1
|
Quinn CH, Beierle AM, Williams AP, Marayati R, Bownes LV, Market HR, Erwin ME, Aye JM, Stewart JE, Mroczek-Musulman E, Yoon KJ, Beierle EA. Preclinical evidence for employing MEK inhibition in NRAS mutated pediatric gastroenteropancreatic neuroendocrine-like tumors. Transl Oncol 2024; 47:102045. [PMID: 38959709 PMCID: PMC11269785 DOI: 10.1016/j.tranon.2024.102045] [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: 04/11/2023] [Revised: 01/22/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Pediatric gastroenteropancreatic neuroendocrine tumors are exceedingly rare, resulting in most pediatric treatment recommendations being based on data derived from adults. Trametinib is a kinase inhibitor that targets MEK1/2 and has been employed in the treatment of cancers harboring mutations in the Ras pathway. METHODS We utilized an established human pediatric gastroenteropancreatic neuroendocrine-like tumor patient-derived xenograft (PDX) with a known NRAS mutation to study the effects of MEK inhibition. We evaluated the effects of trametinib on proliferation, motility, and tumor growth in vivo. We created an intraperitoneal metastatic model of this PDX, characterized both the phenotype and the genotype of the metastatic PDX and again, investigated the effects of MEK inhibition. RESULTS We found target engagement with decreased ERK1/2 phosphorylation with trametinib treatment. Trametinib led to decreased in vitro cell growth and motility, and decreased tumor growth and increased animal survival in a murine flank tumor model. Finally, we demonstrated that trametinib was able to significantly decrease gastroenteropancreatic neuroendocrine intraperitoneal tumor metastasis. CONCLUSIONS The results of these studies support the further investigation of MEK inhibition in pediatric NRAS mutated solid tumors.
Collapse
Affiliation(s)
- Colin H Quinn
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, Birmingham, AL 35205, USA
| | - Andee M Beierle
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL 35222, USA
| | - Adele P Williams
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, Birmingham, AL 35205, USA
| | - Raoud Marayati
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, Birmingham, AL 35205, USA
| | - Laura V Bownes
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, Birmingham, AL 35205, USA
| | - Hooper R Market
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, Birmingham, AL 35205, USA
| | - Michael E Erwin
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, Birmingham, AL 35205, USA
| | - Jamie M Aye
- Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Alabama, Birmingham, Birmingham, AL 35233, USA
| | - Jerry E Stewart
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, Birmingham, AL 35205, USA
| | | | - Karina J Yoon
- Department of Pharmacology and Toxicology, University of Alabama, Birmingham, Birmingham, AL 35233, USA
| | - Elizabeth A Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, Birmingham, AL 35205, USA.
| |
Collapse
|
2
|
Sharma AE, Dermawan JK, Chiang S, Wexler LH, Antonescu CR. Botryoid-type Embryonal Rhabdomyosarcoma: A Comprehensive Clinicopathologic and Molecular Appraisal With Cross-comparison to its Conventional-type Counterpart. Am J Surg Pathol 2024:00000478-990000000-00408. [PMID: 39210566 DOI: 10.1097/pas.0000000000002300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Embryonal rhabdomyosarcoma (ERMS) is the most common subtype of RMS, occurring in soft tissue and visceral sites of young children, and is associated with favorable outcomes. A subset occurs in mucosal-lined luminal structures, displaying a unique grape-like growth termed as "botryoid-type." To further delineate the differences between conventional (cERMS) and botryoid-type (bERMS) RMS, we performed a comparative histologic review and comprehensive molecular profiling of 48 cases (25 bERMS and 23 cERMS). All tumors were subjected to a hybridization capture-based targeted matched tumor-normal DNA NGS assay. The mean age was 17 and 7 years for bERMS and cERMS, respectively. Most bERMS were female with a predilection for the gynecologic tract (75%), while cERMS had a slight male predominance and were preferentially located in abdominopelvic and paratesticular sites (30%, each). All bERMS exhibited an exophytic, bulbous architecture accompanied by a subepithelial "cambium layer." Distinctive germline alterations were detected, with DICER1 (18%) and FH (6%) mutations only in bERMS, and rare TP53, VHL, and APC mutations in cERMS. Similarly, contrasting somatic genomic landscapes were observed, with frequent DICER1 (52%, P**<0.0001) and TP53 (36%, P*<0.05) alterations exclusively in bERMS. Cartilaginous differentiation was only observed in DICER1-mutated bERMS. All patients had longitudinal follow-up. bERMS patients with somatic/germline DICER1 mutations showed significantly improved recurrence-free survival compared with that of DICER1-wild type patients (P*<0.05). Moreover, bERMS showed improved disease-specific survival compared with that of cERMS, with 8% versus 30% (P*<0.05) dead of disease, respectively. In summary, we compare the molecular underpinnings of the largest cohort of bERMS and cERMS with targeted DNA sequencing and long-term follow-up data. Our findings reveal divergent genomic topographies between the 2 groups, with bERMS showing unique germline and somatic abnormalities, including enrichment in DICER1 and TP53 alterations, and a trend towards improved survival.
