1
|
Fan TWM, Higashi RM, Lane AN. Metabolic Reprogramming in Human Cancer Patients and Patient-Derived Models. Cold Spring Harb Perspect Med 2025; 15:a041552. [PMID: 39009444 PMCID: PMC12047743 DOI: 10.1101/cshperspect.a041552] [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: 07/17/2024]
Abstract
Stable isotope-resolved metabolomics delineates reprogrammed intersecting metabolic networks in human cancers. Knowledge gained from in vivo patient studies provides the "benchmark" for cancer models to recapitulate. It is particularly difficult to model patients' tumor microenvironment (TME) with its complex cell-cell/cell-matrix interactions, which shapes metabolic reprogramming crucial to cancer development/drug resistance. Patient-derived organotypic tissue cultures (PD-OTCs) represent a unique model that retains an individual patient's TME. PD-OTCs of non-small-cell lung cancer better recapitulated the in vivo metabolic reprogramming of patient tumors than the patient-derived tumor xenograft (PDTX), while enabling interrogation of immunometabolic response to modulators and TME-dependent resistance development. Patient-derived organoids (PDOs) are also good models for reconstituting TME-dependent metabolic reprogramming and for evaluating therapeutic responses. Single-cell based 'omics on combinations of PD-OTC and PDO models will afford an unprecedented understanding on TME dependence of human cancer metabolic reprogramming, which should translate into the identification of novel metabolic targets for regulating TME interactions and drug resistance.
Collapse
Affiliation(s)
- Teresa W-M Fan
- Center for Environmental and Systems Biochemistry; Markey Cancer Center; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Richard M Higashi
- Center for Environmental and Systems Biochemistry; Markey Cancer Center; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Andrew N Lane
- Center for Environmental and Systems Biochemistry; Markey Cancer Center; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| |
Collapse
|
2
|
McHenry A, Monsrud A, Pors J, Folkins A, Longacre T, Hodan R. Prospective Fumarate Hydratase Tumor Predisposition Syndrome Screening in Patients With Uterine Smooth Muscle Tumors: Age, Morphology, Fumarate Hydratase/S-(2-succino) Cysteine Immunohistochemistry, and Germline Testing. Am J Surg Pathol 2025; 49:315-327. [PMID: 39835370 DOI: 10.1097/pas.0000000000002362] [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: 01/22/2025]
Abstract
Fumarate hydratase tumor predisposition syndrome (FHTPS) is caused by germline fumarate hydratase (FH) pathogenic variants (PVs). Most women with FHTPS develop FH-deficient (FHD) uterine leiomyomas (ULs), which arise 10 to 15 years earlier than aggressive FHD-renal cell carcinoma. We evaluate a previously proposed FHTPS screening strategy for women with ULs. This 5-year, prospective and retrospective study performed FH and later S-(2-succino) cysteine immunohistochemistry (IHC) on all uterine smooth muscle (USM) tumors in patients 40 (later ≤30) years or younger and on all USM tumors with suggestive FHD morphology regardless of age. Patients with FHD tumors by IHC were referred to genetic counseling. Of 840 USM tumors, 112 FHD-tumors by IHC (13%) were identified, all with suggestive FHD-morphology; 44 patients (39%) underwent germline testing, and 15 harbored germline FH PVs (34.1% of germline tested, 13.4% of all FHD-tumors). While FHD tumors were seen across a wide age range (24 to 73 y), those with germline FH PVs were significantly younger (median 33 vs 44 years wild-type, P = 0.0032). Few (12.5%) patients ≥40 and no patients ≥50 had a germline FH PV, whereas a majority (60%) of patients <40 (86% of those <30) had a germline FH PV. We demonstrate that previously proposed resource-conscious screening involving morphology and IHC is effective for identifying women with FHTPS. We provide prospective data confirming patients presenting with FHD-ULs over age 50 are unlikely to harbor germline FH PVs and argue that for germline testing without consideration of other factors, a threshold of younger than 50 years may be appropriate.
Collapse
Affiliation(s)
- Austin McHenry
- Department of Pathology, School of Medicine, Stanford University, Stanford
| | - Ashley Monsrud
- Department of Pathology, School of Medicine, Stanford University, Stanford
| | - Jennifer Pors
- Department of Cancer Genetics and Genomics, Stanford Health Care, Palo Alto, CA
| | - Ann Folkins
- Department of Pathology, School of Medicine, Stanford University, Stanford
| | - Teri Longacre
- Department of Pathology, School of Medicine, Stanford University, Stanford
| | - Rachel Hodan
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver BC, Canada
| |
Collapse
|
3
|
Uzianbaeva L, Moustafa S, Paczos T, Suskin E, Said-Delgado S, Rabin-Havt S, Wang P. Leiomyoma with Bizarre Nuclei and Hereditary Leiomyomatosis and Renal Cell Carcinoma. CRSLS : MIS CASE REPORTS FROM SLS 2025; 12:e2025.00015. [PMID: 40321900 PMCID: PMC12046535 DOI: 10.4293/crsls.2025.00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Introduction Leiomyoma with bizarre nuclei (LBN) is a leiomyoma variant that can be associated with fumarate hydratase (FH) deficiency. Germline pathogenic variants in the FH gene are linked to hereditary leiomyomatosis and renal cell carcinoma (HLRCC), which presents with cutaneous leiomyomata, aggressive renal cell carcinomas (RCCs), and symptomatic uterine leiomyomas. Case Description A 22-year-old nulligravida female presented with multiple uterine fibroids, heavy menstrual bleeding and pelvic pain, lasting over two years. The patient subsequently underwent laparoscopic myomectomy. Histological analysis of the leiomyomas indicated the presence of bizarre nuclei. Immunohistochemical studies confirmed FH deficiency, characterized by loss of FH expression and overexpression of S-(2-succino)-cysteine (2SC). Genetic testing revealed a likely pathogenic variant (c. 1176_1181delTGCTGT) in the FH gene. Discussion Due to potentially devastating consequence and the occult nature of RCCs, the discovery of LBN should be followed with further investigation for HLRCC.
Collapse
Affiliation(s)
- Liaisan Uzianbaeva
- Department of Obstetrics and Gynecology, BronxCare Health System, Bronx, NY, USA. (Drs. Uzianbaeva, Moustafa, Said-Delgado, and Wang)
| | - Salma Moustafa
- Department of Obstetrics and Gynecology, BronxCare Health System, Bronx, NY, USA. (Drs. Uzianbaeva, Moustafa, Said-Delgado, and Wang)
| | - Tamera Paczos
- Department of Pathology, BronxCare Health System, Bronx, NY, USA. (Dr. Paczos)
| | - Emily Suskin
- Department of Reproductive and Medical Genetics, Montefiore Einstein, Bronx, NY, USA. (Drs. Suskin and Rabin-Havt)
| | - Sara Said-Delgado
- Department of Obstetrics and Gynecology, BronxCare Health System, Bronx, NY, USA. (Drs. Uzianbaeva, Moustafa, Said-Delgado, and Wang)
| | - Sara Rabin-Havt
- Department of Reproductive and Medical Genetics, Montefiore Einstein, Bronx, NY, USA. (Drs. Suskin and Rabin-Havt)
| | - Pengfei Wang
- Department of Obstetrics & Gynecology and Reproductive Science, The Icahn School of Medicine at Mt Sinai, New York, NY, USA. (Dr. Wang)
- Department of Obstetrics and Gynecology, BronxCare Health System, Bronx, NY, USA. (Drs. Uzianbaeva, Moustafa, Said-Delgado, and Wang)
| |
Collapse
|
4
|
Suzuki S, Yamamoto Y, Kato T, Hatano K, Matsui T, Hashimoto K, Miyamura T, Nagashima Y, Nonomura N, Kawashima A. Postmortem genetic diagnosis of hereditary leiomyomatosis and renal cell carcinoma syndrome: Identification through normal kidney tissues after surgical removal. IJU Case Rep 2025; 8:109-113. [PMID: 40034895 PMCID: PMC11872217 DOI: 10.1002/iju5.12820] [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: 09/17/2024] [Accepted: 11/26/2024] [Indexed: 03/05/2025] Open
Abstract
Introduction Hereditary leiomyomatosis and renal cell cancer is an autosomal dominant disorder caused by germline mutations in the FH gene and is associated with poor prognosis of aggressive renal cancer. Case presentation A 33-year-old man presented with asymptomatic gross hematuria and was diagnosed with a right renal tumor, cT3aN1M0. He underwent open radical nephrectomy, and pathological examination revealed papillary renal cell carcinoma. Despite aggressive treatment, the disease progressed rapidly, and discussions regarding genetic testing could not take place during his lifetime, although circulating-tumor DNA showed mutation of FH gene. After death, his wife requested postmortem genetic testing. Genetic analysis using DNA extracted from normal kidney tissues in surgical specimens (blood sample absence) confirmed the FH mutation, and hereditary leiomyomatosis and renal cell cancer was diagnosed posthumously. Conclusion This highlights the utility of postmortem genetic testing of surgical specimens to diagnose hereditary leiomyomatosis and renal cell cancer and provide genetic counseling to families, despite limitations during the patient's life.
Collapse
Affiliation(s)
- Shodai Suzuki
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Yoshiyuki Yamamoto
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Taigo Kato
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Koji Hatano
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Takahiro Matsui
- Department of PathologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Kae Hashimoto
- Department of Genetic CounselingOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Takako Miyamura
- Department of PediatricsOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Yoji Nagashima
- Department of Surgical PathologyTokyo Women's Medical University HospitalTokyoJapan
| | - Norio Nonomura
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Atsunari Kawashima
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| |
Collapse
|
5
|
Najera SS, Ricketts CJ, Schmidt LS, Medina JI, Saito K, Ileva L, Brender JR, James AM, Peer CJ, Gouker B, Karim BO, Chernova O, Wells C, Wei MH, Yang Y, Zhang X, Klumpp-Thomas C, Travers J, Chen L, Wilson KM, Issaq SH, Figg WD, Difilippantonio S, Kalen JD, Krishna MC, Thomas CJ, Ceribelli M, Heske CM, Crooks DR, Meier JL. Targeting NAD+ Metabolism Vulnerability in FH-Deficient Hereditary Leiomyomatosis and Renal Cell Carcinoma with the Novel NAMPT Inhibitor OT-82. Mol Cancer Ther 2025; 24:200-213. [PMID: 39397296 DOI: 10.1158/1535-7163.mct-24-0225] [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] [Received: 03/28/2024] [Revised: 08/05/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
Hereditary leiomyomatosis and renal cell cancer (HLRCC) is an inherited cancer syndrome caused by germline pathogenic variants in the fumarate hydratase (FH) gene. Affected individuals are at risk for developing cutaneous and uterine leiomyomas and aggressive FH-deficient renal cell carcinoma (RCC) with a papillary histology. Due to a disrupted tricarboxylic acid cycle, FH-deficient kidney cancers rely on aerobic glycolysis for energy production, potentially creating compensatory metabolic vulnerabilities. This study conducted a high-throughput drug screen in HLRCC cell lines, which identified a critical dependency on nicotinamide adenine dinucleotide (NAD), a redox cofactor produced by the biosynthetic enzyme nicotinamide phosphoribosyltransferase (NAMPT). Human HLRCC tumors and HLRCC-derived cell lines exhibited elevated NAMPT expression compared with controls. FH-deficient HLRCC cells, but not FH-restored HLRCC or normal kidney cells, were sensitive to NAMPT inhibition. HLRCC cell line viability was significantly decreased in both 2D and 3D in vitro cultures in response to the clinically relevant NAMPT inhibitor OT-82. NAMPT inhibition in vitro significantly decreased the total amount of NAD+, NADH, NADP, NADPH, and poly-ADP-ribose levels, and the effects of NAMPT inhibition could be rescued by the downstream NAD precursor nicotinamide mononucleotide (NMN), confirming the on-target activity of OT-82. Moreover, NAMPT inhibition by OT-82 in two HLRCC xenograft models resulted in severely reduced tumor growth. OT-82 treatment of HLRCC xenograft tumors in vivo inhibited glycolytic flux as demonstrated by reduced lactate/pyruvate ratio in hyperpolarized 13C-pyruvate magnetic resonance spectroscopic imaging experiments. Overall, our data define NAMPT inhibition as a potential therapeutic approach for FH-deficient HLRCC-associated RCC.
Collapse
Affiliation(s)
- Susana S Najera
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland
| | - Christopher J Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Laura S Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Julia I Medina
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Keita Saito
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Lilia Ileva
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jeffrey R Brender
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Amy M James
- Animal Research Technical Support, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Cody J Peer
- Genitourinary Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Brad Gouker
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Baktiar O Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | | | - Catherine Wells
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Ming-Hui Wei
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Youfeng Yang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Carleen Klumpp-Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Jameson Travers
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Lu Chen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Kelli M Wilson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Sameer H Issaq
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - William D Figg
- Genitourinary Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Simone Difilippantonio
- Animal Research Technical Support, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Joseph D Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Murali C Krishna
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Michele Ceribelli
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Christine M Heske
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Daniel R Crooks
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jordan L Meier
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland
| |
Collapse
|
6
|
Michaeli O, Kim SY, Mitchell SG, Jongmans MCJ, Wasserman JD, Perrino MR, Das A, MacFarland SP, Scollon SR, Greer MLC, Sobreira N, Gallinger B, Lupo PJ, Malkin D, Schneider KW, Schultz KAP, Foulkes WD, Woodward ER, Stewart DR. Update on Cancer Screening in Children with Syndromes of Bone Lesions, Hereditary Leiomyomatosis and Renal Cell Carcinoma Syndrome, and Other Rare Syndromes. Clin Cancer Res 2025; 31:457-465. [PMID: 39601780 PMCID: PMC11790369 DOI: 10.1158/1078-0432.ccr-24-2171] [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: 08/17/2024] [Revised: 10/27/2024] [Accepted: 11/26/2024] [Indexed: 11/29/2024]
Abstract
The management of children with syndromes associated with an increased risk of benign and malignant neoplasms is a complex challenge for health care professionals. The 2023 American Association for Cancer Research Childhood Cancer Predisposition Workshop provided updated consensus guidelines on cancer surveillance in these syndromes, aiming to improve early detection and intervention and reduce morbidity associated with such neoplasms. In this article, we review several of the rare conditions discussed in this workshop. Ollier disease and Maffucci syndrome are enchondromatoses (disorders featuring benign bone lesions) with up to 50% risk of malignancy, including chondrosarcoma. These patients require surveillance with baseline whole-body MRI and routine monitoring of potential malignant transformation of bony lesions. Hereditary multiple osteochondromas carry a lower risk of chondrosarcoma (<6%) but still require lifelong surveillance and baseline imaging. Related syndromes of benign bone lesions are also described. Hereditary leiomyomatosis and renal cell carcinoma syndrome, associated with fumarate hydratase pathogenic variants, is discussed in detail. Surveillance for renal cell carcinoma in pediatric age is recommended, as well as prompt intervention when a lesion is detected. Schinzel-Giedion syndrome and Rubinstein-Taybi syndrome are described for their associated malignancies and other complications, as well as expert consensus on the need for childhood cancer surveillance. Clinical recommendations, including imaging modalities and frequency of screenings, are proposed and are tailored to each syndrome's age-specific tumor risk profile. In all syndromes, patients and their families should be educated about the potential malignancy risk and advised to seek medical care for rapid growth of a mass, persistent pain, or other unexplained symptoms.
