Carlos Rojas
- Department of Medical Oncology, Bradford Hill Clinical Research Center, Santiago, Chile
| | - Mauricio Burotto
- Department of Medical Oncology, Bradford Hill Clinical Research Center, Santiago, Chile
| |
Collapse
|
14
|
Borkum JM. Cluster Headache and Hypoxia: Breathing New Life into an Old Theory, with Novel Implications. Neurol Int 2024; 16:1691-1716. [PMID: 39728749 DOI: 10.3390/neurolint16060123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Cluster headache is a severe, poorly understood disorder for which there are as yet virtually no rationally derived treatments. Here, Lee Kudrow's 1983 theory, that cluster headache is an overly zealous response to hypoxia, is updated according to current understandings of hypoxia detection, signaling, and sensitization. It is shown that the distinctive clinical characteristics of cluster headache (circadian timing of attacks and circannual patterning of bouts, autonomic symptoms, and agitation), risk factors (cigarette smoking; male gender), triggers (alcohol; nitroglycerin), genetic findings (GWAS studies), anatomical substrate (paraventricular nucleus of the hypothalamus, solitary tract nucleus/NTS, and trigeminal nucleus caudalis), neurochemical features (elevated levels of galectin-3, nitric oxide, tyramine, and tryptamine), and responsiveness to treatments (verapamil, lithium, melatonin, prednisone, oxygen, and histamine desensitization) can all be understood in terms of hypoxic signaling. Novel treatment directions are hypothesized, including repurposing pharmacological antagonists of hypoxic signaling molecules (HIF-2; P2X3) for cluster headache, breath training, physical exercise, high-dose thiamine, carnosine, and the flavonoid kaempferol. The limits of current knowledge are described, and a program of basic and translational research is proposed.
Collapse
Affiliation(s)
- Jonathan M Borkum
- Department of Psychology, University of Maine, 301 Williams Hall, Orono, ME 04469-5742, USA
| |
Collapse
|
15
|
Lanzolla G, Sabini E, Beigel K, Khan MP, Sherry Liu X, Wang D, Laslow B, Taylor D, Bellido T, Giaccia A, Schipani E. Pharmacological inhibition of HIF2 protects against bone loss in an experimental model of estrogen deficiency. Proc Natl Acad Sci U S A 2024; 121:e2416004121. [PMID: 39602268 PMCID: PMC11626196 DOI: 10.1073/pnas.2416004121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/02/2024] [Indexed: 11/29/2024] Open
Abstract
Estrogen deficiency, which is linked to various pathological conditions such as primary ovarian insufficiency and postmenopausal osteoporosis, disrupts the delicate balance between bone formation and resorption. This imbalance leads to bone loss and an increased risk of fractures, primarily due to a significant reduction in trabecular bone mass. Trabecular osteoblasts, the cells responsible for bone formation within the trabecular compartment, originate from skeletal progenitors located in the bone marrow. The microenvironment of the bone marrow contains hypoxic (low oxygen) regions, and the hypoxia-inducible factor-2α (HIF2) plays a crucial role in cellular responses to these low-oxygen conditions. This study demonstrates that the loss of HIF2 in skeletal progenitors and their derivatives during development enhances trabecular bone mass by promoting bone formation. More importantly, PT2399, a small molecule that specifically inhibits HIF2, effectively prevents trabecular bone loss in ovariectomized adult mice, a model for estrogen-deficient bone loss. Both the genetic and pharmacological approaches result in an increase in osteoblast number, which is linked to the expansion of the pool of skeletal progenitor cells. This expansion either by loss or inhibition of HIF2 uncovers a pivotal mechanism for increasing osteoblast numbers and bone formation, resulting in greater trabecular bone mass.
Collapse
Affiliation(s)
- Giulia Lanzolla
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA19104
| | - Elena Sabini
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA19104
| | - Katherine Beigel
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Mohd Parvez Khan
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA19104
| | - Xiaowei Sherry Liu
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA19104
| | - Dian Wang
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA19104
| | - Brittany Laslow
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA19104
| | - Deanne Taylor
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Teresita Bellido
- Department of Physiology and Cell Biology, University of Arkansas, School of Medicine, Little Rock, AR72205
- Central Arkansas Veterans Healthcare System, John L. McClellan, Little Rock, AR72205
| | - Amato Giaccia
- Department of Oncology, University of Oxford, Division of Medical Sciences, OxfordOX37DQ, United Kingdom
| | - Ernestina Schipani
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA19104
| |
Collapse
|
16
|
Zhu Y, Li Y, Li X, Yu Y, Zhang L, Zhang H, Chen C, Chen D, Wang M, Xing N, Yang F, Wasilijiang W, Ye X. Targeting Hypoxia and Autophagy Inhibition via Delivering Sonodynamic Nanoparticles With HIF-2α Inhibitor for Enhancing Immunotherapy in Renal Cell Carcinoma. Adv Healthc Mater 2024; 13:e2402973. [PMID: 39396375 DOI: 10.1002/adhm.202402973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/28/2024] [Indexed: 10/15/2024]
Abstract
Immune checkpoint blockers (ICBs) therapy stands as the first-line treatment option for advanced renal cell carcinoma (RCC). However, its effectiveness is hindered by the immunosuppressive tumor microenvironment (TME). Sonodynamic therapy (SDT) generates tumor cell fragments that can prime the host's antitumor immunity. Nevertheless, the hypoxic microenvironment and upregulated autophagy following SDT often lead to cancer cell resistance. In response to these challenges, a hypoxia-responsive polymer (Poly(4,4'-azobisbenzenemethanol-PMDA)-mPEG5k, P-APm) encapsulating both a HIF-2α inhibitor (belzutifan) and the ultrasonic sensitize (Chlorin e6, Ce6) is designed, to create the nanoparticle APm/Ce6/HIF. APm/Ce6/HIF combined with ultrasound (US) significantly suppresses tumor growth and activates antitumor immunity in vivo. Moreover, this treatment effectively transforms the immunosuppressive microenvironment from "immune-cold" to "immune-hot", thereby enhancing the response to ICBs therapy. The findings indicate that APm/Ce6/HIF offers a synergistic approach combining targeted therapy with immunotherapy, providing new possibilities for treating RCC.
Collapse
Affiliation(s)
- Yihao Zhu
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yajian Li
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xuwen Li
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuan Yu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, 310022, China
| | - Lingpu Zhang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hanchen Zhang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Can Chen
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China
| | - Dong Chen
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Mingshuai Wang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Nianzeng Xing
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Feiya Yang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wahafu Wasilijiang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Urology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Shanxi, 030013, China
| | - Xiongjun Ye
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| |
Collapse
|
17
|
Aykut A, Else T, Demirci H. Belzutifan as the first-line treatment for a challenging von Hippel-Lindau-related retinal hemangioblastoma: successful treatment of a case and review of the literature. CANADIAN JOURNAL OF OPHTHALMOLOGY 2024; 59:e862-e865. [PMID: 39043253 DOI: 10.1016/j.jcjo.2024.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/07/2024] [Accepted: 05/20/2024] [Indexed: 07/25/2024]
Affiliation(s)
- Aslan Aykut
- Kellogg Eye Center, University of Michigan, Ann Arbor, MI; Marmara University Medical School, Istanbul, Turkey
| | - Tobias Else
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI; Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI
| | - Hakan Demirci
- Kellogg Eye Center, University of Michigan, Ann Arbor, MI.
| |
Collapse
|
18
|
Agarwal N, Brugarolas J, Ghatalia P, George S, Haanen JB, Gurney H, Ravilla R, Van der Veldt A, Beuselinck B, Pokataev I, Suelmann BBM, Tuthill MH, Vaena D, Zagouri F, Wu J, Perini RF, Liu Y, Merchan J, Atkins MB. Randomized phase II dose comparison LITESPARK-013 study of belzutifan in patients with advanced clear cell renal cell carcinoma. Ann Oncol 2024; 35:1148-1156. [PMID: 39233312 DOI: 10.1016/j.annonc.2024.08.2338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Belzutifan is a first-in-class hypoxia-inducible factor subunit 2α (HIF-2α) inhibitor approved at a dose of 120 mg once daily for certain adults with VHL disease and adults with advanced renal cell carcinoma (RCC) following therapy with a programmed cell death protein 1 (PD-1) [or programmed death ligand 1 (PD-L1)] inhibitor and a vascular endothelial growth factor tyrosine kinase inhibitor. However, whether the belzutifan dose could be optimized is unclear. PATIENTS AND METHODS The phase II LITESPARK-013 study (NCT04489771) enrolled patients with advanced clear cell RCC whose disease progressed after one to three prior systemic therapies, including an anti-PD-(L)1 regimen. Patients were randomly assigned 1 : 1 to receive belzutifan 120 or 200 mg once daily. The primary endpoint was the objective response rate (ORR) per RECIST version 1.1. The secondary endpoints were duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety. RESULTS Overall, 154 patients were enrolled (120 mg: n = 76; 200 mg: n = 78). The median follow-up was 20.1 months (range 14.8-28.4). The ORR was 23.7% versus 23.1% for the 120 mg and 200 mg groups, respectively [P = 0.5312; -0.5%, 95% confidence interval (CI) -14.0% to 12.9%]. The median DOR was not reached for the 120 mg arm and was 16.1 months (2.1+ to 23.5+) for the 200 mg arm. No between-group differences were observed for PFS [hazard ratio (HR) 0.94, 95% CI 0.63-1.40] or OS (medians not reached; HR 1.11, 95% CI 0.65-1.90). Grade 3 or 4 treatment-related adverse events were observed in 35 patients (46.1%) in the 120 mg group and 36 patients (46.2%) in the 200 mg group. CONCLUSIONS The efficacy of belzutifan was similar between the 120 mg dose and the 200 mg dose for previously treated clear cell RCC. Safety at both doses was consistent with the known safety profile of belzutifan. These results further support 120 mg once daily as the preferred dose for belzutifan.
