Abstract
Ongoing discoveries in cancer genomics and epigenomics have revolutionized clinical oncology and precision health care. This knowledge provides unprecedented insights into tumor biology and heterogeneity within a single tumor, among primary and metastatic lesions, and among patients with the same histologic type of cancer. Large-scale genomic sequencing studies also sparked the development of new tumor classifications, biomarkers, and targeted therapies. Because of the central role of imaging in cancer diagnosis and therapy, radiologists need to be familiar with the basic concepts of genomics, which are now becoming the new norm in oncologic clinical practice. By incorporating these concepts into clinical practice, radiologists can make their imaging interpretations more meaningful and specific, facilitate multidisciplinary clinical dialogue and interventions, and provide better patient-centric care. This review article highlights basic concepts of genomics and epigenomics, reviews the most common genetic alterations in cancer, and discusses the implications of these concepts on imaging by organ system in a case-based manner. This information will help stimulate new innovations in imaging research, accelerate the development and validation of new imaging biomarkers, and motivate efforts to bring new molecular and functional imaging methods to clinical radiology.
Keywords: Oncology, Cancer Genomics, Epignomics, Radiogenomics, Imaging Markers
Supplemental material is available for this article.
© RSNA, 2023
Keywords: Oncology, Cancer Genomics, Epignomics, Radiogenomics, Imaging Markers
Summary
In the era of precision health care, basic genomic concepts and the associated implications in diagnostic imaging will help radiologists expand their impact on patient management decisions for patients with cancer.
Essentials
■ Clinical cancer care has entered an era of genomics and precision health care, with radiologists positioned to play key roles in improving outcomes for patients.
■ Radiogenomics represents a fast-evolving field aimed at correlating cancer genotypes with imaging phenotypes in the search for new noninvasive biomarkers.
■ Radiologists need to understand basic genomic concepts and their impact on imaging in cancer.
Introduction
Clinical oncology has entered an era where diagnosis, treatment options, and prognostication rely heavily on genomic profiles for each patient. Due to the central role of imaging in cancer, imaging specialists need to know the basic concepts of genomics that now underlie clinical oncology. Advances in high-throughput parallel technologies have brought about the “omics revolution,” where omics broadly means “study of the whole.” Cancer genomics focuses on genome-wide DNA aberrations, while transcriptomics and proteomics analyze messenger RNA and proteins, respectively. The growing field of epigenomics uses methods such as methylation profiling to detect reversible modifications in DNA or histones, such as methylation and acetylation, that affect gene expression without altering DNA sequences (Fig 1). Omics-level information continues to advance the understanding of genetic and molecular pathogenesis of cancers (1) and better informs patient stratification at all stages of health care, including prevention, screening, diagnosis, treatment, and follow-up.
Figure 1:
Recent advances in high-throughput parallel technologies have brought about the omics revolution. Epigenomics uses methylation profiling to study reversible modifications in a cell’s DNA or histones, such as methylation and acetylation, which affect gene expression without altering the DNA sequence. Genomics focuses on genome-wide mutations at the level of DNA, using whole-genome, whole-exome, or oncopanel sequencing. Transcriptomics and proteomics allow for consideration of the entire cellular messenger RNA (mRNA) or protein composition, respectively, with technologies including RNA-sequencing (RNA-seq), microarray, and mass spectrometry. Ac = acetylation, Me = methylation.
Cancer originates from genetic mutations in DNA, leading to hallmarks including uncontrollable cell proliferation and escape from cell death. Tumors can be broadly classified as hereditary or sporadic, depending on the predisposing mutations. Hereditary cancer arises from germline DNA mutations inherited from one parent. Although germline mutations exist in all cells, hereditary cancers occur only in characteristic organs, indicating that local environments regulate tumorigenesis. Cancer more commonly arises sporadically from acquired (or somatic) DNA mutations that develop during an individual’s lifetime. DNA replication errors during divisions of normal tissue stem cells and carcinogens like radiation, chemicals, or chronic inflammation increase somatic mutations (2,3). Some cancers arise from either germline or sporadic mutations. For example, 40% of retinoblastomas are heritable, often manifesting with bilateral and early onset disease (4), in opposition to the de novo mutation.
Companies continue to develop newer, faster, and more cost-effective next-generation DNA and RNA sequencing methods. Current sequencing technologies offer options for nontargeted characterization of the whole genome, methylation profiling (5) (epigenomics), and transcriptomics (messenger RNA) (Fig 1). To reduce costs and analysis, oncologists may sequence only a predefined panel of oncogenes, transcripts, or antibodies tailored for a specific malignancy (ie, oncopanel DNA or RNA sequencing, proteome microarrays) (Fig 1). Investigators also may sequence only known protein-coding segments of DNA, called exons (exome sequencing) (Fig 1). Omics tests are performed on primary tumors, metastases, or circulating tumor cells. Sequencing cell-free circulating DNA provides a “liquid biopsy“ for mutations in oncogenes, response to therapy, and disease recurrence (6).
As oncology shifts to omics-informed precision medicine, radiologists must understand how certain cancer genotypes manifest at imaging. Precision medicine also propels radiology reports to move toward more quantitative, reproducible metrics. Numerous standardized reporting systems, such as the Breast Imaging Reporting and Data System, or BI-RADS, and response criteria measures, including the Response Evaluation Criteria in Solid Tumors version 1.1, or RECIST 1.1, provide more quantitative assessments of disease risk and response to therapies. Ongoing advances in pixel-level analysis and artificial intelligence also offer new approaches to extracting quantitative tumor imaging features from conventional imaging studies (7).
In this review article, we demonstrate how a radiologist can advance cancer clinical care and research in the era of omics-informed medicine. We present the most common genetic and epigenetic mutations and pathways by organ system in a case-based manner. A list of genes mentioned in this review but not discussed in detail can be found in Table S1. We also present key imaging findings and implications in oncologic care.
New Cancer Classification and Review of Common Cancer Genomics and Epigenetics Alterations, Clinical Implications, and Imaging Manifestations by Organ
Current oncology practice is progressively moving toward genomics-informed management. Cancers from different sites of origin but harboring the same genetic or epigenetic alterations may be treated using similar therapy (8). For example, lapatinib, an inhibitor of human epidermal growth factor 2 (HER2) that was initially approved by the U.S. Food and Drug Administration for breast cancer in 2007, also shows efficacy in clinical trials for gastric cancer and colorectal cancers with HER2 mutations (9,10) (Fig 2).
Figure 2:
HER2 mutation in colon cancer metastasis in liver. (A–D) Axial intravenous contrast–enhanced CT images in the portal venous phase in a 70-year-old man with metastatic colon cancer show multiple liver metastases (arrow in A, B) that progressed under multiple treatment regimens. Tumor molecular profiling showed an HER2 mutation, and the patient was offered treatment with trastuzumab and lapatinib (selective HER2 inhibitors). On follow-up scans, there was partial response to treatment, with substantial size reduction and necrosis of the liver metastases (arrow in C, D).
Germline TP53 mutations in Li-Fraumeni syndrome predispose to early-onset cancers in diverse tissues, most commonly breast cancer, soft-tissue sarcoma, brain tumors, adrenocortical carcinoma, and bone sarcomas. Early identification and testing of these individuals is essential for adequate screening (11). Current screening protocols indicate that carriers should undergo abdominal US every 3–4 months, annual whole-body and brain MRI, and annual breast MRI for female carriers. Prophylactic mastectomy has been advocated in some instances. Patients require tailored cancer treatment, as radiation therapy and conventional chemotherapy increase the risk of subsequent malignancies (12). TP53 mutations occur very commonly in many other types of cancers. These mutations inform prognosis, such as in hepatocellular cancer, endometrial cancer, and bladder cancer (13,14). We review the most common cancer mutations, mechanisms of oncogenesis, and implications of these concepts on imaging by organ system.
Brain Cancer
Glioma is the most common type of primary brain tumor. Historically, gliomas were classified by histopathologic features into astrocytomas, oligodendrogliomas, oligodendroastrocytomas, ependymomas, and gangliogliomas. Gliomas can be broadly divided into low-grade gliomas and high-grade gliomas by histopathologic features. Treatment of low-grade gliomas entails surgical resection and adjuvant radiation and/or chemotherapy, with a life expectancy of 6–13 years (15). High-grade gliomas include anaplastic astrocytomas (grade III) and glioblastomas (grade IV). Treatment of glioblastomas remains challenging, with surgical resection followed by radiochemotherapy producing a median overall survival of only 14 months (16).
The Cancer Genome Atlas Research Network and the 2016 World Health Organization classification (17) revolutionized the diagnosis and classification of gliomas by using genomic, epigenomic, and molecular profiles. The Cancer Genome Atlas provides open access clinical, genomic, epigenomic, and molecular data from patients with glioblastomas, with corresponding imaging data in the Cancer Imaging Archive (18). These data supported several exploratory radiogenomics studies. The Visually Accessible Rembrandt Images (or, VASARI) project analyzed 24 semantic imaging observations (https://www.cancerimagingarchive.net/) at MRI against clinicogenomic features. This study produced radiogenomic signatures of key molecular markers of gliomas, including IDH1/2 mutation, 1p/19q codeletion, O6-methylguanine DNA methyltransferase (MGMT) methylation, amplification and mutation of the EGFR gene, and histone H3 K27M mutation (19,20) (Table 1), described as follows:
Table 1:
Summary of Most Clinically and Imaging-Relevant Genetic and Epigenetic Alterations in Glioblastoma
Isocitrate dehydrogenases (IDHs) regulate cellular metabolism and epigenetic modifications. Both wild-type mitochondrial (IDH2) and cytosolic (IDH1) IDH forms convert isocitrate into α-ketoglutarate. Mutant IDHs generate an oncometabolite, 2-hydroxyglutarate. Given structural similarity to α-ketoglutarate, 2-hydroxyglutarate competes with and inhibits pathways such as DNA methylation, leading to aberrant gene expression and oncogenesis (21). All patients with glioblastoma undergo genetic testing for IDH. Patients with glioblastomas with mutant IDH show better survival than do those with wild-type IDH1/2 tumors (22).
IDH-mutant glioblastoma localizes predominantly to the frontal lobe or subventricular region of the lateral ventricles, with rare invasion of the eloquent areas of brain (23). It generally has an indistinct tumor border, cortical involvement, less peritumoral edema, and limited or no enhancement (Fig 3) (24–26). Mean apparent diffusion coefficient (ADC) values are higher in IDH-mutant glioblastoma compared with IDH wild-type glioblastoma (27), with values of 0.9 x 10−3 mm2/sec and higher more likely representing IDH-mutant glioblastoma (26). IDH-mutant glioblastomas accumulate high amounts of 2-hydroxyglutarate and low choline to creatinine ratios at MR spectroscopy (28). IDH wild-type glioblastomas occur preferentially at the subcortical white matter of the subventricular zones of both hemispheres, mostly in the anterior horns. These glioblastomas exhibit marked peripheral enhancement, central necrosis, and substantial surrounding vasogenic edema (Fig 3).
1p/19q codeletion impacts migration, apoptosis, and cell cycle regulation. Once identified clinically using fluorescence in situ hybridization (29), it indicates low-grade gliomas, supporting the diagnosis of an oligodendroglial tumor (30). 1p/19q codeletion is associated with improved patient survival and treatment response. If associated with IDH mutations, the survival rate is even higher (median survival of 8 years) when compared with patients with IDH mutations without codeletion (median survival of 6.3 years) or patients with IDH wild-type tumors (median survival of 1.7 years) (31). 1p/19q codeletion is often found together with IDH1/2 mutation, therefore sharing similar imaging features.