Collapse
Affiliation(s)
- Aarti E Sharma
- Department of Pathology & Laboratory Medicine, Memorial Sloan Kettering Cancer Center
- Department of Pathology and Laboratory Medicine, Hospital for Special Surgery
| | - Josephine K Dermawan
- Department of Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Sarah Chiang
- Department of Pathology & Laboratory Medicine, Memorial Sloan Kettering Cancer Center
| | - Leonard H Wexler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Cristina R Antonescu
- Department of Pathology & Laboratory Medicine, Memorial Sloan Kettering Cancer Center
| |
Collapse
|
3
|
de Traux de Wardin H, Dermawan JK, Vanoli F, Jiang SC, Singer S, Chi P, Tap W, Wexler LH, Antonescu CR. NF1-Driven Rhabdomyosarcoma Phenotypes: A Comparative Clinical and Molecular Study of NF1-Mutant Rhabdomyosarcoma and NF1-Associated Malignant Triton Tumor. JCO Precis Oncol 2024; 8:e2300597. [PMID: 38603649 PMCID: PMC11161258 DOI: 10.1200/po.23.00597] [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: 10/31/2023] [Revised: 01/28/2024] [Accepted: 02/16/2024] [Indexed: 04/13/2024] Open
Abstract
PURPOSE Alterations of the NF1 tumor suppressor gene is the second most frequent genetic event in embryonal rhabdomyosarcoma (ERMS), but its associations with clinicopathologic features, outcome, or coexisting molecular events are not well defined. Additionally, NF1 alterations, mostly in the setting of neurofibromatosis type I (NF1), drive the pathogenesis of most malignant peripheral nerve sheath tumor with divergent RMS differentiation (also known as malignant triton tumor [MTT]). Distinguishing between these entities can be challenging because of their pathologic overlap. This study aims to comprehensively analyze the clinicopathologic and molecular spectrum of NF1-mutant RMS compared with NF1-associated MTT for a better understanding of their pathogenesis. METHODS We investigated the clinicopathologic and molecular landscape of a cohort of 22 NF1-mutant RMS and a control group of 13 NF1-associated MTT. Cases were tested on a matched tumor-normal hybridization capture-based targeted DNA next-generation sequencing. RESULTS Among the RMS group, all except one were ERMS, with a median age of 17 years while for MTT the mean age was 39 years. Three MTTs were misdiagnosed as ERMS, having clinical impact in one. The most frequent coexisting alteration in ERMS was TP53 abnormality (36%), being mutually exclusive from NRAS mutations (14%). MTT showed coexisting CDKN2A/B and PRC2 complex alterations in 38% cases and loss of H3K27me3 expression. Patients with NF1-mutant RMS exhibited a 70% 5-year survival rate, in contrast to MTT with a 33% 5-year survival. All metastatic NF1-mutant ERMS were associated with TP53 alterations. CONCLUSION Patients with NF1-mutant ERMS lacking TP53 alterations may benefit from dose-reduction chemotherapy. On the basis of the diagnostic challenges and significant treatment and prognostic differences, molecular profiling of challenging tumors with rhabdomyoblastic differentiation is recommended.
