Skip to main content
The Oncologist logoLink to The Oncologist
. 2021 Aug 26;26(12):e2102–e2109. doi: 10.1002/onco.13927

Next‐Generation Sequencing in the Diagnosis of Metastatic Lesions: Reclassification of a Glioblastoma as an Endometrial Cancer Metastasis to the Brain

Shuk On Annie Leung 1,, Olivia Foley 2, David Chapel 3, Annacarolina Da Silva 4, Marisa Nucci 4, Michael G Muto 2, Susana Campos 5
PMCID: PMC8649002  PMID: 34355460

Abstract

Endometrial cancer is the most common gynecologic cancer in the U.S., but metastasis to the brain is rare, and diagnosis can be challenging. Traditional tools for determining if a tumor is a primary or metastatic lesion include pan‐imaging, histopathologic studies, and immunohistochemistry. Molecular testing with next‐generation sequencing has been increasingly used to augment these tests. We present a case of a patient who initially presented with a brain lesion diagnosed as glioblastoma on histology and immunohistochemistry, but whose diagnosis was later changed to metastasis from an endometrial primary based on molecular findings. The two tumors shared a common microsatellite instability signature and 51 DNA variants, including oncogenic driver mutations KRAS p.G13D, PIK3CA p.E545A, and PTEN p.I135V and p.K267Rfs*9. This highlights the power of molecular analysis in making the diagnosis in cases of rare metastases.

Key Points

  • Brain metastasis from endometrial primary is rare, and histopathological features may be augmented with molecular analysis to aid in diagnosis.

  • Comparison of the molecular makeup of the primary endometrial lesion with the metastatic lesion may reveal high‐risk molecular features that may be indicative of metastatic potential.

Keywords: Endometrial cancer, Brain metastases, KRAS, PTEN, PIK3CA

Short abstract

Metastasis of endometrial cancer to the brain is rare, and diagnosis can be challenging. This case report highlights the power of molecular analysis in making the diagnosis in cases of rare metastases.

Patient Story

A healthy 64‐year‐old gravida 0 woman presented with worsening short‐term memory, visual changes, and word finding difficulty over several weeks and a 60‐lb unintentional weight loss over the preceding year. Pan–computed tomography (CT) revealed an 8‐by‐5 cm left temporal lobe mass with surrounding edema, midline shift, and subfalcine herniation (Fig. 1A), as well as a heterogenous mass‐like expansion of the uterus. The patient underwent an urgent left frontal parietotemporal craniotomy and microsurgical gross total resection of her brain tumor. Pathology demonstrated a high‐grade malignant neoplasm, composed of areas with sheet‐like growth of poorly differentiated epithelioid cells with nuclear pleomorphism, multiple mitoses and apoptosis, and geographic coagulative necrosis. A few foci showed sarcomatous features, with malignant cells placed singly in myxoid stroma (Fig. 2A–C). Vascular proliferation was present. These morphologic features are suggestive but not specific for gliomas, although what initially prompted the diagnosis of glioma was the presence of focal glial differentiation. The tumor cells were focally positive for glial fibrillary acidic protein (GFAP) and SOX2, which are indicative of glial and neuronal cells, and negative for IDH1 R132H mutation‐specific immunomarker, which is more common in low‐grade, less aggressive, gliomas. There was loss of MLH1 and PMS2 expression and retention of MSH2 and MSH6. As discussed below, MLH1 promoter methylation was performed in the uterine tumor and not the brain lesion; however, confirmation of microsatellite instability was observed in both tumors by next‐generation sequencing. A reticulin stain highlighted the sarcomatous elements. On molecular testing, MGMT methylation was present, which is observed in approximately 50% of grade 4 glioblastomas. Based on the immunohistochemical staining and molecular testing, the pathologic diagnosis was grade 4 glioblastoma, gliosarcoma variant.

Figure 1.