Collapse
Affiliation(s)
- N Agarwal
- Director, Genitourinary Oncology Program, Huntsman Cancer Institute, University of Utah (NCI-CCC), Salt Lake City.
| | - J Brugarolas
- Director, Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas; Department of Internal Medicine, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas
| | - P Ghatalia
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia
| | - S George
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, USA
| | - J B Haanen
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - H Gurney
- Director of Medical Oncology and Clinical Trials, MQ Health, Macquarie University, Sydney, Australia
| | - R Ravilla
- Department of Medical Oncology, New York Oncology Hematology, Albany, USA
| | - A Van der Veldt
- Department of Radiology & Nuclear Medicine, Medical Oncology, Erasmus MC, Rotterdam, Netherlands
| | - B Beuselinck
- Department of General Medical Oncology, University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - I Pokataev
- S. S. Yudin City Clinical Hospital, Department of Health of Moscow, Moscow, Russia
| | - B B M Suelmann
- Department of Medical Oncology, Universitair Medisch Centrum Utrecht, Utrecht, Netherlands
| | - M H Tuthill
- Department of Oncology and Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - D Vaena
- Department of Medical Oncology/Hematology, West Cancer Center and Research Institute, Germantown, USA
| | - F Zagouri
- Clinical Therapeutics, Alexandra Regional General Hospital Athens, Athens, Greece
| | - J Wu
- Merck & Co., Inc., Rahway
| | | | - Y Liu
- Merck & Co., Inc., Rahway
| | - J Merchan
- Department of Medicine, Medical Oncology Division, University of Miami - Sylvester Comprehensive Center Cancer, Miami
| | - M B Atkins
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, USA
| |
Collapse
|
19
|
Jimenez C, Baudrand R, Uslar T, Bulzico D. Perspective review: lessons from successful clinical trials and real-world studies of systemic therapy for metastatic pheochromocytomas and paragangliomas. Ther Adv Med Oncol 2024; 16:17588359241301359. [PMID: 39574494 PMCID: PMC11580098 DOI: 10.1177/17588359241301359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/04/2024] [Indexed: 11/24/2024] Open
Abstract
Pheochromocytomas and paragangliomas (PPGLs) are orphan tumors with the potential to spread to distant organs such as the lymph nodes, the skeleton, the lungs, and the liver. These metastatic tumors exhibit high rates of morbidity and mortality due to their frequently large tumor burden, the progression of the disease, and the excessive secretion of catecholamines that lead to cardiovascular disease and gastrointestinal dysmotility. Several molecular drivers responsible for the development of PPGLs have been described over the last 30 years. Although therapeutic options are limited, substantial progress has been made in the recognition of effective systemic therapies for these tumors. Successful clinical trials with radiopharmaceuticals such as high-specific-activity meta-iodobenzylguanidine and tyrosine kinase inhibitors such as cabozantinib and sunitinib have been recently published. This review will discuss the results of these studies and their impact on current clinical practices. In addition, this review will provide valuable information on how to design clinical trials to treat patients with metastatic PPGLs with novel medications.
Collapse
Affiliation(s)
- Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1461, Houston, TX 77030, USA
| | - Rene Baudrand
- Department of Endocrinology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Thomas Uslar
- Department of Endocrinology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Daniel Bulzico
- Department of Nuclear Medicine and Endocrine Oncology, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Ummarino A, Calà N, Allavena P. Extrinsic and Cell-Intrinsic Stress in the Immune Tumor Micro-Environment. Int J Mol Sci 2024; 25:12403. [PMID: 39596467 PMCID: PMC11594858 DOI: 10.3390/ijms252212403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
In continuously progressive tumor tissues, the causes of cellular stress are multiple: metabolic alterations, nutrient deprivation, chronic inflammation and hypoxia. To survive, tumor cells activate the stress response program, a highly conserved molecular reprogramming proposed to cope with challenges in a hostile environment. Not only cancer cells are affected, but stress responses in tumors also have a profound impact on their normal cellular counterparts: fibroblasts, endothelial cells and infiltrating immune cells. In recent years, there has been a growing interest in the interaction between cancer and immune cells, especially in difficult conditions of cellular stress. A growing literature indicates that knowledge of the molecular pathways activated in tumor and immune cells under stress conditions may offer new insights for possible therapeutic interventions. Counter-regulating the stress caused by the presence of a growing tumor can therefore be a weapon to limit disease progression. Here, we review the main pathways activated in cellular stress responses with a focus on immune cells present in the tumor microenvironment.
Collapse
Affiliation(s)
- Aldo Ummarino
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| | - Nicholas Calà
- Etromapmacs Pole, Agorà Biomedical Sciences, 71010 Foggia, Italy;
| | | |
Collapse
|
21
|
Barragan-Carrillo R, Saad E, Saliby RM, Sun M, Albiges L, Bex A, Heng D, Mejean A, Motzer RJ, Plimack ER, Powles T, Rini BI, Zhang T, Choueiri TK. First and Second-line Treatments in Metastatic Renal Cell Carcinoma. Eur Urol 2024:S0302-2838(24)02681-2. [PMID: 39505582 DOI: 10.1016/j.eururo.2024.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/18/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND AND OBJECTIVE The treatment landscape for metastatic renal cell carcinoma (mRCC) has evolved significantly in recent years, leading to improved outcomes. The aim of this review is to provide clinicians with a practical guide for selecting first- and second-line treatments on the basis of current evidence. METHODS We critically evaluated systemic treatment strategies for mRCC. A comprehensive literature search was conducted in PubMed and Embase, alongside manual searches of guidelines and conference proceedings up to October 2024. A narrative review was performed to reach a consensus, with voting used to resolve differing opinions among authors. KEY FINDINGS AND LIMITATIONS First-line treatment options include immune checkpoint inhibitor (ICI)-based combinations or tyrosine kinase inhibitors (TKIs). Four combination regimens have been approved internationally. Owing to the lack of head-to-head trials and standardized biomarkers, treatment decisions rely on factors such as International Metastatic RCC Database Consortium (IMDC) risk score, functional status, safety profiles, sarcomatoid features, use of immunosuppressive drugs, and need for immediate response. Despite advances, many patients will experience disease progression on ICI-based therapy, necessitating further treatment. The need for standardized second-line approaches remains unmet. TKIs, alone or with everolimus, show promising efficacy, while HIF2a inhibitors offer newer options with a favorable toxicity profile. Rechallenge with ICIs after early progression is not recommended. CONCLUSIONS AND CLINICAL IMPLICATIONS For optimal mRCC treatment selection, clinicians must carefully balance efficacy, toxicity, and patient preferences, especially when transitioning between first- and second-line therapies, to provide individualized care.
Collapse
Affiliation(s)
| | - Eddy Saad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Renee-Maria Saliby
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maxine Sun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Axel Bex
- Specialist Centre for Kidney Cancer, Royal Free NHS Foundation Trust, University College London Division of Surgery and Interventional Science, London, UK
| | - Daniel Heng
- Department of Oncology, Tom Baker Cancer Centre, Calgary, Canada
| | - Arnaud Mejean
- Department of Urology, Hôpital Européen Georges Pompidou, Paris, France
| | - Robert J Motzer
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth R Plimack
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Thomas Powles
- Barts Health NHS Trust and the Royal Free NHS Foundation Trust, Barts Cancer Institute, London, UK; Queen Mary University of London, London, UK
| | - Brian I Rini
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Tian Zhang
- Division of Hematology and Oncology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
22
|
Villatore A, Bosi C, Pomaranzi C, Cigliola A, Tateo V, Mercinelli C, Vignale D, Rizzo S, Necchi A, Peretto G. Myocarditis Following Pembrolizumab Plus Axitinib, and Belzutifan Plus Lenvatinib for Renal Cell Carcinoma: A Case Report. Cardiovasc Toxicol 2024; 24:1168-1173. [PMID: 39085529 DOI: 10.1007/s12012-024-09906-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Cardiac toxicity is an adverse event of several classes of anti-cancer drugs. Herein, we present the case of a 52-year-old woman with metastatic renal cell carcinoma (RCC), previously treated with debulking surgery, pembrolizumab (immune checkpoint inhibitor) in combination with axitinib (tyrosine kinase inhibitor (TKI)), followed by lenvatinib (TKI) and belzutifan (HIF-2α inhibitor), who developed myocarditis proven by cardiac magnetic resonance and endomyocardial biopsy. The case was notable for reporting a not-yet described adverse event during treatment with belzutifan plus lenvatinib, the etiology of which was of unobvious determination given the pre-exposure to pembrolizumab, a known cause of drug-related myocarditis. We surmise that myocarditis was a delayed adverse event related to pembrolizumab (8 months after treatment interruption), although we emphasize that only attentive monitoring of cardiac adverse events of patients exposed to belzutifan and lenvatinib in the context of large clinical trials may rule out any causal implication of these drugs.
Collapse
Affiliation(s)
- Andrea Villatore
- Vita-Salute San Raffaele University, Milan, Italy.
- Disease Unit for Myocarditis & Arrhythmogenic Cardiomyopathies, IRCCS San Raffaele Hospital, Milan, Italy.