Collapse
Affiliation(s)
- Henry de Traux de Wardin
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Brussels University Hospital, Academic Children's Hospital Queen Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Josephine K. Dermawan
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Fabio Vanoli
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Samuel C. Jiang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Samuel Singer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ping Chi
- Department of Medicine, Sarcoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - William Tap
- Department of Medicine, Sarcoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Leonard H. Wexler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Cristina R. Antonescu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
4
|
Zeng L, Wang J, Zhu H, Huang Y, Deng Y, Wei P, Nie J, Tang B, Chen A, Zhu S. The RRAS2 pathogenic variant (c.67G>T; p. Gly23Cys) produces Noonan syndrome with embryonal rhabdomyosarcoma. Mol Genet Genomic Med 2024; 12:e2313. [PMID: 37942564 PMCID: PMC10767593 DOI: 10.1002/mgg3.2313] [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: 04/13/2023] [Revised: 08/01/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Noonan syndrome (NS) due to the RRAS2 gene, the pathogenic variant is an extremely rare RASopathies. Our objective was to identify the potential site of RRAS2, combined with the literature review, to find the correlation between clinical phenotype and genotype. De novo missense mutations affect different aspects of the RRAS2 function, leading to hyperactivation of the RAS-MAPK signaling cascade. METHODS Conventional G-banding was used to analyze the chromosome karyotype of the patient. Copy number variation sequencing (CNV-seq) was used to detect the chromosomal gene microstructure of the patient and her parents. The exomes of the patient and her parents were sequenced using trio-based whole exome sequencing (trio-WES) technology. The candidate variant was verified by Sanger sequencing. The pathogenicity of the variant was predicted with a variety of bioinformatics tools. RESULTS Chromosome analysis of the proband revealed 46, XX, and no abnormality was found by CNV-seq. After sequencing and bioinformatics filtering, the variant of RRAS2(c.67G>T; p. Gly23Cys) was found in the proband, while the mutation was absent in her parents. To the best of our knowledge, our patient was with the typical Noonan syndrome, such as short stature, facial dysmorphism, and developmental delay. Furthermore, our study is the first case of NS with embryonal rhabdomyosarcoma (ERMS) caused by the RRAS2 gene mutation reported in China. CONCLUSIONS Our investigations suggested that the heterozygous missense of RRAS2 may be a potential causal variant in a rare cause of Noonan syndrome, expanding our understanding of the causally relevant mutations for this disorder.
Collapse
Affiliation(s)
- Lan Zeng
- Department of Medical Genetics and Prenatal DiagnosisSichuan Provincial Maternity and Child Health Care HospitalChengduChina
| | - Jin Wang
- Department of Medical Genetics and Prenatal DiagnosisSichuan Provincial Maternity and Child Health Care HospitalChengduChina
| | - Hui Zhu
- Department of PediatricsSichuan Provincial Maternity and Child Health Care HospitalChengduChina
| | - Yu Huang
- Department of PediatricsSichuan Provincial Maternity and Child Health Care HospitalChengduChina
| | - Yi Deng
- Department of Medical Genetics and Prenatal DiagnosisSichuan Provincial Maternity and Child Health Care HospitalChengduChina
| | - Ping Wei
- Department of Medical Genetics and Prenatal DiagnosisSichuan Provincial Maternity and Child Health Care HospitalChengduChina
| | - Jing Nie
- Department of Children's Health CareSichuan Provincial Maternity and Child Health Care HospitalChengduChina
| | - Bei Tang
- Department of UltrasoundSichuan Provincial Maternity and Child Health Care HospitalChengduChina
| | - Ai Chen
- Department of PediatricsSichuan Provincial Maternity and Child Health Care HospitalChengduChina
| | - Shuyao Zhu
- Department of PediatricsSichuan Provincial Maternity and Child Health Care HospitalChengduChina
| |
Collapse
|
5
|
Vicha A, Jencova P, Novakova-Kodetova D, Stolova L, Voriskova D, Vyletalova K, Broz P, Drahokoupilova E, Guha A, Kopecká M, Krskova L. Changes on chromosome 11p15.5 as specific marker for embryonal rhabdomyosarcoma? Genes Chromosomes Cancer 2023; 62:732-739. [PMID: 37530573 DOI: 10.1002/gcc.23194] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/04/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023] Open