Figure 1

(A): Brain magnetic resonance imaging (MRI) at time of initial presentation demonstrating midline shift, ring‐enhancement, and surrounding edema. Metastatic brain lesions on MRI often appear as small, well‐defined, ring‐enhancing lesions surrounded by edema; they may also show central necrosis or hemorrhage, or both. However, nonspecific gliomas may also demonstrate central necrosis surrounded by a ring of contrast enhancement along with edema causing mass effect. (B): Pelvic MRI performed as follow‐up for pelvic mass.

Figure 2.

Figure 2

Histopathologic features of brain and endometrial tumors. The left parietal lesion showed a poorly differentiated malignant neoplasm in brain parenchyma (40×) (A), composed of sheets of poorly differentiated epithelioid cells (200×) (B) and rarer foci with sarcomatous features in myxoid stroma (200×) (C). The endometrial adenocarcinoma showed few well‐differentiated gland‐forming areas (200×) (D) but predominantly comprised more poorly differentiated sheets of poorly differentiated carcinoma with only occasional glandular lumina (200×) (E). Metastatic poorly differentiated carcinoma was present in the ovary (200×) (F). All photomicrographs are hematoxylin and eosin stains.

The patient was referred to Neuro‐Oncology with a plan to initiate radiation and chemotherapy. Prior to beginning adjuvant treatment, she had pelvic magnetic resonance imaging (MRI) to characterize the uterus that demonstrated a diffuse heterogeneous soft tissue mass replacing the myometrium with extension to the peritoneal reflection of the pelvis, left adnexa, and sigmoid colon with mildly enlarged pelvic lymph nodes (Fig. 1B). Endometrial biopsy demonstrated grade 1 endometrial adenocarcinoma.

She underwent a radical hysterectomy, bilateral salpingo‐oophorectomy, pelvic lymph node dissection, omentectomy, and resection of tumor from the cul‐de‐sac, left ureter, and rectosigmoid to no evidence of residual disease. The uterus was densely adherent to the left pelvic sidewall with tumor extending into the rectosigmoid colon and encasing the left ureter (pT3bN1M1). The final pathology demonstrated grade 3 endometrioid endometrial adenocarcinoma with focal well‐differentiated areas corresponding to the tumor seen on preoperative biopsy (Fig. 2D–F). The tumor invaded through the myometrium to the uterine serosa as well as the cervical stroma involving the paracervical soft tissues at the radial resection margin; lymphovascular invasion was present. Metastatic endometrial adenocarcinoma was identified in the left ovary, pelvic peritoneum, perirectal fat, and a single obturator node. Immunohistochemistry demonstrated loss of MLH1/PMS2 with MLH1 promoter methylation, consistent with a sporadic microsatellite instability.

Molecular Tumor Board

Given the poorly differentiated morphology of both the endometrial adenocarcinoma and the brain tumor and the shared MLH1 and PMS2 deficiency, the molecular profiles of the two tumors were compared by a previously validated targeted next‐generation sequencing panel designed for the detection of single‐nucleotide variants, insertions and deletions, copy number alterations, and structural variants across 447 genes [1]. Mutation analysis for single‐nucleotide variants was performed using MuTect [2] and annotated by Oncotator [3], and insertions and deletions were called using Indelocator (https://software.broadinstitute.org/cancer/cga/indelocator). Integrative Genomics Viewer (version 2.0.16) and an internally developed application were used for visualization and interpretation. Variants were filtered to exclude those that occur at a populational frequency of greater than 0.1% in the Exome Sequencing Project database (http://evs.gs.washington.edu/EVS/). During the process of test validation, the filtering criteria were determined as follows: Each sequencing run, non‐neoplastic, formalin‐fixed, paraffin‐embedded liver and blood samples were included as controls. Variants identified in these control samples due to sequencing artifacts were filtered. Any filtered variants that were reported in the Catalogue of Somatic Mutations in Cancer (COSMIC v94, United Kingdom) more than twice were rescued and presented for manual review [1]. The two tumors shared a shared microsatellite instability signature and 46 DNA variants, including oncogenic driver mutations, such as KRAS p.G13D, PIK3CA p.E545A, and PTEN p.I135V and p.K267Rfs*9. Upon manual review, there was a smaller number of variants unique to each tumor, mostly present at low allelic fraction (Table 1; 17 variants in the brain lesion and 26 variants in the endometrial lesion). PTEN and KRAS are well‐established mutational drivers in endometrial cancer. However, whereas PTEN mutation is also commonly implicated in brain tumors, KRAS is rarely identified in gliomas [4]. ARID1A alterations are also frequently seen in endometrial carcinomas but are rarely part of the pathogenesis of gliomas [5].

