Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
1.
Medicina (Kaunas) ; 60(5)2024 May 17.
Article in English | MEDLINE | ID: mdl-38793008

ABSTRACT

Background and Objectives: Mutations in succinate dehydrogenase (SDH) and fumarate hydratase (FH) give rise to various familial cancer syndromes, with these alterations being characteristic of certain types of histomorphologically specific leiomyomas that hold significant predictive value. Materials and Methods: This study presents two cases of uterine leiomyomas exhibiting rare histomorphological and genetic characteristics, which are crucial for prognosis and further treatment. Results: Distinct histopathological features such as marked nuclear atypia, intracellular eosinophilic globules, and abnormal intratumoral vessels raise suspicion for specific leiomyoma subtypes, which carry predictive significance for additional hereditary cancer syndromes. Immunohistochemical analysis confirmed FH/SDH deficiency in both patients, who underwent careful follow-up. Conclusions: This study describes two cases involving unusual leiomyomas, the histopathological characteristics of which may easily go unrecognized. These features hold predictive significance because their specific mutations point to additional hereditary cancer syndromes, highlighting the need for further examinations.


Subject(s)
Fumarate Hydratase , Leiomyoma , Succinate Dehydrogenase , Uterine Neoplasms , Humans , Female , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , Succinate Dehydrogenase/deficiency , Succinate Dehydrogenase/genetics , Adult , Leiomyoma/genetics , Leiomyoma/pathology , Middle Aged
2.
Cancer Prev Res (Phila) ; 17(5): 201-208, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38638033

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.


Subject(s)
Fumarate Hydratase , Genetic Testing , Germ-Line Mutation , Leiomyoma , Uterine Neoplasms , Humans , Female , Fumarate Hydratase/genetics , Fumarate Hydratase/deficiency , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , Uterine Neoplasms/diagnosis , Middle Aged , Retrospective Studies , Adult , Leiomyoma/genetics , Leiomyoma/pathology , Leiomyoma/diagnosis , Genetic Predisposition to Disease , Genetic Counseling , Leiomyomatosis/genetics , Leiomyomatosis/pathology , Leiomyomatosis/diagnosis
3.
Clin Cancer Res ; 30(11): 2571-2581, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38512114

ABSTRACT

PURPOSE: Fumarate hydratase-deficient renal cell carcinoma (FH-deficient RCC) is a rare and lethal subtype of kidney cancer. However, the optimal treatments and molecular correlates of benefits for FH-deficient RCC are currently lacking. EXPERIMENTAL DESIGN: A total of 91 patients with FH-deficient RCC from 15 medical centers between 2009 and 2022 were enrolled in this study. Genomic and bulk RNA-sequencing (RNA-seq) were performed on 88 and 45 untreated FH-deficient RCCs, respectively. Single-cell RNA-seq was performed to identify biomarkers for treatment response. Main outcomes included disease-free survival (DFS) for localized patients, objective response rate (ORR), progression-free survival (PFS), and overall survival (OS) for patients with metastasis. RESULTS: In the localized setting, we found that a cell-cycle progression signature enabled to predict disease progression. In the metastatic setting, first-line immune checkpoint inhibitor plus tyrosine kinase inhibitor (ICI+TKI) combination therapy showed satisfactory safety and was associated with a higher ORR (43.2% vs. 5.6%), apparently superior PFS (median PFS, 17.3 vs. 9.6 months, P = 0.016) and OS (median OS, not reached vs. 25.7 months, P = 0.005) over TKI monotherapy. Bulk and single-cell RNA-seq data revealed an enrichment of memory and effect T cells in responders to ICI plus TKI combination therapy. Furthermore, we identified a signature of memory and effect T cells that was associated with the effectiveness of ICI plus TKI combination therapy. CONCLUSIONS: ICI plus TKI combination therapy may represent a promising treatment option for metastatic FH-deficient RCC. A memory/active T-cell-derived signature is associated with the efficacy of ICI+TKI but necessitates further validation.


Subject(s)
Carcinoma, Renal Cell , Fumarate Hydratase , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Male , Female , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/immunology , Kidney Neoplasms/mortality , Kidney Neoplasms/therapy , Middle Aged , Aged , Adult , Lymphocyte Activation/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/adverse effects , Immunologic Memory , Prognosis , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Immunotherapy/methods , Memory T Cells/immunology , T-Lymphocytes/immunology
4.
Int J Surg Pathol ; 32(2): 340-355, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37312573

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.