MGMT is a DNA repair enzyme. Methylation of the promoter region of the gene decreases levels of this enzyme, resulting in less efficient DNA repair following cytotoxic chemotherapy. MGMT methylation frequently occurs with IDH mutations and implicates a better prognosis. MGMT methylation strongly predicts a positive response to temozolomide in new and recurrent high-grade gliomas (32). Pseudoprogression following treatment occurs more commonly in glioblastomas with both MGMT and IDH mutations (33). Imaging phenotypes of MGMT-mutant glioblastomas include a temporal or frontal lobe distribution (34), higher permeability parameters (transfer constant) at dynamic contrast-enhanced MRI, and higher minimum ADC values (35).
Amplification and mutation of EGFR occurs more commonly in primary than in secondary glioblastoma, inducing rapid cellular proliferation and resultant poor prognosis. EGFR-mutant tumors most frequently localize to the left temporal lobe (34), demonstrate complete enhancement or incomplete ring enhancement (36), and show increased perfusion at MRI (37).
Histone H3 K27M mutation, a newly defined entity in the World Health Organization group of grade IV diffuse gliomas, correlates with a poor prognosis. K27M mutation occurs on the histone gene encoding H3.3 and causes epigenetic changes by altering chromatin accessibility. This malignancy can mimic gliomatosis cerebri with additional midline structure involvement. Imaging features include a central or midline location and predominant involvement of the brainstem, thalamus, and spine. K27M-mutant tumors show partial to no, diffuse, or irregular peripheral enhancement. These tumors commonly are solid with mildly restricted diffusion (38).
Figure 3:
IDH-mutant and IDH wild-type gliomas. Axial (A, E) T2-weighted, (B) diffusion-weighted, (C) apparent diffusion coefficient map, and (D, F) T1-weighted MR images with extracellular gadolinium. (A–D) Images in a 65-year-old male patient diagnosed with IDH-mutant glioma demonstrate a typical imaging phenotype. The patient presented with a large, predominantly frontal mass, with indistinct borders, minimal peritumoral edema, and involvement of the cortex (arrow in A). At diffusion-weighted imaging, the mass showed high signal intensity (arrow in B), with high apparent diffusion coefficient values (arrow in C). The contrast-enhanced image demonstrates a poorly enhancing mass (arrow in D). (E, F) Images in a 68-year-old male patient with IDH wild-type glioma show a large left temporal central necrotic mass with marked peripheral enhancement (arrow in F) and surrounding vasogenic edema (arrow in E). (G) Isocitrate dehydrogenase (IDH) function and oncogenic pathway. IDHs play central roles in cellular metabolism and epigenetic regulation. Wild-type mitochondrial (IDH2) and cytosolic (IDH1) IDH forms convert isocitrate into α-ketoglutarate (αKG). When mutated, IDHs generate an oncometabolite, 2-hydroxyglutarate (2HG), which leads to epigenetic changes with aberrant histone and DNA methylation, causing oncogenesis.
Lung Cancer
Genomic characterization of lung cancer classifies tumors according to distinct mutations and matched options for targeted, rather than conventional, cytotoxic chemotherapies. According to current guidelines, patients with advanced or metastatic non–small cell lung cancer (NSCLC) should undergo genetic testing for driver mutations, especially for adenocarcinomas by histologic analysis or if patients have a light or no smoking history. Testing includes EGFR, ALK, KRAS, ROS1, RET, MET, BRAF, and HER2 because activating mutations in these genes have potential targeted therapies (39). National Comprehensive Cancer Network guidelines on NSCLC recommend that all patients with metastatic disease undergo NTRK1/2/3 gene fusion testing, which may indicate a response to tropomyosin receptor kinase inhibitors (larotrectinib or entrectinib) (40). Unfortunately, tumor genomic sequencing is not standard of care worldwide. Furthermore, many patients with advanced disease and compromised status are not suitable for invasive diagnostic procedures. By identifying surrogate imaging markers, radiogenomics can complement genetic testing, predict certain driver mutations, and help direct targeted therapy (41).
NSCLC, the most common type of lung cancer, commonly shows mutations in TP53 (50%), KRAS (30%), and EGFR (15%). ALK, MET, and HER2 genetic alterations are less frequent (<10%) but are important for radiologists to know about, given the availability of targeted therapies and distinct imaging features (Table 2). Interestingly, mutations in EGFR, KRAS, and ALK are typically mutually exclusive in patients with NSCLC (42).
Table 2:
Summary of Most Clinically and Imaging-Relevant Genetic Alterations in Non–Small Cell Lung Cancer
EGFR and HER2 are two of the most frequently dysregulated oncogenes in solid cancers (43). Epidermal growth factor receptor (EGFR) and HER2 are members of the ErbB family of receptor tyrosine kinases that activate downstream signaling cascades driving proliferation, survival, and differentiation (44). Kinase-activating mutations in EGFR occur primarily in cancers like NSCLC and glioblastoma or secondary to acquired drug resistance. EGFR mutations occur in approximately 15% of NSCLC cases overall and in 50% of females with adenocarcinoma who do not smoke (41); it is more common in women than in men and has a higher incidence in Asian populations (45). Targeted molecular therapy involves EGFR tyrosine kinase inhibitors like erlotinib (46). Imaging features associated with EGFR mutations include the following: pure or mixed ground-glass opacity (odds ratio, 1.86) (47), with higher volume ground-glass opacity in patients with exon 21 missense EGFR mutation compared with exon 19 mutation or EGFR wild-type tumors (48); air bronchograms (odds ratio, 1.6); pleural retraction (odds ratio, 1.99); small size; and absence of fibrosis (49). For detection of an EGFR mutation, the previous studies found an odds ratio of 1.58 for early disease stage. Furthermore, emerging PET imaging probes are being developed to detect total EGFR and even activate mutations of this receptor, with initial clinical translation (50,51).
ALK mutations occur in approximately 4% of NSCLC cases, more frequently occurring in young patients who do not smoke (41). Targeted treatment involves ALK tyrosine kinase inhibitors like crizotinib and alectinib (52). Radiologic features associated with ALK mutations in NSCLC include central solid tumors with pleural effusions (49,53), lymphangitic spread, and advanced nodal, pleural, or pericardial metastases (54) (Fig 4). Treating patients with ALK-mutant NSCLC using the targeted therapy, crizotinib, commonly (45%) causes new or increasing simple or complex renal cysts, which often regress after cessation of treatment (55) (Fig 4). Awareness of these treatment-related changes will allow for a more confident and appropriate diagnosis.
Figure 4:
ALK mutation in non–small cell lung cancer. Axial images from (A, B) soft-tissue and lung window CT, (C, D) fusion PET/CT, and (E, F) postintravenous contrast CT of right kidney. Patient is a 53-year-old female who does not smoke, with non–small cell lung cancer with an ALK mutation who presented with (A–D) a central solid fluorodeoxyglucose-avid tumor (blue arrow) accompanied by pleural effusion (green arrow), metastatic mediastinal and hilar lymphadenopathy (yellow arrows), pulmonary lymphangitic carcinomatosis (red arrow in B) and vertebral bone metastasis (white arrow in D). Patients treated with crizotinib (ALK and ROS1 inhibitor) often develop new or increased renal cysts (orange arrow in E, F) with simple or complicated features as part of their treatment. These cysts often regress after stopping crizotinib.
Exon 14 mutations in MET, a receptor tyrosine kinase, occur in approximately 3%–4% of patients with NSCLC (56). U.S. Food and Drug Administration–approved molecular therapies against MET-mutated NSCLC include cabozantinib and crizotinib (57,58). Imaging features associated with MET mutations in NSCLC include peripheral masses typically located in the upper lobes, cavitation, and cystic changes. Mutations in MET increase metastases to the lungs, brain, and adrenal glands (59) (Fig 5).
Figure 5:
MET mutation in non–small cell lung cancer. (A) Axial soft-tissue window CT, (B) axial fusion PET/CT, and (C) coronal soft-tissue window CT images in a 35-year-old nonsmoking male patient with non–small cell lung cancer with a MET exon 14 mutation who presented with an aggressive peripheral fluorodeoxyglucose-avid tumor (green arrow) located in the upper zone of the left lower lobe, with a heterogenic appearance including cystic changes, septations, and calcifications. The tumor invaded the chest wall, compromising the 6th left rib and intercostal muscles. A distant brain metastasis was found during staging MRI (not shown).
HER2 mutations occur in 1%–3% of patients with NSCLC, predominantly in adenocarcinomas arising in people who do not smoke and in females (60). Patients may respond to trastuzumab-based regimens, which target the HER2 receptor (61). A pattern of disseminated lung nodules has been observed frequently with HER2 mutations in lung cancer (41).
Breast Cancer
Genetic mutations driving breast cancer can be somatic (such as TP53 and RB) or inherited germline mutations (62). Germline mutations, most commonly in BRCA1 or BRCA2, account for up to 5%–10% of breast cancers (63–66). BRCA 1 and 2 genes maintain genomic stability by promoting efficient and precise repair of DNA double-strand breaks. BRCA1 is one of the most commonly mutated tumor suppressor genes, accounting for 5% of unselected breast cancers. Patients with a BRCA mutation have up to an 80% lifetime risk of developing breast cancer and a 50% lifetime risk of ovarian cancer (67). BRCA mutations have also been found in several other cancers, including pancreatic, prostate, and peritoneal malignancies (41). Identifying BRCA mutations in breast cancer helps to stratify the risk for breast and other cancers (68) and manage patients by either intense screening or prophylactic mastectomy (69). BRCA mutations confer susceptibility to targeted therapies (poly [adenosine diphosphate-ribose] polymerase inhibitors, ie, olaparib), and patients carrying these mutations have a better response to immunotherapy (70,71).
Clinical guidelines categorize breast cancer into four major, biologically distinct subtypes on the basis of the immunohistochemical expression of hormone receptors (estrogen receptor [ER], progesterone receptor [PR], and HER2 status) and distinct molecular and genetic profiles (Table 3) (72).
Table 3:
Summary of Clinical Features, Implications, and Imaging Findings of Current Clinically Used Breast Cancer Molecular and Genomic Subtypes
As many as 75% of invasive breast carcinomas are associated with significantly higher ER expression (ER+), rendering it an important diagnostic and treatment biomarker. PR expression is modulated by ERs, and as such, provides information about the current functionality of the ER pathway. Higher levels of PR expression (PR+) are associated with improved overall survival and time to recurrence, while lower levels (PR−) are associated with a more aggressive course and worse prognosis (73). HER2 overexpression (HER2+) occurs in approximately 20% of breast cancers and has historically been associated with decreased survival (74).