Table 1.

Pathogenic elements of the molecular profile of patient's endometrial tumor compared with left parietal tumor

Endometrial adenocarcinoma Left parietal tumor
Tumor mutational burden/megabase: 43.343 Tumor mutational burden/megabase: 35.739
Mismatch repair status: Deficient (MMR‐D / MSI‐H) Mismatch repair status: Deficient (MMR‐D / MSI‐H)
Tumor %: 70% Tumor %: 70%
Gene Coverage V.A.F. AA change Gene Coverage V.A.F. AA change
APC 263 0.395 p.E1317Q APC 326 0.475 p.E1317Q
AR 128 0.328 Intron AR 130 0.354 Intron
ARID1A 280 0.357 p.P224Rfs*8 ARID1A 260 0.408 p.P224Rfs*8
ARID1A 310 0.248 p.M1564* ARID1A 310 0.38 p.M1564*
ATM 281 0.32 p.S941Ifs*28 ATM 241 0.427 p.S941Ifs*28
ATR 169 0.373 p.S1965C ATR 129 0.302 p.S1965C
ATR 245 0.322 p.P315H
ATRX 82 0.378 Splice region
ASXL1 304 0.309 p.G645Vfs*58 ASXL1 280 0.371 p.G645Vfs*58
AURKB 270 0.322 p.P226H AURKB 249 0.422 p.P226H
B2M 263 0.034 p.L7*
B2M 186 0.392 splice aceptor
B2M 434 0.295 p.V69Wfs*34
BCL11B 82 0.329 p.P314Q BCL11B 68 0.515 p.P314Q
BCOR 402 0.281 p.V432I BCOR 484 0.434 p.V432I
BCORL1 246 0.228 p.R1292Q BCORL1 424 0.186 p.P1007H
C1orf86 79 0.342 p.A107T
CREBBP 358 0.081 p.A298T
CSF3R 194 0.325 p.A543T CSF3R 160 0.419 p.A543T
CTCF 279 0.326 p.T204Qfs*18 CTCF 274 0.438 p.T204Qfs*18
DICER1 225 0.307 p.L1408P DICER1 247 0.389 p.L1408P
DIS3L2 379 0.061 p.E523K
DKC1 154 0.052 p.K498del
ERBB3 297 0.481 Splice region ERBB3 300 0.473 Splice region
ERCC6 282 0.436 p.R557H ERCC6 265 0.479 p.R557H
FAT1 439 0.314 p.E160G FAT1 439 0.246 p.D2270N
FLCN 235 0.272 p.H429Tfs*39 FLCN 172 0.39 p.H429Tfs*39
HELQ 266 0.44 p.M1036I HELQ 252 0.528 p.M1036I
IKZF1 325 0.308 p.A138V
IKZF1 413 0.414 Intron
JAZF1 454 0.198 p.P232Q
KAT6A 407 0.236 p.P1905H
KDM5A 499 0.299 p.G1200Dfs*9
KRAS 260 0.35 p.G13D KRAS 259 0.425 p.G13D
KMT2D 523 0.415 p.G1235Vfs*95
MDM2 158 0.063 Splice region
MED12 263 0.179 p.R1214H
MET 343 0.504 Intron MET 363 0.54 Intron
MET 151 0.298 Intron MET 200 0.425 Intron