Subject(s)
Carcinoma, Renal Cell , Fumarate Hydratase/deficiency , Kidney Neoplasms , Leiomyomatosis , Metabolism, Inborn Errors , Muscle Hypotonia , Psychomotor Disorders , Skin Neoplasms , Uterine Neoplasms , Humans , Female , Fumarate Hydratase/genetics , Leiomyomatosis/diagnosis , Leiomyomatosis/genetics , Germ-Line Mutation , Edema , Germ Cells , Uterine Neoplasms/genetics
5.
Zhonghua Fu Chan Ke Za Zhi ; 57(6): 435-441, 2022 Jun 25.
Article in Chinese | MEDLINE | ID: mdl-35775251

ABSTRACT

Objective: To investigate the clinicopathological features of fumarate hydratase (FH) deficiency uterine leiomyoma. Methods: The data of 38 patients with FH deficiency uterine leiomyoma were screened and analyzed. The expressions of FH, S-(2-succino)-cysteine (2SC), desmin, p16, p53, CD10 and cell proliferation associated nuclear antigen (Ki-67) proteins were detected by immunohistochemistry, and their clinicopathological features were analyzed retrospectively. Results: (1) Clinical features: the median age of the patients was (42.5±7.4) years old. Twenty-one cases (55%) of them were myomas found in physical examination, and the median maximum diameter of the tumor was 6.0 cm (range: 5.0-7.5 cm); myomectomy was performed in 23 cases (61%), total hysterectomy with or without bilateral appendages in 15 cases (39%); laparoscopic surgery in 27 cases (71%), open surgery in 11 cases (29%); none of the patients had renal cell carcinoma. (2) Histological features: atypical nuclear cells were distributed locally or diffusely, eosinophilic nucleoli and intranuclear inclusion bodies could be seen, glass like globules could be seen in the cytoplasm, nuclear division was 0-4/10 high power field (HPF), and antler like blood vessels and pulmonary edema-like changes could be seen in the stroma. Among 38 patients with FH deficiency uterine leiomyoma, FH was negative in 37 cases (97%), and positive in 1 case (3%); 2SC, desmin, p16, p53, CD10 and Ki-67 showed focal positive expression in 38 cases (100%), including 35 cases (92%) with Ki-67 index<10% and 3 cases (8%) with Ki-67 index ≥10%. (3) Follow-up: 4 cases (11%) recurred, and there was no death. There were significant differences in age, family history, distribution of atypical nuclei and mitosis number between recurrent group and non-recurrent group (all P<0.05). Conclusions: FH deficiency uterine leiomyoma is a rare tumor, which needs pathological examination,immunohistochemical examination and clinical history. Patients younger than 43 years old, with family history, histologically atypical diffuse nuclear distribution and mitotic number ≥3/10 HPF should be alert to the risk of recurrence.


Subject(s)
Fumarate Hydratase , Leiomyoma , Uterine Neoplasms , Adult , Desmin/metabolism , Female , Fumarate Hydratase/deficiency , Fumarate Hydratase/metabolism , Humans , Ki-67 Antigen/metabolism , Leiomyoma/enzymology , Leiomyoma/pathology , Leiomyoma/surgery , Metabolism, Inborn Errors/enzymology , Middle Aged , Muscle Hypotonia/enzymology , Psychomotor Disorders/enzymology , Retrospective Studies , Tumor Suppressor Protein p53 , Uterine Neoplasms/diagnosis
6.
Ars pharm ; 63(2)abr.-jun. 2022.
Article in English | IBECS | ID: ibc-202811

ABSTRACT

Introduction: Fumarase deficiency is a rare autosomal metabolic disease that curse with hypotonia, hyperlacticaemia and seizures. Diagnosis based in laboratory test might be done carefully, as most of metabolic diseases present similar symptomatology: hypotony, convulsions, lactic and pyruvic acidemia. Method: The objective is to analyse the pharmacological and nutritional approach of a neonate who presented symptoms of metabolic congenital disorders.Results:The patient is a three-month girl with heterozygote mutation in fumarase gene, who presented clinical manifestations of the altered enzyme function. She presented hyperlacticaemia, organic aciduria and alterations of amino acid levels. First diagnosis suspected was pyruvate dehydrogenase deficiency, so nutritional treatment with ketogenic diet was initiated. After medical discharge, she was hospitalized in emergency basis with cardiac arrest and metabolic decompensation. Genetic test revealed a heterozygote mutation in fumarase. Clinical symptomatology could have worsened because of the difficult diagnosis. Conclusions: Nutritional and pharmacological treatment of fumarase deficiency is considered essential for the patient’s evolution, but further researchers must be carried out to profoundly understand the mechanism underlying metabolic inborn errors. Multidisciplinary teams would manage the disease from the point of view of diverse clinician experts so the correct diagnosis and treatment would be decided with precision(AU)