Subtypes of breast cancer provide essential predictive information for therapy. Patients in the luminal A (ER+, PR+, HER2−) and luminal B (ER+, PR− or low, HER2± ) subgroups receive targeted antiestrogen therapies, such as selective ER modulators (eg, tamoxifen), selective ER degraders (eg, fulvestrant), or aromatase inhibitors (eg, anastrozole and letrozole) (75). Anti-HER2 therapies, such as trastuzumab and lapatinib, improve survival in patients with the HER2+ subtype (76,77). HER2 overexpression is not unique to breast cancer, and overexpression and/or amplifications or other mutations of HER2 occur in gastric, biliary, colorectal, lung, and bladder cancer (10). Checkpoint inhibitor immunotherapy and poly (adenosine diphosphate-ribose) polymerase inhibitors remain the only targeted therapies for patients with the basal or triple-negative subtype, and conventional chemotherapy remains a mainstay of care. Patients with basal or triple-negative breast cancer have increased risk for metastases and poorer prognosis relative to those with other subtypes (78,79).
Distinct imaging features have been associated with different molecular subtypes of breast cancer (Table 3). Luminal A tumors have irregular shapes, spiculated margins, clustered calcifications, and size less than 2 cm (80,81). At MRI, luminal A tumors usually lack peritumoral enhancement. The luminal B subtype shows irregular shape, noncircumscribed margins, and skin or nipple extension at mammography and lower ADC values at MRI. Imaging findings associated with the HER2 subtype include irregular or round shape, spiculated or noncircumscribed margins, higher ADC values, peritumoral edema, and persistent inflow type enhancement on delayed MR images. Branching or fine linear calcifications, microcalcifications, and increased breast density also occur commonly in tumors with HER2 overexpression (81,82). Basal-like or triple-negative tumors frequently have BRCA mutations. These tumors frequently present as round or oval well-circumscribed lesions with smooth margins and no calcifications (83). US often reveals a hypoechoic mass with microlobulated or angular margins with parallel orientation, with one-third of cases showing posterior acoustic enhancement (83). At MRI, this cancer subtype commonly shows high internal signal intensity on T2-weighted images (84), rim enhancement, higher ADC value than other subtypes, and peritumoral edema (81) (Fig 6). When irregular margins and intratumoral necrosis are present, these features correlate with a poor response to neoadjuvant chemotherapy (85). Peritumoral edema has also been related to reduced disease-free survival (86). Kinetic curves for contrast enhancement and tumor size in basal-like tumors have variable reporting, excluding these indicators from reliably differentiating the basal-like subtype from other subtypes (81). While imaging features aid in breast cancer molecular or genetic subtype differentiation, biopsy still serves as the reference standard.
Figure 6:
Non–triple-negative and triple-negative breast cancer with BRCA1 mutation. (A, G) Axial diffusion-weighted images (b = 850 sec/mm2 and b = 800 sec/mm2, respectively), (B, H) apparent diffusion coefficient maps, (C, I) maximum intensity projection images, (D, J) high-resolution dynamic contrast-enhanced subtraction images, (E, K) fat-saturated images, and (F, L) fat-saturated T2-weighted images. (A–F) Images in a 31-year-old female patient with BRCA1 overexpression and cT2N1 triple-negative left breast cancer who presented with an oval-shaped, rim-enhancing, centrally necrotic, and well-circumscribed mass (yellow arrow) with restricted diffusivity (apparent diffusion coefficient, 0.86 × 10−3 mm2/sec) in the upper outer quadrant of the left breast mid depth. This patient was treated with neoadjuvant chemotherapy. (G–L) Images in a 45-year-old female patient with a 3.2-cm biopsy-proven non–triple-negative estrogen receptor– and progesterone receptor–positive, human epidermal growth factor receptor 2–negative left breast cancer with a positive axillary lymph node. MR images show multicentric disease, with nonnecrotic enhancing foci (yellow arrow).
Gastrointestinal System
Hepatocellular carcinoma.— Hepatocellular carcinoma (HCC) represents about 90% of all primary liver cancers. Genetic and molecular pathogenesis of HCC is heterogeneous as outlined by data in The Cancer Genome Atlas. The two most common mutations in HCC are in the TP53 (31%) and the CTNNB1 (27%) genes (87). The genomic and histologic features classify HCC into two main groups: (a) proliferative, poorly differentiated tumors characterized by TP53 mutation and (b) nonproliferative, well-differentiated tumors characterized by CTNNB1 mutations (Table 4) (88).
Table 4:
Summary of Genetic and Histologic Subtypes of Hepatocellular Carcinoma and Key Imaging Features
The nonproliferative, well-differentiated HCC subtype has good prognosis, with low levels of α-fetoprotein and a nonaggressive phenotype. Imaging usually shows a single small lesion with smooth margins and a capsule (89), mild hyperintensity on T2-weighted images, and high ADC at diffusion-weighted imaging (90). Other imaging biomarkers include intratumoral fat and low fluorine 18 (18F)–labeled fluorodeoxyglucose (FDG) accumulation (89). The value of intratumoral fat remains controversial as it may also occur in poorly differentiated HCCs (91). Nonproliferative HCCs comprise the β-catenin (CTNNB1 gene) and the steatohepatitic subgroups. CTNNB1–mutant HCCs are associated with alcohol consumption (92) and are characterized by activation of the Wnt/β-catenin pathway. CTNNB1-mutant HCC shows hyperintensity at the hepatobiliary phase of gadoxetic acid–enhanced MRI because of overexpression of the OATP1B3 transporter (Fig 7) (93,94). Steatohepatitic HCCs demonstrate activation of the JAK/STAT3 pathway and lack of a CTNNB1 mutation. These HCCs correlate with metabolic syndromes and steatosis in the background liver. At imaging, steatohepatitic HCCs show abundant fat infiltration with loss of signal at T1-weighted MRI in the opposed phase, no necrosis, and hypoenhancement in the hepatobiliary phase. At dynamic CT, this subtype exhibits hyperenhancement in the arterial phase, isoenhancement in the portal phase, and washout in the late phase (95).
Figure 7:
CTNNB1-mutant and wild-type, biopsy-proven hepatocellular carcinoma (HCC). Images from axial contrast-enhanced liver MRI, using an intravenous hepatobiliary-specific contrast agent (gadoxetate disodium): (A, D) arterial, (B, E) portal venous, and (C, F) hepatobiliary phase. (A–C) Images in a 67-year-old man with a background of liver cirrhosis who presented with a focal liver lesion (arrow), with (A) nonrim arterial phase hyperenhancement, (B) washout in the portal venous phase, (C) and hyperintensity in the hepatobiliary phase. (D–F) Images in a 59-year-old man with a background of liver cirrhosis who presented with a focal liver lesion (arrow) with (D) nonrim arterial phase hyperenhancement, (E) washout in the portal venous phase, (F) and hypointensity in the hepatobiliary phase. Hyperintensity at hepatobiliary phase can distinguish CTNNB1-mutant HCC from CTNNB1 wild-type HCC.
The proliferative, poorly differentiated HCC subtype includes the macrotrabecular massive and scirrhous subgroups, which are associated with high serum α-fetoprotein levels and poor survival (96). Macrotrabecular massive HCC frequently occurs in the setting of chronic hepatitis B virus infection, TP53 gene mutation, VEGFA overexpression, and FGF19 gene amplifications (88,97), with patients often presenting with early relapse. At imaging, the macrotrabecular massive subgroup characteristically shows greater than 20% necrosis (98) and other aggressive features, such as large lesions, multifocality, nonsmooth tumor margins, arterial phase hyperenhancement, confluent multinodularity, infiltrative behavior, macro- and microvascular invasion, bile duct dilatation, and increased 18F-FDG uptake (89,99). Several MRI features have been reported to predict microvascular invasion, namely the presence of internal arteries, absence of a hypoattenuating halo, infiltrative behavior, decreased ADC, peritumoral enhancement, nonsmooth tumor margins, peritumoral hypointensity during the hepatobiliary phase of gadoxetic acid–enhanced MRI, and high 18F-FDG accumulation (100–104).
Scirrhous HCCs resemble intrahepatic cholangiocarcinomas, with significant desmoplasia, progenitor cell phenotype, activation of the transforming growth factor β pathway (88,97), and common expression of cytokeratin 19 (105). Imaging differentiation from intrahepatic cholangiocarcinoma is usually the main challenge. Ancillary findings can help make the diagnosis, including central hypointensity on T2-weighted images, intratumoral blood products, and the presence of multifocal intratumoral septa, an enhancing capsule, and a nontargetoid appearance at diffusion-weighted imaging for scirrhous HCC (106,107).
Further studies are still required to establish new and optimized imaging-based stratification of HCCs as new morphologic-molecular HCC subtypes emerge (13). Such classification will enhance patient care by tailoring treatment decisions and monitoring to individual patients.
Colorectal cancer.— Colorectal cancer (CRC) evolves through a stepwise progression from normal tissue epithelium to precursor lesions and then to carcinoma. Three main molecular mechanisms have been documented in CRC tumorigenesis: (a) chromosomal instability (85%), (b) microsatellite instability (MSI) (15%), and (c) CpG island methylator phenotype (17%) (108).
CRC tumors driven by chromosomal instability, defined by alterations in number and structure of chromosomes, are often associated with mutations in the APC, TP53, KRAS, and BRAF genes (109), or via Wnt and mitogen-activating protein kinase, or MAPK, pathway activation. These molecular alterations underlie the classic adenoma-carcinoma sequence. CRC tumors with mutant KRAS and BRAF tend to be more aggressive, with a poorer prognosis. In metastatic CRC, anti-EGFR monoclonal antibodies such as cetuximab and panitumumab demonstrate some clinical benefit. These antibodies block the extracellular domain of EGFR and subsequent downstream activation of the RAS/RAF/MEK/ERK pathway (110) (Fig 8E). However, if downstream signaling molecules are mutated, most commonly KRAS and BRAF mutations in 30%–50% and 10% of CRC cases, respectively, the anti-EGFR antibodies get bypassed, with resulting drug resistance (108,111,112). All patients with metastatic CRC who are candidates for anti-EGFR antibody therapy should be tested for mutant KRAS and BRAF as standard of care (108,111). Because these mutations increase membrane glucose transporter levels, several articles have reported the possible use of 18F-FDG PET/CT uptake as a surrogate marker for KRAS/BRAF mutations (113). However, a recent meta-analysis found low sensitivity and specificity of 18F-FDG PET/CT findings for this purpose (114). Other studies have shown the following additional characteristics of BRAF-mutated colonic tumors: right-sided predominance involving the cecum and up to the proximal two-thirds of the transverse colon, larger primary tumors, heterogeneous enhancement, lack of nonperitoneal metastasis, higher rates of serrated polyp formation, mucinous pathologic features, poor differentiation, and female predominance, particularly in individuals more than 70 years old (115) (Fig 8).
Figure 8:
BRAF-mutant and BRAF wild-type colon cancer. (A, C) Axial and (B, D, E) coronal portal venous phase intravenous contrast–enhanced CT images. (A–D) Images in an 82-year-old woman with a BRAF mutation who presented with a right-sided colonic mass with heterogeneous enhancement (yellow arrow). (E) Image in a 75-year-old man who presented with a BRAF wild-type left colonic mass with overall homogeneous enhancement (yellow arrow). (F) BRAF function and oncogenic pathway. BRAF is a serine/threonine protein kinase involved in the signaling cascade of the mitogen-activated protein (MAP) kinase/extracellular signal-regulated (ERK) pathway, which is crucial for cell proliferation, differentiation, apoptosis, and survival. Activation of this pathway can be triggered by substrate engagement with the receptors on the cell membrane. This leads to subsequent activation of the Raf kinase through direct interaction with Ras and phosphorylation of the immediate downstream mitogen-activated protein kinase (MEK), which in turn activates ERKs. There are various subtypes of BRAF mutations, with the most common at codon 600 (BRAF V600E), accounting for about 95% of BRAF-mutant cases. This pathway has been the subject for the development of many new targeted therapies.