| | - Carlo Bosi
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Antonio Cigliola
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Valentina Tateo
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Chiara Mercinelli
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Davide Vignale
- Experimental Imaging Center, Radiology Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Stefania Rizzo
- Cardiovascular Pathology Unit, Azienda Ospedaliera Di Padova, Padua, Italy
| | - Andrea Necchi
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Giovanni Peretto
- Vita-Salute San Raffaele University, Milan, Italy
- Disease Unit for Myocarditis & Arrhythmogenic Cardiomyopathies, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
23
|
Wiley HE, Srinivasan R, Maranchie JK, Chhablani J, Iversen ABB, Kruse A, Jonasch E, Gombos DS, Else T, Demirci H, Maughan BL, Hartnett ME, Coleman HR, Fu W, Perini RF, Liu Y, Linehan WM, Chew EY. Oral Hypoxia-Inducible Factor 2α Inhibitor Belzutifan in Ocular von Hippel-Lindau Disease: Subgroup Analysis of the Single-Arm Phase 2 LITESPARK-004 Study. Ophthalmology 2024; 131:1324-1332. [PMID: 38849055 DOI: 10.1016/j.ophtha.2024.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/09/2024] Open
Abstract
PURPOSE To report the efficacy of the oral hypoxia-inducible factor 2α inhibitor belzutifan in participants with von Hippel-Lindau disease-associated retinal hemangioblastomas in the LITESPARK-004 study. DESIGN Subgroup analysis of the phase 2, single-arm, open-label LITESPARK-004 study. PARTICIPANTS Adults with 1 or more von Hippel-Lindau disease-associated measurable renal cell carcinoma tumors not requiring immediate surgical intervention were eligible. METHODS Participants received oral belzutifan 120 mg once daily until disease progression or unacceptable treatment-related toxicity. MAIN OUTCOME MEASURES Efficacy of belzutifan in retinal hemangioblastomas was a secondary end point, measured as response (improved, stable, or progressed) by independent reading center-certified graders based on color fundus imaging performed every 12 weeks using the investigator's preferred imaging standards. Additional assessments, where available, included OCT and ultra-widefield fluorescein angiography. RESULTS Among 61 participants in LITESPARK-004, 12 had 1 or more evaluable active retinal hemangioblastomas in 16 eyes at baseline per independent reading center. As of April 1, 2022, the median follow-up for participants with ocular von Hippel-Lindau disease at baseline was 37.3 months. All 16 eyes were graded as improved, with a response rate of 100.0% (95% confidence interval, 79.4%-100%). No new retinal hemangioblastomas or ocular disease progression were reported as of data cutoff date. Eight participants underwent additional multimodal eye assessments performed at the National Institutes of Health study site. Among this subgroup, 10 of 24 hemangioblastomas in 8 eyes of 6 participants measured 500 μm or more in greatest linear dimension at baseline and were analyzed further. All 10 hemangioblastomas had a mean area reduction of 15% or more by month 12 and of 30% or more by month 24. CONCLUSIONS Belzutifan showed promising activity against ocular von Hippel-Lindau disease, including capacity to control retinal hemangioblastomas, with effects sustained for more than 2 years while treatment is ongoing. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Henry E Wiley
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Ramaprasad Srinivasan
- Molecular Cancer Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jodi K Maranchie
- Department of Urology, University of Pittsburgh/UPMC, Pittsburgh, Pennsylvania
| | - Jay Chhablani
- Department of Ophthalmology, University of Pittsburgh/UPMC, Pittsburgh, Pennsylvania
| | | | - Anders Kruse
- Department of Ophthalmology, Aalborg University Hospital, Aalborg, Denmark
| | - Eric Jonasch
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dan S Gombos
- Section of Ophthalmology, Department of Head & Neck Surgery, Division of Surgery, MD Anderson Cancer Center, Houston, Texas
| | - Tobias Else
- Department of Internal Medicine, MEND, Division of Genetic Medicine, University of Michigan, Ann Arbor, Michigan
| | - Hakan Demirci
- Department of Ophthalmology, University of Michigan, Ann Arbor, Michigan
| | - Benjamin L Maughan
- Division of Medical Oncology at Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - M Elizabeth Hartnett
- Department of Ophthalmology, University of Utah, Salt Lake City, Utah; Department of Ophthalmology, Byers Eye Institute at Stanford University, Palo Alto, California
| | - Hanna R Coleman
- VOIANT (Independent Reading Center), Boston, Massachusetts; Department of Ophthalmology, Columbia University, New York, NY
| | - Wei Fu
- Merck & Co., Inc., Rahway, New Jersey
| | | | | | - W Marston Linehan
- Urologic Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - Emily Y Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
24
|
Thapa B, Shreenivas A, Bylow K, Chen HZ, George B, Kurzrock R. Successful Targeting of Somatic VHL Alterations With Belzutifan in Two Cases. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2024; 7:308-313. [PMID: 39524464 PMCID: PMC11541926 DOI: 10.36401/jipo-24-13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 11/16/2024]
Abstract
Clear cell renal cell carcinoma (RCC) is commonly associated with alterations in the VHL tumor suppressor gene, resulting in upregulation of hypoxia-inducible factor pathways. Immune checkpoint inhibitors and vascular endothelial growth factor inhibitors are the mainstays of systemic treatment for metastatic RCC; however, most patients encounter disease progression after the initial response. The phase 3 clinical trial LITESPARK-005-belzutifan (HIF-2α inhibitor) demonstrated improvement in progression-free survival compared with everolimus in heavily pretreated patients unselected for somatic/germline VHL alterations (an objective response rate of 23% and a median time on therapy of 7.6 months in the belzutifan cohort), resulting in U.S. FDA approval for patients with advanced RCC. Herein, we present two cases of refractory metastatic RCC (including one with brain metastases) with somatic VHL mutations who received belzutifan after discussion in the institutional Molecular Tumor Board. Both patients had an excellent clinical response (partial remissions ongoing at >12 and >20 months). Future studies should assess the merits of biomarker selection for belzutifan treatment.
Collapse
Affiliation(s)
- Bicky Thapa
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Aditya Shreenivas
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kathryn Bylow
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hui-Zi Chen
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ben George
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
- LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Razelle Kurzrock
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
- MCW Cancer Center and Mellowes Center for Genomic Sciences and Precision Medicine Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
25
|
Slingo ME. Oxygen-sensing pathways and the pulmonary circulation. J Physiol 2024; 602:5619-5629. [PMID: 37843154 DOI: 10.1113/jp284591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/29/2023] [Indexed: 10/17/2023] Open
Abstract
The unique property of the pulmonary circulation to constrict in response to hypoxia, rather than dilate, brings advantages in both health and disease. Hypoxic pulmonary vasoconstriction (HPV) acts to optimise ventilation-perfusion matching - this is important clinically both in focal disease (such as pneumonia) and in one-lung ventilation during anaesthesia for thoracic surgery. However, during global hypoxia such as that encountered at high altitude, generalised pulmonary vasoconstriction can lead to pulmonary hypertension. There is now a growing body of evidence that links the hypoxia-inducible factor (HIF) pathway and pulmonary vascular tone - in both acute and chronic settings. Genetic and pharmacological alterations to all key components of this pathway (VHL - von Hippel-Lindau ubiquitin E3 ligase; PHD2 - prolyl hydroxylase domain protein 2; HIF1 and HIF2) have clear effects on the pulmonary circulation, particularly in hypoxia. Furthermore, knowledge of the molecular biology of the prolyl hydroxylase enzymes has led to an extensive and ongoing body of research into the importance of iron in both HPV and pulmonary hypertension. This review will explore these relationships in more detail and discuss future avenues of research.
Collapse
Affiliation(s)
- Mary E Slingo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
26
|
Bartram MP, Beck BB, Müller RU. [Genetics in nephrology - any news?]. Dtsch Med Wochenschr 2024; 149:1361-1366. [PMID: 39437829 DOI: 10.1055/a-2198-0363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
While genetic kidney diseases were long regarded as a rare cause of kidney failure, it has been shown in recent years that they account for a relevant proportion of cases. In cohorts of kidney transplant recipients, a monogenic cause is found in up to 30% of cases. Identifying the genetic cause of kidney disease has become much easier thanks to technological advances in DNA sequencing. The focus has now shifted to understanding the significance of the findings and identifying diagnostic gaps. It is still not possible to clarify all CKD cases of unclear aetiology. Besides very effective generic treatments for monogenic kidney disease (e.g., ACE-inhibitor use in Alport Syndrome), increasing knowledge of the pathophysiology of genetic kidney diseases has led to a growing number of targeted therapies. These include the treatment of ADPKD with Tolvaptan, which has now been in use for 10 years. Recently, exciting, and completely new approaches have been added, such as the first siRNA therapies in nephrology for primary hyperoxaluria type 1, the targeted treatment of hyperphagia in Bardet-Biedl syndrome, the therapy of APOL1-associated kidney disease or the use of the HIF-2 antagonist Belzutifan for renal cell carcinoma associated with Von-Hippel-Lindau syndrome. The new possibilities in the treatment of patients with genetic kidney diseases have also clearly revealed deficits in current patient care. Centers of excellence with extensive experience in this area therefore play an important role in improving care. This also applies to the further training of colleagues in the field. In Germany, the National Action Alliance for People with Rare Diseases (NAMSE) and the nationwide establishment of - to date - 36 centers for rare diseases play an important role in this regard.
Collapse
Affiliation(s)
- Malte P Bartram
- Klinik II für Innere Medizin - Nephrologie, Rheumatologie, Diabetologie und Allgemeine Innere Medizin, Uniklinik Köln, Köln, Deutschland
| | - Bodo B Beck
- Institut für Humangenetik, Uniklinik Köln, Köln, Deutschland
| | - Roman-Ulrich Müller
- Klinik II für Innere Medizin - Nephrologie, Rheumatologie, Diabetologie und Allgemeine Innere Medizin, Uniklinik Köln, Köln, Deutschland
| |
Collapse
|
27
|
Bigot P, Boissier R, Khene ZE, Albigès L, Bernhard JC, Correas JM, De Vergie S, Doumerc N, Ferragu M, Ingels A, Margue G, Ouzaïd I, Pettenati C, Rioux-Leclercq N, Sargos P, Waeckel T, Barthelemy P, Rouprêt M. French AFU Cancer Committee Guidelines - Update 2024-2026: Management of kidney cancer. THE FRENCH JOURNAL OF UROLOGY 2024; 34:102735. [PMID: 39581661 DOI: 10.1016/j.fjurol.2024.102735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVE To update the French recommendations for the management of kidney cancer. METHODS A systematic review of the literature was conducted for the period from 2014 to 2024. The most relevant articles concerning the diagnosis, classification, surgical treatment, medical treatment, and follow-up of kidney cancer were selected and incorporated into the recommendations. The recommendations have been updated specifying the level of evidence (strong or weak). RESULTS Kidney cancer following prolonged occupational exposure to trichloroethylene should be considered an occupational disease. The reference examination for the diagnosis and staging of kidney cancer is the contrast-enhanced thoraco-abdominal CT scan. PET scans are not indicated in the staging of kidney cancer. Percutaneous biopsy is recommended in situations where its results will influence therapeutic decisions. It should be used to reduce the number of surgeries for benign tumors, particularly avoiding unnecessary radical nephrectomies. Kidney tumors should be classified according to the pTNM 2017 classification, the WHO 2022 classification, and the ISUP nucleolar grade. Metastatic kidney cancers should be classified according to IMDC criteria. Surveillance of tumors smaller than 2cm should be prioritized and can be offered regardless of patient age. Robot-assisted laparoscopic partial nephrectomy is the reference surgical treatment for T1 tumors. Ablative therapies and surveillance are options for elderly patients with comorbidities for tumors larger than 2cm. Stereotactic radiotherapy is an option to discuss for treating localized kidney tumors in patients not eligible for other treatments. Radical nephrectomy is the first-line treatment for locally advanced localized cancers. Pembrolizumab is recommended for patients at high risk of recurrence after surgery for localized kidney cancer. In metastatic patients, cytoreductive nephrectomy can be immediate in cases of good prognosis, delayed in cases of intermediate or poor prognosis for patients stabilized by medical treatment, or as "consolidation" in patients with complete or major partial response at metastatic sites after systemic treatment. Surgical or local treatment of metastases can be proposed for single lesions or oligometastases. Recommended first-line drugs for metastatic clear cell renal carcinoma are combinations of axitinib/pembrolizumab, nivolumab/ipilimumab, nivolumab/cabozantinib, and lenvatinib/pembrolizumab. Patients with non-clear cell metastatic kidney cancer should be presented to the CARARE Network and prioritized for inclusion in clinical trials. CONCLUSION These updated recommendations are a reference that will enable French and French-speaking practitioners to optimize their management of kidney cancer.