MITF 294 0.333 p.R74T MITF 364 0.426 p.R74T
MYC 316 0.345 p.H374R MYC 350 0.454 p.H374R
NTRK1 331 0.263 p.G248W NTRK1 404 0.292 p.G248W
NSD1 362 0.318 p.G1800Wfs*3 NSD1 335 0.549 p.G1800Wfs*3
NOTCH1 257 0.233 p.D1502V
NFKBIA 228 0.057 p.E55*
NF1 305 0.357 p.G1190C
NF1 346 0.37 p.I679Dfs*21
NF1 368 0.457 p.Q1841Nfs*22
NT5C2 208 0.298 p.S251N
PAX5 310 0.319 p.V26Gfs*49 PAX5 324 0.358 p.V26Gfs*49
PAX5 148 0.25 p.P330S
PDGFRA 241 0.058 p.V129A
PIK3CA 134 0.276 p.E545A PIK3CA 154 0.331 p.E545A
PIK3R1 220 0.341 p.K567E
POLD1 206 0.063 p.Y894C
POLE 317 0.483 p.Y623C POLE 338 0.453 p.Y623C
POLQ 148 0.358 Splice region POLQ 106 0.406 Splice region
PRKDC 233 0.047 p.R1136L PRKDC 298 0.064 Splice donor
PRSS1 468 0.064 p.A173G PRSS1 445 0.043 p.A173G
PRF1 373 0.067 p.L478*
PTCH1 124 0.137 p.R1308Efs*64
PTEN 276 0.322 p.I135V PTEN 274 0.394 p.I135V
PTEN 230 0.383 p.K267Rfs*9 PTEN 228 0.447 p.K267Rfs*9
RARA 91 0.066 p.P440Rfs*203
RASA1 237 0.35 p.M718T RASA1 285 0.435 p.M718T
RB1 174 0.115 p.W78*
RBBP8 243 0.412 p.K357Nfs*3
SETD2 421 0.314 p.S2382Lfs*47 SETD2 366 0.331 p.S2382Lfs*47
SH2B3 332 0.476 p.E292Q SH2B3 455 0.479 p.E292Q
SLX4 278 0.299 p.R204H SLX4 219 0.484 p.R204H
SMARCE1 236 0.22 p.R322M
SMARCA4 250 0.296 p.T910M SMARCA4 311 0.354 p.T910M
STAG2 182 0.253 p.A550T STAG2 184 0.386 p.A550T
TDG 408 0.304 p.S352N TDG 464 0.332 p.S352N
TERT 142 0.493 p.V251I TERT 140 0.529 p.V251I
TOPBP1 341 0.34 p.Q1233H TOPBP1 330 0.47 p.Q1233H
TSC1 372 0.341 p.A616T
TSC1 202 0.045 p.Y195C
TSC2 61 0.508 p.C32Vfs*24
TRIM37 241 0.261 p.R152W TRIM37 217 0.438 p.R152W
XPO1 113 0.097 Splice region
XRCC4 166 0.373 p.P119T XRCC4 173 0.509 p.P119T
ZNF217 463 0.313 p.D528N ZNF217 539 0.44 p.D528N
Intergenic variant 421 0.29 p.E48del
Intergenic variant 197 0.508 p.T193S
Intergenic variant 411 0.365 p.W343Gfs*7
Intergenic variant 404 0.396 Intergenic variant
Intergenic variant 150 0.513 Intergenic variant
Intergenic variant 469 0.452 Intergenic variant