Introducción: El déficit de fumarasa es una enfermedad rara del metabolismo, autosómica, que cursa con hipotonía, hiperlactacidemia y convulsiones. El diagnóstico basado en pruebas de laboratorio debe realizarse con precaución ya que la mayoría de enfermedades relacionadas con el metabolismo presentan la misma sintomatología: hipotonía, convulsiones y acidemia láctica y pirúvica. Método: Analizar retrospectivamente el manejo farmacológico y nutricional de un neonato con síntomas relacionados con errores del metabolismo congénitos.Resultados:La paciente de 3 meses de vida presentaba una mutación heterocigota en el gen de la fumarasa y síntomas relacionados con la alteración de la función enzima. La paciente presentaba hiperlactacidemia, aciduria orgánica y alteraciones analíticas de los aminoácidos. El primer diagnóstico supuesto fue un déficit de piruvato deshidrogenasa, por lo que se inició tratamiento nutricional con dieta cetogénica. Tras el alta de la paciente, volvió a ingresar por urgencias sufriendo una parada cardíaca y descompensación metabólica. El test genético reveló la presencia de una mutación heterocigota en el gen de la fumarasa. La sintomatología clínica pudo haber empeorado debido al difícil diagnóstico. Conclusiones: El tratamiento farmacológico y nutricional del déficit de fumarasa es esencial para la buena evolución del paciente, pero es necesario que se realicen más estudios para entender con profundidad el mecanismo de los errores congénitos del metabolismo Los equipos multidisciplinares permiten manejar la enfermedad desde distintos puntos de vista clínicos para un diagnóstico correcto y poder decidir el tratamiento adecuado con precisión(AU)


Subject(s)
Humans , Female , Infant , Fumarate Hydratase/deficiency , Metabolism, Inborn Errors/diet therapy , Metabolism, Inborn Errors/diagnosis , Diet, Ketogenic , Retrospective Studies
8.
Lab Med ; 53(3): e48-e50, 2022 May 05.
Article in English | MEDLINE | ID: mdl-34643235

ABSTRACT

Fumaric aciduria resulting from fumarate hydratase deficiency is a rare inherited disorder of the Krebs tricarboxylic acid cycle that is characterized by neurologic manifestations, a spectrum of brain abnormalities, and the excretion of fumaric acid in urine. We describe a 3 year old Sri Lankan boy who was referred at age 10 months with poor weight gain and hypotonia for further laboratory investigations. In addition to global developmental delay, there were noticeable dysmorphic features with a prominent forehead, low-set ears, micrognathia, and hypertelorism with persistent neutropenia. Urine organic acid assay revealed a massive elevation of fumaric acid on 2 occasions. Molecular analysis revealed a homozygous likely pathogenic missense variant, NM000143.3:c.1048C>T p. (Arg350Trp), in the FH gene, confirming the biochemical diagnosis. Our patient was the first patient in Sri Lanka molecularly diagnosed with fumaric aciduria. This case study highlights the importance of performing organic acid assays in children presenting with neurologic manifestations especially when these are suspected to have a metabolic basis.


Subject(s)
Diagnostic Tests, Routine , Muscle Hypotonia , Child , Child, Preschool , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Humans , Infant , Male , Metabolism, Inborn Errors , Muscle Hypotonia/diagnosis , Muscle Hypotonia/genetics , Psychomotor Disorders , Sri Lanka
9.
Int J Gynecol Pathol ; 41(3): 268-275, 2022 May 01.
Article in English | MEDLINE | ID: mdl-34108400

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.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Leiomyomatosis , Skin Neoplasms , Smooth Muscle Tumor , Uterine Neoplasms , Carcinoma, Renal Cell/diagnosis , Diagnosis, Differential , Female , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Humans , Kidney Neoplasms/diagnosis , Leiomyomatosis/diagnosis , Leiomyomatosis/genetics , Leiomyomatosis/pathology , Male , Metabolism, Inborn Errors , Muscle Hypotonia , Psychomotor Disorders , Retrospective Studies , Skin Neoplasms/pathology , Smooth Muscle Tumor/diagnosis , Uterine Neoplasms/pathology
10.
Medicine (Baltimore) ; 100(49): e28142, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34889279

