Abstract
High mobility group A protein-2 (HMGA2) is an architectural transcription factor that binds to the A/T-rich DNA minor groove and is responsible for regulating transcriptional activity of multiple genes indirectly through chromatin change and assembling enhanceosome. HMGA2 is overexpressed in multiple tumor types, suggesting its involvement in cancer initiation and progression, thus, making it an ideal candidate for cancer diagnostic and prognostic. We performed a systematic review to examine the role of HMGA2 as a universal tumor cancer diagnostic and prognostic marker. We used Reporting Recommendations for Tumor Marker Prognostic Studies to systematically search OvidMedline, PubMed, and the Cochrane Library for English language studies, published between 1995 and June 2019. Meta-analysis provided pooled risk estimates and their 95% confidence intervals (CIs) for an association between overall survival and recurrence of cancers for studies with available estimates. We identified 42 eligible studies with a total of 5123 tumor samples in 15 types of cancer. The pooled percentage of HMGA2 gene expression in tumor samples was 65.14%. Meta-analysis showed that cancer patients with HMGA2 positive have significantly reduced survival, compared to patients without HMGA2 gene [pooled-hazard ratio (HR) = 1.85, 95% CI 1.48–2.22]. There was a positive association between cancer patients with HMGA2 overexpression and cancer recurrence though this association did not reach significance (pooled-HR = 1.44, 95% CI 0.80–2.07). Overexpression of HMGA2 was found in 15 types of cancer. There was an association between HMGA2 overexpression with reduced survival of cancer patients.
Keywords: cancer diagnostic and prognostic, HMGA2, tumor marker
Introduction
Tumor markers are substances that are produced by cancer cells or by other cells of the body in response to cancer, that are found in body fluids (i.e. blood and urine) and tissue (Bigbee and Herberman, 2003). Two main types of tumor markers that can be used in clinical settings: (1) circulating tumor markers or tumor markers that are associated with tumor cells and (2) tumor tissue markers that are derived from tumor cells. In several cancer patients, they are mainly represented by protein macromolecules (Bigbee and Herberman, 2003). Tumor markers can be associated with a specific cancer site or with multiple cancers; however, up to date there is no marker that is specifically associated with a certain type of cancer.
Even though the National Cancer Institute does not have a guideline for the use of tumor markers in clinical practice, several organizations have such guidelines. Accordingly, the American Society for Clinical Oncology has published different clinical practice guidelines of tumor markers for breast cancer (Hammond et al., 2010; Ramakrishna et al., 2018), colorectal cancer (Locker et al., 2006; Sepulveda et al., 2017), lung cancer (Keedy et al., 2011), and others (Gilligan et al., 2010) while the National Academic of Clinical Biochemistry has also published the guideline entitled the ‘Use of Tumor Makers in Clinical Practice: Quality Requirements’, focusing on the appropriate use of tumor markers for specific cancers (Sturgeon and Diamandis, 2008). Currently, about 35 tumor markers have been characterized and are being used in clinical practice, including BRCA1 and BRCA2 gene mutations, CA19–9, CA-125, carcinoembryonic antigen, EGFR gene mutation analysis or prostate-specific antigen, etc.
More than 30 years ago, the high mobility group A (HMGA) proteins were suggested potential tumor markers for cancer (Giancotti et al., 1987). The HMGA family includes HMGA1a, HMGA1b, HMGA1c, and HMGA2 (formerly called HMGI-C). Since the first publication implicating high mobility group proteins in neoplastic transformation in 1987 (Giancotti et al., 1987) and identification of HMGA2 (HMGI-C) in 1991 (Giancotti et al., 1991), the evidence of the involvement of HMGA2 in cell cycle, neurogenesis, and carcinogenesis is steadily growing.