MSI is defined by instability of short, tandemly repeated DNA sequences known as microsatellites. MSI tumors result from mutations in mismatch repair genes, including MLH1, MSH2, MSH6, and PMS2, or from epigenetic silencing of the MLH1 promoter (108). MSI causes the most common hereditary CRC syndrome, Lynch syndrome, which accounts for 3% of all CRCs (108). Other syndromes with increased risk of CRCs include familial adenomatous polyposis, MUTYH-associated polyposis, juvenile polyposis syndrome, Peutz-Jeghers syndrome, and Cowden syndrome (116). MSI can also occur in 15% of sporadic CRC tumors, often by methylation of the CpG-rich promoter sequence of MLH1. These sporadic MSI tumors tend to arise in the proximal colon and exhibit poor differentiation, mucinous histologic findings, and prominent lymphocytic infiltration. At imaging, more than 70% of MSI-driven CRC develop proximally to the splenic flexure and typically show high lymphocyte infiltration at histopathologic analysis (108,111,116). However, no specific radiogenomic signature has been reported to date. The excellent treatment response and prognosis of MSI-driven tumors treated with checkpoint inhibitor antibodies supports routine testing for MSI status in CRC (108,111).
The CpG island methylator phenotype features hypermethylation of promoter CpG island loci, resulting in silencing of tumor suppressor genes. It is considered a distinct molecular subtype of sporadic CRC and the major driver for the serrated neoplasia pathway in CRC tumorigenesis (108). It often correlates with female sex, older age, proximal and right location, MSI-high status, BRAF mutation, and mucinous histologic findings (117). At imaging, these tumors often have similar characteristics to BRAF tumors.
Genitourinary System
Pheochromocytoma.— Pheochromocytomas and paragangliomas are rare neuroendocrine cancers arising from either the adrenal medulla or extra-adrenal paraganglia, respectively. Paragangliomas occur along the sympathetic nervous system of the chest, abdomen, or pelvis or in the parasympathetic ganglia of the head and neck. Contrary to head and neck paragangliomas, both pheochromocytomas and sympathetic paragangliomas commonly release catecholamines. Approximately 10%–15% of paragangliomas are metastatic at the time of diagnosis, with metastases generally involving lymph nodes, bones, lungs, or liver (118).
Pheochromocytomas and paragangliomas harbor somatic mutations in 30% of cases and germline mutations in about 40% of cases (119). Germline mutations affecting subunits of the Krebs cycle enzyme, succinate dehydrogenase (SDH), are among the most prevalent, occurring in the SDHA, SDHB, SDHC, SDHD, or SDHAF2 genes. Dysfunction of SDH enzymes leads to impaired conversion of succinate to fumarate and consequent substantial accumulation of succinate. As with 2-hydroxyglutarate, succinate acts as an oncometabolite and inhibits several α-ketoglutarate dioxygenases, inducing pseudohypoxia and epigenetic changes (Fig 9E). These tumors often express a pseudohypoxic phenotype, reflected by avid contrast enhancement on imaging studies because of extensive angiogenesis caused by stabilization of hypoxia-inducible factor-1α, a key regulator of angiogenesis and metabolic reprogramming.
Figure 9:
SDHD mutation in paraganglioma. (A) Axial and (B) coronal intravenous contrast–enhanced CT images in the neck and upper abdomen in the (C) arterial and (D) portal venous phases in a 24-year-old man with an SDHD mutation who presented with an arterially enhancing left cervical paraganglioma (yellow arrow). One year after resection, the patient developed a left adrenal pheochromocytoma (red arrow) showing (C) arterial enhancement and (D) washout in the portal venous phase. (E) Succinate dehydrogenase (SDH) function in the pseudohypoxia pathway. Hypoxia inducible factor (HIF)–1 is a key player in reprogramming metabolism in the lack of oxygen. In normoxia, HIF-1 rapidly undergoes degradation by prolyl-hydroxylases and proteasomal degradation facilitated by von Hippel−Lindau (VHL) protein. However, under hypoxia or pseudohypoxia, HIF-1 gets stabilized and transcribed, leading to the activation of multiple downstream pathways such as angiogenesis. This can occur in cancer cells, either by accumulation of oncometabolites (ie, succinate or 2-hydroxyglutarate) or other oncogenic activations, such as VHL mutations. ARNT = aryl hydrocarbon receptor nuclear translocator, HRE = hypoxia response element, OH = hydroxylation, TCA = tricarboxylic acid.
Comprehensive molecular analysis of pheochromocytomas and paragangliomas has unveiled four genomic clusters, each with specific molecular signatures (120): (a) cluster 1, pseudohypoxia; (b) cluster 2, abnormal kinase signaling and activation of the mTOR pathway, including mutations in RET, NF1, TMEM127, MAX, MET, and HRAS; (c) cluster 3, activation of the Wnt-signaling pathway with MAML3 anomalies; and (d) cluster 4, cortical admixture.
The pseudohypoxic cluster can be divided into two subtypes: (a) tricarboxylic acid cycle–related mutations, accounting for 10%–15% of all pheochromocytomas and paragangliomas, and (b) VHL/EPAS1-related mutations, which occur in 15%–20% of cases. The tricarboxylic acid cycle–related subtype involves mutations in SDH gene subunits (SDHx) and other mitochondrial enzymes (120,121). SDHB and SDHD mutations are the most common. SDHB and SDHD mutations predominately occur in abdominal and head and neck paragangliomas, respectively (122). Among all SDH mutations, SDHD has the highest penetrance, with 75% of affected patients manifesting disease by 40 years of age, often with a multifocal distribution. Head and neck paragangliomas occur in more than 90% of patients with SDHD mutations, and multifocal disease occurs most commonly in patients with these mutations compared with those having mutations in other SDH subunits (Fig 9) (123).
Metabolic imaging alongside functional anatomic imaging (MRI and CT) have enhanced phenotypic understanding of the different genomic clusters of pheochromocytomas and paragangliomas. However, more research is needed in this field, with the few available studies focusing on the pseudohypoxic cluster (124–126). Based on their molecular profiles, most paragangliomas exhibit moderate to low 18F-FDG uptake, except cluster 1 paragangliomas (SDHx-related and VHL-related tumors) (124,126,127). This is particularly true for SDHB- and SDHD-related tumors (126). Additionally, a high expression of somatostatin receptor 2A in the tricarboxylic acid cycle–related cluster represents a good imaging target for diagnosis, staging, and surveillance of paragangliomas with gallium 68 DOTATATE PET/CT (128). More studies are required to better understand how this probe may help define the different genetic clusters.
Renal cell carcinoma.— Clear cell renal cell carcinoma (ccRCC) accounts for 70% of kidney cancers. Unlike many other cancers, a few well-recognized genetic mutations drive ccRCC. Most mutations occur on the short arm of chromosome 3, which contains the VHL, PBRM1, BAP1, and SETD2 genes; additional relevant mutations include KDM5C and MUC-4 (129):
Germline or somatic DNA mutation of VHL occur in familial or sporadic ccRCC, respectively. VHL encodes part of a protein complex that degrades hypoxia-inducible factor-1α, a key orchestrator of angiogenesis and metabolic reprogramming (Fig 9E). Inactivation of VHL often produces hypervascular and hypoxic tumors. Imaging features include tumors with well-defined margins, nodular enhancement, intra-tumoral vascularity, and solid appearance (rather than cystic) (130) (Fig 10). Germline mutations in VHL also predispose to adrenal pheochromocytomas, pancreatic cysts, and central nervous system hemangioblastomas (Fig 10). Despite being mutated in greater than 90% of ccRCC, a VHL mutation bears no prognostic value (131). Effects of mutant VHL have motivated development of antiangiogenic therapies like tyrosine kinase inhibitors targeting vascular endothelial growth factor receptor 2 and neutralizing VEGF antibodies.
PBRM1, an epigenetic regulator, encodes a nucleosome remodeling complex that limits DNA accessibility to RNA polymerase and transcription factors. PBRM1 mutations occur in 40%–50% of ccRCC cases, often with mutant VHL (132). Imaging shows tumors with solid structure. Mutant PBRM1 correlates with poor survival and higher stage disease. Mutations may predict response to immune checkpoint therapy (133).
BAP1, a tumor suppressor gene mutated in both hereditary and sporadic ccRCC, regulates cell breakdown and replication (132). Imaging features include ill-defined margins, calcifications, and renal vein invasion. Loss of BAP1 in multicystic ccRCC correlates with higher histopathologic grade and more aggressive disease.
KDM5C mutations occur in 6%–7% of ccRCC and are consistently absent in multicystic ccRCC (132). Imaging features include hypoenhancement relative to the renal cortex at nephrographic phase CT and renal vein invasion. Patients with this mutation often show higher total and visceral abdominal fat (134). KDMC5 mutations paradoxically correlate with unfavorable prognosis for localized cancer and prolonged survival in advanced metastatic disease (135).
SETD2 is a tumor suppressor gene mutated in approximately 10%–15% of ccRCC. Loss of this protein occurs in multicystic ccRCC, with tumors showing predominately exophytic growth (132). SETD2 mutations are associated with a poor prognosis in localized disease.
MUC4 is a transmembrane mucin. MUC4 mutations confer ccRCC tumors a preferential exophytic tumor growth pattern and a decreased overall survival.
Figure 10:
Clear cell renal cell carcinoma (ccRCC) with a germline VHL mutation. (A, B, D) Axial and (C) coronal soft-tissue CT images, as well as sagittal (E) T1-weighted image with extracellular gadolinium and (F) T2-weighted image of the cervicothoracic spine in a 35-year-old man with a germline VHL mutation (von Hippel−Lindau syndrome) who presented with bilateral arterially enhancing renal lesions, consistent with ccRCC (yellow arrows in A and C), and bilateral renal cysts. Other syndromic lesions were also identified in this patient, namely an avidly enhancing left adrenal pheochromocytoma (yellow arrow in B), multiple pancreatic cysts (yellow arrows in D), and several spinal hemangioblastomas (yellow arrows in E, F).
Bladder cancer.— Urothelial carcinoma, the most common urinary tract malignancy, involves the bladder in 90% of cases and the upper urinary tract in 10% of cases (136). Bladder cancer occurs as a superficial tumor (70%), which often has good prognosis, or as a muscle-invasive urothelial carcinoma (MIUC) (30%) (137) with poor survival due to metastases (138). Superficial tumors are routinely treated by transurethral resection, followed, in cases of high-grade histologic features and/or early subepithelial invasion, by bacillus Calmette–Guerin intravesical immunotherapy. Patients with invasive bladder cancer generally undergo radical cystectomy, often preceded by neoadjuvant chemotherapy (139). Given the incremental risk for metastases with certain genetic subtypes, patients will inevitably require more imaging tests and follow-up. Hence, it is important for a radiologist to understand the phenotypic translation of key genetic mutations.