Abstract
Rhabdomyosarcomas (RMS) constitute a heterogeneous spectrum of tumors with respect to clinical behavior and tumor morphology. The paternal uniparental disomy (pUPD) of 11p15.5 is a molecular change described mainly in embryonal RMS. In addition to LOH, UPD, the MLPA technique (ME030kit) also determines copy number variants and methylation of H19 and KCNQ1OT1 genes, which have not been systematically investigated in RMS. All 127 RMS tumors were divided by histology and PAX status into four groups, pleomorphic histology (n = 2); alveolar RMS PAX fusion-positive (PAX+; n = 39); embryonal RMS (n = 70) and fusion-negative RMS with alveolar pattern (PAX-RMS-AP; n = 16). The following changes were detected; negative (n = 21), pUPD (n = 75), gain of paternal allele (n = 9), loss of maternal allele (n = 9), hypermethylation of H19 (n = 6), hypomethylation of KCNQ1OT1 (n = 6), and deletion of CDKN1C (n = 1). We have shown no difference in the frequency of pUPD 11p15.5 in all groups. Thus, we have proven that changes in the 11p15.5 are not only specific to the embryonal RMS (ERMS), but are often also present in alveolar RMS (ARMS). We have found changes that have not yet been described in RMS. We also demonstrated new potential diagnostic markers for ERMS (paternal duplication and UPD of whole chromosome 11) and for ARMS PAX+ (hypomethylation KCNQ1OT1).
Collapse
Affiliation(s)
- Ales Vicha
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Pavla Jencova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Daniela Novakova-Kodetova
- Department of Pathology and Molecular Medicine, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Lucie Stolova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Dagmar Voriskova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Kristyna Vyletalova
- Department of Pathology and Molecular Medicine, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Petr Broz
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
- BIOXSYS, Ústí nad Labem, Czech Republic
| | - Eva Drahokoupilova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Anasuya Guha
- Department of Otorhinolaryngology, 3rd Faculty of Medicine, Charles University in Prague and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Marie Kopecká
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Lenka Krskova
- Department of Pathology and Molecular Medicine, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
6
|
Zuntini R, Cattani C, Pedace L, Miele E, Caraffi SG, Gardini S, Ficarelli E, Pizzi S, Radio FC, Barone A, Piana S, Bertolini P, Corradi D, Marinelli M, Longo C, Motolese A, Zuffardi O, Tartaglia M, Garavelli L. Case Report: Sequential postzygotic HRAS mutation and gains of the paternal chromosome 11 carrying the mutated allele in a patient with epidermal nevus and rhabdomyosarcoma: evidence of a multiple-hit mechanism involving HRAS in oncogenic transformation. Front Genet 2023; 14:1231434. [PMID: 37636262 PMCID: PMC10447906 DOI: 10.3389/fgene.2023.1231434] [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: 05/30/2023] [Accepted: 08/01/2023] [Indexed: 08/29/2023] Open
Abstract
We report a 7-year-old boy born with epidermal nevi (EN) arranged according to Blaschko's lines involving the face and head, right upper limb, chest, and left lower limb, who developed a left paratesticular embryonal rhabdomyosarcoma at 18 months of age. Parallel sequencing identified a gain-of-function variant (c.37G>C, p.Gly13Arg) of HRAS in both epidermal nevus and tumor but not in leukocytes or buccal mucosal epithelial cells, indicating its postzygotic origin. The variant accounted for 33% and 92% of the total reads in the nevus and tumor DNA specimens, respectively, supporting additional somatic hits in the latter. DNA methylation (DNAm) profiling of the tumor documented a signature consistent with embryonal rhabdomyosarcoma and CNV array analysis inferred from the DNAm arrays and subsequent MLPA analysis demonstrated copy number gains of the entire paternal chromosome 11 carrying the mutated HRAS allele, likely as the result of paternal unidisomy followed by subsequent gain(s) of the paternal chromosome in the tumor. Other structural rearrangements were observed in the tumours, while no additional pathogenic variants affecting genes with role in the RAS-MAPK and PI3K-AKT-MTOR pathways were identified. Our findings provide further evidence of the contribution of "gene dosage" to the multistep process driving cell transformation associated with hyperactive HRAS function.