Collapse
Affiliation(s)
- Orli Michaeli
- Division of Hematology/Oncology, Schneider Children’s Medical Center of Israel, Petach Tikva, Israel
| | - Sun Young Kim
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH, United States
| | - Sarah G. Mitchell
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatric Hematology/Oncology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Marjolijn C. J. Jongmans
- Princess Máxima Center for Pediatric Oncology, Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jonathan D. Wasserman
- Division of Endocrinology, The Hospital for Sick Children / Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Melissa R. Perrino
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Anirban Das
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Suzanne P MacFarland
- Division of Oncology, The Children’s Hospital of Philadelphia, and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sarah R Scollon
- Department of Pediatrics, Division of Hematology/Oncology, Texas Children’s Cancer and Hematology Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, United States
| | - Mary-Louise C. Greer
- Department of Diagnostic and Interventional Radiology, The Hospital for Sick Children, Department of Medical Imaging, University of Toronto, Toronto, Canada
| | - Nara Sobreira
- McKusick-Nathans Department of Genetic Medicine Johns Hopkins University, Baltimore, MD, United States
| | - Bailey Gallinger
- Clinical and Metabolic Genetics, The Hospital for Sick Children. Toronto, ON. Canada. Department of Molecular Genetics, The University of Toronto. Toronto, ON. Canada
| | - Philip J. Lupo
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - David Malkin
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Kami Wolfe Schneider
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children’s Hospital Colorado, Aurora, CO
| | - Kris Ann P. Schultz
- International Pleuropulmonary Blastoma/DICER1 Registry, Cancer and Blood Disorders, Children’s Minnesota, Minneapolis, MN
| | | | - Emma R Woodward
- Faculty of Biology, Medicine and Health, University of Manchester and Manchester Centre for Genomic Medicine, Manchester, United Kingdom
| | - Douglas R. Stewart
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD; United States
| |
Collapse
|
7
|
Shaker N, Li Z, Shaker N, Poombal F, Sangueza OP, Pradhan D. Fumarate Hydratase-Deficient Abdominal Wall Leiomyoma Presenting a Decade Post-Fumarate Hydratase-Deficient Uterine Leiomyoma Excision: An Incidental or Syndromic Association? Int J Surg Pathol 2025; 33:99-103. [PMID: 38847132 DOI: 10.1177/10668969241256118] [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] [Indexed: 01/28/2025]
Abstract
BACKGROUND The occurrence of fumarate hydratase-deficient leiomyoma of the abdominal wall is exceptionally rare. CASE PRESENTATION A 50-year-old female patient with a past medical history of fumarate hydratase-deficient uterine leiomyoma presented with a left lower quadrant abdominal mass that has been present for the past 2 years. An ultrasound revealed a 3.5 cm oval hypoechoic mass. A subsequent CT scan showed a 3.5 cm hyperdense mass within the left internal oblique musculature. No family history is noted. A biopsy of the mass exhibited bundles of spindle cell neoplasm exhibiting bizarre ovoid nuclei and eosinophilic cytoplasm. No evidence of mitotic figures or tumor necrosis was noted. Immunohistochemical staining showed positive staining for desmin and smooth muscle actin (SMA), but negative staining for MART-1, S100, and CD34. Lesional cells showed expression of 2-succinocysteine and loss of fumarate hydratase expression. A diagnosis of fumarate hydratase-deficient leiomyoma was rendered. CONCLUSION This report reinforces the importance of considering genetic testing for fumarate hydratase mutations in the evaluation of extra-uterine leiomyomatous lesions. Comprehensive follow-up and clinical screening in individuals with new lesions and a known history of fumarate hydratase-deficient neoplasms is mandatory. Recent recommendations support the integration of morphology-based evaluation along with immunohistochemical staining and genetic testing as a part of the standard evaluation for all uterine leiomyomas.
Collapse
Affiliation(s)
- Nada Shaker
- Pathology and Laboratory Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Zaibo Li
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Nuha Shaker
- Department of Pathology, University of Pittsburgh Medical Center Health System, Pittsburgh, PA, USA
| | - Fnu Poombal
- Department of Pathology, Nishtar Medical College and Hospital, Multan, Punjab, Pakistan
| | - Omar P Sangueza
- Departments of Dermatology and Dermatopathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Dinesh Pradhan
- Department of Pathology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
8
|
Khamaiseh S, Äyräväinen A, Arffman M, Reinikka S, Mehine M, Härkki P, Bützow R, Pasanen A, Vahteristo P. Clinical and molecular risk factors for repeat interventions due to symptomatic uterine leiomyomas. Am J Obstet Gynecol 2025; 232:110.e1-110.e23. [PMID: 39094728 DOI: 10.1016/j.ajog.2024.06.051] [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: 02/22/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Repeat leiomyoma occurrence or even reintervention is common after myomectomy. Little is known about the factors related to repeat interventions. OBJECTIVE This study aimed to determine the frequency of leiomyoma-related reintervention after an initial laparoscopic or abdominal myomectomy and to analyze both clinical and molecular risk factors for reinterventions. Another objective was to define the frequency of clonally related tumors from repeat operations. STUDY DESIGN This retrospective cohort study included 234 women who had undergone laparoscopic or abdominal myomectomy in 2009 to 2014. Information on repeat leiomyoma-related interventions as well as on other clinical factors was collected from medical records after a median follow-up time of 11.4 years (range 7.9-13.8 years) after the index procedure. The effect of clinical risk factors on the risk of reintervention was analyzed by the Kaplan-Meier estimator and the Cox proportional hazards model. For molecular analyses, we examined the mutation profiles of 133 formalin-fixed paraffin-embedded leiomyoma samples from 33 patients with repeat operations. We screened the tumors for the 3 primary leiomyoma driver alterations-mediator complex subunit 12 mutations, high mobility group AT-hook 2 overexpression, and fumarate hydratase-deficiency-utilizing Sanger sequencing and immunohistochemistry. To further assess the clonal relationship of the tumors, we executed whole-exome sequencing for 52 leiomyomas from 21 patients who exhibited the same driver alteration in tumors obtained from multiple procedures. RESULTS Reintervention rate at 11.4 years after myomectomy was 20% (46/234). Number of leiomyomas removed at the index myomectomy was a risk factor (hazard ratio 1.21; 95% confidence interval 1.09-1.34). Age at index myomectomy (hazard ratio 0.94; 95% confidence interval 0.89-0.99) and postoperative parity (hazard ratio 0.23; 95% confidence interval 0.09-0.60) were protective factors. Molecular characterization of tumors from index and nonindex operations confirmed a clonal relationship of the tumors in 3/33 (9%) patients. None of the leiomyomas harboring a mediator complex subunit 12 mutation-the most common leiomyoma driver-were confirmed clonally related. Fumarate hydratase-deficiency was detected in repeat leiomyomas from 3/33 (9%) patients. All these patients harbored a germline fumarate hydratase mutation, which is distinctive for the hereditary leiomyomatosis and renal cell cancer syndrome. Finally, we identified 3 (3/33; 9%) patients with 2 tumors each displaying somatic mutations in a recently identified novel leiomyoma driver gene, YEATS domain-containing protein 4. All YEATS domain-containing protein 4 mutations were different and thus the tumors were not clonally related. CONCLUSION Our study shows that reintervention is common after surgical myomectomy. Uterine leiomyomas typically develop independently, but some share a clonal origin. Repeat leiomyoma occurrence may be due to genetic predisposition, such as a germline fumarate hydratase mutation. Distinct somatic YEATS domain-containing protein 4 mutations identified in multiple leiomyomas from the same patient indicate a possible role for YEATS domain-containing protein 4 in repeat leiomyomas.
Collapse
Affiliation(s)
- Sara Khamaiseh
- Applied Tumor Genomics Research Program, University of Helsinki, Helsinki, Finland; Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Anna Äyräväinen
- Applied Tumor Genomics Research Program, University of Helsinki, Helsinki, Finland; Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland; Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
| | - Maare Arffman
- Applied Tumor Genomics Research Program, University of Helsinki, Helsinki, Finland; Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Siiri Reinikka
- Applied Tumor Genomics Research Program, University of Helsinki, Helsinki, Finland; Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Miika Mehine
- Applied Tumor Genomics Research Program, University of Helsinki, Helsinki, Finland; Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Päivi Härkki
- Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
| | - Ralf Bützow
- Applied Tumor Genomics Research Program, University of Helsinki, Helsinki, Finland; Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland; Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Annukka Pasanen
- Applied Tumor Genomics Research Program, University of Helsinki, Helsinki, Finland; Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Pia Vahteristo
- Applied Tumor Genomics Research Program, University of Helsinki, Helsinki, Finland; Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| |
Collapse
|
9
|
Batra N, Rekhi B, Menon S, Mittal N, Deodhar KK. An octad of fumarate hydratase-deficient uterine leiomyomas: Case series with review of literature from a single institution. INDIAN J PATHOL MICR 2025; 68:69-78. [PMID: 39011604 DOI: 10.4103/ijpm.ijpm_356_24] [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: 05/04/2024] [Accepted: 06/04/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Fumarate hydratase (FH)-deficient (FH-d) leiomyomas are included in the recent World Health Organization fascicle of the female genital tumors. These are known to be associated with hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome. The tumors can be diagnosed based on certain histopathological features, along with loss of immunohistochemical expression of FH immunostain in most tumors. Currently, there is no documentation on these tumors from our subcontinent. AIMS We analyzed eight FH-d leiomyomas diagnosed at our institute. RESULTS The most common presentation was vaginal bleeding (menorrhagia). Pelvic ultrasonogram revealed multiple fibroids in most patients except in two, who harbored a single fibroid. The size of these fibroids ranged from 3 to 19 cm. Five patients underwent myomectomies, while three underwent a total abdominal hysterectomy and bilateral salphino-ophorectomy. The most consistently observed histopathological features were hemangiopericytomatous vascular patterns, cytoplasmic globules, increased cellularity, distinct eosinophilic nucleoli, and cytological atypia (8/8, 100% tumors), followed by multinucleate giant cells and perivascular edema, seen in 62% and 50% tumors, respectively. Immunohistochemically, all tumors were positive for desmin, smooth muscle actin, and h-caldesmon and showed loss of FH immunostain, along with low Ki-67/MIB1. None of those patients had any renal or cutaneous manifestations. CONCLUSIONS This constitutes the first such study from the Indian subcontinent and reinforces that although uterine leiomyomas constitute an integral component of the diagnosis of HLRCC syndrome, these occur in the absence of renal or cutaneous manifestations. FH-d uterine leiomyomas are more likely sporadic and could be a false alarm to raise the possibility of HLRCC with their exclusive presence.
Collapse
Affiliation(s)
- Nishtha Batra
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI) University, Parel, Mumbai, Maharashtra, India
| | | | | | | | | |
Collapse
|
10
|
Wei G, Chen J, Gong X, Zhang D. Fumarate Hydratase-Deficient Leiomyoma with Double Mutation Sites in the FH Gene: A Rare Case Report and Literature Review. Int J Womens Health 2024; 16:2137-2141. [PMID: 39677552 PMCID: PMC11640030 DOI: 10.2147/ijwh.s444069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024] Open
Abstract
Background Fumarate Hydratase (FH)-deficient uterine leiomyomas are a rare type of uterine fibroid associated with somatic or germline mutations in the FH gene. Herein, we report a case of FH-deficient uterine leiomyoma with a double-site mutation of FH in a 41-year-old woman. Case Presentation The woman was found to have an intrauterine mass during a routine physical examination two years prior. She had no previous medical history or family history of genetic diseases. Ultrasound examination revealed a slightly hypoechoic mass on the posterior wall of the uterus, approximately 4 cm × 4.1 cm in size, suggesting the possibility of a uterine fibroid. The patient opted for regular annual follow-ups and received no specific treatment. However, during the subsequent two years of follow-up, the mass was found to increase in size annually. The patient then came to our hospital and underwent laparoscopic myomectomy. Postoperative pathology indicated that the tumor was negative for FH but positive for 2-succinocysteine (2SC), suggesting a potential diagnosis of FH-deficient leiomyoma. Sanger sequencing analysis demonstrated that the leiomyoma harbored the c.724C>T (p.L242F) mutation in exon 5 and the c.1292C>T (p.T431I) mutation in exon 9 of the FH gene, further confirming the diagnosis of FH-deficient leiomyoma. Conclusion We report a rare case of FH-deficient uterine leiomyoma with double mutation sites in the FH gene. Pathological examination and genetic testing are crucial for a definitive diagnosis.
Collapse
Affiliation(s)
- Gang Wei
- Department of Oncology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, 441000, People’s Republic of China
| | - Jie Chen
- Department of Orthopedic, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, 441000, People’s Republic of China
| | - Xing Gong
- Department of Oncology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, 441000, People’s Republic of China
| | - Dongdong Zhang
- Department of Oncology, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, 441000, People’s Republic of China
| |
Collapse
|
11
|
Guberovic I, Frezza C. Functional implications of fumarate-induced cysteine succination. Trends Biochem Sci 2024; 49:775-790. [PMID: 38876954 DOI: 10.1016/j.tibs.2024.05.003] [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: 02/15/2024] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 06/16/2024]
Abstract
Mutations in metabolic enzymes are associated with hereditary and sporadic forms of cancer. For example, loss-of-function mutations affecting fumarate hydratase (FH), the tricarboxylic acid (TCA) cycle enzyme, result in the accumulation of millimolar levels of fumarate that cause an aggressive form of kidney cancer. A distinct feature of fumarate is its ability to spontaneously react with thiol groups of cysteines in a chemical reaction termed succination. Although succination of a few proteins has been causally implicated in the molecular features of FH-deficient cancers, the stoichiometry, wider functional consequences, and contribution of succination to disease development remain largely unexplored. We discuss the functional implications of fumarate-induced succination in FH-deficient cells, the available methodologies, and the current challenges in studying this post-translational modification.