HMGA2 is an architectural transcription factor that binds to the A/T-rich DNA minor groove using so-called AT-hook sequences, changes its conformation and consequently facilitates binding of a group of transcription factors. It regulates transcriptional activity of multiple genes indirectly through chromatin change and assembling enhanceosome (Reeves, 2010). Accordingly, two mechanisms that have been identified involving in this process. The first mechanism related to the transcription of the IFN-β gene that is activated in virus infected cells where HMGA binds to and coordinates the formation of an enhanceosome on ‘naked’ promoter DNA. Noted that there are two positioned nucleosomes cause the flank of this ‘naked’ promoter DNA. The IFN-β enhancesome would then enroll chromatin modifying and remodeling complexes. The formation of remodeling complex induced sliding of the inhibitory nucleosome and introduced TATA box which then leading to the binding of TBP/TFIIB and initiating Poll transcription. The second mechanism is involved the activation of different promoters, including IL-2, IL-Rα, CRYAB, and the 5′ LTR of the HIV-1 virus prior to transcriptional activation. For each of the activation of the above promoters, a nucleosome is stably positioned on a regulatory DNA element, containing biding sites for transcriptional factors, including HMGA, Elf-1, or AP-1. One of the important hallmarks of these positioned nucleosomes is that there are one or more stretches of A/T-rich DNA position on the surface of the nucleosome and adjacent to one of its edges (Reeves, 2010).
While HMGA2 is abundantly expressed during embryogenesis and re-expressed in pre-malignant or malignant tissues, the level of expression is very low or undetectable in adult tissues. However, HMGA2 is overexpressed in multiple tumor types, suggesting its involvement in cancer initiation and progression (Pallante et al., 2015). This makes HMGA2 unique, along with other embryonic biomarkers and an ideal candidate for cancer diagnostic and prognostic. Recently, we described a new prognostic biomarker of melanoma progression, transcription factor HMGA2 (Raskin et al., 2013) associated with development of metastases and patient survival. Specifically, we used transcriptome profiling of 46 primary melanomas, 12 melanoma metastasized and 16 normal skin samples and replicated in an independent set of 330 melanomas using AQUA analysis of tissue microarray. We found that transcriptional factor HMGA2 is significantly upregulated in primary melanomas and metastases (P = 1.2 × 10−7 and 9 × 10−5, respectively), compared with normal samples. We also found that HMGA2 overexpression is associated with BRAF/NRAS mutation (P = 0.0002) and that HMGA2 is independently associated with disease-free survival (DFS) [hazard ratio (HR) = 6.3, 95% confidence interval (CI) 1.8–22.3] and overall survival (OS; stratified log-rank P = 0.008) as well as distant metastases-free survival (DMFS) (HR = 6.4, 95% CI 1.4–29.7).
The oncogenic role of HMGA2 has been well documented in almost all cancer types, where it can be overexpressed, amplified, or fused with other proteins (Fusco and Fedele, 2007). HMGA2 can also become an excellent therapy target, since only tumor cells express this protein in adults. For example, inhibition of HMGA2 has been demonstrated to reduce ovarian cancer growth both in vitro and in vivo (Malek et al., 2008).
Different mechanisms of HMGA2 oncogenicity have been documented previously. For example, Fedele et al. (2006) found the activation of transcription factor E2F1 through binding HMGA2 to pRB. Specifically, they reported that HMGA2 interacts with pRB, leading to the induction of E2F1 activity in mouse pituitary adenomas by displacing HDAC1 from pRB/E2F1 complex, which later resulted in E2F1 acetylation. Other mechanisms include direct or indirect induction of cyclin A (Hammond et al., 2010) or negative regulation of nucleotide excision repair gene (Ramakrishna et al., 2018), the ERCC1 gene, causing DNA bending.
In terms of prognostic value, it is also observed that the transcription of human telomerase reverse transcriptase is enhanced by HMGA2 to upgrade carcinogenesis, a necessity for cancer cell development and self-renewal (Sepulveda et al., 2017). In addition, HMGA2 plays an important role in the epithelial-to-mesenchymal transition by activating the TGFβ signaling pathway, leading to the invasion and metastasis of human epithelial cancers (Locker et al., 2006).
More than half of the publications on HMGA2 in cancer have been published in the last 5 years, an indication of increasing interest to this oncogene. In addition to the research on HMGA2 regulation in cancer, there is a growing number of studies demonstrating that expression of HMGA2 in neoplasm is associated with metastatic phenotype and inferior patient survival. While the current understanding of HMGA2 involvement in carcinogenesis and tumor invasiveness has been reviewed, to our knowledge, no effort has been made to systematically examine the role of HMGA2 overexpression as a diagnostic and prognostic biomarker in multiple cancer types. We, therefore, performed this systematic review to address this gap and to present future perspectives of HMGA2 in the era of precision medicine.