Collapse
Affiliation(s)
- Roberta Zuntini
- Medical Genetics Unit, Azienda USL, IRCCS, Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Chiara Cattani
- Medical Genetics Unit, Azienda USL, IRCCS, Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Lucia Pedace
- Department of Pediatric Hematology, Oncology and Cellular and Gene Therapy, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Evelina Miele
- Department of Pediatric Hematology, Oncology and Cellular and Gene Therapy, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | | | - Stefano Gardini
- Dermatology Unit, Azienda USL, IRCCS, Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Elena Ficarelli
- Dermatology Unit, Azienda USL, IRCCS, Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Simone Pizzi
- Molecular Genetics and Functional Genomics Research Unit, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Francesca Clementina Radio
- Molecular Genetics and Functional Genomics Research Unit, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Angelica Barone
- Paediatric Hematology Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Simonetta Piana
- Department of Oncology and Advanced Technologies, Pathology Unit, Azienda USL, IRCCS, Arcispedale S Maria Nuova, Reggio Emilia, Italy
| | - Patrizia Bertolini
- Paediatric Hematology Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Domenico Corradi
- Department of Medicine and Surgery, Unit of Pathology, University of Parma, Parma, Italy
| | - Maria Marinelli
- Medical Genetics Unit, Azienda USL, IRCCS, Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Caterina Longo
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
- Department of Oncology and Advanced Technologies, Unit of Dermatology, Azienda USL, IRCCS, Arcispedale S Maria Nuova, Reggio Emilia, Italy
| | - Alberico Motolese
- Dermatology Unit, Azienda USL, IRCCS, Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Orsetta Zuffardi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics Research Unit, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Livia Garavelli
- Medical Genetics Unit, Azienda USL, IRCCS, Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| |
Collapse
|
7
|
Hebron KE, Wan X, Roth JS, Liewehr DJ, Sealover NE, Frye WJ, Kim A, Stauffer S, Perkins OL, Sun W, Isanogle KA, Robinson CM, James A, Awasthi P, Shankarappa P, Luo X, Lei H, Butcher D, Smith R, Edmondson EF, Chen JQ, Kedei N, Peer CJ, Shern JF, Figg WD, Chen L, Hall MD, Difilippantonio S, Barr FG, Kortum RL, Robey RW, Vaseva AV, Khan J, Yohe ME. The Combination of Trametinib and Ganitumab is Effective in RAS-Mutated PAX-Fusion Negative Rhabdomyosarcoma Models. Clin Cancer Res 2023; 29:472-487. [PMID: 36322002 PMCID: PMC9852065 DOI: 10.1158/1078-0432.ccr-22-1646] [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: 05/23/2022] [Revised: 09/22/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022]
Abstract
PURPOSE PAX-fusion negative rhabdomyosarcoma (FN RMS) is driven by alterations in the RAS/MAP kinase pathway and is partially responsive to MEK inhibition. Overexpression of IGF1R and its ligands is also observed in FN RMS. Preclinical and clinical studies have suggested that IGF1R is itself an important target in FN RMS. Our previous studies revealed preclinical efficacy of the MEK1/2 inhibitor, trametinib, and an IGF1R inhibitor, BMS-754807, but this combination was not pursued clinically due to intolerability in preclinical murine models. Here, we sought to identify a combination of an MEK1/2 inhibitor and IGF1R inhibitor, which would be tolerated in murine models and effective in both cell line and patient-derived xenograft models of RAS-mutant FN RMS. EXPERIMENTAL DESIGN Using proliferation and apoptosis assays, we studied the factorial effects of trametinib and ganitumab (AMG 479), a mAb with specificity for human and murine IGF1R, in a panel of RAS-mutant FN RMS cell lines. The molecular mechanism of the observed synergy was determined using conventional and capillary immunoassays. The efficacy and tolerability of trametinib/ganitumab was assessed using a panel of RAS-mutated cell-line and patient-derived RMS xenograft models. RESULTS Treatment with trametinib and ganitumab resulted in synergistic cellular growth inhibition in all cell lines tested and inhibition of tumor growth in four of six models of RAS-mutant RMS. The combination had little effect on body weight and did not produce thrombocytopenia, neutropenia, or hyperinsulinemia in tumor-bearing SCID beige mice. Mechanistically, ganitumab treatment prevented the phosphorylation of AKT induced by MEK inhibition alone. Therapeutic response to the combination was observed in models without a mutation in the PI3K/PTEN axis. CONCLUSIONS We demonstrate that combined trametinib and ganitumab is effective in a genomically diverse panel of RAS-mutated FN RMS preclinical models. Our data also show that the trametinib/ganitumab combination likely has a favorable tolerability profile. These data support testing this combination in a phase I/II clinical trial for pediatric patients with relapsed or refractory RAS-mutated FN RMS.