Collapse
Affiliation(s)
- Iva Guberovic
- Institute for Metabolomics in Ageing, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Christian Frezza
- Institute for Metabolomics in Ageing, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Institute of Genetics, Faculty of Mathematics and Natural Sciences, Faculty of Medicine, University of Cologne, Cologne, Germany.
| |
Collapse
|
12
|
O'Connor M, Paul M, Wylie G. Cutaneous leiomyosarcoma in a case of hereditary leiomyomatosis and renal cell carcinoma syndrome. BMJ Case Rep 2024; 17:e261618. [PMID: 39179269 DOI: 10.1136/bcr-2024-261618] [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] [Indexed: 08/26/2024] Open
Abstract
Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) syndrome is an autosomal-dominant disorder that results from a germline pathogenic variant in the fumarate hydratase (FH) gene on chromosome 1, characterised by renal cell carcinoma (RCC), cutaneous leiomyoma and uterine leiomyoma. Leiomyosarcomas are reported in less than 1% of those with HLRCC. We report a case of a man in his 30s who had a long-standing plaque excised from the left upper arm after undergoing a radical nephrectomy for a fumarate-deficient RCC, with histological exam revealing a grade 1 leiomyosarcoma. Genetic testing confirmed a heterozygous pathogenic variant in the FH gene. This is a rare case of leiomyosarcoma associated with HLRCC, and our patient remains under surveillance with interval abdominal imaging and skin examination. Leiomyosarcomas are difficult to distinguish clinically from their benign counterpart; therefore, histopathological examination is paramount with a low threshold for excision.
Collapse
Affiliation(s)
| | - Maxine Paul
- Pathology Department, Queen Elizabeth University Hospital, Glasgow, UK
| | - Grant Wylie
- Dermatology Department, Queen Elizabeth University Hospital, Glasgow, UK
| |
Collapse
|
13
|
Cicchetti R, Basconi M, Litterio G, Mascitti M, Tamborino F, Orsini A, Digiacomo A, Ferro M, Schips L, Marchioni M. Advances in Molecular Mechanisms of Kidney Disease: Integrating Renal Tumorigenesis of Hereditary Cancer Syndrome. Int J Mol Sci 2024; 25:9060. [PMID: 39201746 PMCID: PMC11355026 DOI: 10.3390/ijms25169060] [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: 07/11/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Renal cell carcinoma (RCC) comprises various histologically distinct subtypes, each characterized by specific genetic alterations, necessitating individualized management and treatment strategies for each subtype. An exhaustive search of the PubMed database was conducted without any filters or restrictions. Inclusion criteria encompassed original English articles focusing on molecular mechanisms of kidney cancer. On the other hand, all non-original articles and articles published in any language other than English were excluded. Hereditary kidney cancer represents 5-8% of all kidney cancer cases and is associated with syndromes such as von Hippel-Lindau syndrome, Birt-Hogg-Dubè syndrome, succinate dehydrogenase-deficient renal cell cancer syndrome, tuberous sclerosis complex, hereditary papillary renal cell carcinoma, fumarate hydratase deficiency syndrome, BAP1 tumor predisposition syndrome, and other uncommon hereditary cancer syndromes. These conditions are characterized by distinct genetic mutations and related extra-renal symptoms. The majority of renal cell carcinoma predispositions stem from loss-of-function mutations in tumor suppressor genes. These mutations promote malignant advancement through the somatic inactivation of the remaining allele. This review aims to elucidate the main molecular mechanisms underlying the pathophysiology of major syndromes associated with renal cell carcinoma. By providing a comprehensive overview, it aims to facilitate early diagnosis and to highlight the principal therapeutic options available.
Collapse
Affiliation(s)
- Rossella Cicchetti
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Martina Basconi
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Giulio Litterio
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Marco Mascitti
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Flavia Tamborino
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Angelo Orsini
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Alessio Digiacomo
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Matteo Ferro
- Division of Urology, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20141 Milan, Italy;
| | - Luigi Schips
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Michele Marchioni
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| |
Collapse
|
14
|
Kotecha RR, Doshi SD, Knezevic A, Chaim J, Chen Y, Jacobi R, Zucker M, Reznik E, McHugh D, Shah NJ, Feld E, Aggen DH, Rafelson W, Xiao H, Carlo MI, Feldman DR, Lee CH, Motzer RJ, Voss MH. A Phase 2 Trial of Talazoparib and Avelumab in Genomically Defined Metastatic Kidney Cancer. Eur Urol Oncol 2024; 7:804-811. [PMID: 37945488 PMCID: PMC11074239 DOI: 10.1016/j.euo.2023.10.017] [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: 05/21/2023] [Revised: 08/07/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Although different kidney cancers represent a heterogeneous group of malignancies, multiple subtypes including Von Hippel-Lindau (VHL)-altered clear cell renal cell carcinoma (ccRCC), fumarate hydratase (FH)- and succinate dehydrogenase (SDH)-deficient renal cell carcinoma (RCC), and renal medullary carcinoma (RMC) are affected by genomic instability. Synthetic lethality with poly ADP-ribose polymerase inhibitors (PARPis) has been suggested in preclinical models of these subtypes, and paired PARPis with immune checkpoint blockade (ICB) may achieve additive and/or synergistic effects in patients with previously treated advanced kidney cancers. OBJECTIVE To evaluate combined PARPi + ICB in treatment-refractory metastatic kidney cancer. DESIGN, SETTING, AND PARTICIPANTS We conducted a single-center, investigator-initiated phase 2 trial in two genomically selected advanced kidney cancer cohorts: (1) VHL-altered RCC with at least one prior ICB agent and one vascular endothelial growth factor (VEGF) inhibitor, and (2) FH- or SDH-deficient RCC with at least one prior ICB agent or VEGF inhibitor and RMC with at least one prior line of chemotherapy. INTERVENTION Patients received talazoparib 1 mg daily plus avelumab 800 mg intravenously every 14 d in 28-d cycles. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The primary endpoint was objective response rate (ORR) by Immune Response Evaluation Criteria in Solid Tumors at 4 mo, and the secondary endpoints included progression-free survival (PFS), overall survival, and safety. RESULTS AND LIMITATIONS Cohort 1 consisted of ten patients with VHL-altered ccRCC. All patients had previously received ICB. The ORR was 0/9 patients; one patient was not evaluable due to missed doses. In this cohort, seven patients achieved stable disease (SD) as the best response. The median PFS was 3.5 mo (95% confidence interval [CI] 1.0, 3.9 mo). Cohort 2 consisted of eight patients; four had FH-deficient RCC, one had SDH-deficient RCC, and three had RMC. In this cohort, six patients had previously received ICB. The ORR was 0/8 patients; two patients achieved SD as the best response and the median PFS was 1.2 mo (95% CI 0.4, 2.9 mo). The most common treatment-related adverse events of all grades were fatigue (61%), anemia (28%), nausea (22%), and headache (22%). There were seven grade 3-4 and no grade 5 events. CONCLUSIONS The first clinical study of combination PARPi and ICB therapy in advanced kidney cancer did not show clinical benefit in multiple genomically defined metastatic RCC cohorts or RMC. PATIENT SUMMARY We conducted a study to look at the effect of two medications, talazoparib and avelumab, in patients with metastatic kidney cancer who had disease progression on standard treatment. Talazoparib blocks the normal activity of molecules called poly ADP-ribose polymerase, which then prevents tumor cells from repairing themselves and growing, while avelumab helps the immune system recognize and kill cancer cells. We found that the combination of these agents was safe but not effective in specific types of kidney cancer.
Collapse
Affiliation(s)
- Ritesh R Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| | - Sahil D Doshi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Knezevic
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joshua Chaim
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yingbei Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rachel Jacobi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark Zucker
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ed Reznik
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Deaglan McHugh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neil J Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Emily Feld
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - David H Aggen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - William Rafelson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Han Xiao
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Maria I Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Darren R Feldman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
15
|
Wang L, Du R, Han L, Yang R, Li Y. A new missense mutation c.1240A>G in fumarate hydratase gene leads to uterine leiomyoma associated hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome in Chinese. Transl Oncol 2024; 45:101963. [PMID: 38663218 PMCID: PMC11063639 DOI: 10.1016/j.tranon.2024.101963] [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: 02/29/2024] [Revised: 03/27/2024] [Accepted: 04/15/2024] [Indexed: 05/05/2024] Open
Abstract
OBJECTIVE This study presents a detailed analysis of the clinical and genetic characteristics of uterine leiomyoma associated with Hereditary Leiomyomatosis and Renal Cell Cancer (HLRCC), combined with exploration of family history, pathology, and management procedures, supported by thorough evidence collection. METHODS Blood samples were collected from the proband, and the pathogenic variant was verified using Sanger sequencing. A comprehensive review of family history, FH deficiency pathology, FH and 2SC immunohistochemistry staining was conducted. Functional evidence was derived from clinical and genetic information, supplemented by a literature collection and mutation was reclassified based on ACMG/AMP guidelines. RESULTS The study successfully identifies a novel missense mutation (c.1240A>G; p.Lys414Glu) in exon 9 of FH, with no prior reports in existing databases. The patient's phenotype and family history, coupled with evidence collected from the literature, contribute to the preliminary determination of the variant as likely pathogenic. We also emphasize that the importance of combining FH-deficient morphology and immunohistochemical staining with 2SC for enhanced sensitivity. CONCLUSION This research adds a novel missense mutation to the repertoire of FH gene variants, emphasizing its likely pathogenic nature based on a multidimensional analysis of phenotype, family history, and literature evidence. The findings enhance our understanding of the genetic landscape associated with FH and underscore the importance of thorough characterization for accurate variant classification.
Collapse
Affiliation(s)
- Li Wang
- Department of Gynecology & Obstetrics, Liaocheng People's Hospital, School of Medicine, Liaocheng University, PR China
| | - Ran Du
- Department of Pathology, Liaocheng People's Hospital, PR China
| | - Lin Han
- Department of Pathology, Liaocheng People's Hospital, PR China
| | - Rui Yang
- Biomedical Laboratory, School of Medicine, Liaocheng University, PR China
| | - Yingxue Li
- Department of Pathology, Liaocheng People's Hospital, PR China.
| |
Collapse
|
16
|
Glennon KI, Endo M, Usui Y, Iwasaki Y, Breau RH, Kapoor A, Lathrop M, Tanguay S, Momozawa Y, Riazalhosseini Y. Germline Susceptibility to Renal Cell Carcinoma and Implications for Genetic Screening. JCO Precis Oncol 2024; 8:e2400094. [PMID: 39088769 DOI: 10.1200/po.24.00094] [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: 02/09/2024] [Revised: 03/31/2024] [Accepted: 06/11/2024] [Indexed: 08/03/2024] Open
Abstract
PURPOSE Genetic susceptibility to nonsyndromic renal cell carcinoma (RCC) remains poorly understood, especially for different histological subtypes, as does variations in genetic predisposition in different populations. The objectives of this study were to identify risk genes for RCC in the Canadian population, investigate their clinical significance, and evaluate variations in germline pathogenic variants (PVs) among patients with RCC across the globe. MATERIALS AND METHODS We conducted targeted sequencing of 19 RCC-related and 27 cancer predisposition genes for 960 patients with RCC from Canada and identified genes enriched in rare germline PVs in RCC compared with cancer-free controls. We combined our results with those reported for patients from Japan, the United Kingdom, and the United States to investigate PV variations in different populations. Furthermore, we evaluated the performance of referral criteria for genetic screening for including patients with rare PVs. RESULTS We identified 39 germline PVs in 56 patients (5.8%) from the Canadian cohort. Compared with cancer-free controls, PVs in CHEK2 (odds ratio [OR], 4.8 [95% CI, 2.7 to 7.9], P = 3.94 × 10-5) and ATM (OR, 4.5 [95% CI, 2.0 to 8.7], P = .016) were significantly enriched in patients with clear cell, whereas PVs in FH (OR, 215.1 [95% CI, 64.4 to 597.8], P = 6.14 × 10-9) were enriched in patients with non-clear cell RCCs. PVs in BRCA1, BRCA2, and ATM were associated with metastasis (P = .003). Comparative analyses showed an enrichment of TP53 PVs in patients from Japan, of CHEK2 and ATM in patients from Canada, the United States and the United Kingdom, and of FH and BAP1 in the United States. CONCLUSION CHEK2, ATM, and FH are risk genes for RCC in the Canadian population, whereas PVs in BRCA1/2 and ATM are associated with risk of metastasis. Globally, clinical guidelines for genetic screening in RCC fail to include more than 70% of patients with rare germline PVs.
Collapse
Affiliation(s)
- Kate I Glennon
- Department of Human Genetics, McGill University, Montreal, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Canada
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mikiko Endo
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yoshiaki Usui
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yusuke Iwasaki
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | | | - Anil Kapoor
- Juravinski Cancer Centre, McMaster University, Hamilton, Canada
- Deceased
| | - Mark Lathrop
- Department of Human Genetics, McGill University, Montreal, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Canada
| | - Simon Tanguay
- Department of Surgery, Division of Urology, McGill University, Montreal, Canada
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yasser Riazalhosseini
- Department of Human Genetics, McGill University, Montreal, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Canada
| |
Collapse
|
17
|
Carmona-Rocha E, Rusiñol L, Yélamos O. Painful flesh-coloured papules in a middle-aged woman. Clin Exp Dermatol 2024; 49:658-660. [PMID: 38183665 DOI: 10.1093/ced/llae007] [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: 10/07/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 01/08/2024]
Abstract
We report a woman in her 60s who presented with multiple firm, flesh-coloured papules progressively developing on her neckline and chest. This case illustrates the constellation of clinical signs that should draw the attention of the dermatologist to suspect a tumour predisposition syndrome, highlighting the importance of early diagnosis and management for these patients.