ABSTRACT

RATIONALE: Hereditary leiomyomatosis and renal cell carcinoma is an uncommon autosomal dominant disease caused by mutations in the fumarate hydratase (FH) gene. They usually demonstrated multiple uterine myomas and preformed surgical procedures for myomectomy and/or hysterectomy 10 years earlier than sporadic myomas due to early development. This case report describes a woman with multiple uterine leiomyomas diagnosed with FH deficiency. PATIENT CONCERNS: A 37-year-old woman visited a gynecological clinic for the discovery of uterine leiomyoma for more than 1 year. The size of the largest grew from 42 × 27 × 46 to 98 × 85 × 113 mm in 1 year. She had a history of surgery for breast cancer and thyroid cancer but denied a history of uterine leiomyoma in her family. DIAGNOSIS AND INTERVENTIONS: The patient underwent successful transabdominal hysterectomy. The pathological results showed multiple uterine leiomyomas (partly cellular leiomyomas) with scattered large bizarre giant cells. Immunohistochemistry results demonstrated FH deficiency. OUTCOMES: On follow-up, the patient did not have any complications. She was finally referred to the oncologists and urologists for follow-up. LESSONS: Gynecologists should be aware that early onset uterine leiomyoma presenting as large, multiple, and symptomatic lesion, may be associated with FH deficiency.


Subject(s)
Fumarate Hydratase/genetics , Leiomyoma/surgery , Uterine Neoplasms/surgery , Adult , Carcinoma, Renal Cell , Female , Fumarate Hydratase/blood , Fumarate Hydratase/deficiency , Humans , Leiomyoma/genetics , Metabolism, Inborn Errors/blood , Muscle Hypotonia/blood , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/surgery , Psychomotor Disorders/blood , Uterine Neoplasms/genetics
11.
Int J Mol Sci ; 22(15)2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34360727

ABSTRACT

Hereditary leiomyomatosis and renal cell carcinoma (HL (RCC)) entails cutaneous and uterine leiomyomatosis with aggressive type 2 papillary RCC-like histology. HLRCC is caused by pathogenic variants in the FH gene, which encodes fumarate hydratase (FH). Here, we describe an episode of young-onset RCC caused by a genomic FH deletion that was diagnosed via clinical sequencing. A 35-year-old woman was diagnosed with RCC and multiple metastases: histopathological analyses supported a diagnosis of FH-deficient RCC. Although the patient had neither skin tumors nor a family history of HLRCC, an aggressive clinical course at her age and pathological diagnosis of FH-deficient RCC suggested a germline FH variant. After counseling, the patient provided written informed consent for germline genetic testing. She was simultaneously subjected to paired tumor profiling tests targeting the exome to identify a therapeutic target. Although conventional germline sequencing did not detect FH variants, exome sequencing revealed a heterozygous germline FH deletion. As such, paired tumor profiling, not conventional sequencing, was required to identify this genetic deletion. RCC caused by a germline FH deletion has hitherto not been described in Japan, and the FH deletion detected in this patient was presumed to be of maternal European origin. Although the genotype-phenotype correlation in HLRCC-related tumors is unclear, the patient's family was advised to undergo genetic counseling to consider additional RCC screening.


Subject(s)
Fumarate Hydratase/deficiency , Gene Deletion , Germ-Line Mutation , Leiomyomatosis/genetics , Metabolism, Inborn Errors/genetics , Muscle Hypotonia/genetics , Neoplastic Syndromes, Hereditary/genetics , Psychomotor Disorders/genetics , Skin Neoplasms/genetics , Uterine Neoplasms/genetics , Adult , Female , Fumarate Hydratase/genetics , Genetic Testing , Humans
12.
J Clin Pathol ; 74(10): 615-619, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34353877

ABSTRACT

Fumarate hydratase (FH), encoded by the FH gene, is an enzyme which catalyses the conversion of fumarate to L-malate as part of the tricarboxylic acid cycle. Biallelic germline mutations in FH result in fumaric aciduria, a metabolic disorder resulting in severe neurological and developmental abnormalities. Heterozygous germline mutations in FH result in hereditary leiomyomatosis and renal cell carcinoma, a cancer predisposition syndrome. FH deficiency has multiple oncogenic mechanisms including through promotion of aerobic glycolysis, induction of pseudohypoxia, post-translational protein modification and impairment of DNA damage repair by homologous recombination. FH-deficient neoplasms can present with characteristic morphological features that raise suspicion for FH alterations and also frequently demonstrate loss of FH immunoreactivity and intracellular accumulation of 2-succinocysteine, also detected by immunohistochemistry.