Collapse
Affiliation(s)
- Katie E. Hebron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892,Laboratory of Cell and Developmental Signaling, Center for Cancer Research, 8560 Progress Drive, Frederick, MD 21701
| | - Xiaolin Wan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Jacob S. Roth
- Early Translation Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850
| | - David J. Liewehr
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Nancy E. Sealover
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Services, Bethesda, MD 20814
| | - William J.E. Frye
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892
| | - Angela Kim
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, 8560 Progress Drive, Frederick, MD 21701
| | - Stacey Stauffer
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, 8560 Progress Drive, Frederick, MD 21701
| | - Olivia L. Perkins
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Wenyue Sun
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892
| | - Kristine A. Isanogle
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Christina M. Robinson
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Amy James
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Parirokh Awasthi
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Priya Shankarappa
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Xiaoling Luo
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Haiyan Lei
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Donna Butcher
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Roberta Smith
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Elijah F. Edmondson
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Jin-Qiu Chen
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Noemi Kedei
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Cody J. Peer
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Jack F. Shern
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - W. Douglas Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892
| | - Lu Chen
- Early Translation Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850
| | - Matthew D. Hall
- Early Translation Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Frederic G. Barr
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892
| | - Robert L. Kortum
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Services, Bethesda, MD 20814
| | - Robert W. Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892
| | - Angelina V. Vaseva
- Greehey Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, Texas, USA
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892,Co-corresponding authors Correspondence: Marielle Yohe, M.D., Ph.D., Center for Cancer Research, National Cancer Institute, 8560 Progress Drive Room D3026, Frederick, MD 27101, Phone: (240) 760-7436,
| | - Marielle E. Yohe
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892,Laboratory of Cell and Developmental Signaling, Center for Cancer Research, 8560 Progress Drive, Frederick, MD 21701,Co-corresponding authors Correspondence: Marielle Yohe, M.D., Ph.D., Center for Cancer Research, National Cancer Institute, 8560 Progress Drive Room D3026, Frederick, MD 27101, Phone: (240) 760-7436,
| |
Collapse
|
8
|
Hindi N, Carrillo-García J, Blanco-Alcaina E, Renshaw M, Luna P, Durán J, Jiménez N, Sancho P, Ramos R, Moura DS, Martín-Broto J. Platinum-Based Regimens Are Active in Advanced Pediatric-Type Rhabdomyosarcoma in Adults and Depending on HMGB1 Expression. Int J Mol Sci 2023; 24:ijms24010856. [PMID: 36614297 PMCID: PMC9821763 DOI: 10.3390/ijms24010856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023] Open
Abstract