The Cancer Genome Atlas Research Network has reported 58 significantly mutated genes in MIUC (140), with many of these genes involved in chromatin structure, cell cycle regulation, and kinase signaling. Molecular profiling defines five subtypes of MIUC: (a) luminal-papillary (35%), strongly associated with FGFR3 mutations; (b) luminal-infiltrated (19%), with best response rate to immunotherapy using checkpoint inhibitors; (c) luminal (6%), with high expression of urothelial differentiation markers and uroplakins; (d) basal-squamous (35%), with high expression of basal markers, CD44 stem cell-like markers, and TP53 mutations; and (e) neuronal (5%), featuring TP53 and RB1 mutations and elevated expression of neuronal and developmental genes (140). The best and worst 5-year overall survival rates are for the luminal-papillary and the neuronal subtypes, respectively.
The few radiogenomic studies have focused on specific low-risk (ARID1A, FGFR3, PIK3CA, STAG2, and TSC1) and high-risk (TP53, RB1, and KDM6A) mutations rather than molecular subtypes and clusters. Low-risk mutations demonstrate better outcomes, particularly in the setting of FGFR3 mutations (corresponding to the luminal-papillary subtype) (141), lower tumor grade and stage for STAG2 mutations (142), and reduced recurrence in non–muscle-invasive bladder cancer with a PIK3CA mutation (143). These mutations are associated with longer metastasis-free and greater overall survival in MIUC (144).
For high-risk mutations, TP53 mutations correlate with higher grade tumors, more invasive behavior, greater risk of recurrence, worse outcome, higher frequency of nodal and osseous metastases, and shorter metastasis-free and overall survival (Fig 11) (144–146). RB1 and KDM6A mutations correlate with an increased risk of metastatic disease and recurrence rates, as well as a greater likelihood of early progression and lower survival rates (Fig 12) (144,147).
Figure 11:
TP53 mutation in bladder cancer. (A, C) Axial soft-tissue and (B, D) lung window CT images and (E) sagittal T1-weighted image of the lumbar spine in a 75-year-old man diagnosed with resectable TP53-mutant bladder cancer (not shown) and no metastatic spread at initial diagnosis (A, B). After 3 months, the patient developed multiple lung and nodal metastases (arrows in C, D), followed by bone metastases 1 month later (arrow in E). This patient had a metastasis-free survival of 3 months and an overall survival of 6.6 months. TP53-mutant bladder cancers usually have an aggressive phenotype with early metastatic spread and poor survival compared with TP53 wild-type bladder cancers.
Figure 12:
RB1 mutation in bladder cancer. (A–C) Coronal oral contrast–enhanced images of the abdomen and pelvis without (A) and with intravenous contrast media (B, C) in a 61-year-old man diagnosed with a large, resectable RB1-mutant bladder mass (arrow in A) and no metastatic spread. After 2 months, the patient developed multiple nodal metastases (arrows in B), which further progressed at the subsequent CT scan performed 2 months later (arrows in C). This patient had a metastasis-free survival of 2 months and an overall survival of 4.7 months. Like bladder cancers with TP53 mutations, bladder cancers with RB1 mutations have aggressive phenotypes with early metastatic spread and poor survival.
Understanding the clinical significance of certain bladder cancer mutations can aid interpretation of imaging scans, particularly for patients with MIUC, given their higher risk of metastasis. When reporting a scan in a patient with TP53 or RB1 mutations, the radiologist should be more critical about equivocally enlarged or heterogeneous small lymph nodes, particularly in the pelvis, and search carefully for early signs of metastasis.
Conclusions
Omics technologies are now part of clinical care in oncology, underscoring the need for imaging experts to understand how these technologies integrate into risk assessment, diagnosis, and treatment. We highlighted key genetic and epigenetic regulators of cancer pathogenesis and treatment response with examples in different organ systems to familiarize radiologists with the basic concepts of cancer genomics. Applying these concepts will position radiologists to provide better patient-centered cancer care.
Authors declared no funding for this work.
Disclosures of conflicts of interest: E.M.S. Member of Radiology: Imaging Cancer Trainee Editorial Board. M.K. Associate editor for Radiology: Imaging Cancer; Radiology: Imaging Cancer Trainee Editorial Board alum. B.M.M. No relevant relationships. A.J. No relevant relationships. R.L.G. No relevant relationships. K.P. Associate editor for Radiology: Imaging Cancer; research grants: Digital hybrid breast PET/MRI for enhanced diagnosis of breast cancer (HYPMED) H2020 – Research and Innovation Framework Programme PHC-11-2015 number 667211-1, A body scan for cancer detection using quantum technology (CANCERSCAN) H2020-FETOPEN number 828978, Multiparametric 18F-fluoroestradiol PET/MRI coupled with radiomics analysis and machine learning for prediction and assessment of response to neoadjuvant endocrine therapy in patients with hormone receptor+/HER2− invasive breast cancer Jubiläumsfonds of the Austrian National Bank number Nr:18207, Deciphering breast cancer heterogeneity and tackling the hypoxic tumor microenvironment challenge with PET/MRI, MSI, and radiomics –The Vienna Science and Technology Fund LS19-046, MSCKK 2020 Molecularly Targeted Intra-Operative Imaging Award (July 2020–June 2021), Breast Cancer Research Foundation (June 2019–May 2021), National Institutes of Health (NIH) R01 Breast Cancer Intravoxel-Incoherent-Motion MRI Multisite (BRIMM) (September 1, 2020–August 30, 2025), NIH R01 subaward: Abbreviation non–contrast-enhanced MRI for breast cancer screening (September 1, 2023–August 31, 2025); supported in part through the NIH/National Cancer Institute Cancer Center Support Grant number P30 CA008748; Memorial Sloan-Kettering has institutional financial interests relative to GRAIL; consulting fees from Genentech (May 2019 to present), Merantix Healthcare (May 2020 to present), and AURA Health Technologies (April 2021 to present); payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from the European Society of Breast Imaging (active), Bayer (active), Siemens Healthineers (ended), IDKD 2019 (ended), and Olea Medical (ended); support for attending meetings and/or travel from the European Society of Breast Imaging IDKD 2019; participation on a Data Safety Monitoring Board or Advisory Board for Guerbet (2022); leadership or fiduciary role on the executive board of the European Society of Breast Imaging. G.L. Editor of Radiology: Imaging Cancer. M.A.H. No relevant relationships. A.B.S. Consulting fees from Virtualscopics and Imaging Endpoints; speaker honorarium from Yuhan and Samjim Pharmaceutics. X.L. Associate editor for Radiology: Imaging Cancer; Radiology: Imaging Cancer Trainee Editorial Board alum.
Abbreviations:
- ADC
- apparent diffusion coefficient
- ccRCC
- clear cell renal cell carcinoma
- CRC
- colorectal cancer
- EGFR
- epidermal growth factor receptor
- ER
- estrogen receptor
- FDG
- fluorodeoxyglucose
- HCC
- hepatocellular carcinoma
- HER2
- human epidermal growth factor 2
- IDH
- isocitrate dehydrogenase
- MGMT
- O6-methylguanine DNA methyltransferase
- MIUC
- muscle-invasive urothelial carcinoma
- MSI
- microsatellite instability
- NSCLC
- non–small cell lung cancer
- PR
- progesterone receptor
- SDH
- succinate dehydrogenase
References
- 1. Hanahan D , Weinberg RA . Hallmarks of cancer: the next generation . Cell 2011. ; 144 ( 5 ): 646 – 674 . [DOI] [PubMed] [Google Scholar]
- 2. Pelengaris SKM . The molecular biology of cancer: A bridge from bench to bedside . 2nd ed. Chichester, England: : Wiley-Blackwell; , 2013. . [Google Scholar]
- 3. Tomasetti C , Li L , Vogelstein B . Stem cell divisions, somatic mutations, cancer etiology, and cancer prevention . Science 2017. ; 355 ( 6331 ): 1330 – 1334 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Retinoblastoma . NORD – National Organization for Rare Disorders . https://rarediseases.org/rare-diseases/retinoblastoma/. Published 2017. Accessed February 20, 2023 .
- 5. Laird PW . Principles and challenges of genomewide DNA methylation analysis . Nat Rev Genet 2010. ; 11 ( 3 ): 191 – 203 . [DOI] [PubMed] [Google Scholar]
- 6. Ignatiadis M , Sledge GW , Jeffrey SS . Liquid biopsy enters the clinic - implementation issues and future challenges . Nat Rev Clin Oncol 2021. ; 18 ( 5 ): 297 – 312 . [DOI] [PubMed] [Google Scholar]
- 7. Aerts HJ . The potential of radiomic-based phenotyping in precision medicine: A review . JAMA Oncol 2016. ; 2 ( 12 ): 1636 – 1642 . [DOI] [PubMed] [Google Scholar]
- 8. Tsimberidou AM , Hong DS , Fu S , et al . Precision medicine: Preliminary results from the initiative for molecular profiling and advanced cancer therapy 2 (impact2) study . NPJ Precis Oncol 2021. ; 5 ( 1 ): 21 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. El Gharib K , Khoury M , Kourie HR . HER2 in gastric adenocarcinoma: where do we stand today? Per Med 2022. ; 19 ( 1 ): 67 – 78 . [DOI] [PubMed] [Google Scholar]
- 10. Oh DY , Bang YJ . HER2-targeted therapies - a role beyond breast cancer . Nat Rev Clin Oncol 2020. ; 17 ( 1 ): 33 – 48 . [DOI] [PubMed] [Google Scholar]
- 11. Guha T , Malkin D . Inherited tp53 mutations and the Li-Fraumeni syndrome . Cold Spring Harb Perspect Med 2017. ; 7 ( 4 ): a026187 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Frebourg T , Bajalica Lagercrantz S , Oliveira C , Magenheim R , Evans DG ; European Reference Network GENTURIS . Guidelines for the Li-Fraumeni and heritable TP53-related cancer syndromes . Eur J Hum Genet 2020. ; 28 ( 10 ): 1379 – 1386 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Shimada S , Mogushi K , Akiyama Y , et al . Comprehensive molecular and immunological characterization of hepatocellular carcinoma . EBioMedicine 2019. ; 40 : 457 – 470 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Berggren P , Steineck G , Adolfsson J , et al . p53 mutations in urinary bladder cancer . Br J Cancer 2001. ; 84 ( 11 ): 1505 – 1511 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Lombardi G , Barresi V , Castellano A , et al . Clinical management of diffuse low-grade gliomas . Cancers (Basel) 2020. ; 12 ( 10 ): 3008 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Delgado-López PD , Corrales-García EM . Survival in glioblastoma: a review on the impact of treatment modalities . Clin Transl Oncol 2016. ; 18 ( 11 ): 1062 – 1071 . [DOI] [PubMed] [Google Scholar]