Collapse
Affiliation(s)
- Elena Carmona-Rocha
- Dermatology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Lluís Rusiñol
- Dermatology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Oriol Yélamos
- Dermatology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| |
Collapse
|
18
|
Kipnis LM, Breen KM, Koeller DR, Levine AS, Yang Z, Jun H, Tayob N, Stokes SM, Hayes CP, Ghazani AA, Hill SJ, Rana HQ. Germline and Somatic Fumarate Hydratase Testing in Atypical Uterine Leiomyomata. Cancer Prev Res (Phila) 2024; 17:201-208. [PMID: 38638033 PMCID: PMC11439430 DOI: 10.1158/1940-6207.capr-23-0535] [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] [Received: 12/30/2023] [Revised: 02/20/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024]
Abstract
Women with germline pathogenic variants (PV) in the fumarate hydratase (FH) gene develop cutaneous and uterine leiomyomata and have an increased risk of developing aggressive renal cell carcinomas. Many of these women are unaware of their cancer predisposition until an atypical uterine leiomyoma is diagnosed during a myomectomy or hysterectomy, making a streamlined genetic counseling process after a pathology-based atypical uterine leiomyoma diagnosis critical. However, the prevalence of germline pathogenic/likely PVs in FH among atypical uterine leiomyomata cases is unknown. To better understand FH germline PV prevalence and current patterns of genetic counseling and germline genetic testing, we undertook a retrospective review of atypical uterine leiomyomata cases at a single large center. We compared clinical characteristics between the FH PV, FH wild-type (WT), and unknown genetic testing cohorts. Of the 144 cases with atypical uterine leiomyomata with evaluable clinical data, only 49 (34%) had documented genetic test results, and 12 (8.3%) had a germline FH PV. There were 48 IHC-defined FH-deficient cases, of which 41 (85%) had FH testing and nine had a germline FH PV, representing 22% of the tested cohort and 18.8% of the FH-deficient cohort. Germline FH PVs were present in 8.3% of evaluable patients, representing 24.5% of the cohort that completed genetic testing. These data highlight the disconnect between pathology and genetic counseling, and help to refine risk estimates that can be used when counseling patients with atypical uterine leiomyomata. PREVENTION RELEVANCE Women diagnosed with fumarate hydratase (FH)-deficient uterine leiomyomata are at increased risk of renal cancer. This work suggests a more standardized pathology-genetic counseling referral pathway for these patients, and that research on underlying causes of FH-deficient uterine leiomyomata in the absence of germline FH pathogenic/likely pathogenic variants is needed.
Collapse
Affiliation(s)
- Lindsay M. Kipnis
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Katelyn M. Breen
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Diane R. Koeller
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alison Schwartz Levine
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Zelei Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hyeji Jun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nabihah Tayob
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Samantha M. Stokes
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Connor P. Hayes
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA, USA
| | - Arezou A. Ghazani
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA, USA
| | - Sarah J. Hill
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Huma Q. Rana
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Przybycin CG. Papillary Renal Cell Carcinoma: Evolving Classification by Combined Morphologic and Molecular Means. Adv Anat Pathol 2024; 31:147-156. [PMID: 38329413 DOI: 10.1097/pap.0000000000000434] [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: 02/09/2024]
Abstract
Papillary renal cell carcinoma classification has evolved as a result of attentive morphologic observations by pathologists coupled with specific immunohistochemical, molecular, and clinical data. Refinement of this relatively common diagnostic category of renal neoplasia has resulted in the parsing out of specific renal cell carcinoma subtypes that no longer belong in the papillary renal cell carcinoma category and can have distinct familial and prognostic implications (eg, fumarate hydratase (FH)-deficient renal cell carcinomas). In addition, evolving classification has enabled more accurate diagnosis by surgical pathologists (through the description of recognizable morphologic variants). In many cases, molecular findings have aided and confirmed morphologic categorization. The combination of morphologic and molecular findings continues to provide important prognostic information for patients and their clinicians.
Collapse
Affiliation(s)
- Christopher G Przybycin
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
20
|
Shi W, Liu Y, Aisagbonhi O, Roma AA, Hasteh F, Zare SY, Fadare O. Fumarate Hydratase-Deficient Leiomyoma of the Uterine Corpus: Comparative Morphologic Analysis of Protein-Deficient Tumors With and Without Pathogenic Germline Fumarate Hydratase Gene Mutations. Int J Surg Pathol 2024; 32:340-355. [PMID: 37312573 DOI: 10.1177/10668969231180285] [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] [Indexed: 06/15/2023]
Abstract
Deficiency of fumarate hydratase (FH) protein expression in uterine corpus leiomyomas may be attributable to either germline or somatic mutations of the FH gene, the former being definitional for the hereditary leiomyomatosis and renal cell cancer syndrome. The authors assess whether, using previously reported FH-associated morphologic features, FH protein-deficient uterine corpus leiomyomas associated with a pathogenic germline mutations of the FH gene (group 1) are distinguishable from FH protein-deficient uterine corpus leiomyomas without such mutations (and whose FH protein loss is presumed to be attributable to somatic/epigenetic inactivation or other unknown phenomena: group 2). Groups 1 and 2 were compared regarding a variety of clinicopathologic features, including 7 core "FH-associated" tumoral morphologic features: staghorn vasculature; alveolar-type edema; bizarre nuclei; chain-like tumor nuclei; hyaline cytoplasmic globules; prominent nucleoli, intranuclear inclusions, and perinucleolar halos; and prominent eosinophilic/fibrillary cytoplasm. Among 2418 patients diagnosed with uterine corpus leiomyoma during the study period, FH-associated morphologic features were reported in 1.5% (37 patients), and FH immunohistochemistry was performed in 29 (1.19%). Fourteen (48.27%) of the 29 patients showed FH protein deficiency by immunohistochemistry. Twelve patients underwent germline testing, of which 8 (66.7%) were classified as group 1 and 4 (33.3%) as group 2. FH protein-deficient tumors were larger (10.44 vs 4.08 cm, P = 0.01) and associated with younger patients (42.05 vs 47.97, P = 0.004) than 370 randomly selected uterine leiomyoma controls. Groups 1 and 2 showed no significant differences in patient age and tumor size. In group 1 tumors, the FH-associated morphologic features were generally present diffusely; all group 1 tumors displayed ≥5 FH-associated features, whereas all group 2 tumors displayed <5 FH-associated features (means 6.5 ± 0.53 vs 3.5 ± 1.00, P < 0.001). Notably, eosinophilic/fibrillary cytoplasm and alveolar-type edema were each significantly more prevalent in group 1 tumors than group 2 tumors (P = 0.018 for both). No single morphologic feature was found to be completely sensitive and specific in making the distinction between group 1 and 2 tumors. Our findings suggest that groups 1 and 2 are unlikely to be morphologically distinguishable by individual morphologic features. Whether there is a combination of features that can reliably make this distinction is unclear and will require additional studies with larger cohorts.
Collapse
Affiliation(s)
- Wangpan Shi
- Department of Pathology, University of California San Diego Health System, San Diego, CA, USA
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA, USA
- Accelerated Clinical Experience Scholar, Health Sciences International, University of California San Diego School of Medicine, La Jolla, CA, USA
- Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Yu Liu
- Department of Pathology, University of California San Diego Health System, San Diego, CA, USA
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA, USA
- Accelerated Clinical Experience Scholar, Health Sciences International, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Omonigho Aisagbonhi
- Department of Pathology, University of California San Diego Health System, San Diego, CA, USA
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Andres A Roma
- Department of Pathology, University of California San Diego Health System, San Diego, CA, USA
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Farnaz Hasteh
- Department of Pathology, University of California San Diego Health System, San Diego, CA, USA
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Somaye Y Zare
- Department of Pathology, University of California San Diego Health System, San Diego, CA, USA
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Oluwole Fadare
- Department of Pathology, University of California San Diego Health System, San Diego, CA, USA
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
21
|
Upadhyay S, Dubey PK. Gene variants polymorphisms and uterine leiomyoma: an updated review. Front Genet 2024; 15:1330807. [PMID: 38572418 PMCID: PMC10987786 DOI: 10.3389/fgene.2024.1330807] [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: 10/31/2023] [Accepted: 02/22/2024] [Indexed: 04/05/2024] Open
Abstract
Uterine leiomyoma, commonly referred to as fibroids, is a benign tumor that develops in the muscular wall of the uterus. These growths are non-cancerous and can vary in size, ranging from tiny nodules to larger masses. Uterine leiomyomas often occur during a woman's reproductive years and can lead to symptoms such as heavy menstrual bleeding, pelvic pain, and pressure on nearby organs. While the exact cause is not fully understood, hormonal factors, particularly estrogen and progesterone, are believed to play a role in their development. The exploration of connections between genetic variants and uterine leiomyoma has captivated scientific attention for numerous years. The results from investigations remain a subject of intrigue within the scientific community. To date, the findings regarding the relationships between single nucleotide polymorphisms (SNPs) and uterine leiomyoma have exhibited some inconsistencies. However, amidst these inconsistencies, several promising outcomes have emerged that hold the potential to shape future research endeavors. These promising leads could pave the way for the development of innovative targeted therapies and novel prognostic biomarkers. This review specifically centers on accentuating the existing literature data concerning genetic variants that have been explored for their potential connections to uterine leiomyoma. Additionally, it underscores the prospects of employing genetic variations as diagnostic and prognostic biomarkers for individuals diagnosed with uterine leiomyoma.
Collapse
Affiliation(s)
| | - Pawan K. Dubey
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
22
|
Ono A, Nakamura M, Takada T, Miura S, Tsuru I, Izumi T, Kusakabe M, Mitarai S, Nagashima Y, Kume H, Morikawa T, Shiga Y. Bilateral fumarate hydratase deficient renal cell carcinoma in a patient with hereditary leiomyomatosis and renal cell cancer syndrome. IJU Case Rep 2024; 7:144-147. [PMID: 38440695 PMCID: PMC10909150 DOI: 10.1002/iju5.12688] [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: 09/20/2023] [Accepted: 12/17/2023] [Indexed: 03/06/2024] Open
Abstract
Introduction Patients with hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome have high risks of uterine and cutaneous leiomyomas and renal cell carcinoma (RCC), which are caused by germline mutation of the fumarate hydratase (FH) gene. RCC lesions are mostly high-grade tumors with a poor prognosis. Case presentation A 37-year-old man who had previously undergone treatment for a left RCC was referred to our hospital with a diagnosis of right RCC. Robot-assisted partial nephrectomy was performed, and the pathological diagnosis revealed fumarate hydratase (FH)-deficient RCC. The left RCC, which was originally diagnosed as mucinous tubular and spindle cell carcinoma, was reviewed and diagnosed as FH-deficient RCC. The patient's father and uncle both died of RCC, and the father's tumor was also immunohistochemically proven to be FH-deficient RCC. Conclusion HLRCC-related RCC should be considered in a differential diagnosis of young patients with a family history of RCC.
Collapse
Affiliation(s)
- Akihiro Ono
- Department of UrologyNTT Medical Center TokyoTokyoJapan
| | | | - Takuya Takada
- Department of Diagnostic PathologyNTT Medical Center TokyoTokyoJapan
| | - Sakiko Miura
- Department of Diagnostic PathologyNTT Medical Center TokyoTokyoJapan
| | - Ibuki Tsuru
- Department of UrologyNTT Medical Center TokyoTokyoJapan
| | - Taro Izumi
- Department of UrologyNTT Medical Center TokyoTokyoJapan
| | | | | | - Yoji Nagashima
- Department of Surgical PathologyTokyo Women's Medical UniversityTokyoJapan
| | - Haruki Kume
- Department of Urology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Teppei Morikawa
- Department of Diagnostic PathologyNTT Medical Center TokyoTokyoJapan
| | | |
Collapse
|
23
|
Sánchez-Heras AB, Dámaso E, Castillejo A, Robledo M, Teulé A, Lázaro C, Sánchez-Martínez R, Zúñiga Á, López-Fernández A, Balmaña J, Robles L, Ramon Y Cajal T, Castillejo MI, Ibañez RP, Sevila CM, Sánchez-Mira A, Escandell I, Gómez L, Berbel P, Soto JL. Genetic and clinical characterization of a novel FH founder mutation in families with hereditary leiomyomatosis and renal cell cancer syndrome. Orphanet J Rare Dis 2024; 19:26. [PMID: 38279137 PMCID: PMC10811853 DOI: 10.1186/s13023-024-03017-z] [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: 09/22/2022] [Accepted: 01/11/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND Hereditary leiomyomatosis and renal cell cancer syndrome is a rare autosomal dominant hereditary syndrome. Previously, we published the largest cohort of FH mutation carriers in Spain and observed a highly recurrent missense heterozygous variant, FH(NM_000143.4):c.1118A > G p.(Asn373Ser), in 104 individuals from 31 apparently unrelated families. Here, we aimed to establish its founder effect and characterize the associated clinical phenotype. RESULTS Haplotype analysis confirmed that families shared a common haplotype (32/38 markers) spanning 0.61-0.82 Mb, indicating this recurrent variant was inherited from a founder ancestor. Cutaneous and uterine leiomyomatosis were diagnosed in 64.6% (64/99) and 98% (50/51) of patients, respectively, and renal cell cancer was present in 10.4% (10/96). The pathogenic FH_c.1118A > G variant is a Spanish founder mutation that originated 12-26 generations ago. We estimate that the variant may have appeared between 1370 and 1720. Individuals carrying this founder mutation had similar frequency of renal cell cancer and a higher frequency of renal cysts and leiomyomas than those in other cohorts of this syndrome. CONCLUSIONS In the Spanish province of Alicante there is a high prevalence of HLRCC because of the founder mutation FH c.1118A > G; p.(Asn373Ser). The characterization of founder mutations provides accurate and specific information regarding their penetrance and expressivity. In individuals with suspected HLRCC from the province of Alicante, genetic testing by direct analysis of the founder FH c.1118A > G; p.(Asn373Ser) mutation may be a faster and more efficient diagnostic tool compared with complete gene sequencing.