Collapse
Affiliation(s)
- Pierre Bigot
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Angers University Hospital, Angers, France.
| | - Romain Boissier
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology and Kidney Transplantation, Conception University Hospital, Aix-Marseille University, AP-HM, Marseille, France
| | - Zine-Eddine Khene
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Rennes University Hospital, Rennes, France
| | - Laurence Albigès
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Cancer Medicine, Gustave-Roussy, Paris-Saclay University, Villejuif, France
| | - Jean-Christophe Bernhard
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Hôpital Pellegrin, Bordeaux University Hospital, Bordeaux, France
| | - Jean-Michel Correas
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Adult Radiology, Hôpital Necker, University of Paris, AP-HP Centre, Paris, France
| | - Stéphane De Vergie
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Nantes University Hospital, Nantes, France
| | - Nicolas Doumerc
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology and Renal Transplantation, Toulouse University Hospital, Toulouse, France
| | - Matthieu Ferragu
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Angers University Hospital, Angers, France
| | - Alexandre Ingels
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, UPEC, Hôpital Henri-Mondor, Créteil, France
| | - Gaëlle Margue
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Hôpital Pellegrin, Bordeaux University Hospital, Bordeaux, France
| | - Idir Ouzaïd
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Bichat University Hospital, AP-HP, Paris, France
| | - Caroline Pettenati
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Hôpital Foch, University of Versailles - Saint-Quentin-en-Yvelines, 40, rue Worth, 92150 Suresnes, France
| | - Nathalie Rioux-Leclercq
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Pathology, Rennes University Hospital, Rennes, France
| | - Paul Sargos
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Radiotherapy, Hôpital Pellegrin, Bordeaux University Hospital, Bordeaux, France
| | - Thibaut Waeckel
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Caen University Hospital, Caen, France
| | - Philippe Barthelemy
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Medical Oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Morgan Rouprêt
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Urology, Hôpital Pitié-Salpêtrière, Predictive Onco-Urology, GRC 5, Sorbonne University, AP-HP, 75013 Paris, France
| |
Collapse
|
28
|
Liao C, Hu L, Zhang Q. Von Hippel-Lindau protein signalling in clear cell renal cell carcinoma. Nat Rev Urol 2024; 21:662-675. [PMID: 38698165 DOI: 10.1038/s41585-024-00876-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/05/2024]
Abstract
The distinct pathological and molecular features of kidney cancer in adaptation to oxygen homeostasis render this malignancy an attractive model for investigating hypoxia signalling and potentially developing potent targeted therapies. Hypoxia signalling has a pivotal role in kidney cancer, particularly within the most prevalent subtype, known as renal cell carcinoma (RCC). Hypoxia promotes various crucial pathological processes, such as hypoxia-inducible factor (HIF) activation, angiogenesis, proliferation, metabolic reprogramming and drug resistance, all of which contribute to kidney cancer development, growth or metastasis formation. A substantial portion of kidney cancers, in particular clear cell RCC (ccRCC), are characterized by a loss of function of Von Hippel-Lindau tumour suppressor (VHL), leading to the accumulation of HIF proteins, especially HIF2α, a crucial driver of ccRCC. Thus, therapeutic strategies targeting pVHL-HIF signalling have been explored in ccRCC, culminating in the successful development of HIF2α-specific antagonists such as belzutifan (PT2977), an FDA-approved drug to treat VHL-associated diseases including advanced-stage ccRCC. An increased understanding of hypoxia signalling in kidney cancer came from the discovery of novel VHL protein (pVHL) targets, and mechanisms of synthetic lethality with VHL mutations. These breakthroughs can pave the way for the development of innovative and potent combination therapies in kidney cancer.
Collapse
Affiliation(s)
- Chengheng Liao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lianxin Hu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Qing Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
29
|
Silvestrini ML, Solazzo R, Boral S, Cocco MJ, Closson JD, Masetti M, Gardner KH, Chong LT. Gating residues govern ligand unbinding kinetics from the buried cavity in HIF-2α PAS-B. Protein Sci 2024; 33:e5198. [PMID: 39467204 PMCID: PMC11516114 DOI: 10.1002/pro.5198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024]
Abstract
While transcription factors have been generally perceived as "undruggable," an exception is the HIF-2 hypoxia-inducible transcription factor, which contains an internal cavity that is sufficiently large to accommodate a range of small-molecules, including the therapeutically used inhibitor belzutifan. Given the relatively long ligand residence times of these small molecules and the lack of any experimentally observed pathway connecting the cavity to solvent, there has been great interest in understanding how these drug ligands exit the buried receptor cavity. Here, we focus on the relevant PAS-B domain of hypoxia-inducible factor 2α (HIF-2α) and examine how one such small molecule (THS-017) exits from the buried cavity within this domain on the seconds-timescale using atomistic simulations and ZZ-exchange NMR. To enable the simulations, we applied the weighted ensemble path sampling strategy, which generates continuous pathways for a rare-event process [e.g., ligand (un)binding] with rigorous kinetics in orders of magnitude less computing time compared to conventional simulations. Results reveal the formation of an encounter complex intermediate and two distinct classes of pathways for ligand exit. Based on these pathways, we identified two pairs of conformational gating residues in the receptor: one for the major class (N288 and S304) and another for the minor class (L272 and M309). ZZ-exchange NMR validated the kinetic importance of N288 for ligand unbinding. Our results provide an ideal simulation dataset for rational manipulation of ligand unbinding kinetics.
Collapse
Affiliation(s)
| | - Riccardo Solazzo
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐Università di BolognaBolognaItaly
| | - Soumendu Boral
- Structural Biology InitiativeCUNY Advanced Science Research CenterNew YorkNew YorkUSA
| | - Melanie J. Cocco
- Department of Pharmaceutical SciencesUniversity of California, IrvineIrvineCaliforniaUSA
- Department of Molecular Biology and BiochemistryUniversity of California, IrvineIrvineCaliforniaUSA
| | - Joseph D. Closson
- Structural Biology InitiativeCUNY Advanced Science Research CenterNew YorkNew YorkUSA
- PhD Program in BiochemistryCUNY Graduate CenterNew YorkNew YorkUSA
| | - Matteo Masetti
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐Università di BolognaBolognaItaly
| | - Kevin H. Gardner
- Structural Biology InitiativeCUNY Advanced Science Research CenterNew YorkNew YorkUSA
- Department of Chemistry and BiochemistryCity College of New YorkNew YorkNew YorkUSA
- PhD Programs in Biochemistry, Biology, and ChemistryCUNY Graduate CenterNew YorkNew YorkUSA
| | - Lillian T. Chong
- Department of ChemistryUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
30
|
Alkaissi H, Nazari MA, Hadrava Vanova K, Uher O, Gordon CM, Talvacchio S, Diachenko N, Mukvich O, Wang H, Glod J, Zhuang Z, Pacak K. Rapid Cardiovascular Response to Belzutifan in HIF2A-Mediated Paraganglioma. N Engl J Med 2024; 391:1552-1555. [PMID: 39442048 DOI: 10.1056/nejmc2409427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Affiliation(s)
- Hussam Alkaissi
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | | | | | | | | | | | - Nina Diachenko
- Institute of Pediatrics, Obstetrics, and Gynecology, Kyiv, Ukraine
| | - Olena Mukvich
- Institute of Pediatrics, Obstetrics, and Gynecology, Kyiv, Ukraine
| | | | - John Glod
- National Cancer Institute, Bethesda, MD
| | | | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| |
Collapse
|
31
|
Ahmad S, Muhlebner A, Snijders TJ, de Leng WW, Seute T, van Leeuwaarde RS. Somatostatin receptor 2A expression in von Hippel-Lindau-related hemangioblastomas. Cancer 2024; 130:3473-3479. [PMID: 38824656 DOI: 10.1002/cncr.35418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/01/2024] [Accepted: 05/03/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Central nervous system hemangioblastomas are the most prevalent manifestation of von Hippel-Lindau (VHL) disease and remain the main cause of mortality. Surgical resection is the primary treatment strategy, but is not always possible, and should be used as restrictively as possible. There is an unmet need for less invasive treatment strategies, such as targeted therapy. Expression of somatostatin receptor 2A (SSTR2A) in VHL-related hemangioblastomas has been described earlier, but the extent of expression in a larger population has yet to be determined. The authors hypothesize that a substantial subset of VHL-related hemangioblastomas show SSTR2A expression, which may serve as a potential new treatment target. METHODS Patients who were surgically treated for a VHL-related hemangioblastoma from 1990 until 2021 at the UMC Utrecht were included. Clinical data was derived from a clinical database. Tissue samples were histopathologically examined with use of hematoxylin and eosin staining, and immunohistochemical analysis of SSTR2A expression was performed. RESULTS Forty-three tissue samples were obtained from 26 patients. Nine showed strong positivity for SSTR2A expression, whereas 13 showed moderate and 15 sparse expression. Three samples showed no expression of SSTR2A. The distribution showed right-skewedness favoring a strong expression. SSTR2A expression colocalized with endothelial markers and not with stromal cells. Additionally, within-patient variability for SSTR2A expression was described in 14 patients. CONCLUSION SSTR2A is expressed in varying degrees in the majority of VHL-related hemangioblastomas. Future treatment with somatostatin analogues or even peptide receptor radionuclide treatment may be considered for SSTR2A-positive cases.