Gray shading indicates the unique variants present in each tumor sample. The minimal coverage for each region was at least 50 reads.

Abbreviations: AA, amino acid; MMR‐D, mismatch repair deficient; MSI‐H, microsatellite instability high; VAF, variant allele fraction.

Copy number alterations are estimated using an equation to calculate copy numbers based on the normalized log2 ratio and tumor percentage: number of copies = (2avg. log2ratio) × (100 ÷ [tumor percentage]) × 2. The avg.log2ratio is calculated across all of the segments represented a single gene. In assessing copy number alterations, including gains and losses, the results highlighted the clonal relationship between the primary endometrial and metastasis to the brain (Fig. 3). There was an isolated gain of chromosome arm 1q in the primary endometrial carcinoma. Endometrioid carcinomas are generally slightly hyperdiploid with chromosome gains involving mainly the long arm of chromosome 1 (70% of the cases), although isochromosomes or unbalanced translocations can be also seen [6]. The metastatic brain tumor shows additional loss of short arm of chromosome 5 that could also represent a sign of tumor evolution. This was interpreted as strong molecular evidence for a clonal relationship between the endometrial and brain tumors.

Figure 3.

Figure 3

Copy number plots of primary endometrial and brain tumors. (A): Endometrial carcinoma showing isolated gain of chromosome arm 1q. (B): Metastatic brain tumor with gain along 1q and a broad loss of 5q denoting some degree of tumor evolution with the additional loss on chromosome 5. The similarity of copy number alterations is remarkable and reinforces that the tumors are related.

Additional immunohistochemical stains were then performed on the brain tumor, which showed rare cells positive for pan‐cytokeratin markers and for OLIG2 (a marker of glial differentiation). The tumor cells were negative for PAX8. On integration of the molecular findings, despite multiple unusual morphologic features and a nonspecific immunophenotype, the brain tumor was classified as a poorly differentiated malignant neoplasm with glial differentiation, most compatible with metastasis from the endometrial primary. This ultimately changed the diagnosis from two separate primaries to metastatic poorly differentiated endometrial cancer.

KRAS is a proto‐oncogene located at chromosome 12p12.1 that encodes a signaling protein that moderates response to extracellular signals via downregulation of the MAPK or PI3K/AKT pathways [7]. KRAS plays a role in both an early checkpoint of transition from endometrial hyperplasia to carcinoma (present in 6%–16% of endometrial hyperplasia) and is a marker of invasive potential in the case of grade 1 endometrioid carcinoma [7]. KRAS mutation has also been associated with increased activation of estrogen signaling that is mediated through the Ras/MAPK pathway and provides a route to circumvent MEK inhibition to ultimately upregulate MAPK signaling. From a targetable therapy standpoint, this suggests that combination treatment of MEK inhibitors (e.g., trametinib) with antiestrogen therapy is required for efficacy in patients with KRAS mutant tumors; other combinations of MEK inhibitors, such as AKT inhibitors, are currently under investigation [8].

PIK3CA mutation is present in approximately 50% of endometrial cancers, and PTEN acts as a negative regulator of the PI3K‐AKT pathway and has been identified in up to 55% of endometrial cancers [9]. PTEN protein loss has also been found to be correlated with improved overall survival [10]. Several PI3K pathway inhibitors have been developed and are currently under investigation in preclinical studies and early clinical trials. PTEN mutations also trigger cellular senescence through a TP53‐dependent pathway associated with mTOR activation; modest effects have been shown with mTOR inhibitors in patients with these mutations [11].

Lastly, microsatellite instability (MSI) was also one of the key molecular alterations in this tumor. MSI is caused by aberrant mismatch repair gene and is identified in 20%–30% of patients with endometrial cancer, with Lynch syndrome accounting for approximately 25% of these cases [12]. A recent study showed that MSI analysis is effective as a predictive biomarker for the effect of immune checkpoint inhibitors, which are new anticancer drugs, including anti–programmed cell death protein 1 (PD‐1)/programmed cell death ligand 1 (PD‐L1) antibody [13].

The mutational profile in this case, including mutations in KRAS, PTEN, and PIK3CA as well as microsatellite instability, is characteristic of endometrioid neoplasia and therefore in keeping with the histologic findings in the endometrial tumor. The biological basis for dedifferentiation and distant metastasis in this tumor is not entirely clear, although next‐generation sequencing did reveal a hotspot (pT910M) mutation in the SWI/SNF protein SMARCA4, which is reported in a subset of dedifferentiated endometrial carcinomas and is associated with aggressive behavior [14]. SMARCA4‐deficient tumors, such as small cell carcinoma of the ovary and non‐small cell lung cancer, have been reported to respond to PD‐L1 inhibition and other novel immunotherapies [15].

Patient Update

The patient recovered well from her abdominal surgery and shortly afterward began adjuvant treatment for her presumed glioblastoma with temozolomide and brain radiation. Unfortunately, repeat imaging after 6 weeks demonstrated recurrent/residual tumor at the vaginal cuff and new peritoneal carcinomatosis. Temozolomide was discontinued based upon the change in diagnosis from primary glioblastoma to metastatic endometrial cancer. She completed pelvic radiation and two cycles of carboplatin/paclitaxel but tolerated chemotherapy poorly with weight loss, malnutrition, and cytopenia. She received one cycle of single agent paclitaxel with a subsequent rapid decline. Given her poor performance status, targeted treatments were not initiated. She was admitted with failure to thrive, transitioned to comfort measures only, and passed away 6 months after her initial presentation.