Collapse
Affiliation(s)
- Ana Beatriz Sánchez-Heras
- Cancer Genetic Counselling Unit, Medical Oncology Department, Hospital General Universitario de Elche, 03203, Elche, Alicante, Spain.
| | - Estela Dámaso
- Molecular Genetics Laboratory, Hospital General Universitario de Elche, Elche, Alicante, Spain
- Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), FISABIO-Elche Health Department, Elche, Spain
| | - Adela Castillejo
- Molecular Genetics Laboratory, Hospital General Universitario de Elche, Elche, Alicante, Spain
- Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), FISABIO-Elche Health Department, Elche, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer, Human Cancer Genetics Programme Spanish National Cancer Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, 28029, Madrid, Spain
| | - Alexandre Teulé
- Hereditary Cancer Program, Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Catalan Institute of Oncology, L'Hospitalet del Llobregat, Barcelona, Spain
| | - Conxi Lázaro
- Hereditary Cancer Program, Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Catalan Institute of Oncology, L'Hospitalet del Llobregat, Barcelona, Spain
| | - Rosario Sánchez-Martínez
- Multidisciplinary Rare Disease Unit, Internal Medicine Department, Alicante University General Hospital, Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Ángel Zúñiga
- Clinical Genetics Unit, Hospital Politécnico y Universitario La Fe, Valencia, Spain
| | - Adrià López-Fernández
- Hereditary Cancer Genetics Group, VHIO, and Medical Oncology Department, Hospital Vall D'Hebron, Barcelona, Spain
| | - Judith Balmaña
- Hereditary Cancer Genetics Group, VHIO, and Medical Oncology Department, Hospital Vall D'Hebron, Barcelona, Spain
| | - Luis Robles
- Medical Oncology Department. Hospital 12 de Octubre, Madrid, Spain
| | - Teresa Ramon Y Cajal
- Familiar Cancer Clinic, Medical Oncology Department, Santa Creu i Sant Pau Hospital, Barcelona, Spain
| | - M Isabel Castillejo
- Molecular Genetics Laboratory, Hospital General Universitario de Elche, Elche, Alicante, Spain
- Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), FISABIO-Elche Health Department, Elche, Spain
| | - Raquel Perea Ibañez
- Cancer Genetic Counselling Unit, Medical Oncology Department, Hospital General Universitario de Elche, 03203, Elche, Alicante, Spain
| | - Carmen Martínez Sevila
- Cancer Genetic Counselling Unit, Medical Oncology Department, Hospital General Universitario de Elche, 03203, Elche, Alicante, Spain
| | - Andrea Sánchez-Mira
- Molecular Genetics Laboratory, Hospital General Universitario de Elche, Elche, Alicante, Spain
- Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), FISABIO-Elche Health Department, Elche, Spain
| | - Inés Escandell
- Servicio de Dermatología, Hospital General Universitario de Elda, Elda, Alicante, Spain
| | - Luís Gómez
- Urology Department, Hospital General Universitario de Elche, Elche, Alicante, Spain
| | - Pere Berbel
- Departamento de Histología y Anatomía, Facultad de Medicina, Universidad Miguel Hernández, Sant Joan d'Alacant, Alicante, Spain
| | - José Luis Soto
- Molecular Genetics Laboratory, Hospital General Universitario de Elche, Elche, Alicante, Spain
- Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), FISABIO-Elche Health Department, Elche, Spain
| |
Collapse
|
24
|
Zhang X, Wang C, Shen D. The use of Clinicopathological, immunohistochemistry and molecular detection in the diagnosis of fumarate hydratase-deficient uterine leiomyomas. Pathol Res Pract 2024; 253:154916. [PMID: 38029712 DOI: 10.1016/j.prp.2023.154916] [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: 06/17/2023] [Revised: 09/22/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Fumarate hydratase-deficient uterine leiomyomas (FH-dUL) are rare, accounting for only 0.4-1.6% of uterine leiomyomas. FH germline mutation gene is associated with hereditary leiomyomatosis and renal cell carcinoma syndrome (HLRCC). METHODS In this study, we aim to investigate Clinicopathological features and FH mutation in FH-dUL. We performed a retrospective analysis of 300 cases of uterine leiomyoma, diagnosed from January 2017 to December 2021, within the archives of the Department of Pathology at Peking University People's Hospital. In our review of the immunohistochemical(IHC) staining was performed on 300 uSMTs to detect FH deficiency. RESULTS We identified 21cases (21/300,7%) of FH-dUL. Nineteen cases (6.33%) displayed negative FH. Twenty-one cases (7%) displayed 2SC diffuse plasma and nuclear staining. The most common FH-d morphological features included staghorn vasculature ( 100%,21/21), alveolar-pattern oedema (71.43%, 15/21), scattered bizarre nuclei (23.81%, 5/21), eosinophilic cytoplasmic (rhabdoid) inclusions (47.62%, 10/21), significant eosinophilic nucleolus with peri-nucleolus hollowing (23.81%, 5/21), ovoid nuclei sometimes arranged in chains (9.52%, 2/21). DNA sequencing for the 21 cases was performed using Next Generation Sequencing (NGS). 6 cases were detected significant variations for the FH gene, 11 cases were detected FH gene mutation forvariants of uncertain significance (VUS), and 2 cases were detected a TP53 gene mutation. No related mutations were detected in the other two cases. CONCLUSIONS FH-dUL is rare. The combination of predictive Clinicopathological evaluation,FH and 2SC IHC test, and molecular test were helpful for the screening of FH-dUL from uSMTs,or even the screening of HLRCC.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Pathology, Peking University People's Hospital, Beijing 100044, PR China
| | - Chen Wang
- Department of Pathology, Peking University People's Hospital, Beijing 100044, PR China
| | - Danhua Shen
- Department of Pathology, Peking University People's Hospital, Beijing 100044, PR China.
| |
Collapse
|
25
|
Ruan Y, Feng W, Yang C. A novel nonsense mutation in the fumarate hydratase gene in a Chinese patient with recurrent leiomyomas. F S Rep 2023; 4:410-415. [PMID: 38204953 PMCID: PMC10774885 DOI: 10.1016/j.xfre.2023.08.007] [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: 05/14/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 01/12/2024] Open
Abstract
Objective To describe a novel nonsense mutation in the fumarate hydratase (FH) gene in a Chinese patient with recurrent multiple leiomyomas. Design Case report. Setting Medical school-affiliated tertiary hospital. Patients A nulligravida patient aged 30 years with large uterine leiomyomas (ULMs) and severe anemia. Interventions Clinical evaluation, abdominal myomectomy, targeted next-generation sequencing. Main outcome measures Fumarate hydratase gene mutation in ULMs. Results A novel nonsense mutation (c.771T>G) in the FH gene was identified in this patient. This mutation is located in exon 6, which encodes the N-terminal fumarate lyase domain. It leads to a predicted truncated protein with loss of the majority of the lyase domain, resulting in FH deficiency. Conclusions Because of the recurrent multiple leiomyomas, this patient received 2 myomectomies within 5 years. On immunostaining the leiomyoma, FH deficiency was detected, and targeted next-generation sequencing revealed a novel mutation of the FH gene. This patient was at risk for early disease relapse and developing renal cancer, and close disease monitoring is recommended. Meanwhile, the expanded mutation database should benefit patients in diagnosing FH gene-associated ULMs.
Collapse
Affiliation(s)
- Yiyin Ruan
- Department of Gynecology and obstetrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Weiwei Feng
- Department of Gynecology and obstetrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Chenmin Yang
- Department of Gynecology and obstetrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
26
|
Crooks DR, Cawthon GM, Fitzsimmons CM, Perez M, Ricketts CJ, Vocke CD, Yang Y, Middelton L, Nielsen D, Schmidt LS, Tandon M, Merino MJ, Ball MW, Meier JL, Batista PJ, Linehan WM. Cryptic splice mutation in the fumarate hydratase gene in patients with clinical manifestations of Hereditary Leiomyomatosis and Renal Cell Cancer. Hum Mol Genet 2023; 32:3135-3145. [PMID: 37561409 PMCID: PMC10630246 DOI: 10.1093/hmg/ddad131] [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: 07/07/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023] Open
Abstract
Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is an autosomal dominant condition characterized by the development of cutaneous and uterine leiomyomas and risk for development of an aggressive form of papillary renal cell cancer. HLRCC is caused by germline inactivating pathogenic variants in the fumarate hydratase (FH) gene, which encodes the enzyme that catalyzes the interconversion of fumarate and L-malate. We utilized enzyme and protein mobility assays to evaluate the FH enzyme in a cohort of patients who showed clinical manifestations of HLRCC but were negative for known pathogenic FH gene variants. FH enzyme activity and protein levels were decreased by 50% or greater in three family members, despite normal FH mRNA expression levels as measured by quantitative PCR. Direct Nanopore RNA sequencing demonstrated 57 base pairs of retained intron sequence between exons 9 and 10 of polyadenylated FH mRNA in these patients, resulting in a truncated FH protein. Genomic sequencing revealed a heterozygous intronic alteration of the FH gene (chr1: 241498239 T/C) resulting in formation of a splice acceptor site near a polypyrimidine tract, and a uterine fibroid obtained from a patient showed loss of heterozygosity at this site. The same intronic FH variant was identified in an unrelated patient who also showed a clinical phenotype of HLRCC. These data demonstrate that careful clinical assessment as well as biochemical characterization of FH enzyme activity, protein expression, direct RNA sequencing, and genomic DNA sequencing of patient-derived cells can identify pathogenic variants outside of the protein coding regions of the FH gene.
Collapse
Affiliation(s)
- Daniel R Crooks
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, United States
| | - Geetha Mariah Cawthon
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, United States
| | - Christina M Fitzsimmons
- RNA Metabolism and Epitranscriptomics Unit, Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892, United States
| | - Minervo Perez
- Chemical Biology Laboratory, National Cancer Institute, 1050 Boyles St., Frederick, MD 21072, United States
| | - Christopher J Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, United States
| | - Cathy D Vocke
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, United States
| | - Ye Yang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, United States
| | - Lindsay Middelton
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, United States
| | - Debbie Nielsen
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, United States
| | - Laura S Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, 1050 Boyles St. Frederick, MD 21701, United States
| | - Mayank Tandon
- CCR Collaborative Bioinformatics Resource (CCBR), Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., 1050 Boyles St., Frederick, MD 21072, United States
| | - Maria J Merino
- Translational Surgical Pathology, Laboratory of Pathology Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, United States
| | - Mark W Ball
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, United States
| | - Jordan L Meier
- Chemical Biology Laboratory, National Cancer Institute, 1050 Boyles St., Frederick, MD 21072, United States
| | - Pedro J Batista
- RNA Metabolism and Epitranscriptomics Unit, Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892, United States
| | - William Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, United States
| |
Collapse
|
27
|
Audet SE, Koleva E, Alhameedi HN, Ashcroft S, Imran A. Unusual Cutaneous Lesions in a 53-Year-Old Female: A Potential Indicator of Underlying Renal Cell Carcinoma. Cureus 2023; 15:e48783. [PMID: 38098923 PMCID: PMC10719877 DOI: 10.7759/cureus.48783] [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] [Accepted: 11/14/2023] [Indexed: 12/17/2023] Open
Abstract
Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a rare autosomal dominant genetic disorder resulting from mutations in the fumarate hydratase (FH) gene. It is characterised by a predisposition to cutaneous and uterine leiomyomas (fibroids) and an aggressive form of renal cell carcinoma (RCC). We report the case of a 53-year-old female who presented with an unusual rash in the context of a personal and family history of uterine leiomyomas requiring hysterectomy. A skin biopsy confirmed cutaneous leiomyomas and subsequent genetic testing demonstrated a pathogenic heterozygous mutation on exon 7 of the FH gene, confirming a diagnosis of HLRCC. Due to the recognised association with renal cell carcinoma in this syndrome, abdominal imaging was performed, which excluded RCC, and the patient was commenced on lifelong surveillance with annual abdominal magnetic resonance imaging.
Collapse
Affiliation(s)
- Samuel E Audet
- Dermatology, Mid and South Essex NHS Foundation Trust, Basildon, GBR
| | - Elena Koleva
- Dermatology, Mid and South Essex NHS Foundation Trust, Southend, GBR
| | - Hayder N Alhameedi
- Internal Medicine, Mid and South Essex NHS Foundation Trust, Southend, GBR
| | - Samuel Ashcroft
- Dermatology, Mid and South Essex NHS Foundation Trust, Basildon, GBR
| | - Ayesha Imran
- Dermatology, Mid and South Essex NHS Foundation Trust, Southend, GBR
| |
Collapse
|
28
|
Bayram A, Bagbudar S, Sozen H, Onder S, Yavuz E. The Role of Morphology in Predicting Fumarate Hydratase-deficient Uterine Leiomyomas in Young Women. Appl Immunohistochem Mol Morphol 2023; 31:657-660. [PMID: 37751278 DOI: 10.1097/pai.0000000000001161] [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: 05/09/2023] [Accepted: 08/28/2023] [Indexed: 09/27/2023]
Abstract
Hereditary leiomyomatosis and renal cell carcinoma is caused by germline mutations in the fumarate hydratase (FH) gene and is associated with an increased incidence of leiomyomas and a potentially aggressive variant of renal cell carcinoma. Pathologic evaluation of uterine leiomyoma can provide an opportunity for early recognition of the syndrome. We reviewed all archived slides of the cases to identify the characteristic morphologic features described for FH-deficient leiomyomas. We performed immunohistochemistry on whole sections of patients with uterine leiomyoma to evaluate for both FH and 2-succinocysteine (2SC) expression. Of the 106 cases, 19 showed the characteristic eosinophilic nucleoli with perinuclear halos, and 24 revealed a characteristic eosinophilic cytoplasmic inclusion consisting of pink globules present within the cytoplasm. Both of these morphologic findings were present together in 15 cases, and hemangiopericytomatous vessels were detected in 23 cases. The loss of FH protein expression was detected in 14 out of 106 cases (13%), and 13 out of 106 cases (12%) were positive for 2SC. We detected 10 cases with both 2SC-positive and FH expression loss. The presence of eosinophilic nucleoli with perinuclear halos and eosinophilic cytoplasmic inclusion was associated with both loss of FH protein expression and 2SC positivity ( P < 0.001). These findings underscore the importance of hematoxylin and eosin-based predictive morphology in FH-deficient uterine leiomyomas. Therefore, morphologic assessment of uterine leiomyomas for features of FH deficiency can serve as a screening tool for hereditary leiomyomatosis and renal cell carcinoma syndrome, allowing patients to be divided according to their hereditary risk assessment.
Collapse
Affiliation(s)
| | | | - Hamdullah Sozen
- Department of Gynecological Oncology, Istanbul University, Istanbul, Turkey
| | | | | |
Collapse
|
29
|
Alkhrait S, Ali M, Kertowidjojo E, Romero IL, Hathaway F, Madueke-Laveaux OS. Investigating Fumarate Hydratase-Deficient Uterine Fibroids: A Case Series. J Clin Med 2023; 12:5436. [PMID: 37685503 PMCID: PMC10487707 DOI: 10.3390/jcm12175436] [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: 06/28/2023] [Revised: 07/27/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Uterine leiomyomas or uterine fibroids are the most common benign soft tissue tumor in reproductive-aged women. Fumarate hydratase deficient (FH-d) uterine fibroids are a rare subtype that is diagnosed only on pathologic evaluation. FH-d uterine fibroids may be the first indicator of hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome. Therefore, identifying and understanding the clinical implication and diagnosis of FH-d uterine fibroids is critical for early diagnosis of HLRCC. This case series investigates the uncommon yet significant condition of FH-d uterine fibroids. We examined the clinical manifestation, diagnostic imaging, and histopathological characteristics of FH-d uterine fibroids in five cases identified at our institution over the last ten years. All diagnoses were confirmed by pathologic evaluation after surgical treatment. Gynecologists and pathologists play a critical role in the early diagnosis of FH-d uterine fibroids and must recognize the relevant clinical and pathologic findings that raise suspicion about this diagnosis. The detection of these cases is largely dependent on the pathologist's ability to recognize unique histopathologic features. Once these characteristics are identified, it should prompt a referral to a gynecologist to consider conducting germline genetic testing. The management of FH-d uterine fibroids necessitates a multidisciplinary approach, including proper genetic screening and regular surveillance, especially for renal tumors.