- 17. Louis DN , Perry A , Reifenberger G , et al . The 2016 World Health Organization classification of tumors of the central nervous system: A summary . Acta Neuropathol (Berl) 2016. ; 131 ( 6 ): 803 – 820 . [DOI] [PubMed] [Google Scholar]
- 18. Prior F , Smith K , Sharma A , et al . The public cancer radiology imaging collections of The Cancer Imaging Archive . Sci Data 2017. ; 4 ( 1 ): 170124 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Schwartzentruber J , Korshunov A , Liu XY , et al . Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma . Nature 2012. ; 482 ( 7384 ): 226 – 231 . [Published correction appears in Nature 2012;484(7392):130.] [DOI] [PubMed] [Google Scholar]
- 20. Liu XY , Gerges N , Korshunov A , et al . Frequent ATRX mutations and loss of expression in adult diffuse astrocytic tumors carrying IDH1/IDH2 and TP53 mutations . Acta Neuropathol (Berl) 2012. ; 124 ( 5 ): 615 – 625 . [DOI] [PubMed] [Google Scholar]
- 21. Pirozzi CJ , Yan H . The implications of IDH mutations for cancer development and therapy . Nat Rev Clin Oncol 2021. ; 18 ( 10 ): 645 – 661 . [DOI] [PubMed] [Google Scholar]
- 22. Jiao Y , Killela PJ , Reitman ZJ , et al . Frequent ATRX, CIC, FUBP1 and IDH1 mutations refine the classification of malignant gliomas . Oncotarget 2012. ; 3 ( 7 ): 709 – 722 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Carrillo JA , Lai A , Nghiemphu PL , et al . Relationship between tumor enhancement, edema, IDH1 mutational status, MGMT promoter methylation, and survival in glioblastoma . AJNR Am J Neuroradiol 2012. ; 33 ( 7 ): 1349 – 1355 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Johnson DR , Diehn FE , Giannini C , et al . Genetically defined oligodendroglioma is characterized by indistinct tumor borders at MRI . AJNR Am J Neuroradiol 2017. ; 38 ( 4 ): 678 – 684 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Ding H , Huang Y , Li Z , et al . Prediction of IDH status through MRI features and enlightened reflection on the delineation of target volume in low-grade gliomas . Technol Cancer Res Treat 2019. ; 18 : 1533033819877167 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Villanueva-Meyer JE , Wood MD , Choi BS , et al . MRI features and IDH mutational status of grade II diffuse gliomas: Impact on diagnosis and prognosis . AJR Am J Roentgenol 2018. ; 210 ( 3 ): 621 – 628 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Hong EK , Choi SH , Shin DJ , et al . Radiogenomics correlation between MR imaging features and major genetic profiles in glioblastoma . Eur Radiol 2018. ; 28 ( 10 ): 4350 – 4361 . [DOI] [PubMed] [Google Scholar]
- 28. Choi C , Ganji SK , DeBerardinis RJ , et al . 2-hydroxyglutarate detection by magnetic resonance spectroscopy in IDH-mutated patients with gliomas . Nat Med 2012. ; 18 ( 4 ): 624 – 629 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Reifenberger J , Reifenberger G , Liu L , James CD , Wechsler W , Collins VP . Molecular genetic analysis of oligodendroglial tumors shows preferential allelic deletions on 19q and 1p . Am J Pathol 1994. ; 145 ( 5 ): 1175 – 1190 . [PMC free article] [PubMed] [Google Scholar]
- 30. Cancer Genome Atlas Research Network ; Brat DJ , Verhaak RG , et al . Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas . N Engl J Med 2015. ; 372 ( 26 ): 2481 – 2498 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Cairncross G , Wang M , Shaw E , et al . Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402 . J Clin Oncol 2013. ; 31 ( 3 ): 337 – 343 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Herrlinger U , Tzaridis T , Mack F , et al . Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): a randomised, open-label, phase 3 trial . Lancet 2019. ; 393 ( 10172 ): 678 – 688 . [DOI] [PubMed] [Google Scholar]
- 33. Kálovits F , Tompa M , Nagy Á , Kálmán B . Isocitrate dehydrogenase mutations in defining the biology of and supporting clinical decision making in glioblastoma . Ideggyogy Sz 2018. ; 71 ( 7-08 ): 237 – 247 . [DOI] [PubMed] [Google Scholar]
- 34. Ellingson BM , Lai A , Harris RJ , et al . Probabilistic radiographic atlas of glioblastoma phenotypes . AJNR Am J Neuroradiol 2013. ; 34 ( 3 ): 533 – 540 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Romano A , Calabria LF , Tavanti F , et al . Apparent diffusion coefficient obtained by magnetic resonance imaging as a prognostic marker in glioblastomas: correlation with MGMT promoter methylation status . Eur Radiol 2013. ; 23 ( 2 ): 513 – 520 . [DOI] [PubMed] [Google Scholar]
- 36. Pope WB , Chen JH , Dong J , et al . Relationship between gene expression and enhancement in glioblastoma multiforme: exploratory DNA microarray analysis . Radiology 2008. ; 249 ( 1 ): 268 – 277 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Liu TT , Achrol AS , Mitchell LA , et al . Magnetic resonance perfusion image features uncover an angiogenic subgroup of glioblastoma patients with poor survival and better response to antiangiogenic treatment . Neuro-oncol 2017. ; 19 ( 7 ): 997 – 1007 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Qiu T , Chanchotisatien A , Qin Z , et al . Imaging characteristics of adult H3 K27M-mutant gliomas . J Neurosurg 2019. ; 133 ( 6 ): 1662 – 1670 . [DOI] [PubMed] [Google Scholar]
- 39. Lindeman NI , Cagle PT , Aisner DL , et al . Updated molecular testing guideline for the selection of lung cancer patients for treatment with targeted tyrosine kinase inhibitors: Guideline from the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology . J Mol Diagn 2018. ; 20 ( 2 ): 129 – 159 . [DOI] [PubMed] [Google Scholar]
- 40. Ettinger DS , Wood DE , Aisner DL , et al . Non-small cell lung cancer, version 3.2022, nccn clinical practice guidelines in oncology . J Natl Compr Canc Netw 2022. ; 20 ( 5 ): 497 – 530 . [DOI] [PubMed] [Google Scholar]
- 41. Cox VL , Bhosale P , Varadhachary GR , et al . Cancer genomics and important oncologic mutations: A contemporary guide for body imagers . Radiology 2017. ; 283 ( 2 ): 314 – 340 . [DOI] [PubMed] [Google Scholar]
- 42. Gainor JF , Varghese AM , Ou SH , et al . ALK rearrangements are mutually exclusive with mutations in EGFR or KRAS: an analysis of 1,683 patients with non-small cell lung cancer . Clin Cancer Res 2013. ; 19 ( 15 ): 4273 – 4281 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Santarius T , Shipley J , Brewer D , Stratton MR , Cooper CS . A census of amplified and overexpressed human cancer genes . Nat Rev Cancer 2010. ; 10 ( 1 ): 59 – 64 . [DOI] [PubMed] [Google Scholar]
- 44. Schlessinger J . Receptor tyrosine kinases: legacy of the first two decades . Cold Spring Harb Perspect Biol 2014. ; 6 ( 3 ): a008912 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Shi Y , Au JS , Thongprasert S , et al . A prospective, molecular epidemiology study of EGFR mutations in Asian patients with advanced non-small-cell lung cancer of adenocarcinoma histology (PIONEER) . J Thorac Oncol 2014. ; 9 ( 2 ): 154 – 162 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Soria JC , Ohe Y , Vansteenkiste J , et al . Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer . N Engl J Med 2018. ; 378 ( 2 ): 113 – 125 . [DOI] [PubMed] [Google Scholar]
- 47. Aoki T , Hanamiya M , Uramoto H , Hisaoka M , Yamashita Y , Korogi Y . Adenocarcinomas with predominant ground-glass opacity: correlation of morphology and molecular biomarkers . Radiology 2012. ; 264 ( 2 ): 590 – 596 . [DOI] [PubMed] [Google Scholar]
- 48. Lee HJ , Kim YT , Kang CH , et al . Epidermal growth factor receptor mutation in lung adenocarcinomas: relationship with CT characteristics and histologic subtypes . Radiology 2013. ; 268 ( 1 ): 254 – 264 . [DOI] [PubMed] [Google Scholar]
- 49. Rizzo S , Petrella F , Buscarino V , et al . CT radiogenomic characterization of EGFR, K-RAS, and ALK mutations in non-small cell lung cancer . Eur Radiol 2016. ; 26 ( 1 ): 32 – 42 . [DOI] [PubMed] [Google Scholar]
- 50. Sun X , Xiao Z , Chen G , et al . A PET imaging approach for determining EGFR mutation status for improved lung cancer patient management . Sci Transl Med 2018. ; 10 ( 431 ): eaan8840 . [DOI] [PubMed] [Google Scholar]
- 51. Su X , Cheng K , Jeon J , et al . Comparison of two site-specifically (18)F-labeled affibodies for PET imaging of EGFR positive tumors . Mol Pharm 2014. ; 11 ( 11 ): 3947 – 3956 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Peters S , Camidge DR , Shaw AT , et al . Alectinib versus crizotinib in untreated alk-positive non-small-cell lung cancer . N Engl J Med 2017. ; 377 ( 9 ): 829 – 838 . [DOI] [PubMed] [Google Scholar]
- 53. Yamamoto S , Korn RL , Oklu R , et al . ALK molecular phenotype in non-small cell lung cancer: CT radiogenomic characterization . Radiology 2014. ; 272 ( 2 ): 568 – 576 . [DOI] [PubMed] [Google Scholar]
- 54. Choi CM , Kim MY , Hwang HJ , Lee JB , Kim WS . Advanced adenocarcinoma of the lung: comparison of CT characteristics of patients with anaplastic lymphoma kinase gene rearrangement and those with epidermal growth factor receptor mutation . Radiology 2015. ; 275 ( 1 ): 272 – 279 . [DOI] [PubMed] [Google Scholar]
- 55. Lin YT , Wang YF , Yang JC , et al . Development of renal cysts after crizotinib treatment in advanced ALK-positive non-small-cell lung cancer . J Thorac Oncol 2014. ; 9 ( 11 ): 1720 – 1725 . [DOI] [PubMed] [Google Scholar]
- 56. Awad MM , Oxnard GR , Jackman DM , et al . MET exon 14 mutations in non-small-cell lung cancer are associated with advanced age and stage-dependent MET genomic amplification and c-Met overexpression . J Clin Oncol 2016. ; 34 ( 7 ): 721 – 730 . [DOI] [PubMed] [Google Scholar]
- 57. Wolf J , Seto T , Han JY , et al . Capmatinib in MET exon 14-mutated or MET-amplified non-small-cell lung cancer . N Engl J Med 2020. ; 383 ( 10 ): 944 – 957 . [DOI] [PubMed] [Google Scholar]