Discussion and Conclusion

The diagnostic dilemma of identifying a primary versus metastatic brain lesion has been well summarized in the literature [16]. Gliomas are the most common type of primary brain tumor (5 in 100,000), but overall, metastasis is more common than primary tumor (12 in 100,000). Presenting symptoms in brain metastases are similar to those of primary brain tumors (e.g., headache, aphasia, localized neurological deficits, and seizures). CT and MRI are commonly used for detecting brain lesions. Perfusion MRI has been used to differentiate primary gliomas and brain metastases, where measurements of relative cerebral blood volume of the edema are lower in metastases compared with glioma. However, radiologic appearance alone will not always lead to a specific diagnosis, as was the challenge in the present case. In general, multiple brain lesions are more suspicious for metastatic process than are single lesions.

The brain is a relatively uncommon site of metastasis for endometrial cancer (0.3%–1.16%), making this diagnosis difficult [17, 18, 19]. Although deep myometrial invasion has been suggested to be the strongest predictor of hematogenous dissemination and thus risk of spread to lung, liver, bone, and brain, the majority of patients with brain metastases do not have high‐risk factors (e.g., lymphovascular space invasion; microcystic, elongated, and fragmented (MELF) pattern; positive cytology), which suggests that despite early stage and low‐grade disease, there may be other inherent high‐risk features [19].

The use of molecular genetic testing has been increasingly used to assist in diagnosis, prognosis, and tailored treatment. In the case presented, we highlight the power of molecular testing as a diagnostic tool, especially in identifying tissue of origin for metastatic cancer to a rare site. Traditional tools for identifying tissue of origin in the setting of metastases with unknown or uncertain primary site include pan‐imaging, histopathologic studies, and immunohistochemistry. Initial accurate histopathologic diagnosis in this case was limited by the poorly differentiated nature of the patient's brain lesion and the lack of clinical and radiological correlation to the occult uterine primary at the time of brain biopsy. Molecular genetic testing has been increasingly used to augment these traditional tools. In one prospective study, molecular testing predicted tissue of origin in 247 of 252 patients with metastatic cancer of unknown primary site (98%) [20]. In this case, although immunohistochemistry gave the initial clue to the relationship between the tumors (as both had MLH1 and PMS2 deficiencies), the similarities in molecular signature led to the ultimate diagnosis.

Integrated genomic and proteomic analysis from The Cancer Genome Atlas (TCGA) has expanded our understanding of disease biology and diagnostic classification of endometrial cancer [21]. TCGA established four distinct endometrial cancer subclasses based on the extent of mutational load and somatic copy number alterations, each with its own prognostic significance: POLE‐mutant/ultramutated, MSI high/hypermutated, copy number low, and copy number high [21]. Since the development of TCGA, various next‐generation sequencing methods have been used to explore the genomic makeup of endometrial cancer. TCGA focused primarily on primary disease; 75% of samples were collected from patients who did not develop recurrent disease. Thus, less is known about the genomic landscape of tumors that metastasize or recur at extrapelvic sites. Furthermore, as highlighted in the present case, confounding elements are present in distinguishing primary from metastatic brain lesion even in the molecular mutational profile. For example, glioblastomas have been reported to harbor PTEN and PIK3CA alterations, among others, in whole‐exome sequencing of 543 glioblastoma samples [22]. In contrast, the MGMT promoter methylation seen in the patient's brain lesion is infrequent in endometrial cancer [23]. The variation in molecular signatures would be clarified as the genomic landscape of tumors that metastasize versus those that do not is further elucidated.