Methods
Search strategy
From January 2017 to June 2019, an experienced librarian (Allison M. Howard) and two investigators (Y.T.-H.P. and O.U.) conducted a systematic search to identify published studies on HMGA2 from January 1995 to June 2019. Three main biomedical databases (i.e. OvidMedline, PubMed, and Cochrane Library) were searched using the following terms: (HMGA2 protein) OR (‘high mobility group A2’ OR HMGA2) OR (HMGI-C OR HMGIC OR STQTL9) AND (humans OR not animals) AND (cancer) AND (limit to years = ‘1995–2019’).
Study screening and selection
Inclusion criteria for the present systematic review were English language reports of the studies that determined the association between gene or protein expression levels of HMGA2 in tumor tissues/biospecimens and overall or progression-free survival (PFS) in any cancer types. All studies met Reporting Recommendations for Tumor Marker Prognostic Studies (REMARK) criteria during period 1995 to June 2019. The following exclusion criteria were applied: (1) no cancer outcomes; (2) not in English language; (3) not original research (i.e. review, commentary and editorial) or case report; (4) not using tumor tissues/biospecimens; and (5) unmet REMARK criteria (Altman et al., 2012). All extracted reports were reviewed independently by two investigators (i.e. Y.T.-H.P. and O.U.). We also requested additional information from corresponding authors of four articles (Sarhadi et al., 2006; Piscuoglio et al., 2012; Rizzi et al, 2013; Chang et al., 2015) that have reported P values without information on HRs or relative risks, and 95% CIs.
Data abstraction and coding
All eligible studies were abstracted independently by two reviewers (Y.T.-H.P. and O.U.) using coding system based on three guidelines: the REMARK criteria (Altman et al., 2012), the Strengthening the Reporting of Observational studies in Epidemiology-Molecular Epidemiology (Gallo et al., 2011), and the Standards for Reporting Diagnostic Accuracy (Bossuyt et al., 2003). Any discrepancies were resolved by discussion and consensus between the two investigators. The abstracted information for each study included: first author’s name, year of publication, country of origin, study design (i.e. cross-sectional, case-control, cohort, and randomized controlled trial), and patient/biospecimen characteristics. We also extracted additional information regarding preservation methods [i.e. frozen or formalin-fixed paraffin embedded (FFPE)], quantification methods [i.e. immunohistochemistry (IHC) and real-time PCR (RT-PCR)], primary antibody and dilution for IHC and RNA-isolation for RT-PCR, HMGA2 expression levels in tumor cells for diagnosis (i.e. proportion of cells expressing HMGA2) and survival estimates for prognosis (i.e. multivariable HR and respective 95% CI).
Systematic review and statistical analysis
Because of the study heterogeneity and limited data for each cancer type, except for thyroid cancer, we first reported the results of a systematic review of the expression of HMGA2 as a biomarker for cancer diagnosis and prognosis. Then, we performed a pooled analysis of HMGA2 expression in two types of estimates (percentage and fold change) and meta-analysis of OS/recurrence using the studies with available estimates as described below.
While HMGA2 expression levels were reported in percentage format in 38 out of 42 identified articles, four articles (Jones et al., 2008; Arora et al., 2009; Klemke et al, 2014; Nagar et al., 2014) reported fold change as an estimate. We grouped these four articles to calculate pooled-adjusted fold change of HMGA2 gene expression in cancers. Additionally, two articles (Miyazawa et al, 2004; Meyer et al., 2007) reported both types of estimate (i.e. percentage and fold change), we, therefore, included them in both analyses.
We also calculated the pooled-adjusted percentage of HMGA2 gene expression as a weighted average of study-specific rates in which the weights were proportions of those study-specific sample sizes to the pooled-sample size, as described below:
Where = individual study (from 1 to k);
= sample size of individual study;
= pooled-sample size;
= percentage of HMGA2 gene expression at individual study level.
We used the same formula to calculate pooled-fold change of HMGA2 expression in six studies included in the current analysis.