Rhabdomyosarcoma (RMS) in adults is a rare and aggressive disease, which lacks standard therapies for relapsed or advanced disease. This retrospective study aimed to describe the activity of BOMP-EPI (bleomycin, vincristine, methotrexate and cisplatin alternating with etoposide, cisplatin and ifosfamide), an alternative platinum-based regimen, in adult patients with relapsed/metastatic RMS. In the study, 10 patients with RMS with a median age at diagnosis of 20.8 years and a female/male distribution of 6/4 received a mean of 2.5 cycles of BOMP-EPI. The best RECIST response was a complete response in 1/10 (10%) patients, a partial response in 5/10 (50%), stable disease in 3/10 (30%) and progression in 1/10 (10%). With a median follow-up in the alive patients from the start of therapy of 30.5 months (15.7-258), all patients progressed with a median progression-free survival of 8.47 months (95% CI 8.1-8.8), and 7/10 patients died with a median overall survival of 24.7 months (95% CI 13.7-35.6). BOMP-EPI was an active chemotherapy regimen in adults with pediatric-type metastatic RMS, with outcomes in terms of survival that seem superior to what was expected for this poor-prognosis population. Low HMGB1 expression level was identified as a predictive factor of better response to this treatment.
Collapse
Affiliation(s)
- Nadia Hindi
- Health Research Institute Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
- Medical Oncology Department, University Hospital General de Villalba, 28400 Madrid, Spain
- Medical Oncology Department, University Hospital Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Jaime Carrillo-García
- Health Research Institute Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
- Medical Oncology Department, University Hospital General de Villalba, 28400 Madrid, Spain
| | - Elena Blanco-Alcaina
- Institute of Biomedicine of Seville (IBIS), HUVR-CSIC-University of Seville, 41013 Seville, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marta Renshaw
- Health Research Institute Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
- Medical Oncology Department, University Hospital General de Villalba, 28400 Madrid, Spain
| | - Pablo Luna
- Medical Oncology Department, University Hospital Son Espases, 07210 Palma, Spain
| | - José Durán
- Medical Oncology Department, University Hospital Son Espases, 07210 Palma, Spain
| | - Natalia Jiménez
- Medical Oncology Department, San Vicente de Paúl Hospital, Heredia 40101, Costa Rica
| | - Pilar Sancho
- Medical Oncology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain
| | - Rafael Ramos
- Pathology Department, University Hospital Son Espases, 07210 Palma, Spain
| | - David S. Moura
- Health Research Institute Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
- Medical Oncology Department, University Hospital General de Villalba, 28400 Madrid, Spain
| | - Javier Martín-Broto
- Health Research Institute Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
- Medical Oncology Department, University Hospital General de Villalba, 28400 Madrid, Spain
- Medical Oncology Department, University Hospital Fundación Jiménez Díaz, 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-910-908-102 (ext. 52831)
| |
Collapse
|
9
|
Li NM, Jiang SH, Zhou P, Li XH. Case Report: An NTRK1 fusion-positive embryonal rhabdomyosarcoma: clinical presentations, pathological characteristics and genotypic analyses. Front Oncol 2023; 13:1178945. [PMID: 37188172 PMCID: PMC10175838 DOI: 10.3389/fonc.2023.1178945] [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: 03/03/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is a prevalent form of soft tissue sarcoma that primarily affects children. Pediatric RMS is characterized by two distinct histological variants: embryonal (ERMS) and alveolar (ARMS). ERMS is a malignant tumor with primitive characteristics resembling the phenotypic and biological features of embryonic skeletal muscles. With the widespread and growing application of advanced molecular biological technologies, such as next-generation sequencing (NGS), it has been possible to determine the oncogenic activation alterations of many tumors. Specifically for soft tissue sarcomas, the determination of tyrosine kinase gene and protein related changes can be used as diagnostic aids and may be used as predictive markers for targeted tyrosine kinase inhibition therapy. Our study reports a rare and exceptional case of an 11-year-old patient diagnosed with ERMS, who tested positive for MEF2D-NTRK1 fusion. The case report presents a comprehensive overview of the clinical, radiographic, histopathological, immunohistochemical, and genetic characteristics of a palpebral ERMS. Furthermore, this study sheds light on an uncommon occurrence of NTRK1 fusion-positive ERMS, which may provide theoretical basis for therapy and prognosis.