- 58. Paik PK , Felip E , Veillon R , et al . Tepotinib in non-small-cell lung cancer with MET exon 14 skipping mutations . N Engl J Med 2020. ; 383 ( 10 ): 931 – 943 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Digumarthy SR , Mendoza DP , Zhang EW , Lennerz JK , Heist RS . Clinicopathologic and imaging features of non-small-cell lung cancer with met exon 14 skipping mutations . Cancers (Basel) 2019. ; 11 ( 12 ): 2033 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Pillai RN , Behera M , Berry LD , et al . HER2 mutations in lung adenocarcinomas: A report from the Lung Cancer Mutation Consortium . Cancer 2017. ; 123 ( 21 ): 4099 – 4105 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Mazieres J , Lafitte C , Ricordel C , et al . Combination of trastuzumab, pertuzumab, and docetaxel in patients with advanced non-small-cell lung cancer harboring HER2 mutations: Results from the ifct-1703 r2d2 trial . J Clin Oncol 2022. ; 40 ( 7 ): 719 – 728 . [DOI] [PubMed] [Google Scholar]
- 62. Walker-Smith TL , Peck J . Genetic and genomic advances in breast cancer diagnosis and treatment . Nurs Womens Health 2019. ; 23 ( 6 ): 518 – 525 . [DOI] [PubMed] [Google Scholar]
- 63. Claus EB , Schildkraut JM , Thompson WD , Risch NJ . The genetic attributable risk of breast and ovarian cancer . Cancer 1996. ; 77 ( 11 ): 2318 – 2324 . [DOI] [PubMed] [Google Scholar]
- 64. Tung NM , Boughey JC , Pierce LJ , et al . Management of hereditary breast cancer: American Society of Clinical Oncology, American Society for Radiation Oncology, and Society of Surgical Oncology guideline . J Clin Oncol 2020. ; 38 ( 18 ): 2080 – 2106 . [DOI] [PubMed] [Google Scholar]
- 65. Magnoni F , Sacchini V , Veronesi P , et al . Surgical management of inherited breast cancer: Role of breast-conserving surgery . Cancers (Basel) 2022. ; 14 ( 13 ): 3245 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Fu X , Tan W , Song Q , Pei H , Li J . BRCA1 and breast cancer: Molecular mechanisms and therapeutic strategies . Front Cell Dev Biol 2022. ; 10 : 813457 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Zhong Q , Peng HL , Zhao X , Zhang L , Hwang WT . Effects of BRCA1- and BRCA2-related mutations on ovarian and breast cancer survival: a meta-analysis . Clin Cancer Res 2015. ; 21 ( 1 ): 211 – 220 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Evans DG , Shenton A , Woodward E , Lalloo F , Howell A , Maher ER . Penetrance estimates for BRCA1 and BRCA2 based on genetic testing in a Clinical Cancer Genetics service setting: risks of breast/ovarian cancer quoted should reflect the cancer burden in the family . BMC Cancer 2008. ; 8 ( 1 ): 155 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Riedl CC , Luft N , Bernhart C , et al . Triple-modality screening trial for familial breast cancer underlines the importance of magnetic resonance imaging and questions the role of mammography and ultrasound regardless of patient mutation status, age, and breast density . J Clin Oncol 2015. ; 33 ( 10 ): 1128 – 1135 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Samstein RM , Krishna C , Ma X , et al . Mutations in BRCA1 and BRCA2 differentially affect the tumor microenvironment and response to checkpoint blockade immunotherapy . Nat Can 2021. ; 1 ( 12 ): 1188 – 1203 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Cortesi L , Rugo HS , Jackisch C . An overview of PARP inhibitors for the treatment of breast cancer . Target Oncol 2021. ; 16 ( 3 ): 255 – 282 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Perou CM , Sørlie T , Eisen MB , et al . Molecular portraits of human breast tumours . Nature 2000. ; 406 ( 6797 ): 747 – 752 . [DOI] [PubMed] [Google Scholar]
- 73. Nicolini A , Ferrari P , Duffy MJ . Prognostic and predictive biomarkers in breast cancer: Past, present and future . Semin Cancer Biol 2018. ; 52 ( Pt 1 ): 56 – 73 . [DOI] [PubMed] [Google Scholar]
- 74. Vaz-Luis I , Winer EP , Lin NU . Human epidermal growth factor receptor-2-positive breast cancer: Does estrogen receptor status define two distinct subtypes? Ann Oncol 2013. ; 24 ( 2 ): 283 – 291 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Lumachi F , Brunello A , Maruzzo M , Basso U , Basso SM . Treatment of estrogen receptor-positive breast cancer . Curr Med Chem 2013. ; 20 ( 5 ): 596 – 604 . [DOI] [PubMed] [Google Scholar]
- 76. Xu ZQ , Zhang Y , Li N , et al . Efficacy and safety of lapatinib and trastuzumab for HER2-positive breast cancer: A systematic review and meta-analysis of randomised controlled trials . BMJ Open 2017. ; 7 ( 3 ): e013053 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Hynes NE , Lane HA . ERBB receptors and cancer: The complexity of targeted inhibitors . Nat Rev Cancer 2005. ; 5 ( 5 ): 341 – 354 . [DOI] [PubMed] [Google Scholar]
- 78. Székely B , Silber AL , Pusztai L . New therapeutic strategies for triple-negative breast cancer . Oncology (Williston Park) 2017. ; 31 ( 2 ): 130 – 137 . [PubMed] [Google Scholar]
- 79. Foulkes WD , Smith IE , Reis-Filho JS . Triple-negative breast cancer . N Engl J Med 2010. ; 363 ( 20 ): 1938 – 1948 . [DOI] [PubMed] [Google Scholar]
- 80. Galati F , Rizzo V , Moffa G , et al . Radiologic-pathologic correlation in breast cancer: Do MRI biomarkers correlate with pathologic features and molecular subtypes? Eur Radiol Exp 2022. ; 6 ( 1 ): 39 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Ab Mumin N , Ramli Hamid MT , Wong JHD , Rahmat K , Ng KH . Magnetic resonance imaging phenotypes of breast cancer molecular subtypes: A systematic review . Acad Radiol 2022. ; 29 ( Suppl 1 ): S89 – S106 . [DOI] [PubMed] [Google Scholar]
- 82. Elias SG , Adams A , Wisner DJ , et al . Imaging features of HER2 overexpression in breast cancer: A systematic review and meta-analysis . Cancer Epidemiol Biomarkers Prev 2014. ; 23 ( 8 ): 1464 – 1483 . [DOI] [PubMed] [Google Scholar]
- 83. Karbasian N , Sohrabi S , Omofoye TS , et al . Imaging features of triple negative breast cancer and the effect of BRCA mutations . Curr Probl Diagn Radiol 2021. ; 50 ( 3 ): 303 – 307 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Youk JH , Son EJ , Chung J , Kim JA , Kim EK . Triple-negative invasive breast cancer on dynamic contrast-enhanced and diffusion-weighted MR imaging: comparison with other breast cancer subtypes . Eur Radiol 2012. ; 22 ( 8 ): 1724 – 1734 . [DOI] [PubMed] [Google Scholar]
- 85. Ab Mumin N , Ramli Hamid MT , Wong JHD , Rahmat K , Ng KH . Magnetic resonance imaging phenotypes of breast cancer molecular subtypes: A systematic review . Acad Radiol 2022. ; 29 ( Suppl 1 ): S89 – S106 . [DOI] [PubMed] [Google Scholar]
- 85. Kawashima H , Inokuchi M , Furukawa H , Kitamura S . Triple-negative breast cancer: Are the imaging findings different between responders and nonresponders to neoadjuvant chemotherapy? Acad Radiol 2011. ; 18 ( 8 ): 963 – 969 . [DOI] [PubMed] [Google Scholar]
- 86. Bae MS , Shin SU , Ryu HS , et al . Pretreatment MR imaging features of triple-negative breast cancer: Association with response to neoadjuvant chemotherapy and recurrence-free survival . Radiology 2016. ; 281 ( 2 ): 392 – 400 . [DOI] [PubMed] [Google Scholar]
- 87. Teramoto T , Satonaka K , Kitazawa S , Fujimori T , Hayashi K , Maeda S . p53 gene abnormalities are closely related to hepatoviral infections and occur at a late stage of hepatocarcinogenesis . Cancer Res 1994. ; 54 ( 1 ): 231 – 235 . [PubMed] [Google Scholar]
- 88. Calderaro J , Ziol M , Paradis V , Zucman-Rossi J . Molecular and histological correlations in liver cancer . J Hepatol 2019. ; 71 ( 3 ): 616 – 630 . [DOI] [PubMed] [Google Scholar]
- 89. Yoneda N , Matsui O , Kobayashi S , et al . Current status of imaging biomarkers predicting the biological nature of hepatocellular carcinoma . Jpn J Radiol 2019. ; 37 ( 3 ): 191 – 208 . [DOI] [PubMed] [Google Scholar]
- 90. Kitao A , Matsui O , Yoneda N , et al . Hepatocellular carcinoma with β-catenin mutation: imaging and pathologic characteristics . Radiology 2015. ; 275 ( 3 ): 708 – 717 . [DOI] [PubMed] [Google Scholar]
- 91. Asayama Y , Nishie A , Ishigami K , et al . Fatty change in moderately and poorly differentiated hepatocellular carcinoma on MRI: a possible mechanism related to decreased arterial flow . Clin Radiol 2016. ; 71 ( 12 ): 1277 – 1283 . [DOI] [PubMed] [Google Scholar]
- 92. Schulze K , Imbeaud S , Letouzé E , et al . Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets . Nat Genet 2015. ; 47 ( 5 ): 505 – 511 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Kitao A , Matsui O , Yoneda N , et al . Gadoxetic acid-enhanced MR imaging for hepatocellular carcinoma: molecular and genetic background . Eur Radiol 2020. ; 30 ( 6 ): 3438 – 3447 . [DOI] [PubMed] [Google Scholar]
- 94. Ueno A , Masugi Y , Yamazaki K , et al . OATP1B3 expression is strongly associated with Wnt/β-catenin signalling and represents the transporter of gadoxetic acid in hepatocellular carcinoma . J Hepatol 2014. ; 61 ( 5 ): 1080 – 1087 . [DOI] [PubMed] [Google Scholar]
- 95. Yamaoka K , Saitoh S , Kinowaki K , et al . Clinicopathological assessment of steatohepatitic hepatocellular carcinoma . Clin Res Hepatol Gastroenterol 2022. ; 46 ( 8 ): 101799 . [DOI] [PubMed] [Google Scholar]
- 96. Fowler KJ , Burgoyne A , Fraum TJ , et al . Pathologic, molecular, and prognostic radiologic features of hepatocellular carcinoma . RadioGraphics 2021. ; 41 ( 6 ): 1611 – 1631 . [DOI] [PubMed] [Google Scholar]
- 97. Calderaro J , Couchy G , Imbeaud S , et al . Histological subtypes of hepatocellular carcinoma are related to gene mutations and molecular tumour classification . J Hepatol 2017. ; 67 ( 4 ): 727 – 738 . [DOI] [PubMed] [Google Scholar]
- 98. Mulé S , Galletto Pregliasco A , Tenenhaus A , et al . Multiphase liver MRI for identifying the macrotrabecular-massive subtype of hepatocellular carcinoma . Radiology 2020. ; 295 ( 3 ): 562 – 571 . [DOI] [PubMed] [Google Scholar]
- 99. Izuishi K , Yamamoto Y , Mori H , et al . Molecular mechanisms of [18F]fluorodeoxyglucose accumulation in liver cancer . Oncol Rep 2014. ; 31 ( 2 ): 701 – 706 . [DOI] [PubMed] [Google Scholar]
- 100. Ahn SY , Lee JM , Joo I , et al . Prediction of microvascular invasion of hepatocellular carcinoma using gadoxetic acid-enhanced MR and (18)F-FDG PET/CT . Abdom Imaging 2015. ; 40 ( 4 ): 843 – 851 . [DOI] [PubMed] [Google Scholar]
- 101. Segal E , Sirlin CB , Ooi C , et al . Decoding global gene expression programs in liver cancer by noninvasive imaging . Nat Biotechnol 2007. ; 25 ( 6 ): 675 – 680 . [DOI] [PubMed] [Google Scholar]