Collapse
Affiliation(s)
- Samar Alkhrait
- Department of OBGYN, University of Chicago Medicine, Chicago, IL 60637, USA;
| | - Munira Ali
- College of Medicine, University of Illinois, Chicago, IL 60607, USA;
| | | | - Iris L Romero
- Department of OBGYN, University of Chicago Medicine, Chicago, IL 60637, USA;
| | - Feighanne Hathaway
- Department of Medicine, University of Chicago Medicine, Chicago, IL 60637, USA;
| | | |
Collapse
|
30
|
Mahé M, Rios-Fuller TJ, Karolin A, Schneider RJ. Genetics of enzymatic dysfunctions in metabolic disorders and cancer. Front Oncol 2023; 13:1230934. [PMID: 37601653 PMCID: PMC10433910 DOI: 10.3389/fonc.2023.1230934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Inherited metabolic disorders arise from mutations in genes involved in the biogenesis, assembly, or activity of metabolic enzymes, leading to enzymatic deficiency and severe metabolic impairments. Metabolic enzymes are essential for the normal functioning of cells and are involved in the production of amino acids, fatty acids and nucleotides, which are essential for cell growth, division and survival. When the activity of metabolic enzymes is disrupted due to mutations or changes in expression levels, it can result in various metabolic disorders that have also been linked to cancer development. However, there remains much to learn regarding the relationship between the dysregulation of metabolic enzymes and metabolic adaptations in cancer cells. In this review, we explore how dysregulated metabolism due to the alteration or change of metabolic enzymes in cancer cells plays a crucial role in tumor development, progression, metastasis and drug resistance. In addition, these changes in metabolism provide cancer cells with a number of advantages, including increased proliferation, resistance to apoptosis and the ability to evade the immune system. The tumor microenvironment, genetic context, and different signaling pathways further influence this interplay between cancer and metabolism. This review aims to explore how the dysregulation of metabolic enzymes in specific pathways, including the urea cycle, glycogen storage, lysosome storage, fatty acid oxidation, and mitochondrial respiration, contributes to the development of metabolic disorders and cancer. Additionally, the review seeks to shed light on why these enzymes represent crucial potential therapeutic targets and biomarkers in various cancer types.
Collapse
Affiliation(s)
| | | | | | - Robert J. Schneider
- Department of Microbiology, Grossman NYU School of Medicine, New York, NY, United States
| |
Collapse
|
31
|
Schwartz JW, Peyser A, Tarrash M, Goldman RH. Fumarase Deficiency and Its Effect on Infertility: A Case Series. J Reprod Infertil 2023; 24:206-211. [PMID: 37663422 PMCID: PMC10471946 DOI: 10.18502/jri.v24i3.13277] [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: 05/24/2023] [Accepted: 06/24/2023] [Indexed: 09/05/2023] Open
Abstract
Background Fumarase deficiency is an autosomal recessive condition characterized by severe neurologic abnormalities due to homozygous mutations in the fumarate hydratase (FH) gene. Heterozygous carriers of FH mutations have increased risk of developing uterine fibroids that can be associated with hereditary leiomyomatosis and renal cell cancer (HLRCC). The association between FH mutations and infertility remains uncertain. The objective of our study was to characterize the infertility diagnoses, treatments, and outcomes in women presenting to a fertility center who were found to be carriers of fumarase deficiency based on the presence of heterozygous FH mutations. Case Presentation A retrospective case series was conducted including 10 women presenting to an academic fertility center who were found to be FH carriers based on genetic carrier screening. Of the 9 women who were engaged in further workup, 2 had imaging results consistent with uterine fibroids. One woman underwent hysteroscopic myomectomy prior to two courses of ovulation induction with timed intercourse (OI/TIC) followed by one successful cycle of IVF. Of the remaining patients, only 1 woman successfully delivered after a cycle of ovulation induction with intrauterine insemination (OI/IUI). Other patients pursuing OI/IUI, OI/TIC, or monitored natural cycles had unsuccessful experiences. Conclusion Patients with infertility who are offered genetic testing should be screened for FH mutations, as the carriers are at risk of developing HLRCC-associated uterine fibroids, which can influence fertility and pregnancy. Additional research is needed to investigate the impacts of FH mutations on infertility.
Collapse
Affiliation(s)
- Jessica Wesley Schwartz
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hofstra University, New York, USA
| | - Alexandra Peyser
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, Northwell Health Fertility, North Shore University Hospital, New York, USA
| | - Miriam Tarrash
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, Northwell Health Fertility, North Shore University Hospital, New York, USA
| | - Randi Heather Goldman
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hofstra University, New York, USA
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, Northwell Health Fertility, North Shore University Hospital, New York, USA
| |
Collapse
|
32
|
Helbig D, Dippel E, Erdmann M, Frisman A, Kage P, Leiter U, Mentzel T, Seidel C, Weishaupt C, Ziemer M, Ugurel S. S1-Leitlinie dermales und subkutanes Leiomyosarkom. J Dtsch Dermatol Ges 2023; 21:555-564. [PMID: 37183752 DOI: 10.1111/ddg.14989_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Affiliation(s)
- Doris Helbig
- Klinik und Poliklinik für Dermatologie und Venerologie, Universitätsklinikum Köln
| | - Edgar Dippel
- Klinik für Dermatologie und Venerologie, Klinikum der Stadt Ludwigshafen
| | - Michael Erdmann
- Hautklinik, Universitätsklinikum Erlangen, CCC Erlangen EMN, Erlangen
| | - Alexander Frisman
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Leipzig
| | - Paula Kage
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Leipzig
| | - Ulrike Leiter
- Zentrum für Dermatoonkologie, Universitäts-Hautklinikum, Eberhard-Karls-Universität Tübingen
| | - Thomas Mentzel
- MVZ Dermatopathologie Friedrichshafen/Bodensee, Friedrichshafen
| | - Clemens Seidel
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Leipzig
| | | | - Mirjana Ziemer
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Leipzig
| | - Selma Ugurel
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Essen
| |
Collapse
|
33
|
Iliopoulos O. Diseases of Hereditary Renal Cell Cancers. Urol Clin North Am 2023; 50:205-215. [PMID: 36948667 DOI: 10.1016/j.ucl.2023.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Germline mutations in tumor suppressor genes and oncogenes lead to hereditary renal cell carcinoma (HRCC) diseases, characterized by a high risk of RCC and extrarenal manifestations. Patients of young age, those with a family history of RCC, and/or those with a personal and family history of HRCC-related extrarenal manifestations should be referred for germline testing. Identification of a germline mutation will allow for testing of family members at risk, as well as personalized surveillance programs to detect the early onset of HRCC-related lesions. The latter allows for more targeted and therefore more effective therapy and better preservation of renal parenchyma.
Collapse
Affiliation(s)
- Othon Iliopoulos
- VHL Comprehensive Clinical Care Center and Hemangioblastoma Center; Division of Hematology-Oncology, Department of Medicine, Massachusetts General Hospital; Center for Cancer Research, Massachusetts General Hospital Cancer Center, 149 13th Street, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
McMurtry V, Mahlow J, Coleman JF, Deftereos G, Jattani R, Bastien RRL, Durtschi J, Jarboe E, Lomo L, Sirohi D. Morphologic Characteristics and Mutational Analysis of Fumarate Hydratase Deficient Kidney and Smooth Muscle Tumors. Am J Clin Pathol 2023; 159:164-171. [PMID: 36495298 DOI: 10.1093/ajcp/aqac148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/25/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Fumarate hydratase (FH)-deficient tumors can occur due to germline or somatic mutations and have distinctive morphologic features. The aims of this study are to refine morphologic criteria and identify mutations in FH-deficient smooth muscle tumors (SMTs). METHODS The morphology of SMTs and kidney tumors submitted to a national reference laboratory for FH immunohistochemistry (IHC) was reviewed by two gynecologic and two genitourinary pathologists, respectively. Fisher exact test was used for analysis. Fourteen SMTs were sequenced using the Illumina TruSight Oncology 500 Assay. RESULTS Twenty-two kidney tumors (5 FH deficient) and 51 SMTs (27 FH deficient) were reviewed. FH-deficient kidney tumors exclusively showed cord-like growth, rhabdoid change, and absence of coagulative tumor necrosis and psammoma bodies. FH-deficient SMTs were significantly more likely to have staghorn vessels, eosinophilic cytoplasmic inclusions, schwannoma-like areas, or hereditary leiomyomatosis and renal cell cancer-like nuclei (P < .05 for each). Seven of 14 sequenced SMTs showed mutations of the FH gene and no other driver mutations. CONCLUSIONS FH-deficient SMTs submitted for FH immunohistochemistry (IHC) showed distinct morphology. Although FH IHC is used for screening of FH-deficient tumors, FH mutations were identified in only 50% of FH-deficient SMTs. This highlights the need for additional exploration of mechanisms of FH protein loss in tumors lacking FH mutations.
Collapse
Affiliation(s)
- Valarie McMurtry
- The Institute for Experimental Pathology, ARUP Laboratories, Salt Lake City, UT, USA.,Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Jonathan Mahlow
- The Institute for Experimental Pathology, ARUP Laboratories, Salt Lake City, UT, USA.,Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Joshua F Coleman
- The Institute for Experimental Pathology, ARUP Laboratories, Salt Lake City, UT, USA.,Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Georgios Deftereos
- The Institute for Experimental Pathology, ARUP Laboratories, Salt Lake City, UT, USA.,Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | | | - Roy R L Bastien
- The Institute for Experimental Pathology, ARUP Laboratories, Salt Lake City, UT, USA
| | | | - Elke Jarboe
- The Institute for Experimental Pathology, ARUP Laboratories, Salt Lake City, UT, USA.,Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Lesley Lomo
- The Institute for Experimental Pathology, ARUP Laboratories, Salt Lake City, UT, USA.,Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Deepika Sirohi
- The Institute for Experimental Pathology, ARUP Laboratories, Salt Lake City, UT, USA.,Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| |
Collapse
|
35
|
Hassan IH, Cakirli YA, Scharrer K, Willey A. Multiple painful skin papules, uterine leiomyomas and hysterectomy. Clin Exp Dermatol 2023; 48:54-56. [PMID: 36669188 DOI: 10.1093/ced/llac010] [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: 07/27/2022] [Revised: 09/03/2022] [Accepted: 09/13/2022] [Indexed: 01/21/2023]
Abstract
A 38-year-old woman presented with multiple papules and nodules. Her mother and maternal grandmother had similar lesions, and all three women had undergone hysterectomy at a young age. Histopathogical analysis revealed dense dermal nodules composed of eosinophilic spindle cells, and exposed elongated cells with abundant eosinophilic cytoplasm arranged in fascicles.
Collapse
Affiliation(s)
- Imran Haseeb Hassan
- Department of Dermatology, Pilgrim Hospital, United Lincolnshire Hospital Trusts, Boston, Lincolnshire, UK
| | - Yasar Alp Cakirli
- Department of Dermatology, Pilgrim Hospital, United Lincolnshire Hospital Trusts, Boston, Lincolnshire, UK
| | - Krisztina Scharrer
- Department of Dermatology, Pilgrim Hospital, United Lincolnshire Hospital Trusts, Boston, Lincolnshire, UK
| | - Anu Willey
- Department of Pathology, Pilgrim Hospital, United Lincolnshire Hospital Trusts, Boston, Lincolnshire, UK
| |
Collapse
|
36
|
Scharnitz T, Nakamura M, Koeppe E, Henry ML, Lowe L, Else T, Cha KB. The spectrum of clinical and genetic findings in hereditary leiomyomatosis and renal cell cancer (HLRCC) with relevance to patient outcomes: a retrospective study from a large academic tertiary referral center. Am J Cancer Res 2023; 13:236-244. [PMID: 36777509 PMCID: PMC9906083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 02/24/2022] [Indexed: 02/14/2023] Open
Abstract
Hereditary leiomyomatosis and renal cell cancer (HLRCC) is an autosomal dominant condition attributed to pathogenic variants in fumarate hydratase (FH) and presents with cutaneous leiomyomas (CLMs), uterine leiomyomas (ULMs) and renal cell cancer (RCC). The objective of this study was to characterize the spectrum of clinical and genetic findings in HLRCC at a large academic tertiary care referral center with a focus on dermatologic manifestations. Fifty-seven patients, 41 female and 16 male, with 27 unique pathogenic or likely-pathogenic FH variants were identified from 38 families. Mean age of HLRCC diagnosis was 44.4 years (range 8-82). CLMs were the primary reason for referral in 49.1% (n=28). CLMs were present in 43/56 patients who underwent full skin examination. Three of these 56 patients were diagnosed with cutaneous leiomyosarcoma. Incidence of ULMs was 37/41 female patients; no uterine leiomyosarcomas were observed. RCC was observed in 6/57 patients (mean age of diagnosis: 47.3 years (range 28-79)). CLMs predated RCC in the 3 patients diagnosed with both. Dermatologists have an opportunity to recognize cutaneous manifestations of HLRCC, including cutaneous leiomyomas and rarely cutaneous leiomyosarcomas, and refer for genetic evaluation to provide definitive diagnosis. Identification of HLRCC can promote family cascade testing and screening for RCC.