Collapse
Affiliation(s)
- Saya Ahmad
- Department of Endocrine Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Angelika Muhlebner
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Tom J Snijders
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wendy W de Leng
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Tatjana Seute
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Rachel S van Leeuwaarde
- Department of Endocrine Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
32
|
Tu R, Ma J, Chen Y, Kang Y, Ren D, Cai Z, Zhang R, Pan Y, Liu Y, Da Y, Xu Y, Yu Y, Wang D, Wang J, Dong Y, Lu X, Zhang C. USP7 depletion potentiates HIF2α degradation and inhibits clear cell renal cell carcinoma progression. Cell Death Dis 2024; 15:749. [PMID: 39406703 PMCID: PMC11482519 DOI: 10.1038/s41419-024-07136-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by Von Hippel Lindau (VHL) gene loss of function mutation, which leads to the accumulation of hypoxia-inducible factor 2α (HIF2α). HIF2α has been well-established as one of the major oncogenic drivers of ccRCC, however, its therapeutic targeting remains a challenge. Through an analysis of proteomic data from ccRCCs and adjacent non-tumor tissues, we herein revealed that Ubiquitin-Specific Peptidase 7 (USP7) was upregulated in tumor tissues, and its depletion by inhibitors or shRNAs caused significant suppression of tumor progression in vitro and in vivo. Mechanistically, USP7 expression is activated by the transcription factors FUBP1 and FUBP3, and it promotes tumor progression mainly by deubiquitinating and stabilizing HIF2α. Moreover, the combination of USP7 inhibitors and afatinib (an ERBB family inhibitor) coordinately induce cell death and tumor suppression. In mechanism, afatinib indirectly inhibits USP7 transcription and accelerates the degradation of HIF2α protein, and the combination of them caused a more profound suppression of HIF2α abundance. These findings reveal a FUBPs-USP7-HIF2α regulatory axis that underlies the progression of ccRCC and provides a rationale for therapeutic targeting of oncogenic HIF2α via combinational treatment of USP7 inhibitor and afatinib.
Collapse
Affiliation(s)
- Rongfu Tu
- The First Affiliated Hospital of Xi'an Jiaotong University, Center for Precision Cancer Medicine, MED-X Institute, 710000, Xi'an, China.
| | - Junpeng Ma
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Center for Molecular Diagnosis and Precision Medicine, 1519 Dongyue Dadao, 330209, Nanchang, China
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Department of Clinical Laboratory, 1519 Dongyue Dadao, 330209, Nanchang, China
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Provincial Center for Advanced Diagnostic Technology and Precision Medicine, 1519 DongYue Dadao, 330209, Nanchang, China
| | - Yule Chen
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, 710000, Xi'an, China
| | - Ye Kang
- The First Affiliated Hospital of Xi'an Jiaotong University, Center for Precision Cancer Medicine, MED-X Institute, 710000, Xi'an, China
| | - Doudou Ren
- The First Affiliated Hospital of Xi'an Jiaotong University, Center for Precision Cancer Medicine, MED-X Institute, 710000, Xi'an, China
| | - Zeqiong Cai
- The First Affiliated Hospital of Xi'an Jiaotong University, Center for Precision Cancer Medicine, MED-X Institute, 710000, Xi'an, China
| | - Ru Zhang
- The First Affiliated Hospital of Xi'an Jiaotong University, Center for Precision Cancer Medicine, MED-X Institute, 710000, Xi'an, China
| | - Yiwen Pan
- The First Affiliated Hospital of Xi'an Jiaotong University, Precision Medicine Center, 710000, Xi'an, China
| | - Yijia Liu
- The First Affiliated Hospital of Xi'an Jiaotong University, Precision Medicine Center, 710000, Xi'an, China
| | - Yanyan Da
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Center for Molecular Diagnosis and Precision Medicine, 1519 Dongyue Dadao, 330209, Nanchang, China
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Department of Clinical Laboratory, 1519 Dongyue Dadao, 330209, Nanchang, China
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Provincial Center for Advanced Diagnostic Technology and Precision Medicine, 1519 DongYue Dadao, 330209, Nanchang, China
| | - Yao Xu
- The First Affiliated Hospital of Xi'an Jiaotong University, Center for Precision Cancer Medicine, MED-X Institute, 710000, Xi'an, China
| | - Yahuan Yu
- Department of Nephrology, The Affiliated Hospital of Qingdao University, 266100, Qingdao, China
| | - Donghai Wang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, 430071, Wuhan, China
| | - Jingchao Wang
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, Shenzhen University School of Medicine, 518055, Shenzhen, China
| | - Yang Dong
- Department of Pathology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China
| | - Xinlan Lu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, 710000, Xi'an, China
| | - Chengsheng Zhang
- The First Affiliated Hospital of Xi'an Jiaotong University, Center for Precision Cancer Medicine, MED-X Institute, 710000, Xi'an, China.
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Center for Molecular Diagnosis and Precision Medicine, 1519 Dongyue Dadao, 330209, Nanchang, China.
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Department of Clinical Laboratory, 1519 Dongyue Dadao, 330209, Nanchang, China.
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Provincial Center for Advanced Diagnostic Technology and Precision Medicine, 1519 DongYue Dadao, 330209, Nanchang, China.
- Department of Medical Genetics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 DongYue Dadao, 330209, Nanchang, China.
| |
Collapse
|
33
|
Maluchenko A, Maksimov D, Antysheva Z, Krupinova J, Avsievich E, Glazova O, Bodunova N, Karnaukhov N, Feidorov I, Salimgereeva D, Voloshin M, Volchkov P. Molecular Basis of Pancreatic Neuroendocrine Tumors. Int J Mol Sci 2024; 25:11017. [PMID: 39456803 PMCID: PMC11507569 DOI: 10.3390/ijms252011017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic neuroendocrine tumors (NETs) are rare well-differentiated neoplasms with limited therapeutic options and unknown cells of origin. The current classification of pancreatic neuroendocrine tumors is based on proliferative grading, and guides therapeutic strategies, however, tumors within grades exhibit profound heterogeneity in clinical manifestation and outcome. Manifold studies have highlighted intra-patient differences in tumors at the genetic and transcriptomic levels. Molecular classification might become an alternative or complementary basis for treatment decisions and reflect tumor biology, actionable cellular processes. Here, we provide a comprehensive review of genomic, transcriptomic, proteomic and epigenomic studies of pancreatic NETs to elucidate patterns shared between proposed subtypes that could form a foundation for new classification. We denote four NET subtypes with distinct molecular features, which were consistently reproduced using various omics technologies.
Collapse
Affiliation(s)
- Alesia Maluchenko
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
| | - Denis Maksimov
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
| | - Zoia Antysheva
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
| | - Julia Krupinova
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Ekaterina Avsievich
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Olga Glazova
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Natalia Bodunova
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Nikolay Karnaukhov
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Ilia Feidorov
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Diana Salimgereeva
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Mark Voloshin
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Pavel Volchkov
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| |
Collapse
|
34
|
Choi SH, Chen YW, Panian J, Yuen K, McKay RR. Emerging innovative treatment strategies for advanced clear cell renal cell carcinoma. Oncologist 2024:oyae276. [PMID: 39401004 DOI: 10.1093/oncolo/oyae276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Dramatic advances in biological discoveries, since the 1990s, have continued to reshape the treatment paradigm of metastatic renal cell carcinoma (RCC). Von Hippel Lindau (VHL) gene alterations are associated with pro-angiogenic activity and are central to the pathogenesis of clear cell RCC (ccRCC), the most predominant histologic subtype of RCC. Antiangiogenic strategies revolving around this VHL/HIF/VEGF axis have been shown to improve survival in metastatic ccRCC. The discovery of immune checkpoints and agents that target their inhibition introduced a new treatment paradigm for patients with RCC. While initially approved as monotherapy, studies investigating immune checkpoint inhibitor combinations have led to their approval as the new standard of care, providing durable responses and unprecedented improvements in clinical outcome. Despite these advances, the projected 14 390 deaths in 2024 from RCC underscore the need to continue efforts in expanding and optimizing treatment options for patients with metastatic RCC. This article reviews key findings that have transformed the way we understand and treat metastatic RCC, in addition to highlighting novel treatment strategies that are currently under development.
Collapse
Affiliation(s)
- Sharon H Choi
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Yu-Wei Chen
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Justine Panian
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Kit Yuen
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Rana R McKay
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
- Department of Urology, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
35
|
Stransky LA, Gao W, Schmidt LS, Bi K, Ricketts CJ, Ramesh V, James A, Difilippantonio S, Ileva L, Kalen JD, Karim B, Jeon A, Morgan T, Warner AC, Turan S, Unite J, Tran B, Choudhari S, Zhao Y, Linn DE, Yun C, Dhandapani S, Parab V, Pinheiro EM, Morris N, He L, Vigeant SM, Pignon JC, Sticco-Ivins M, Signoretti S, Van Allen EM, Linehan WM, Kaelin WG. Toward a CRISPR-based mouse model of Vhl-deficient clear cell kidney cancer: Initial experience and lessons learned. Proc Natl Acad Sci U S A 2024; 121:e2408549121. [PMID: 39365820 PMCID: PMC11474080 DOI: 10.1073/pnas.2408549121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 10/06/2024] Open
Abstract
CRISPR is revolutionizing the ability to do somatic gene editing in mice for the purpose of creating new cancer models. Inactivation of the VHL tumor suppressor gene is the signature initiating event in the most common form of kidney cancer, clear cell renal cell carcinoma (ccRCC). Such tumors are usually driven by the excessive HIF2 activity that arises when the VHL gene product, pVHL, is defective. Given the pressing need for a robust immunocompetent mouse model of human ccRCC, we directly injected adenovirus-associated viruses (AAVs) encoding sgRNAs against VHL and other known/suspected ccRCC tumor suppressor genes into the kidneys of C57BL/6 mice under conditions where Cas9 was under the control of one of two different kidney-specific promoters (Cdh16 or Pax8) to induce kidney tumors. An AAV targeting Vhl, Pbrm1, Keap1, and Tsc1 reproducibly caused macroscopic ccRCCs that partially resembled human ccRCC tumors with respect to transcriptome and cell of origin and responded to a ccRCC standard-of-care agent, axitinib. Unfortunately, these tumors, like those produced by earlier genetically engineered mouse ccRCCs, are HIF2 independent.