Subject(s)
Fumarate Hydratase/deficiency , Fumarate Hydratase/metabolism , Metabolism, Inborn Errors/enzymology , Muscle Hypotonia/enzymology , Neoplasms/enzymology , Psychomotor Disorders/enzymology , Animals , Fumarate Hydratase/genetics , Gene Expression Regulation, Enzymologic , Genetic Predisposition to Disease , Humans , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Leiomyomatosis/pathology , Metabolism, Inborn Errors/genetics , Muscle Hypotonia/genetics , Mutation , Neoplasms/genetics , Neoplasms/pathology , Neoplastic Syndromes, Hereditary/enzymology , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Phenotype , Psychomotor Disorders/genetics , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Uterine Neoplasms/enzymology , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
13.
Acta Med Okayama ; 75(3): 397-402, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34176946

ABSTRACT

We report a 62-year-old male with metastatic fumarate hydratase-deficient renal cell carcinoma (FH-deficient RCC) without fumarate hydratase (FH) mutation (FH-deficient-like RCC). The International Metastatic RCC Database Consortium risk score was intermediate, and immunotherapy with nivolumab and ipilimumab (Ipi/ Nivo) was initiated. Four cycles of Ipi/Nivo and 5 cycles of nivolumab resulted in a complete response of the metastases. Hypophysitis occurred as an immune-related adverse event after four cycles of Ipi/Nivo. The prognosis of patients with FH-deficient RCC is generally poor. Few reports of FH-deficient RCC successfully treated with Ipi/Nivo have been published. Ipi/Nivo can be effective for treating FH-deficient RCC.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Carcinoma, Renal Cell/therapy , Ipilimumab/adverse effects , Kidney Neoplasms/drug therapy , Nivolumab/administration & dosage , Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Renal Cell/diagnostic imaging , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Germ-Line Mutation , Humans , Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/pathology , Male , Middle Aged , Nephrectomy , Tomography, X-Ray Computed
14.
Eur J Cancer ; 151: 106-114, 2021 07.
Article in English | MEDLINE | ID: mdl-33975058

ABSTRACT

PURPOSE: Fumarate hydratase-deficient (FHdef) renal cell carcinoma (RCC) is a rare entity associated with the hereditary leiomyomatosis and RCC syndrome with no standard therapy approved. The aim of this retrospective study was to evaluate the efficacy of different systemic treatments in this population. METHODS: We performed a multicentre retrospective analysis of Fhdef RCC patients to determine the response to systemic treatments. The endpoints were objective response rate (ORR), time-to-treatment failure (TTF), and overall survival (OS). The two latter were estimated using the Kaplan-Meier method. RESULTS: Twenty-four Fhdef RCC patients were identified, and 21 under systemic therapy were included in the analysis: ten received cabozantinib, 14 received sunitinib, nine received "other antiangiogenics" (sorafenib, pazopanib, and axitinib), three received erlotinib-bevacizumab (E-B), three received mTOR inhibitors, and 11 received immune checkpoint blockers (ICBs). ORR for treatments were 50% for cabozantinib, 43% for sunitinib, 63% for "other antiangiogenics," and 30% for E-B, whereas ORR was 0% for mTOR inhibitors and 18% for ICBs. The median TTF (mTTF) was significantly higher with antiangiogenics (11.6 months) than with mTOR inhibitors (4.4 months) or ICBs (2.7 months). In the first-line setting, antiangiogenics presented a higher ORR compared with nivolumab-ipilimumab (64% versus 25%) and a significantly superior mTTF (11.0 months vs 2.5 months; p = 0.0027). The median OS from the start of the first systemic treatment was 44.0 months (95% confidence interval: 13.0-95.0). CONCLUSIONS: We report the first European retrospective study of Fhdef RCC patients treated with systemic therapy with a remarkably long median OS of 44.0 months. Our results suggest that antiangiogenics may be superior to ICB/mTOR inhibitors in this population.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Biomarkers, Tumor/genetics , Fumarate Hydratase/genetics , Immune Checkpoint Inhibitors/therapeutic use , Kidney Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Adult , Angiogenesis Inhibitors/adverse effects , Biomarkers, Tumor/deficiency , Disease Progression , Female , France , Fumarate Hydratase/deficiency , Genetic Predisposition to Disease , Humans , Immune Checkpoint Inhibitors/adverse effects , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Male , Middle Aged , Molecular Targeted Therapy , Phenotype , Protein Kinase Inhibitors/adverse effects , Retrospective Studies , Spain , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Time Factors , Treatment Failure , Young Adult
15.
Clin Cancer Res ; 27(10): 2910-2919, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33658299