In the meta-analysis, we calculated HRs and the corresponding 95% CIs for survival and recurrence in cohort studies. Overall pooled HR and its 95% CI was calculated based on the individual estimates from nine cohort studies (Motoyama et al., 2008; Wang et al., 2011; Wu et al., 2012; Zou et al., 2012; Raskin et al., 2013; Kong et al., 2014; Lee et al., 2014; Liu et al., 2015; Xia et al., 2015) for survival analysis and six cohort studies (Miyazawa et al., 2004; Yang et al., 2011; Raskin et al., 2013; Califano et al., 2014; Liu et al., 2015; Jun et al, 2015) for recurrence analysis. We included estimates from both training and validation sets in the study by Wang et al. (2011) for the meta-analysis of survival and recurrence. We also used estimates from training and validation sets from the study by Raskin et al. (2013) for meta-analysis of survival; however, only estimate from the training set was included in the meta-analysis of recurrence. In the meta-analysis, each study was given a weight based on the inverse of the effect variance. Random-effects models that included a study heterogeneity variance component were used in the meta-analysis (DerSimonian and Laird, 1986). To evaluate publication bias, both funnel plots for visualization and Egger’s test for statistical significance were used (Egger et al, 1997). Meta-analysis was performed using the commands metan and metafunnel of the statistical software STATA 14.0 (College Station, Texas, USA). All tests were two-sided, and P < 0.05 was considered statistically significant.
Results
Among 42 eligible articles in the current review (Fig. 1) with a total of 5123 tumor samples, 11 studies were in thyroid cancer (Belge et al., 2008; Chiappetta et al., 2008; Prasad et al., 2008; Arora et al., 2009; Lappinga et al., 2010; Jin et al., 2011; Prasad et al., 2012; Klemke et al., 2014; Nagar et al., 2014; Jin et al., 2015; Jang et al., 2015), five studies in ovarian cancer (Mahajan et al., 2010; Hetland et al, 2012; Califano et al., 2014; Kim et al., 2015; Wu et al, 2015), four studies in gastric cancer (Motoyama et al., 2008; Kong et al., 2014; Jun et al., 2015; Lee et al., 2015), four studies in colorectal cancer (Huang et al., 2009; Wang et al., 2011; Helmke et al., 2012; Rizzi et al., 2013), three studies in liver cancer (Wu et al., 2012; Lee et al., 2013, Lee et al., 2014), two studies in breast cancer (Rogalla et al., 1997; Jones et al., 2008), two studies in lung cancer (Sarhadi et al., 2006; Meyer et al., 2007), two studies in oral cancer (Miyazawa et al., 2004; Chang et al., 2015), two studies in nasopharyngeal cancer (Liu et al., 2015; Xia et al., 2015), one study in pancreatic cancer (Piscuoglio et al., 2012), one study in melanoma (Raskin et al., 2013), one study in bladder cancer (Yang et al., 2011), one study in bile duct carcinoma (Zakharov et al., 2013), one study in gallbladder cancer (Zou et al., 2012), one study in glioma (Liu et al., 2014), and one study in esophageal cancer (Liu et al., 2014).
Fig. 1.
Search strategy and screening for eligibility for current systematic review. REMARK, Reporting Recommendations for Tumor Marker Prognostic Studies.
The majority of studies were conducted in the USA (n = 12), followed by China (n = 7), Germany (n = 5), multi-countries (n = 5), South Korea (n = 4), Italy (n = 3), while each of the following countries – Finland, Japan, Norway, Switzerland, Taiwan, and UK – provided one study. Regarding to study design, 20 studies were cross-sectional studies, one was case-only study and 21 were cohort studies. Also, 20 studies were conducted for the purpose of diagnostic only, one study was for the purpose of prognostic only and 21 studies were for both purposes (Table 1 and Supplemental Table 1, Supplemental digital content 1, http://links.lww.com/EJCP/A297).
Table 1.