A study from Memorial Sloan Kettering assessed the clinical utility of prospective molecular characterization in 189 patients with advanced endometrial cancer in which 51% had tumor samples from metastatic sites [24]. They identified 63% with TP53 mutation, 56% with PTEN alterations, 70% with PI3K/AKT/mTOR pathway alteration, and 65% with RTK/RAS/β‐catenin pathway alterations. Phase II trials have investigated PI3K/AKT/mTOR inhibitors with encouraging preliminary results. Furthermore, high MSI was identified in 16% of the cohort, which has significant clinical implications, as pembrolizumab, a PD‐1 inhibitor, and lenvatinib, a multikinase inhibitor, have promising antitumor activity in MSI‐high/mismatch repair–deficient endometrial cancer [25]. Thus, an in‐depth understanding of the molecular makeup of endometrial carcinomas could have significant treatment and outcome implications among patients with metastases.

Additional high‐risk features may be revealed by molecular analysis. In the Memorial Sloan Kettering cohort mentioned above, the researchers noted four patients with matched primary and metastatic tumors with some genomic heterogeneity that involved potentially actionable hotspot mutations, including mTOR L2427Q and PIK3R1 E468* [24]. This suggests that metastatic lesions may have a higher rate of unique targetable mutations. However, lack of paired samples limited their ability to comment on genetic alterations in the tumor in response to therapy, and metastases were not categorized based on site.

In conclusion, given the importance of correct identification of tissue of origin in developing an appropriate oncologic treatment plan, use of molecular testing in patients with metastatic disease represents a significant opportunity. In cases of unclear tissue of origin, rare metastatic presentation, and/or other diagnostic uncertainty, we advocate for the use of molecular testing to both arrive at a diagnosis and to develop a targeted treatment plan based on specific mutations.

Glossary of Genomic Terms and Nomenclature

AKT: v‐akt murine thymoma viral oncogene.

ARID1: AT‐rich interactive domain‐containing protein 1A.

KRAS: Kirsten rat sarcoma viral oncogene.

MAPK: mitogen‐activated protein kinase.

mTOR: mammalian target of rapamycin.

PTEN: phosphatase and tensin homolog.

PI3K phosphoinositide‐3‐kinase.

PIK3CA: PI3K catalytic subunit α.

Author Contributions

Conception/design: Shuk On Annie Leung

Provision of study material or patients: Marisa Nucci, Michael G. Muto, Susana Campos

Collection and/or assembly of data: David Chapel, Annacarolina DaSilva

Manuscript writing: Shuk On Annie Leung, Olivia Foley, David Chapel, Annacarolina Da Silva

Final approval of manuscript: Shuk On Annie Leung, Olivia Foley, David Chapel, Annacarolina Da Silva, Marisa Nucci, Michael G. Muto, Susana Campos

Disclosures

The authors indicated no financial relationships.

Disclosures of potential conflicts of interest may be found at the end of this article.

No part of this article may be reproduced, stored, or transmitted in any form or for any means without the prior permission in writing from the copyright holder. For information on purchasing reprints contact commercialreprints@wiley.com. For permission information contact permissions@wiley.com.