Collapse
Affiliation(s)
| | - Mio Nakamura
- Department of Dermatology, University of MichiganMI 48109, USA
| | - Erika Koeppe
- Department of Internal Medicine, University of MichiganMI 48109, USA
| | | | - Lori Lowe
- Department of Pathology, University of MichiganMI 48109, USA
| | - Tobias Else
- Department of Internal Medicine, University of MichiganMI 48109, USA
| | - Kelly B Cha
- Department of Dermatology, University of MichiganMI 48109, USA
| |
Collapse
|
37
|
Zhang W, Liu B, Wu S, Zhao L. TMT-based comprehensive proteomic profiling identifies serum prognostic signatures of acute myeloid leukemia. Open Med (Wars) 2023; 18:20220602. [PMID: 37016705 PMCID: PMC10066874 DOI: 10.1515/med-2022-0602] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/19/2022] [Accepted: 10/16/2022] [Indexed: 04/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is classified into favorable-risk, intermediate-risk, and poor-risk subtypes. This study aimed to compare the serum proteomic signatures of the three AML subtypes and identify prognostic biomarkers for AML. Serum samples from patients with favorable-risk (n = 14), intermediate-risk (n = 19), and poor-risk AMLs (n = 18) were used for the analysis of tandem mass tag (TMT) labeling-based quantitative proteomics. Comparative analysis was performed to identify differentially expressed proteins (DEPs) between groups. Prognostic proteins were screened using binary logistics regression analysis. TMT-MS/MS proteomics analysis identified 138 DEPs. Fumarate hydratase (FH), isocitrate dehydrogenase 2 (IDH2), and enolase 1 (ENO1) were significantly upregulated in poor-risk patients compared with favorable-risk patients. ELISA assay confirmed that patients with poor-risk AMLs had higher levels of IDH2, ENO1, and FH compared with intermediate-risk AML patients. Logistics analysis identified that proteins 3-hydroxyacyl-CoA dehydrogenase type-2 (HADH, odds ratio (OR) = 1.035, p = 0.010), glutamine synthetase (GLUL, OR = 1.022, p = 0.039), and lactotransferrin (LTF, OR = 1.1224, p = 0.016) were associated with poor prognosis, and proteins ENO1 (OR = 1.154, p = 0.053), FH (OR = 1.043, p = 0.059), and IDH2 (OR = 3.350, p = 0.055) were associated with AML prognosis. This study showed that AML patients had elevated levels of FH, IDH2, ENO1, LTF, and GLUL proteins and might be at high risk of poor prognosis.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Central Laboratory, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Bei Liu
- Department of Hematology, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Shiwen Wu
- Department of Laboratory Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Li Zhao
- Department of Central Laboratory, The First Hospital of Lanzhou University, #1 Donggang West Road, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
38
|
Kidney tumors associated with germline mutations of FH and SDHB show a CpG island methylator phenotype (CIMP). PLoS One 2022; 17:e0278108. [PMID: 36455002 PMCID: PMC9714951 DOI: 10.1371/journal.pone.0278108] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/10/2022] [Indexed: 12/03/2022] Open
Abstract
Germline mutations within the Krebs cycle enzyme genes fumarate hydratase (FH) or succinate dehydrogenase (SDHB, SDHC, SDHD) are associated with an increased risk of aggressive and early metastasizing variants of renal cell carcinoma (RCC). These RCCs express significantly increased levels of intracellular fumarate or succinate that inhibit 2-oxoglutarate-dependent dioxygenases, such as the TET enzymes that regulate DNA methylation. This study evaluated the genome-wide methylation profiles of 34 RCCs from patients with RCC susceptibility syndromes and 11 associated normal samples using the Illumina HumanMethylation450 BeadChip. All the HLRCC (FH mutated) and SDHB-RCC (SDHB mutated) tumors demonstrated a distinct CpG island methylator phenotype (CIMP). HLRCC tumors demonstrated an extensive and relatively uniform level of hypermethylation that showed some correlation with tumor size. SDHB-RCC demonstrated a lesser and more varied pattern of hypermethylation that overlapped in part with the HLRCC hypermethylation. Combined methylation and mRNA expression analysis of the HLRCC tumors demonstrated hypermethylation and transcription downregulation of genes associated with the HIF pathway, HIF3A and CITED4, the WNT pathway, SFRP1, and epithelial-to-mesenchymal transition and MYC expression, OVOL1. These observations were confirmed in the TCGA CIMP-RCC tumors. A selected panel of probes could identify the CIMP tumors and differentiate between HLRCC and SDHB-RCC tumors. This panel accurately detected all CIMP-RCC tumors within the TCGA RCC cohort, identifying them as HLRCC -like, and could potentially be used to create a liquid biopsy-based screening tool. The CIMP signature in these aggressive tumors could provide both a useful biomarker for diagnosis and a target for novel therapies.
Collapse
|
39
|
Taylor AS, Skala SL. Tumors masquerading as type 2 papillary renal cell carcinoma: pathologists' ever-expanding differential diagnosis for a heterogeneous group of entities. Urol Oncol 2022; 40:499-511. [PMID: 34116938 DOI: 10.1016/j.urolonc.2021.04.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 03/22/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023]
Abstract
Although papillary renal cell carcinoma has historically been classified as either type 1 or type 2, data from The Cancer Genome Atlas (TCGA) has demonstrated significant genomic heterogeneity in tumors classified as "type 2 papillary renal cell carcinoma" (T2PRCC). Papillary renal cell carcinoma is expected to have a favorable clinical course compared to clear cell renal cell carcinoma (CCRCC). However, tumors with poor outcome more similar to CCRCC were included in the T2PRCC cohort studied by the TCGA. The differential diagnosis for T2PRCC includes a variety of other renal tumors, including aggressive entities such as TFE3 translocation-associated renal cell carcinoma, TFEB-amplified renal cell carcinoma, fumarate hydratase-deficient renal cell carcinoma, high-grade CCRCC, and collecting duct carcinoma. Accurate classification of these tumors is important for prognostication and selection of therapy.
Collapse
Affiliation(s)
- Alexander S Taylor
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Stephanie L Skala
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI.
| |
Collapse
|
40
|
Khaleel S, Ricketts C, Linehan WM, Ball M, Manley B, Turajilic S, Brugarolas J, Hakimi A. 2022 WUOF/SIU International Consultation on Urological Diseases: Genetics and Tumor Microenvironment of Renal Cell Carcinoma. SOCIETE INTERNATIONALE D'UROLOGIE JOURNAL : SIUJ 2022; 3:386-396. [PMID: 38840811 PMCID: PMC11151163 DOI: 10.48083/blpv3411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Renal cell carcinoma is a diverse group of diseases that can be distinguished by distinct histopathologic and genomic features. In this comprehensive review, we highlight recent advancements in our understanding of the genetic and microenvironmental hallmarks of kidney cancer. We begin with clear cell renal cell carcinoma (ccRCC), the most common subtype of this disease. We review the chromosomal and genetic alterations that drive initiation and progression of ccRCC, which has recently been shown to follow multiple highly conserved evolutionary trajectories that in turn impact disease progression and prognosis. We also review the diverse genetic events that define the many recently recognized rare subtypes within non-clear cell RCC. Finally, we discuss our evolving understanding of the ccRCC microenvironment, which has been revolutionized by recent bulk and single-cell transcriptomic analyses, suggesting potential biomarkers for guiding systemic therapy in the management of advanced ccRCC.
Collapse
Affiliation(s)
- Sari Khaleel
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Christopher Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Mark Ball
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Brandon Manley
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States
| | - Samra Turajilic
- The Francis Crick Institute, London, United Kingdom
- Renal Unit, The Royal Marsden Hospital, London, United Kingdom
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, United States
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, United States
| | - Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, United States
| |
Collapse
|
41
|
Fakhri S, Abdian S, Moradi SZ, Delgadillo BE, Fimognari C, Bishayee A. Marine Compounds, Mitochondria, and Malignancy: A Therapeutic Nexus. Mar Drugs 2022; 20:md20100625. [PMID: 36286449 PMCID: PMC9604966 DOI: 10.3390/md20100625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022] Open
Abstract
The marine environment is important yet generally underexplored. It contains new sources of functional constituents that can affect various pathways in food processing, storage, and fortification. Bioactive secondary metabolites produced by marine microorganisms may have significant potential applications for humans. Various components isolated from disparate marine microorganisms, including fungi, microalgae, bacteria, and myxomycetes, showed considerable biological effects, such as anticancer, antioxidant, antiviral, antibacterial, and neuroprotective activities. Growing studies are revealing that potential anticancer effects of marine agents could be achieved through the modulation of several organelles. Mitochondria are known organelles that influence growth, differentiation, and death of cells via influencing the biosynthetic, bioenergetic, and various signaling pathways related to oxidative stress and cellular metabolism. Consequently, mitochondria play an essential role in tumorigenesis and cancer treatments by adapting to alterations in environmental and cellular conditions. The growing interest in marine-derived anticancer agents, combined with the development and progression of novel technology in the extraction and cultures of marine life, led to revelations of new compounds with meaningful pharmacological applications. This is the first critical review on marine-derived anticancer agents that have the potential for targeting mitochondrial function during tumorigenesis. This study aims to provide promising strategies in cancer prevention and treatment.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Sadaf Abdian
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Blake E. Delgadillo
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Carmela Fimognari
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
- Correspondence: or
| |
Collapse
|
42
|
Alzahrani AS, Alswailem M, Alghamdi B, Al-Hindi H. Fumarate Hydratase is a Novel Gene for Familial Non-Medullary Thyroid Cancer. J Clin Endocrinol Metab 2022; 107:2539-2544. [PMID: 35751867 DOI: 10.1210/clinem/dgac386] [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: 04/26/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT The majority of cases of epithelial cell-derived thyroid cancer are sporadic. Familial non-medullary thyroid cancer (FNMTC) occurs in about 5% to 9% of cases, either as a part of known syndromes such as Cowden syndrome or in the form of familial clustering of 2 or more affected family members. Hereditary leiomyoma and renal cell cancer (HLRCC) syndrome is a rare familial cancer syndrome. The underlying etiology is heterozygous germline mutations of the fumarate hydratase (FH) gene. In addition to extensive uterine and skin leiomyomas and RCC, other tumors may arise in this syndrome. However, thyroid cancer has never been described as part of HLRCC. Here, we describe a woman who presented with an aggressive poorly differentiated thyroid cancer (PDTC) and was found to have HLRCC syndrome because of a novel heterozygous germline FH mutation. RESULTS A 43-year-old woman presented with a large lower neck mass that was found to be PDTC. During her evaluation, she was found to have extensive uterine leiomyomatosis and bilateral adrenal nodules. Whole exome and subsequent Sanger sequencing of leucocyte DNA revealed a novel monoallelic nonsense FH mutation (c.760C>T, p.Q254*). Sequencing of the thyroid tumor tissue showed a biallelic loss at the same mutation site (loss of heterozygosity) and immunohistochemistry of the PDTC showed loss of FH staining in the tumor tissue, indicating the pathogenic role of this mutation in the development of PDTC in this patient. CONCLUSION Thyroid cancer is a novel feature of the FH-related HLRCC syndrome. This syndrome can be added to the rare genetic causes of syndromic FNMTC.
Collapse
Affiliation(s)
- Ali S Alzahrani
- Department of Medicine, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia
- Department of Molecular Oncology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia
| | - Meshael Alswailem
- Department of Molecular Oncology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia
| | - Balgees Alghamdi
- Department of Molecular Oncology, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia
| | - Hindi Al-Hindi
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
43
|
Major heritable renal cell carcinoma syndromes: novel treatment options and future perspectives. Curr Opin Urol 2022; 32:488-494. [PMID: 35855559 DOI: 10.1097/mou.0000000000001030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW To provide an overview of diagnosis, genetic abnormalities, clinical signs and treatment options for the major heritable renal cell carcinoma (RCC) syndromes. RECENT FINDINGS RCC in major hereditary syndromes are disorders which are typically autosomal dominant. They predispose patients to early onset of RCC and may exhibit other extrarenal manifestations. Early recognition of these diseases allows correct screening at appropriate ages as well as early detection of RCC. Moreover, expedient identification may optimize the management of extra renal manifestations as well as allow for genetic testing and screening of at-risk relatives. SUMMARY The risk of RCC in these major heritable syndromes is higher than sporadic disease. They occur at earlier age groups and can be multifocal or bilateral. Tumours are observed until at least 3 cm before any intervention, while nephron sparing surgery is widely considered as the treatment of choice except for hereditary leiomyomatosis with renal cell cancer, of which radical nephrectomy is treatment of choice. Intervention should be timeous as there is a highly reported incidence of early metastasis. Molecular therapies have been used in the setting of patients with metastasis, some of which show favourable outcomes.
Collapse
|
44
|
Li S, Li W, Yuan J, Bullova P, Wu J, Zhang X, Liu Y, Plescher M, Rodriguez J, Bedoya-Reina OC, Jannig PR, Valente-Silva P, Yu M, Henriksson MA, Zubarev RA, Smed-Sörensen A, Suzuki CK, Ruas JL, Holmberg J, Larsson C, Christofer Juhlin C, von Kriegsheim A, Cao Y, Schlisio S. Impaired oxygen-sensitive regulation of mitochondrial biogenesis within the von Hippel-Lindau syndrome. Nat Metab 2022; 4:739-758. [PMID: 35760869 PMCID: PMC9236906 DOI: 10.1038/s42255-022-00593-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/20/2022] [Indexed: 11/20/2022]
Abstract
Mitochondria are the main consumers of oxygen within the cell. How mitochondria sense oxygen levels remains unknown. Here we show an oxygen-sensitive regulation of TFAM, an activator of mitochondrial transcription and replication, whose alteration is linked to tumours arising in the von Hippel-Lindau syndrome. TFAM is hydroxylated by EGLN3 and subsequently bound by the von Hippel-Lindau tumour-suppressor protein, which stabilizes TFAM by preventing mitochondrial proteolysis. Cells lacking wild-type VHL or in which EGLN3 is inactivated have reduced mitochondrial mass. Tumorigenic VHL variants leading to different clinical manifestations fail to bind hydroxylated TFAM. In contrast, cells harbouring the Chuvash polycythaemia VHLR200W mutation, involved in hypoxia-sensing disorders without tumour development, are capable of binding hydroxylated TFAM. Accordingly, VHL-related tumours, such as pheochromocytoma and renal cell carcinoma cells, display low mitochondrial content, suggesting that impaired mitochondrial biogenesis is linked to VHL tumorigenesis. Finally, inhibiting proteolysis by targeting LONP1 increases mitochondrial content in VHL-deficient cells and sensitizes therapy-resistant tumours to sorafenib treatment. Our results offer pharmacological avenues to sensitize therapy-resistant VHL tumours by focusing on the mitochondria.