Collapse
Affiliation(s)
- Laura A. Stransky
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Wenhua Gao
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Laura S. Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD20892
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Kevin Bi
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02115
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA02115
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
| | - Christopher J. Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD20892
| | - Vijyendra Ramesh
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Amy James
- Animal Research Technical Support, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Simone Difilippantonio
- Animal Research Technical Support, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Lilia Ileva
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Joseph D. Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Albert Jeon
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Tamara Morgan
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Andrew C. Warner
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Sevilay Turan
- National Cancer Institute Center for Cancer Research, Sequencing Facility, Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Joanne Unite
- National Cancer Institute Center for Cancer Research, Sequencing Facility, Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Bao Tran
- National Cancer Institute Center for Cancer Research, Sequencing Facility, Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Sulbha Choudhari
- Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD21701
| | - Yongmei Zhao
- Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD21701
| | | | - Changhong Yun
- Pharmacokinetics, Merck & Co., Inc., Boston, MA02115
| | | | - Vaishali Parab
- Pharmacokinetics, Merck & Co., Inc., South San Francisco, CA94080
| | | | - Nicole Morris
- Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Lixia He
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Sean M. Vigeant
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Jean-Christophe Pignon
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women's Hospital, Boston, MA02115
| | - Maura Sticco-Ivins
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women's Hospital, Boston, MA02115
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA02115
| | - Sabina Signoretti
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women's Hospital, Boston, MA02115
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA02115
| | - Eliezer M. Van Allen
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02115
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA02115
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
| | - W. Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD20892
| | - William G. Kaelin
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
- HHMI, Chevy Chase, MD20815
| |
Collapse
|
36
|
Wang L, Zhang L, Zhang Z, Wu P, Zhang Y, Chen X. Advances in targeting tumor microenvironment for immunotherapy. Front Immunol 2024; 15:1472772. [PMID: 39421736 PMCID: PMC11484021 DOI: 10.3389/fimmu.2024.1472772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
The tumor microenvironment (TME) provides essential conditions for the occurrence, invasion, and spread of cancer cells. Initial research has uncovered immunosuppressive properties of the TME, which include low oxygen levels (hypoxia), acidic conditions (low pH), increased interstitial pressure, heightened permeability of tumor vasculature, and an inflammatory microenvironment. The presence of various immunosuppressive components leads to immune evasion and affects immunotherapy efficacy. This indicates the potential value of targeting the TME in cancer immunotherapy. Therefore, TME remodeling has become an effective method for enhancing host immune responses against tumors. In this study, we elaborate on the characteristics and composition of the TME and how it weakens immune surveillance and summarize targeted therapeutic strategies for regulating the TME.
Collapse
Affiliation(s)
- Lugang Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liubo Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Wu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
37
|
Kreatsoulas DC, Lonser RR. Spinal cord hemangioblastomas in von Hippel-Lindau disease. Neurooncol Adv 2024; 6:iii66-iii72. [PMID: 39430395 PMCID: PMC11485647 DOI: 10.1093/noajnl/vdad153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024] Open
Abstract
Background von Hippel-Lindau disease (VHL) is an autosomal dominant familial neoplasia syndrome. The most common manifestation of VHL is central nervous system hemangioblastomas. VHL patients will often develop multiple hemangioblastomas along their craniospinal axis over their lifetime. Spinal cord hemangioblastomas account for nearly half of all nervous system hemangioblastomas in VHL. Methods The authors conducted a literature review and summation of available articles on spinal cord hemangioblastomas associated with VHL. Results The embryological origins, epidemiology, natural history, surgical outcomes, nonsurgical treatments, and future directions in spinal cord hemangioblastomas are discussed. Conclusions Hemangioblastomas in VHL are optimally managed with a multidisciplinary approach that includes surgical resection of symptomatic lesions. Novel treatments are gaining traction, but must be studied further for efficacy and safety.
Collapse
Affiliation(s)
- Daniel C Kreatsoulas
- Department of Neurological Surgery, Ohio State University Wexner Medical Center, Ohio State University, Columbus, Ohio, USA
| | - Russell R Lonser
- Department of Neurological Surgery, Ohio State University Wexner Medical Center, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
38
|
Palavani LB, Camerotte R, Vieira Nogueira B, Ferreira MY, Oliveira LB, Pari Mitre L, Coelho Nogueira de Castro W, Canto Gomes GL, Fabrini Paleare LF, Batista S, Fim Andreão F, Bertani R, Dias Polverini A. Innovative solutions? Belzutifan therapy for hemangioblastomas in Von Hippel-Lindau disease: A systematic review and single-arm meta-analysis. J Clin Neurosci 2024; 128:110774. [PMID: 39128437 DOI: 10.1016/j.jocn.2024.110774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/31/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Von Hippel-Lindau (VHL) disease is a rare autosomal dominant disorder that predisposes patients to develop multiple cysts and tumors, such as hemangioblastomas (HBs) and clear cell renal cell carcinoma (ccRCC), due to mutations in the VHL tumor suppressor gene. While treatment of HBs varies based on their characteristics and has improved patient survival, it still involves high morbidity and mortality, leading to ongoing debates and studies to refine therapy strategies. Recent developments include the emergence of Belzutifan, a novel inhibitor targeting hypoxia-inducible factor 2α (HIF-2α), which has shown promising results in ongoing trials, particularly for patients not immediately requiring surgery. METHODS This systematic review and meta-analysis aimed to comprehensively evaluate the efficacy and safety of Belzutifan for treating HBs associated with VHL disease. Search was conducted across Medline, Embase, Cochrane, and Web of Science databases. Statistical Analysis was performed, with proportions and 95 % confidence intervals. Statistical analyses were carried out using R Studio. RESULTS Ten studies were selected, comprising 553 patients. The population mean age was 40 (24-65), and 50 % of the population was formed by males. In terms of proportion, 6 analyses were performed: Disease Stability of 31 % [95 %CI:14 %-47 %; I2 = 2 %]; Disease Progression of 2 %[95 %CI:0 %-9 %; I2 = 0 %]; Partial Response of 75 % [95 %CI:54 %-96 %; I2 = 58 %]. Complete response of 1 % [95 %CI:0 %-7 %; I2 = 0 %];and Side effects, anemia 81 % rate [95 % CI:54 %-100 %; I2 = 94 %], and fatigue rate of 79 % [95 % CI:54 %-100 %;I2 = 94 %]. CONCLUSION Results indicate that Belzutifan effectively stabilizes disease, reduces tumor progression, and achieves significant therapeutic responses, although side effects like anemia and fatigue were noted.
Collapse
Affiliation(s)
| | | | | | - Márcio Yuri Ferreira
- Department of Neurosurgery, Lenox Hill Hospital/Northwell Health, New York, NY, USA
| | - Leonardo B Oliveira
- Department of Neurosurgery, State University of Ponta Grossa, Ponta Grossa, PR, Brazil
| | - Lucas Pari Mitre
- Santa Casa de São Paulo School of Medical Sciences, São Paulo, SP, Brazil
| | | | | | | | - Sávio Batista
- Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Raphael Bertani
- Department of Neurosurgery, University of São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
39
|
Varghese J, Skefos CM, Jimenez C. Metastatic pheochromocytoma and paraganglioma: Integrating tumor biology in clinical practice. Mol Cell Endocrinol 2024; 592:112344. [PMID: 39182716 DOI: 10.1016/j.mce.2024.112344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGL) are rare neuroendocrine tumors derived from chromaffin cells in the autonomic nervous system. Depending on their location, these tumors are capable of excessive catecholamine production, which may lead to uncontrolled hypertension and other life-threatening complications. They are associated with a significant risk of metastatic disease and are often caused by an inherited germline mutation. Although surgery can cure localized disease and lead to remission, treatments for metastatic PPGL (mPPGL)-including chemotherapy, radiopharmaceutical agents, multikinase inhibitors, and immunotherapy used alone or in combination- aim to control tumor growth and limit organ damage. Substantial advances have been made in understanding hereditary and somatic molecular signaling pathways that play a role in tumor growth and metastasis. Treatment options for metastatic disease are rapidly evolving, and this paper aims to provide a brief overview of the management of mPPGL with a focus on therapy options.
Collapse
Affiliation(s)
- Jeena Varghese
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Catherine M Skefos
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
40
|
Tan D, Fujiwara RJT, Tan C, Isaacson B, Hunter JB. Endolymphatic Sac Tumors Associated With von Hippel-Lindau: A Case Report Highlighting Opportunity for Novel Orphan Drug Therapy. Otol Neurotol 2024; 45:e644-e646. [PMID: 39165133 DOI: 10.1097/mao.0000000000004307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
OBJECTIVE To discuss the potential benefit of belzutifan therapy in a patient with von Hippel-Lindau (VHL) disease-associated endolymphatic sac tumor (ELST). PATIENTS Case report. INTERVENTIONS Clinical details of a patient with residual ELST after hearing preservation surgery who initiated belzutifan therapy postoperatively for concurrent renal cell carcinoma, as well as literature review of belzutifan and ELST. MAIN OUTCOME MEASURES The patient remained without radiologic evidence of growth of her residual tumor at 17 months post-initiation of belzutifan. It is unknown whether this represents therapeutic drug effect, nonviability of residual tumor, or slow tumor growth not captured radiographically within the duration of follow-up. CONCLUSIONS Belzutifan could have direct therapeutic benefit in patients with VHL-associated ELST.
Collapse
Affiliation(s)
- Donald Tan
- Department of Otolaryngology-Head and Neck Surgery, University of Texas Southwestern Medical Center, Texas
| | - Rance J T Fujiwara
- Department of Otolaryngology-Head and Neck Surgery, University of Texas Southwestern Medical Center, Texas
| | - Christopher Tan
- University of California Irvine School of Engineering, Irvine, California
| | - Brandon Isaacson
- Department of Otolaryngology-Head and Neck Surgery, University of Texas Southwestern Medical Center, Texas
| | - Jacob B Hunter
- Department of Otolaryngology-Head and Neck Surgery, Jefferson Health, Philadelphia, Pennsylvania
| |
Collapse
|
41
|
Marathe DD, Jauslin PM, Jan Kleijn H, De Miranda Silva C, Chain A, Abraham AK, Kauh EA, Liu Y, Perini RF, Alwis DPD, Jain L. Exposure-Response Analyses for Belzutifan to Inform Dosing Considerations and Labeling. J Clin Pharmacol 2024; 64:1246-1258. [PMID: 38752556 DOI: 10.1002/jcph.2459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/26/2024] [Indexed: 09/28/2024]
Abstract
Belzutifan (Welireg, Merck & Co., Inc., Rahway, NJ, USA) is an oral, potent hypoxia-inducible factor-2α inhibitor, recently approved in the United States for the treatment of von Hippel-Lindau (VHL) disease-associated renal cell carcinoma (RCC) and other VHL disease-associated neoplasms. Safety and efficacy were investigated in two clinical studies: a Phase 1 dose escalation/expansion study in solid tumors and RCC and a Phase 2 study in VHL-RCC. A population pharmacokinetic model was used to estimate belzutifan exposures to facilitate exposure-response (E-R) analyses for efficacy and safety endpoints. Relationships between exposure and efficacy (overall response rate, disease control rate, progression-free survival, best overall tumor size response, and other endpoints), safety outcomes (Grade ≥3 anemia, Grade ≥3 hypoxia, and time to first dose reduction/dose interruption), and pharmacodynamic biomarkers (erythropoietin [EPO] and hemoglobin [Hgb]) were evaluated using various regression techniques and time-to-event analyses. Efficacy E-R was generally flat with non-significant positive trends with exposure. The safety E-R analyses demonstrated a lack of relationship for Grade ≥3 hypoxia and a positive relationship for Grade ≥3 anemia, with incidences also significantly dependent on baseline Hgb. Exposure-dependent reductions in EPO and Hgb were observed. Based on the cumulative benefit-risk assessment in VHL disease-associated neoplasms using E-R, no a priori dose adjustment is recommended for any subpopulation. These analyses supported the benefit-risk profile of belzutifan 120 mg once daily dosing in patients with VHL-RCC for labeling and the overall development program.