- 102. Banerjee S , Wang DS , Kim HJ , et al . A computed tomography radiogenomic biomarker predicts microvascular invasion and clinical outcomes in hepatocellular carcinoma . Hepatology 2015. ; 62 ( 3 ): 792 – 800 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Taouli B , Hoshida Y , Kakite S , et al . Imaging-based surrogate markers of transcriptome subclasses and signatures in hepatocellular carcinoma: preliminary results . Eur Radiol 2017. ; 27 ( 11 ): 4472 – 4481 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Renzulli M , Brocchi S , Cucchetti A , et al . Can current preoperative imaging be used to detect microvascular invasion of hepatocellular carcinoma? Radiology 2016. ; 279 ( 2 ): 432 – 442 . [DOI] [PubMed] [Google Scholar]
- 105. Rhee H , Kim H , Park YN . Clinico-radio-pathological and molecular features of hepatocellular carcinomas with keratin 19 expression . Liver Cancer 2020. ; 9 ( 6 ): 663 – 681 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Choi SY , Kim YK , Min JH , et al . Added value of ancillary imaging features for differentiating scirrhous hepatocellular carcinoma from intrahepatic cholangiocarcinoma on gadoxetic acid-enhanced MR imaging . Eur Radiol 2018. ; 28 ( 6 ): 2549 – 2560 . [DOI] [PubMed] [Google Scholar]
- 107. Park HJ , Kim YK , Cha DI , et al . Targetoid hepatic observations on gadoxetic acid-enhanced MRI using LI-RADS version 2018: emphasis on hepatocellular carcinomas assigned to the LR-M category . Clin Radiol 2020. ; 75 ( 6 ): 478.e13 – 478.e23 . [DOI] [PubMed] [Google Scholar]
- 108. Harada S , Morlote D . Molecular pathology of colorectal cancer . Adv Anat Pathol 2020. ; 27 ( 1 ): 20 – 26 . [DOI] [PubMed] [Google Scholar]
- 109. Lengauer C , Kinzler KW , Vogelstein B . Genetic instability in colorectal cancers . Nature 1997. ; 386 ( 6625 ): 623 – 627 . [DOI] [PubMed] [Google Scholar]
- 110. Cunningham D , Humblet Y , Siena S , et al . Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer . N Engl J Med 2004. ; 351 ( 4 ): 337 – 345 . [DOI] [PubMed] [Google Scholar]
- 111. Dekker E , Tanis PJ , Vleugels JLA , Kasi PM , Wallace MB . Colorectal cancer . Lancet 2019. ; 394 ( 10207 ): 1467 – 1480 . [DOI] [PubMed] [Google Scholar]
- 112. Cicenas J , Tamosaitis L , Kvederaviciute K , et al . KRAS, NRAS and BRAF mutations in colorectal cancer and melanoma . Med Oncol 2017. ; 34 ( 2 ): 26 . [DOI] [PubMed] [Google Scholar]
- 113. Kawada K , Nakamoto Y , Kawada M , et al . Relationship between 18F-fluorodeoxyglucose accumulation and KRAS/BRAF mutations in colorectal cancer . Clin Cancer Res 2012. ; 18 ( 6 ): 1696 – 1703 . [DOI] [PubMed] [Google Scholar]
- 114. Kim SJ , Pak K , Kim K . Diagnostic performance of F-18 FDG PET/CT for prediction of KRAS mutation in colorectal cancer patients: a systematic review and meta-analysis . Abdom Radiol (NY) 2019. ; 44 ( 5 ): 1703 – 1711 . [DOI] [PubMed] [Google Scholar]
- 115. Eurboonyanun K , Lahoud RM , Kordbacheh H , et al . Imaging predictors of BRAF mutation in colorectal cancer . Abdom Radiol (NY) 2020. ; 45 ( 8 ): 2336 – 2344 . [DOI] [PubMed] [Google Scholar]
- 116. Katabathina VS , Menias CO , Khanna L , et al . Hereditary gastrointestinal cancer syndromes: Role of imaging in screening, diagnosis, and management . RadioGraphics 2019. ; 39 ( 5 ): 1280 – 1301 . [DOI] [PubMed] [Google Scholar]
- 117. Zhang X , Zhang W , Cao P . Advances in CpG island methylator phenotype colorectal cancer therapies . Front Oncol 2021. ; 11 : 629390 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Lam AK . Update on adrenal tumours in 2017 World Health Organization (WHO) of endocrine tumours . Endocr Pathol 2017. ; 28 ( 3 ): 213 – 227 . [DOI] [PubMed] [Google Scholar]
- 119. Buffet A , Burnichon N , Favier J , Gimenez-Roqueplo AP . An overview of 20 years of genetic studies in pheochromocytoma and paraganglioma . Best Pract Res Clin Endocrinol Metab 2020. ; 34 ( 2 ): 101416 . [DOI] [PubMed] [Google Scholar]
- 120. Fishbein L , Leshchiner I , Walter V , et al . Comprehensive molecular characterization of pheochromocytoma and paraganglioma . Cancer Cell 2017. ; 31 ( 2 ): 181 – 193 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Crona J , Taïeb D , Pacak K . New perspectives on pheochromocytoma and paraganglioma: Toward a molecular classification . Endocr Rev 2017. ; 38 ( 6 ): 489 – 515 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Turchini J , Cheung VKY , Tischler AS , De Krijger RR , Gill AJ . Pathology and genetics of phaeochromocytoma and paraganglioma . Histopathology 2018. ; 72 ( 1 ): 97 – 105 . [DOI] [PubMed] [Google Scholar]
- 123. Withey SJ , Perrio S , Christodoulou D , et al . Imaging features of succinate dehydrogenase-deficient pheochromocytoma-paraganglioma syndromes . RadioGraphics 2019. ; 39 ( 5 ): 1393 – 1410 . [DOI] [PubMed] [Google Scholar]
- 124. Blanchet EM , Gabriel S , Martucci V , et al . 18F-FDG PET/CT as a predictor of hereditary head and neck paragangliomas . Eur J Clin Invest 2014. ; 44 ( 3 ): 325 – 332 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Jansen RW , van Amstel P , Martens RM , et al . Non-invasive tumor genotyping using radiogenomic biomarkers, a systematic review and oncology-wide pathway analysis . Oncotarget 2018. ; 9 ( 28 ): 20134 – 20155 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Timmers HJ , Chen CC , Carrasquillo JA , et al . Staging and functional characterization of pheochromocytoma and paraganglioma by 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography . J Natl Cancer Inst 2012. ; 104 ( 9 ): 700 – 708 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Taïeb D , Pacak K . New insights into the nuclear imaging phenotypes of cluster 1 pheochromocytoma and paraganglioma . Trends Endocrinol Metab 2017. ; 28 ( 11 ): 807 – 817 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Kong G , Schenberg T , Yates CJ , et al . The role of 68Ga-DOTA-octreotate PET/CT in follow-up of SDH-associated pheochromocytoma and paraganglioma . J Clin Endocrinol Metab 2019. ; 104 ( 11 ): 5091 – 5099 . [DOI] [PubMed] [Google Scholar]
- 129. Cancer Genome Atlas Research Network . Comprehensive molecular characterization of clear cell renal cell carcinoma . Nature 2013. ; 499 ( 7456 ): 43 – 49 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Karlo CA , Di Paolo PL , Chaim J , et al . Radiogenomics of clear cell renal cell carcinoma: associations between CT imaging features and mutations . Radiology 2014. ; 270 ( 2 ): 464 – 471 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Kim BJ , Kim JH , Kim HS , Zang DY . Prognostic and predictive value of VHL gene alteration in renal cell carcinoma: a meta-analysis and review . Oncotarget 2017. ; 8 ( 8 ): 13979 – 13985 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Khaleel S , Katims A , Cumarasamy S , et al . Radiogenomics in clear cell renal cell carcinoma: A review of the current status and future directions . Cancers (Basel) 2022. ; 14 ( 9 ): 2085 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Joseph RW , Kapur P , Serie DJ , et al . Clear cell renal cell carcinoma subtypes identified by BAP1 and PBRM1 expression . J Urol 2016. ; 195 ( 1 ): 180 – 187 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Greco F , Mallio CA . Relationship between visceral adipose tissue and genetic mutations (VHL and KDM5C) in clear cell renal cell carcinoma . Radiol Med (Torino) 2021. ; 126 ( 5 ): 645 – 651 . [DOI] [PubMed] [Google Scholar]
- 135. Alessandrino F , Shinagare AB , Bossé D , Choueiri TK , Krajewski KM . Radiogenomics in renal cell carcinoma . Abdom Radiol (NY) 2019. ; 44 ( 6 ): 1990 – 1998 . [DOI] [PubMed] [Google Scholar]
- 136. Babjuk M , Burger M , Zigeuner R , et al . EAU guidelines on non-muscle-invasive urothelial carcinoma of the bladder: update 2013 . Eur Urol 2013. ; 64 ( 4 ): 639 – 653 . [DOI] [PubMed] [Google Scholar]
- 137. Rouprêt M , Babjuk M , Compérat E , et al . European Association of Urology guidelines on upper urinary tract urothelial cell carcinoma: 2015 update . Eur Urol 2015. ; 68 ( 5 ): 868 – 879 . [DOI] [PubMed] [Google Scholar]
- 138. Kaufman DS , Shipley WU , Feldman AS . Bladder cancer . Lancet 2009. ; 374 ( 9685 ): 239 – 249 . [DOI] [PubMed] [Google Scholar]
- 139. Lopez-Beltran A , Cimadamore A , Montironi R , Cheng L . Molecular pathology of urothelial carcinoma . Hum Pathol 2021. ; 113 : 67 – 83 . [DOI] [PubMed] [Google Scholar]
- 140. Robertson AG , Kim J , Al-Ahmadie H , et al . Comprehensive molecular characterization of muscle-invasive bladder cancer . Cell 2018. ; 174 ( 4 ): 1033 . [Published correction appears in Cell 2017;171(3):540-556.e25.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. van Rhijn BW , van der Kwast TH , Liu L , et al . The FGFR3 mutation is related to favorable pT1 bladder cancer . J Urol 2012. ; 187 ( 1 ): 310 – 314 . [DOI] [PubMed] [Google Scholar]
- 142. Balbás-Martínez C , Sagrera A , Carrillo-de-Santa-Pau E , et al . Recurrent inactivation of STAG2 in bladder cancer is not associated with aneuploidy . Nat Genet 2013. ; 45 ( 12 ): 1464 – 1469 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Dueñas M , Martínez-Fernández M , García-Escudero R , et al . PIK3CA gene alterations in bladder cancer are frequent and associate with reduced recurrence in non-muscle invasive tumors . Mol Carcinog 2015. ; 54 ( 7 ): 566 – 576 . [DOI] [PubMed] [Google Scholar]
- 144. Alessandrino F , Williams K , Nassar AH , et al . Muscle-invasive urothelial cancer: Association of mutational status with metastatic pattern and survival . Radiology 2020. ; 295 ( 3 ): 572 – 580 . [DOI] [PubMed] [Google Scholar]
- 145. Nassar AH , Umeton R , Kim J , et al . Mutational analysis of 472 urothelial carcinoma across grades and anatomic sites . Clin Cancer Res 2019. ; 25 ( 8 ): 2458 – 2470 . [DOI] [PubMed] [Google Scholar]
- 146. Esrig D , Elmajian D , Groshen S , et al . Accumulation of nuclear p53 and tumor progression in bladder cancer . N Engl J Med 1994. ; 331 ( 19 ): 1259 – 1264 . [DOI] [PubMed] [Google Scholar]
- 147. He F , Mo L , Zheng XY , et al . Deficiency of pRb family proteins and p53 in invasive urothelial tumorigenesis . Cancer Res 2009. ; 69 ( 24 ): 9413 – 9421 . [DOI] [PMC free article] [PubMed] [Google Scholar]