ABSTRACT

PURPOSE: Fumarate hydratase-deficient renal cell carcinoma (FH-RCC) is a rare, aggressive form of RCC associated with hereditary leiomyomatosis and RCC syndrome. Evidence for systemic therapy efficacy is lacking. EXPERIMENTAL DESIGN: We studied clinical and genomic characteristics of FH-RCC, including response [objective response rate (ORR)] to systemic therapies and next-generation sequencing (NGS). Patients with metastatic FH-RCC, defined by presence of pathogenic germline or somatic FH mutation plus IHC evidence of FH loss, were included. RESULTS: A total of 28 of 32 included patients (median age 46; range, 20-74; M:F, 20:12) underwent germline testing; 23 (82%) harbored a pathogenic FH germline variant. Five (16%) were negative for germline FH mutations; all had biallelic somatic FH loss. Somatic NGS (31/32 patients) revealed co-occurring NF2 mutation most frequently (n = 5). Compared with clear-cell RCC, FH-RCC had a lower mutation count (median 2 vs. 4; P < 0.001) but higher fraction of genome altered (18.7% vs. 10.3%; P = 0.001). A total of 26 patients were evaluable for response to systemic therapy: mTOR/VEGF combination (n = 18, ORR 44%), VEGF monotherapy (n = 15, ORR 20%), checkpoint inhibitor therapy (n = 8, ORR 0%), and mTOR monotherapy (n = 4, ORR 0%). No complete responses were seen. Median overall and progression-free survival were 21.9 months [95% confidence interval (CI): 14.3-33.8] and 8.7 months (95% CI: 4.8-12.3), respectively. CONCLUSIONS: Although most FH-RCC tumors are due to germline FH alterations, a significant portion result from biallelic somatic FH loss. Both somatic and germline FH-RCC have similar molecular characteristics, with NF2 mutations, low tumor mutational burden, and high fraction of genome altered. Although immunotherapy alone produced no objective responses, combination mTOR/VEGF therapy showed encouraging results.


Subject(s)
Biomarkers, Tumor , Carcinoma, Renal Cell/genetics , Fumarate Hydratase/deficiency , Adult , Aged , Alleles , Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/mortality , Computational Biology , DNA Copy Number Variations , Female , Germ-Line Mutation , Humans , Immunohistochemistry , Male , Middle Aged , Mutation , Neoplasm Grading , Neoplasm Staging , Prognosis , Retrospective Studies , Treatment Outcome , Young Adult
16.
Cancer Imaging ; 21(1): 24, 2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33608050

ABSTRACT

BACKGOUND: Fumarate hydratase-deficient renal cell carcinoma (FH-RCC) is a subtype of RCC that is increasingly recognized pathologically. The aim of this study was to evaluate the imaging features of FH-RCC on computed tomography (CT), magnetic resonance imaging (MRI), and fluorodeoxyglucose positron emission tomography (FDG PET), and to determine the pre-operative diagnostic potential of imaging. METHODS: This single-site retrospective study included patients with histologically confirmed FH-RCC or with a renal cancer and known germline FH mutation; imaging of the renal mass before treatment with contrast-enhanced CT, contrast-enhanced MRI, or FDG PET/CT between October 2007 and May 2019. Clinical information, pathological data, and imaging features were analyzed and reported descriptively. RESULTS: Sixteen patients with sixteen tumors were included (median age 46 years, interquartile range 38-53 years; 31 % female). Almost all tumors were unifocal (15/16, 94 %). Most tumors had infiltrative margins (14/16, 88 %); few were circumscribed (2/16, 12 %). A large cystic tumor component (> 75 % of tumor volume) was seen in 8/16 (50 %) of tumors. Involvement of renal sinus fat was seen in 13/16 (81 %) of tumors, involvement of the hilar collecting system in 8/16 (50 %), and renal vein tumor thrombus in 6/16 (38 %). All 12 tumors (100 %) imaged with MRI had heterogenous tumor enhancement and heterogenous T2 signal. Of those patients that had diffusion-weighted imaging, 11/11 (100 %) of tumors had diffusion restriction in the solid portions of the tumor. Of the patients who had PET, 3/3 (100 %) tumors showed high metabolic activity with mean maximum standardized uptake value (SUVmax) of 16.4 (range 9.6-21.9). Patients presented with retroperitoneal nodal metastases in 69 % of cases and distant metastases in 75 %. Of those four patients without metastatic disease at presentation, three (75 %) developed metastases within 4 years of diagnosis. CONCLUSIONS: In our study, the majority of tumors (≥ 75 %) were unifocal, had an infiltrative margin, invaded the renal sinus fat, and presented with distant metastases. On MRI, most tumors had heterogenous T2 signal and diffusion restriction in their solid components. The small number of cases that had PET imaging showed high metabolic activity.