Collapse
|
42
|
Abu-Remaileh M, Persky NS, Lee Y, Root DE, Kaelin WG. Total loss of VHL gene function impairs neuroendocrine cancer cell fitness due to excessive HIF2α activity. Proc Natl Acad Sci U S A 2024; 121:e2410356121. [PMID: 39320914 PMCID: PMC11459182 DOI: 10.1073/pnas.2410356121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024] Open
Abstract
Loss-of-function germline von Hippel-Lindau (VHL) tumor suppressor mutations cause VHL disease, which predisposes individuals to kidney cancer, hemangioblastomas, and paragangliomas. The risk that a given VHL disease family will manifest some or all these tumor types is profoundly influenced by the VHL allele it carries. For example, almost all VHL disease families that develop paraganglioma have missense VHL mutations. VHL families with null VHL alleles develop kidney cancer and hemangioblastomas without a high risk of paraganglioma. The latter is surprising because the VHL gene product, pVHL, suppresses the HIF2 transcription factor and gain-of-function HIF2 mutations are also linked to paraganglioma. Paragangliomas arise from the sympathetic or parasympathetic nervous system. Given the lack of human paraganglioma cell lines, we studied the effects of inactivating VHL in neuroblastoma cell lines, which also arise from the sympathetic nervous system. We found that total loss of pVHL function profoundly impairs the fitness of neuroblastoma cell lines in a HIF2-dependent manner both ex vivo and in vivo. This fitness defect can be rescued by pVHL variants linked to paraganglioma, but not by pVHL variants associated with a low risk of paraganglioma. These findings suggest that HIF2 activity above a critical threshold prevents the development of paraganglioma.
Collapse
Affiliation(s)
- Muhannad Abu-Remaileh
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Nicole S. Persky
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
| | - Yenarae Lee
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
| | - David E. Root
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
| | - William G. Kaelin
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
- HHMI, Chevy Chase, MD20815
| |
Collapse
|
43
|
Iliopoulos O, Iversen AB, Narayan V, Maughan BL, Beckermann KE, Oudard S, Else T, Maranchie JK, Goldberg CM, Fu W, Perini RF, Liu Y, Linehan WM, Srinivasan R, Jonasch E. Belzutifan for patients with von Hippel-Lindau disease-associated CNS haemangioblastomas (LITESPARK-004): a multicentre, single-arm, phase 2 study. Lancet Oncol 2024; 25:1325-1336. [PMID: 39284337 DOI: 10.1016/s1470-2045(24)00389-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND The first-in-class hypoxia-inducible factor-2α inhibitor, belzutifan, showed clinically meaningful antitumour activity in von Hippel-Lindau (VHL) disease-associated neoplasms in the ongoing, single-arm, phase 2 LITESPARK-004 study. We aimed to investigate antitumour activity with an additional 16 months of follow-up and present updated results for the subgroup of patients with CNS haemangioblastomas. METHODS In the multicentre, single-arm, phase 2 LITESPARK-004 study, adults (aged ≥18 years) from 11 cancer centres or hospitals in the USA, Denmark, France, and the UK, with germline VHL alterations, at least one measurable renal cell carcinoma tumour, no renal cell carcinoma tumour greater than 3 cm requiring immediate surgical intervention, an Eastern Cooperative Oncology Group performance status 0 or 1, and no previous systemic therapy received oral belzutifan 120 mg once daily until unacceptable toxicity, disease progression, or patient decision to withdraw. The primary endpoint, evaluated in patients with CNS haemangioblastomas, was the proportion of patients with an objective response per RECIST version 1.1 by an independent review committee. We assessed response using two approaches. In approach 1, we evaluated all measurable (≥1 cm maximum diameter) or non-measurable lesions at baseline, including both the solid lesion and the associated cystic component if present. In approach 2, we evaluated only baseline lesions with a measurable (≥1 cm maximum diameter) solid lesion. Antitumour activity was assessed in all patients who received at least one dose of belzutifan. This study is no longer recruiting but is ongoing, and is registered with Clinicaltrials.gov, NCT03401788. FINDINGS Between May 31, 2018, and March 29, 2019, of 67 patients screened, 61 (32 [52%] male and 29 [48%] female) were enrolled; 50 (82%) had at least one CNS haemangioblastoma evaluable at baseline (184 total lesions). Median follow-up for the 50 patients with CNS haemangioblastomas was 38·0 months (IQR 36·7-40·1). In approach 1, 22 of 50 patients (44% [95% CI 30-59]) had an objective response. In approach 2, 19 of 25 patients (76% [55-91] had an objective response. 23 (46%) of 50 patients had a grade 3-5 all-cause adverse event. 19 (38%) patients reported grade 3 adverse events, the most common of which was anaemia (in 6 [12%] patients). Two of 50 patients (4%) reported grade 4 events (retinal vein occlusion and embolism). Two patients died owing to adverse events not considered treatment-related (suicide and toxicity to various agents). INTERPRETATION Belzutifan showed meaningful antitumour activity in VHL disease-associated CNS haemangioblastomas that was sustained for more than 3 years of treatment. These results continue to support belzutifan as a systemic treatment option for patients with VHL disease-related CNS haemangioblastomas. FUNDING Merck Sharp & Dohme, National Institutes of Health, and National Cancer Institute.
Collapse
Affiliation(s)
- Othon Iliopoulos
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA.
| | | | - Vivek Narayan
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Stephane Oudard
- Hôpital Européen Georges Pompidou, University Paris Cité, Paris, France
| | - Tobias Else
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | - W Marston Linehan
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Eric Jonasch
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
44
|
Larcher A, Belladelli F, Cei F, Re C, Rowe I, Salerno L, Marandino L, Raggi D, Necchi A, Capitanio U, Montorsi F, Salonia A. Removing Barriers to the Use of Systemic Agents for Patients with von Hippel-Lindau Disease. Eur Urol Oncol 2024; 7:1141-1143. [PMID: 38272746 DOI: 10.1016/j.euo.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 12/20/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Affiliation(s)
- Alessandro Larcher
- Department of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Federico Belladelli
- Department of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Cei
- Department of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Chiara Re
- Department of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Isaline Rowe
- Department of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Lucia Salerno
- Department of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Laura Marandino
- Department of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Daniele Raggi
- Department of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Necchi
- Department of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Umberto Capitanio
- Department of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Montorsi
- Department of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Salonia
- Department of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
45
|
Pumford AD, Bauman M, Bouchal S, Riviere-Cazaux C, Jusue-Torres I, Hong S, Neth BJ, Sener U, Parney IF. Neurosurgical Implications of Targeting Hypoxia-Inducible Factor 2α in Hemangioblastomas with Belzutifan. World Neurosurg 2024; 190:291-296. [PMID: 39094939 DOI: 10.1016/j.wneu.2024.07.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE To highlight the neurosurgical implications of the hypoxia-inducible factor-2α- targeting agent belzutifan in the management of both von-Hippel Lindau (VHL)-associated and sporadic hemangioblastomas (HBLs). METHODS The literature was queried for VHL, HBLs, and belzutifan. A summary of recent uses of belzutifan and currently ongoing clinical trials that are investigating the use of belzutifan in the treatment of HBLs is presented. RESULTS VHL disease occurs as a result of germline mutations in the VHL tumor suppressor gene on chromosome 3p25-p26, leading to growth of benign and malignant tumors such as HBLs. The possibility of intermittent growth in HBLs indicates that it is important to avoid hasty surgical interventions. Belzutifan is the first nonsurgical food and drug administration-approved treatment for VHL disease-related tumors that may delay or circumvent the need for surgery or radiation therapy by inhibiting HIF-2α, an important component of cellular hypoxic response. There is limited real-world experience of belzutifan in patients with HBLs as a primary indication, though there are 2 phase II clinical trials investigating the use of belzutifan in the treatment of HBLs. CONCLUSIONS There is limited experience regarding the use of belzutifan for CNS hemangioblastoma. While its application has been limited to a small group of clinical cases, it has exhibited significant efficacy in reducing the size and consequences of HBLs. Based on the promising outcomes observed in individual patient experiences and ongoing clinical trials, we infer that further exploration and integration of belzutifan into neurosurgical treatment plans for both sporadic and VHL-associated HBLs are warranted.