References

  • 1. Garcia EP, Minkovsky A, Jia Y et al. Validation of OncoPanel: A targeted next‐generation sequencing assay for the detection of somatic variants in cancer. Arch Pathol Lab Med 2017;141:751–758. [DOI] [PubMed] [Google Scholar]
  • 2. Cibulskis K, Lawrence MS, Carter SL et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotech 2013;31:213–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Ramos AH, Lichtenstein L, Gupta M et al. Oncotator: Cancer variant annotation tool. Hum Mutat 2015;36:E2423–E2429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Schittenhelm J, Krischker N, Gepfner‐Tuma I et al. Oncogenic KRAS hotspot mutations are rare in IDH‐mutant gliomas. Brain Pathol 2019;29:321–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Umphlett M, Shea S, Tome‐Garcia J et al. Widely metastatic glioblastoma with BRCA1 and ARID1A mutations: A case report. BMC Cancer 2020;20:47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Bardi G, Pandis N, Schousboe K et al. Near‐diploid karyotypes with recurrent chromosome abnormalities characterize early‐stage endometrial cancer. Cancer Genet Cytogenet 1995;80:110–114. [DOI] [PubMed] [Google Scholar]
  • 7. Banno K, Yanokura M, Iida M et al. Carcinogenic mechanisms of endometrial cancer: Involvement of genetics and epigenetics. J Obstet Gynaecol Res 2014;40:1957–1967. [DOI] [PubMed] [Google Scholar]
  • 8. Westin SN, Sill MW, Coleman RL et al. Safety lead‐in of the MEK inhibitor trametinib in combination with GSK2141795, an AKT inhibitor, in patients with recurrent endometrial cancer: An NRG Oncology/GOG study. Gynecol Oncol 2019;155:420–428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Risinger JI, Hayes AK, Berchuck A et al. PTEN/MMAC1 mutations in endometrial cancers. Cancer Res 1997;57:4736–4738. [PubMed] [Google Scholar]
  • 10. Li W, Wang Y, Fang X et al. Differential expression and clinical significance of DNA methyltransferase 3B (DNMT3B), phosphatase and tensin homolog (PTEN) and human MutL homologs 1 (hMLH1) in endometrial carcinomas. Med Sci Monitor 2017;23:938–947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Slomovitz BM, Coleman RL. The PI3K/AKT/mTOR pathway as a therapeutic target in endometrial cancer. Clin Cancer Res 2012;18:5856–5864. [DOI] [PubMed] [Google Scholar]
  • 12. Karamurzin Y, Rutgers JK. DNA mismatch repair deficiency in endometrial carcinoma. Int J Gynecol Pathol 2009;28:239–255. [DOI] [PubMed] [Google Scholar]
  • 13. Le DT, Uram JN, Wang H et al. PD‐1 blockade in tumors with mismatch‐repair deficiency. N Engl J Med 2015;372:2509–2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Tessier‐Cloutier B, Coatham M, Carey M et al. SWI/SNF‐deficiency defines highly aggressive undifferentiated endometrial carcinoma. J Pathol Clin Res 2021;7:144–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Jelinic P, Ricca J, Van Oudenhove E et al. Immune‐active microenvironment in small cell carcinoma of the ovary, hypercalcemic type: Rationale for immune checkpoint blockade. J Natl Cancer Inst 2018;110:787–790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Campos S, Davey P, Hird A et al. Brain metastasis from an unknown primary, or primary brain tumour? A diagnostic dilemma. Curr Oncol 2009;16:62–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Uccella S, Morris JM, Multinu F et al. Primary brain metastases of endometrial cancer: A report of 18 cases and review of the literature. Gynecol Oncol 2016;142:70–75. [DOI] [PubMed] [Google Scholar]
  • 18. Piura E, Piura B. Brain metastases from endometrial carcinoma. ISRN Oncol 2012;2012:581749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Cybulska P, Stasenko M, Alter R et al. Brain metastases in patients with low‐grade endometrial carcinoma. Gynecol Oncol Rep 2018;26:87–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Hainsworth JD, Rubin MS, Spigel DR et al. Molecular gene expression profiling to predict the tissue of origin and direct site‐specific therapy in patients with carcinoma of unknown primary site: A prospective trial of the Sarah Cannon Research Institute. J Clin Oncol 2013;31:217–223. [DOI] [PubMed] [Google Scholar]
  • 21. Levine DA, Getz G, Gabriel SB et al. Integrated genomic characterization of endometrial carcinoma. Nature 2013;497:67–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Brennan Cameron W, Verhaak Roel GW, McKenna A et al. The somatic genomic landscape of glioblastoma. Cell 2013;155:462–477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Rimel BJ, Huettner P, Powell MA et al. Absence of MGMT promoter methylation in endometrial cancer. Gynecol Oncol 2009;112:224–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Soumerai TE, Donoghue MTA, Bandlamudi C et al. Clinical utility of prospective molecular characterization in advanced endometrial cancer. Clin Cancer Res 2018;24:5939–5947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Makker V, Taylor MH, Aghajanian C et al. Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer. J Clin Oncol 2020;38:2981–2992. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Oncologist are provided here courtesy of Oxford University Press

RESOURCES