Subject(s)
Carcinoma, Renal Cell/diagnostic imaging , Fumarate Hydratase/deficiency , Kidney Neoplasms/diagnostic imaging , Positron Emission Tomography Computed Tomography/methods , Adult , Carcinoma, Renal Cell/pathology , Female , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Retrospective Studies , Tumor Burden
17.
Clin Cancer Res ; 27(6): 1734-1743, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33414138

ABSTRACT

PURPOSE: Fumarate hydratase-deficient renal cell carcinoma (FH-deficient RCC) is a rare but lethal subtype of RCC. Little is known about the genomic profile of FH-deficient RCC, and the therapeutic options for advanced disease are limited. To this end, we performed a comprehensive genomics study to characterize the genomic and epigenomic features of FH-deficient RCC. EXPERIMENTAL DESIGN: Integrated genomic, epigenomic, and molecular analyses were performed on 25 untreated primary FH-deficient RCCs. Complete clinicopathologic and follow-up data of these patients were recorded. RESULTS: We identified that FH-deficient RCC manifested low somatic mutation burden (median 0.58 mutations per megabase), but with frequent somatic copy-number alterations. The majority of FH-deficient RCCs were characterized by a CpG sites island methylator phenotype, displaying concerted hypermethylation at numerous CpG sites in genes of transcription factors, tumor suppressors, and tumor hallmark pathways. However, a few cases (20%) with low metastatic potential showed relatively low DNA methylation levels, indicating the heterogeneity of methylation pattern in FH-deficient RCC. Moreover, FH-deficient RCC is potentially highly immunogenic, characterized by increased tumor T-cell infiltration but high expression of immune checkpoint molecules in tumors. Clinical data further demonstrated that patients receiving immune checkpoint blockade-based treatment achieved improved progression-free survival over those treated with antiangiogenic monotherapy (median, 13.3 vs. 5.1 months; P = 0.03). CONCLUSIONS: These results reveal the genomic features and provide new insight into potential therapeutic strategies for FH-deficient RCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/pathology , Epigenesis, Genetic , Fumarate Hydratase/deficiency , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/pathology , Mutation , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , DNA Methylation , Female , Follow-Up Studies , Fumarate Hydratase/genetics , Humans , Immune Checkpoint Inhibitors/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Male , Middle Aged , Neoplasm Metastasis , Prognosis , Retrospective Studies , Survival Rate , Young Adult
18.
Sci Signal ; 14(664)2021 01 05.
Article in English | MEDLINE | ID: mdl-33402335

ABSTRACT

Understanding the mechanisms of the Warburg shift to aerobic glycolysis is critical to defining the metabolic basis of cancer. Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is an aggressive cancer characterized by biallelic inactivation of the gene encoding the Krebs cycle enzyme fumarate hydratase, an early shift to aerobic glycolysis, and rapid metastasis. We observed impairment of the mitochondrial respiratory chain in tumors from patients with HLRCC. Biochemical and transcriptomic analyses revealed that respiratory chain dysfunction in the tumors was due to loss of expression of mitochondrial DNA (mtDNA)-encoded subunits of respiratory chain complexes, caused by a marked decrease in mtDNA content and increased mtDNA mutations. We demonstrated that accumulation of fumarate in HLRCC tumors inactivated the core factors responsible for replication and proofreading of mtDNA, leading to loss of respiratory chain components, thereby promoting the shift to aerobic glycolysis and disease progression in this prototypic model of glucose-dependent human cancer.