Collapse
Affiliation(s)
- Andrew D Pumford
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, Minnesota, USA. @mayo.edu
| | - Megan Bauman
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Samantha Bouchal
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Sukwoo Hong
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Bryan J Neth
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ugur Sener
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ian F Parney
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
46
|
Rose TL, Kim WY. Renal Cell Carcinoma: A Review. JAMA 2024; 332:1001-1010. [PMID: 39196544 DOI: 10.1001/jama.2024.12848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Importance Renal cell carcinoma (RCC) is a common malignancy, with an estimated 434 840 incident cases worldwide in 2022. In the US, it is the sixth most common cancer among males and ninth among females. Observations Clear cell RCC is the most common histologic subtype (75%-80% of cases) and is characterized by inactivation of the von Hippel Lindau (VHL) tumor suppressor gene. Many patients (37%-61%) are diagnosed with RCC incidentally on an abdominal imaging study such as ultrasound or computed tomographic scan, and 70% of patients have stage I RCC at diagnosis. Although its incidence has increased approximately 1% per year from 2015 through 2019, the mortality rate of RCC has declined about 2% per year in the US from 2016 through 2020. Patients with a solid renal mass or complex cystic renal mass should be referred to urology. Treatment options for RCC confined to the kidney include surgical resection with partial or radical nephrectomy, ablative techniques (eg, cryoablation, radiofrequency ablation, radiation), or active surveillance for some patients (especially those with renal masses <2 cm). For patients with renal masses less than 4 cm in size (48% of patients), partial nephrectomy can result in a 5-year cancer-specific survival of more than 94%. For advanced or metastatic RCC, combinations of immune checkpoint inhibitors or the combination of immune checkpoint inhibitors with tyrosine kinase inhibitors are associated with tumor response of 42% to 71%, with a median overall survival of 46 to 56 months. Conclusions and Relevance RCC is a common malignancy that is often diagnosed incidentally on an abdominal imaging study. Seventy percent of patients are diagnosed with stage I RCC and 11% of patients with stage IV. First-line treatments for early-stage RCC are partial or radical nephrectomy, which can result in 5-year cancer-specific survival of more than 94%, ablative techniques, or active surveillance. New treatment options for patients with metastatic RCC include immune checkpoint inhibitors and tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Tracy L Rose
- Division of Oncology, Department of Medicine, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
| | - William Y Kim
- Division of Oncology, Department of Medicine, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
- Department of Genetics, University of North Carolina at Chapel Hill
- Department of Pharmacology, University of North Carolina at Chapel Hill
| |
Collapse
|
47
|
Greco F, Panunzio A, D’Andrea V, Vescovo M, Tafuri A, Carotti S, Beomonte Zobel B, Mallio CA. Exploring Tumor Heterogeneity: Radiogenomic Assessment of ADFP in Low WHO/ISUP Grade Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2024; 16:3164. [PMID: 39335136 PMCID: PMC11430299 DOI: 10.3390/cancers16183164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/11/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
This study aimed to investigate the association between metabolic lipid computed tomography (CT) features and adipose differentiation-related protein (ADFP) expression in clear cell renal cell carcinoma (ccRCC), providing insights into non-invasive methods for assessing ADFP expression and tumor characteristics. This study utilized data from The Cancer Genome Atlas and the Cancer Imaging Archive to analyze genetic alterations and imaging characteristics in ccRCC patients. Tumoral Hounsfield units (HU) analysis and quantification of abdominal adipose tissue compartments were performed using CT images. Statistical analyses were conducted to compare tumoral HU values according to ADFP gene expression and World Health Organization/International Society of Urological Pathology (WHO/ISUP) tumor grade, as well as to explore correlations between tumoral HU values and adipose tissue quantification. Among the 174 identified patients, those with ADFP gene expression showed significantly lower minimum tumoral HU values in low-grade cancers compared to high-grade cancers. Similarly, patients with low-grade cancers expressing ADFP exhibited lower minimum tumoral HU values compared to those without ADFP expression. Negative correlations were observed between minimum tumoral HU values and visceral adipose tissue, subcutaneous adipose tissue, and total adipose tissue in both ccRCC patients with and without ADFP expression. This study reveals a significant association between metabolic lipid CT features and ADFP expression in ccRCC patients. Lower minimum tumoral HU values, suggestive of higher intracellular lipid accumulation, were observed in tumors with low WHO/ISUP grade and ADFP expression.
Collapse
Affiliation(s)
- Federico Greco
- Department of Radiology, Cittadella della Salute, Azienda Sanitaria Locale di Lecce, Piazza Filippo Bottazzi, 2, 73100 Lecce, Italy
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (V.D.); (B.B.Z.); (C.A.M.)
| | - Andrea Panunzio
- Department of Urology, “Vito Fazzi” Hospital, Piazza Filippo Muratore, 1, 73100 Lecce, Italy; (A.P.); (A.T.)
| | - Valerio D’Andrea
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (V.D.); (B.B.Z.); (C.A.M.)
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Mariavittoria Vescovo
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy;
| | - Alessandro Tafuri
- Department of Urology, “Vito Fazzi” Hospital, Piazza Filippo Muratore, 1, 73100 Lecce, Italy; (A.P.); (A.T.)
| | - Simone Carotti
- Microscopic and Ultrastructural Anatomy Research Unit, Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy;
- Predictive Molecular Diagnostics, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Bruno Beomonte Zobel
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (V.D.); (B.B.Z.); (C.A.M.)
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Carlo Augusto Mallio
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (V.D.); (B.B.Z.); (C.A.M.)
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| |
Collapse
|
48
|
Natarajan V, Satalkar V, Gumbart JC, Torres M. Molecular Dynamics Reveals Altered Interactions between Belzutifan and HIF-2 with Natural Variant G323E or Proximal Phosphorylation at T324. ACS OMEGA 2024; 9:37843-37855. [PMID: 39281922 PMCID: PMC11391435 DOI: 10.1021/acsomega.4c03777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/11/2024] [Accepted: 08/16/2024] [Indexed: 09/18/2024]
Abstract
In patients with von-Hippel Lindau (VHL) disease, hypoxia-independent accumulation of HIF-2α leads to increased transcriptional activity of HIF-2α:ARNT that drives cancers such as renal cell carcinoma. Belzutifan, a recently FDA-approved drug, is designed to prevent the transcriptional activity of HIF-2α:ARNT, thereby overcoming the consequences of its unnatural accumulation in VHL-dependent cancers. Emerging evidence suggests that the naturally occurring variant G323E located in the HIF-2α drug binding pocket prevents inhibitory activity of belzutifan analogs, though the mechanism of inhibition remains unclear. Interestingly, proximal phosphorylation at neighboring T324, previously shown to regulate HIF-2 protein interactions, has also been proposed to affect HIF-2 drug binding. Here, we used molecular dynamics (MD) simulations to understand and compare the molecular-level effects of G323E and phospho-T324 (pT324) on the belzutifan bound-HIF-2α:ARNT complex. We find that both G323E and pT324 increase structural flexibility within the drug binding site and reduce the apparent binding affinity for belzutifan. Whereas the effects of G323E are concentrated in the binding pocket Fα helix within the HIF-2α PAS-B domain, pT324 decreased the belzutifan binding affinity and stabilized the HIF-2 heterodimer through an alternate mechanism involving polar interactions between the HIF-2α PAS-B and PAS-A domains. Further analysis via ensemble machine learning uncovered important and distinct interchain residue interactions modified by G323E and pT324. These findings reveal a molecular mechanism of G323E-induced drug resistance and suggest that pT324 may also affect the efficacy of HIF-2 drug binding interactions via allosteric effects.
Collapse
Affiliation(s)
- Vishva Natarajan
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Vardhan Satalkar
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - James C Gumbart
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Matthew Torres
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
49
|
Antony MB, Kozel Z, Gopal N, Loebach L, Metwalli AR, Gurram S, Linehan WM, Ball MW. Cumulative Impact of Serial Partial Nephrectomy for the Treatment of Recurrent Renal Masses. J Urol 2024; 212:431-440. [PMID: 38865696 PMCID: PMC11309881 DOI: 10.1097/ju.0000000000004099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 05/31/2024] [Indexed: 06/14/2024]
Abstract
PURPOSE Reoperative partial nephrectomy (RePN) offers several advantages for the treatment of recurrent, multifocal renal masses. RePN has been previously demonstrated to be technically feasible and delay the need for renal replacement therapy. However, there is still inherent complexity and known risks to reoperative nephrectomy. We studied the largest population of RePNs to characterize renal functional outcomes and the likelihood of intra- and postoperative complications. MATERIALS AND METHODS Query of an institutional surgical registry was conducted. Demographic data, serum creatinine for estimated glomerular filtration rate (eGFR), and protein dipstick results were assessed within 1 week prior to surgery, and postoperative function assessments were studied within a year of surgery. RePN was defined as serial surgical resection of the ipsilateral renal unit. RESULTS A total of 1131 partial nephrectomies performed on 663 patients at a single center were retrospectively evaluated. In reoperative cases, median number of operations per renal unit was 2 (range: 2-6). There was a stepwise decline in eGFR with an average decline of 6.1 with each RePN. With each subsequent nephrectomy, surgical duration, estimated blood loss, and incidence of preoperative anemia increased. Postoperative eGFR showed a significant positive association with preoperative eGFR, while negative associations were found with age, number of previous ipsilateral partial nephrectomies, number of tumors, and largest tumor size. High-grade complications were associated with the number of ipsilateral partial nephrectomies, tumor count, and tumor size. Robotic or laparoscopic procedures exhibited a likelihood of grade 3 or greater complications compared to open surgery. CONCLUSIONS RePN contributes to renal dysfunction and an increased risk of surgical complications. Intraoperative blood loss and surgical duration increase with subsequent nephrectomy. Such risks are dependent on the number of prior operative interventions on the kidney, suggesting a stepwise progression of surgical morbidity.
Collapse
Affiliation(s)
- Maria B. Antony
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, MD, USA
| | - Zach Kozel
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, MD, USA
| | - Nikhil Gopal
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, MD, USA
| | - Lauren Loebach
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, MD, USA
| | - Adam R. Metwalli
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, MD, USA
| | - Sandeep Gurram
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, MD, USA
| | - W. Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, MD, USA
| | - Mark W. Ball
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, MD, USA
| |
Collapse
|
50
|
Wachtel H, Nathanson KL. Molecular Genetics of Pheochromocytoma/Paraganglioma. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2024; 36:100527. [PMID: 39328362 PMCID: PMC11424047 DOI: 10.1016/j.coemr.2024.100527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGL) are neuroendocrine tumors which secrete catecholamines, causing cardiovascular compromise. While isolated tumors and locoregional disease can be treated surgically, treatment options for metastatic disease are limited, and no targeted therapies exist. Approximately 25% of PPGL are causatively associated with germline pathogenic variants, which are known risk factors for multifocal and metastatic PPGL. Knowledge of somatic driver mutations continues to evolve. Molecular classification of PPGL has identified three genomic subtypes: Cluster 1 (pseudohypoxia), Cluster 2 (kinase signaling) and Cluster 3 (Wnt-altered). This review summaries recent studies characterizing the tumor microenvironment, genomic drivers of tumorigenesis and progression, and current research on molecular targets for novel diagnostic and therapeutic strategies in PPGL.
Collapse
Affiliation(s)
- Heather Wachtel
- Hospital of the University of Pennsylvania, Department of Surgery, Division of Endocrine and Oncologic Surgery and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Katherine L Nathanson
- Hospital of the University of Pennsylvania, Department of Medical Genetics, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|