Collapse
|
10
|
Rakheja D, Park JY, Alhasan M, Uddin N. Spindle Cell/Sclerosing Rhabdomyosarcoma With PAX8::PPARG Fusion. Int J Surg Pathol 2022; 30:950-955. [DOI: 10.1177/10668969221095170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The spindle cell/sclerosing subtype of rhabdomyosarcoma is classified based on genetic features into the three categories of MYOD1-mutated, gene fusion-driven, and a subset without a currently identified genetic driver event. The gene fusion-driven spindle cell/sclerosing rhabdomyosarcomas are heterogenous and characterized by increasing numbers of gene fusions, the most common gene partners being VGLL2, NCOA2, and TFCP2. Here we report a spindle cell/sclerosing rhabdomyosarcoma that arose in the orbit of a 4-year-old male. This tumor harbored a unique PAX8::PPARG fusion. PAX8::PPARG fusions have previously only been described in follicular thyroid carcinoma and follicular variant of papillary thyroid carcinoma. Our report adds to the growing number of gene fusions in spindle cell/sclerosing rhabdomyosarcomas.
Collapse
Affiliation(s)
- Dinesh Rakheja
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Children’s Health, Dallas, TX, USA
| | - Jason Y. Park
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Children’s Health, Dallas, TX, USA
| | - Mustafa Alhasan
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Children’s Health, Dallas, TX, USA
| | - Naseem Uddin
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Children’s Health, Dallas, TX, USA
| |
Collapse
|
11
|
Ragab N, Bauer J, Botermann DS, Uhmann A, Hahn H. Oncogenic NRAS Accelerates Rhabdomyosarcoma Formation When Occurring within a Specific Time Frame during Tumor Development in Mice. Int J Mol Sci 2021; 22:ijms222413377. [PMID: 34948179 PMCID: PMC8703790 DOI: 10.3390/ijms222413377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 11/26/2022] Open
Abstract
In the Ptch+/- mouse model for embryonal rhabdomyosarcoma (ERMS), we recently showed that oncogenic (onc) H-, K- or NRAS mutations do not influence tumor growth when induced at the advanced, full-blown tumor stage. However, when induced at the invisible ERMS precursor stage at 4 weeks of age, tumor development was enforced upon oncHRAS and oncKRAS but not by oncNRAS, which instead initiated tumor differentiation. These data indicate that oncRAS-associated processes differ from each other in dependency on the isoform and their occurrence during tumor development. Here, we investigated the outcome of oncNRAS induction at an earlier ERMS precursor stage at 2 weeks of age. In this setting, oncNRAS accelerates tumor growth because it significantly shortens the ERMS-free survival and increases the ERMS incidence. However, it does not seem to alter the differentiation of the tumors. It is also not involved in tumor initiation. Together, these data show that oncNRAS mutations can accelerate tumor growth when targeting immature ERMS precursors within a specific time window, in which the precursors are permissive to the mutation and show that oncNRAS-associated processes differ from each other in dependency on their occurrence during tumor development.
Collapse
Affiliation(s)
| | | | | | | | - Heidi Hahn
- Correspondence: ; Tel.: +49-(0)551-3914010
| |
Collapse
|