Subject(s)
Carcinoma, Renal Cell/genetics , Citric Acid Cycle , DNA Damage , DNA, Mitochondrial/metabolism , Fumarate Hydratase/genetics , Kidney Neoplasms/genetics , Leiomyomatosis/enzymology , Neoplastic Syndromes, Hereditary/enzymology , Skin Neoplasms/enzymology , Uterine Neoplasms/enzymology , Adult , Aged , Carcinoma, Renal Cell/etiology , Carcinoma, Renal Cell/metabolism , DNA Repair , DNA Replication , Female , Fumarate Hydratase/deficiency , Gene Expression Profiling , Humans , Kidney Neoplasms/etiology , Kidney Neoplasms/metabolism , Leiomyomatosis/complications , Male , Middle Aged , Mitochondria/genetics , Mitochondria/metabolism , Mutation , Neoplastic Syndromes, Hereditary/complications , Skin Neoplasms/complications , Uterine Neoplasms/complications , Young Adult
19.
J Child Neurol ; 36(4): 310-323, 2021 03.
Article in English | MEDLINE | ID: mdl-33052056

ABSTRACT

Fumarase deficiency (FD) is a rare and severe autosomal disorder, caused by inactivity of the enzyme fumarase, due to biallelic mutations of the fumarase hydratase (FH) gene. Several pathogenic mutations have been published. The article describes an infant with failure to thrive, microcephaly, axial hypotonia, and developmental retardation with increased excretion of fumarate, no activity of fumarase and a homozygous mutation of the FH gene, which was until recently only known as a variant of unknown significance. Carriers of pathogenic mutations in the FH gene are at risk for developing renal cell carcinoma and should therefore be screened. Both parents were healthy carriers of the mutation and had decreased levels of enzyme activity. In addition, the article presents an overview and analysis of all cases of FD reported thus far in the literature.


Subject(s)
Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/pathology , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Psychomotor Disorders/genetics , Psychomotor Disorders/pathology , Humans , Infant , Male , Metabolism, Inborn Errors/diagnostic imaging , Muscle Hypotonia/diagnostic imaging , Psychomotor Disorders/diagnostic imaging
20.
Ann Diagn Pathol ; 49: 151599, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32977234

ABSTRACT

Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) has been incorporated into the recent international histological classification of renal tumors. However, to date, there are limited studies describing the clinicopathological features of fumarate hydratase (FH)-deficient RCC, including the hereditary (HLRCC) and sporadic forms. Herein, we present a clinicopathological study of seven cases with FH-deficient RCC. The age of patients ranged from 26 to 70 years with mean and median age of 51.7 and 57 years, respectively. The follow-up data of all patients were available. One patient was alive without the disease and five patients were alive with active disease. One patient died of the disease. Family history of RCC, or skin or uterine smooth muscle tumor within second degree of kinship was present in four of seven patients. Metastasis was observed in all tumors. Metastatic sites included bone, lungs, liver, peritoneum, ovaries, tonsils, or lymph nodes. Grossly, the cut surface of the tumor usually showed light brown, brown, or whitish color. Microscopically, the cytoplasm of the tumor cells was predominantly eosinophilic and all tumors displayed various architectural patterns such as papillary, tubular, solid, or microcystic patterns. Furthermore, two tumors demonstrated a tubulocystic pattern. Sarcomatoid change and rhabdoid features were seen in five tumors and two tumors, respectively. Large cytomegaloviral (CMV) inclusion-like eosinophilic nucleoli surrounded by a clear halo were identified in all tumors. All tumors showed negative immunohistochemical reaction for FH protein. False positive results of TFE3 protein were observed in three tumors. Furthermore, a germline mutation of FH gene was identified in one patient with family history of the disease. In conclusion, FH-deficient RCC includes hereditary and sporadic forms. Grossly, this tumor is solitary and occurs unilaterally. Histologically, the tumor is characterized by various patterns such as papillary, tubular, solid, tubulocystic, or microcystic, has eosinophilic cytoplasm and CMV-like high-grade nuclei. FH-deficient RCCs frequently metastasize to other anatomic sites. TFE immunoreactivity may occur in some FH-deficient RCCs, and immunohistochemistry can accurately diagnose these tumors and mutational analysis of FH gene.


Subject(s)
Carcinoma, Renal Cell/pathology , Fumarate Hydratase/deficiency , Kidney Neoplasms/pathology , Adult , Aged , Carcinoma, Renal Cell/enzymology , Female , Humans , Kidney Neoplasms/enzymology , Leiomyomatosis/pathology , Male , Middle Aged , Neoplastic Syndromes, Hereditary/pathology , Skin Neoplasms/pathology , Uterine Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...