Collapse
Affiliation(s)
- Shuijie Li
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
- College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Wenyu Li
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Juan Yuan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Petra Bullova
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jieyu Wu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Xuepei Zhang
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Yong Liu
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Monika Plescher
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Javier Rodriguez
- Edinburgh Cancer Research Centre, IGMM, University of Edinburgh, Edinburgh, UK
| | - Oscar C Bedoya-Reina
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Paulo R Jannig
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Paula Valente-Silva
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Meng Yu
- Department of Medicine, Karolinska University Hospital, Stockholm, Sweden
| | | | - Roman A Zubarev
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Anna Smed-Sörensen
- Department of Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Carolyn K Suzuki
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Johan Holmberg
- Department of Molecular Biology, Faculty of Medicine, Umeå University, Umeå, Sweden
| | - Catharina Larsson
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Alex von Kriegsheim
- Edinburgh Cancer Research Centre, IGMM, University of Edinburgh, Edinburgh, UK
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Schlisio
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
45
|
Li H, Yang W, Tu X, Yu L, Huang D, Cheng Y, Chang B, Tang S, Ge H, Bao L, Zhou X, Bi R. Clinicopathological and molecular characteristics of fumarate-hydratase-deficient uterine smooth muscle tumors: A single-center study of 52 cases. Hum Pathol 2022; 126:136-145. [PMID: 35659509 DOI: 10.1016/j.humpath.2022.05.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 11/04/2022]
Abstract
AIMS The fumarate hydratase (FH) gene germline mutations cause hereditary leiomyomatosis and renal cell carcinoma syndrome (HLRCC), predisposing carriers to uterine and cutaneous leiomyomas and renal cell carcinoma (RCC). In this study, we aim to investigate morphology and the correlation between FH mutation in FH-deficient (FH-d) uterine smooth muscle tumors (uSMTs). METHODS AND RESULTS We conducted immunohistochemical staining in 161 cases of uSMTs to detect FH deficiency. We identified 52 (52/161, 32%) cases of FH-d, including 34 leiomyomas with bizarre nuclei, 10 uSMTs of uncertain malignant potential (STUMPs), 4 cellular leiomyomas, 3 usual type leiomyomas, and 1 leiomyosarcoma. Patients with FH-d were aged 24-67 years (median, 40 years). The most common FH-d morphological features included staghorn-shaped blood vessels (87%), bizarre nuclei (81%), alveolar pattern edema (65%), macronucleoli surrounded by a halo (65%), cytoplasmic eosinophilic globules (56%), and chain-like distribution of smooth muscle cells (52%). A targeted next generation sequence was performed in 11 of 52 FH-d tumors. Five (5/11, 45%) cases were found with FH germline mutations, including four leiomyomas with bizarre nuclei and one STUMP. The median age of patients with germline FH mutation was 30 years. The germline mutations including three pathogenic, one likely pathogenic, and one rare uncertain clinical significance variants. CONCLUSIONS Our results revealed that FH-d uSMTs usually exhibit the distinct morphology features and high frequency of FH germline mutations. The combination of predictive morphology evaluation, FH immunotype and molecular testing is helpful for the screening of HLRCC in uSMTs.
Collapse
Affiliation(s)
- Hui Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, People's Republic of China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, People's Republic of China
| | - Xiaoyu Tu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, People's Republic of China
| | - Lin Yu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, People's Republic of China
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, People's Republic of China
| | - Yufan Cheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, People's Republic of China
| | - Bin Chang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, People's Republic of China
| | - Shaoxian Tang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, People's Republic of China
| | - Huijuan Ge
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, People's Republic of China
| | - Longlong Bao
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, People's Republic of China
| | - Xiaoyan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, People's Republic of China
| | - Rui Bi
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, People's Republic of China.
| |
Collapse
|
46
|
Pors J, Weiel JJ, Devereaux KA, Folkins AK, Longacre TA. Fumarate Hydratase Deficiency Should be Considered in the Differential Diagnosis of Uterine and Extrauterine Smooth Muscle Tumors of Uncertain Malignant Potential (STUMP). Int J Gynecol Pathol 2022; 41:268-275. [PMID: 34108400 DOI: 10.1097/pgp.0000000000000797] [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: 11/25/2022]
Abstract
Fumarate hydratase-deficient leiomyomas (dFH leiomyomas) often display atypical pathologic features yet exhibit a benign clinical course. Recent data suggest that dFH leiomyomas may be misclassified as smooth muscle tumors of uncertain malignant potential, a category that encompasses a heterogenous subgroup of uterine neoplasms with smooth muscle differentiation and atypical features that impart ambiguity regarding their expected clinical behavior. dFH leiomyomas can be seen in the context of hereditary leiomyomatosis and renal cell carcinoma syndrome or in the sporadic setting. In this retrospective study, we sought to examine the prevalence and clinicopathologic characteristics of dFH leiomyomas in 48 tumors previously diagnosed as smooth muscle tumors of uncertain malignant potential from 38 patients. Of these 48 tumors, 3 (6.3%) occurring in 2 patients were found to be deficient for FH by immunohistochemistry, including 1 uterine and 2 extrauterine (abdominopelvic) tumors. The 3 tumors showed histologic features typical of dFH leiomyomas, including hemangiopericytoma-like vessels, edema, macronucleoli, and atypia. Neither patient developed recurrent leiomyomas or renal cell carcinoma, and both were alive without disease at last follow-up. Our data suggest that dFH leiomyomas should be considered in the differential diagnosis of smooth muscle tumors of uncertain malignant potential, even in the context of extrauterine disease. Identification of FH deficiency in these tumors supports their classification as dFH leiomyomas despite their atypical morphologic features and/or clinical presentation. Importantly, detection of dFH in these cases may identify women at increased risk for hereditary leiomyomatosis and renal cell carcinoma who would benefit from genetic counseling and consideration for FH germline testing.
Collapse
Affiliation(s)
- Jennifer Pors
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | | | | | | | | |
Collapse
|
47
|
Dawsey SJ, Gupta S. Hereditary Renal Cell Carcinoma. KIDNEY CANCER 2022. [DOI: 10.3233/kca-210008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Hereditary renal cell carcinoma (RCC) is a complex and rapidly evolving topic as there is a growing body of literature regarding inherited syndromes and mutations associated with an increased risk of RCC. OBJECTIVES: We sought to systematically review 13 hereditary syndromes associated with RCC; von Hippel-Lindau Disease associated RCC (VHLRCC), BAP-1 associated clear cell RCC (BAPccRCC), Familial non-von Hippel Lindau clear cell RCC (FccRCC), Tuberous Sclerosis Complex associated RCC (TSCRCC), Birt-Hogg-Dub e ´ Syndrome associated RCC (BHDRCC), PTEN Hamartoma Tumor Syndrome associated RCC (PHTSRCC), Microphthalmia-associated Transcription Family translocation RCC (MiTFtRCC), RCC with Chromosome 6p Amplification (TFEBRCC), Autosomal Dominant Polycystic Kidney Disease Associated RCC (ADPKDRCC), Hereditary Leiomyomatosis associated RCC (HLRCC), Succinate Dehydrogenase RCC (SDHRCC), Hereditary Papillary RCC (HPRCC), and ALK-Rearrangement RCC (ALKRCC). RESULTS: Hereditary RCC is generally associated with early age of onset, multifocal and/or bilateral lesions, and aggressive disease course. VHLRCC, BAPccRCC, FccRCC, and certain mutations resulting in SDHRCC are associated with clear cell RCC (ccRCC). HPRCC is associated with Type 1 papillary RCC. HLRCC is associated with type 2 papillary RCC. BHDRCC is associated with Chromophobe RCC. TSCRCC, PHTSRCC, MiTFtRCC, TFEBRCC, ADPKDRCC, certain SDHRCC and ALKRCC have variable histology. CONCLUSIONS: There has been tremendous advancement in our understanding of the pathophysiology of hereditary RCC. Ongoing research will refine our understanding of hereditary RCC and its therapeutic targets.
Collapse
Affiliation(s)
- Scott J. Dawsey
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Shilpa Gupta
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
48
|
Ahvenainen T, Kaukomaa J, Kämpjärvi K, Uimari O, Ahtikoski A, Mäkinen N, Heikinheimo O, Aaltonen LA, Karhu A, Bützow R, Vahteristo P. Comparison of 2SC, AKR1B10, and FH Antibodies as Potential Biomarkers for FH-deficient Uterine Leiomyomas. Am J Surg Pathol 2022; 46:537-546. [PMID: 34678832 DOI: 10.1097/pas.0000000000001826] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a tumor predisposition syndrome caused by germline fumarate hydratase (FH) mutations and characterized by uterine and cutaneous leiomyomas and renal cell cancer. Currently, there is no generally approved method to differentiate FH-deficient uterine leiomyomas from other leiomyomas. Here, we analyzed 3 antibodies (S-(2-succino)-cysteine [2SC], aldo-keto reductase family 1, member B10 [AKR1B10], and FH) as potential biomarkers. The study consisted of 2 sample series. The first series included 155 formalin-fixed paraffin-embedded uterine leiomyomas, of which 90 were from HLRCC patients and 65 were sporadic. The second series included 1590 unselected fresh frozen leiomyomas. Twenty-seven tumors were from known HLRCC patients, while the FH status for the remaining 1563 tumors has been determined by copy number analysis and Sanger sequencing revealing 45 tumors with monoallelic (n=33) or biallelic (n=12) FH loss. Altogether 197 samples were included in immunohistochemical analyses: all 155 samples from series 1 and 42 available corresponding formalin-fixed paraffin-embedded samples from series 2 (15 tumors with monoallelic and 7 with biallelic FH loss, 20 with no FH deletion). Results show that 2SC performed best with 100% sensitivity and specificity. Scoring was straightforward with unambiguously positive or negative results. AKR1B10 identified most tumors accurately with 100% sensitivity and 99% specificity. FH was 100% specific but showed slightly reduced 91% sensitivity. Both FH and AKR1B10 displayed also intermediate staining intensities. We suggest that when patient's medical history and/or histopathologic tumor characteristics indicate potential FH-deficiency, the tumor's FH status is determined by 2SC staining. When aberrant staining is observed, the patient can be directed to genetic counseling and mutation screening.
Collapse
Affiliation(s)
- Terhi Ahvenainen
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
- iCAN Digital Precision Cancer Medicine Flagship
| | - Jaana Kaukomaa
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
| | - Kati Kämpjärvi
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
| | - Outi Uimari
- Department of Obstetrics and Gynecology, Oulu University Hospital
- PEDEGO Research Unit, University of Oulu and Oulu University Hospital
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu
| | - Anne Ahtikoski
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu
- Department of Pathology, Oulu University Hospital
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland
| | - Netta Mäkinen
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
| | - Oskari Heikinheimo
- Applied Tumor Genomics Research Program
- Department of Obstetrics and Gynecology, Helsinki University Hospital
| | - Lauri A Aaltonen
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
- iCAN Digital Precision Cancer Medicine Flagship
| | - Auli Karhu
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
- iCAN Digital Precision Cancer Medicine Flagship
| | - Ralf Bützow
- Applied Tumor Genomics Research Program
- Department of Pathology, Laboratory of Helsinki University Hospital (HUSLAB), Helsinki University Hospital and University of Helsinki, Helsinki
| | - Pia Vahteristo
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
- iCAN Digital Precision Cancer Medicine Flagship
| |
Collapse
|
49
|
Abstract
Although tumourigenesis occurs due to genetic mutations, the role of epigenetic dysregulations in cancer is also well established. Epigenetic dysregulations in cancer may occur as a result of mutations in genes encoding histone/DNA-modifying enzymes and chromatin remodellers or mutations in histone protein itself. It is also true that misregulated gene expression without genetic mutations in these factors could also support tumour initiation and progression. Interestingly, metabolic rewiring has emerged as a hallmark of cancer due to gene mutations in specific metabolic enzymes or dietary/environmental factors. Recent studies report an intricate cross-talk between epigenetic and metabolic reprogramming in cancer. This review discusses the role of epigenetic and metabolic dysregulations and their cross-talk in tumourigenesis with a special focus on gliomagenesis. We also discuss the role of recently developed human embryonic stem cells/induced pluripotent stem cells-derived organoid models of gliomas and how these models are proving instrumental in uncovering human-specific cellular and molecular complexities of gliomagenesis.
Collapse
Affiliation(s)
- Bismi Phasaludeen
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Suraiya Anjum Ansari
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| |
Collapse
|
50
|
Denize T, Massa S, Valent A, Militti L, Bertolotti A, Barisella M, Rioux-Leclercq N, Malouf GG, Spreafico F, Verschuur A, van der Beek J, Tytgat L, van den Heuvel-Eibrink MM, Vujanic G, Collini P, Coulomb A. Renal cell carcinoma in children and adolescents: A retrospective study of a French-Italian series of 93 cases. Histopathology 2022; 80:928-945. [PMID: 35238063 DOI: 10.1111/his.14634] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/06/2022] [Accepted: 02/23/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Renal cell carcinomas represent 2 to 5% of kidney malignancies in children and adolescents. Appropriate diagnostic and classification are crucial for the correct management of the patients and in order to avoid inappropriate preoperative chemotherapy, which is usually recommended if a Wilms tumor is suspected. METHODS a French-Italian series of 93 renal cell carcinomas collected from 1990 to 2019 in patients aged less than 18 years old was reclassified according to the 2016 WHO classification and the latest literature. TFE3 and TFEB FISH analyses and a panel of immunohistochemical stains were applied. RESULTS The median age at diagnosis was 11 years (range: 9 months - 17 years). MiT family (MiTF) translocation renal cell carcinomas accounted for 52% of the tumors, followed by papillary renal cell carcinomas (20%) and unclassified renal cell carcinomas (13%). Other subtypes, such as SDHB-deficient and Fumarate hydratase-deficient renal cell carcinomas, represented 1 to 3% of the cases. We also described a case of ALK-rearranged renal cell carcinoma with a metanephric adenoma-like morphology. CONCLUSION A precise histological diagnosis is mandatory as targeted therapy could be applied for some RCC subtypes, i.e., MiTF-translocation and ALK-translocation renal cell carcinomas. Moreover, some RCC subtypes may be associated with a predisposition syndrome that will impact patients' and family's management and genetic counseling. A precise RCC subtype is also mandatory for the clinical management of the patients and the inclusion in new prospective clinical trials.
Collapse
Affiliation(s)
- Thomas Denize
- Department of Pathology, Sorbonne Université, Assistance Publique Hôpitaux de Paris - Hôpital Armand Trousseau, Paris, France
| | - Simona Massa
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,present address: Unit of Pathology, Azienda Ospedaliera Specialistica dei Colli Monaldi-Cotugno-CTO, Naples, Italy
| | - Alexander Valent
- Service de Génétique des tumeurs, Département de Pathologie, Institut Gustave Roussy, Villejuif, France
| | - Lucia Militti
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessia Bertolotti
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marta Barisella
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Gabriel G Malouf
- Service d'Oncologie Médicale, Institut de Cancérologie de Strasbourg, Strasbourg, France
| | - Filippo Spreafico
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Arnauld Verschuur
- Department of Pediatric Oncology, Hôpital d'enfants de la Timone, Marseille, France
| | - Justine van der Beek
- Princess Máxima Center for Pediatric Oncology, and Utrecht University, Utrecht, The Netherlands
| | - Lieve Tytgat
- Princess Máxima Center for Pediatric Oncology, and Utrecht University, Utrecht, The Netherlands
| | | | - Gordan Vujanic
- Department of Pathology, Sidra Medicine / Weill Cornell Medicine, Doha, Qatar.,Sorbonne Université, Assistance Publique Hôpitaux de Paris - Hôpital Armand Trousseau, Paris, France
| | - Paola Collini
- Soft Tissue and Bone Pathology and Pediatric Pathology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Aurore Coulomb
- Department of Pathology, Sorbonne Université, Assistance Publique Hôpitaux de Paris - Hôpital Armand Trousseau, Paris, France
| |
Collapse
|