Characteristics and summary of results of eligible studies for the current systematic review
Author, year (location) | Study design | Sample size and source | Patients | Quantification method | Detection/diagnostic results | Prognostic results |
---|---|---|---|---|---|---|
| ||||||
Thyroid cancer | ||||||
Belge et al. (2008) (Germany) | Cross-sectional | 64 FFPE | 19 adenomas; 28 PTC; 9 FTC; 3 ATC | IHC, RT-PCR | 100% | |
Chiappetta et al. (2008) (Italy) | Cross-sectional | 128 FFPE | 12 hyperplastic lesions; 31 adenomas; 21 FTC; 45 PTC; 12 ATC | IHC | PTC 30/45 = 75.5%; FTC 4/21 = 19.1%; ATC 11/12 = 91.6% | |
RT-PCR | Adenomas 1/7 = 14.3%; PTC 4/37 = 91.9%; FTC 13/16 = 81.2%; Anaplastic carcinoma 4/4 = 100% | |||||
Prasad et al. (2008) (USA) | Cross-sectional | 125 FFPE | 70 benign (20 adenomatoid nodules; 20 adenomas; 17 HA; 13 lymphocytic nodules); 55 malignant (19 PTC; 16 FVPTC; 14 FTC; 6 HC) | IHC, RT-PCR | PTC 26/30 = 87%; FVPTC 13/16 = 81%; FTC 11/14 = 79%; very high in malignant tumor | |
Arora et al. (2009) (USA) | Cross-sectional | 90 | 16 PTC, 22 FVPTC, 15 hyperplastic nodules, 22 FA; 15 borderline | Expression microarray | In tumor 3.56-fold change (P = 0.02) | |
Lappinga et al. (2010) (USA) | Cross-sectional | 115 | 71 benign and 44 malignant (13 FTC, 22 PTC, 9 HC) | RT-PCR | FTC 11/13 = 85%; HC 3/9 = 33%; PTC 17/22 = 33% | |
Jin et al. (2011) (USA) | Cross-sectional | 170 FFPE, 226 FNA | 34 FA, 10 HA, 6 hyperplastic nodules, 4 atypical adenomas, 44 PTC, 29 FVPTC, 23 FTC, 17 HC, 3 ATC | RT-PCR | Overall 71.6–79.8%; FNA 79.8–88.6% | |
Prasad et al. (2012) (USA) | Cross-sectional | 193 FFPE, 95 FNA | FFPE: 36 PTC; 20 FVPTC; 17 FTC; 30 FA; 18 Lymphocytic thyroid nodule; 18 adenomatoid nodule; 11 HA; 4 HC; 30 normal FNA: 67-benign; 28-malignant | IHC, RT-PCR | FTC 6/17 = 35%; FVPTC 12/20 = 60%; PTC 26/36 = 72% | |
Klemke et al. (2014) (Germany) | Cross-sectional | 37 FFPE | 14-FA, 11-PTC, 4-FVPTC, 8 = FTC | RT-PCR | FVPTC 23.9–156.9; PTC 128.2–1207.5 | |
Nagar et al. (2014) (USA) | Cross-sectional | 52 FNA | 21-FA, 12-FTC, 19-FVPTC | RT-PCR | FVPTC 28-fold higher (P < 0.01) | |
Jin et al. (2015) (USA) | Cross-sectional | 80 FFPE, 120 FNA | FFPE: 48 benign and 32 carcinoma; 120 FNA (56 benign and 64 carcinoma) | RT-PCR | PTC 23/24 = 95.8%; FVPTC 8/10 = 80%; FC 15/17 = 88.2%; HC 4/13 = 30.8% | |
Jang et al. (2015) (South Korea) | Cross-sectional | 192 FFPE | 41 goiter, 72 FA, 79 FTC | IHC | FA 30/72 = 41.7%; FTC 44/79 = 55.7% (P = 0.05) | |
Ovarian cancer | ||||||
Mahajan et al. (2010) (USA) | Case-only study | 115 FFPE | 30 HG-PSC; 10 SBT; 15 MMMT; 30 EOC; 15 MOC; 15 CCOC | IHC | HG-PSC 18/30 = 64.3%, MMMT 9/15 = 64.3%, SBT 1/10 = 10%, EOC 2/15 = 7.1%, MOC 1/15 = 6.7%, CCOC 3/15 = 23.1% | |
Hetland et al. (2012) (Norway) | Retrospective cohort | 199 FFPE | 199-effusion samples; 50 Primary tumors; 50 solid metastasis | IHC | Effusion 188/199 = 94.5%, primary tumors 48/50 = 96%, solid metastasis 45/50 = 90% | No association between HMGA2 expression and PFS or OS |
Califano et al. (2014) (Italy) | Retrospective cohort | 117 FFPE | 117 primary advanced ovarian cancer | IHC | 62/117 = 53.0% | DFS HMGA2 only: HR = 0.83 (0.38–1.82); HMGA2 + BMI: HR = 3.17 (1.25–8.03) |
Kim et al. (2015) (South Korea) | Retrospective cohort | 35 frozen, 39 FFPE | 74 ovarian carcinomas treated with Paclitaxel and Cisplatin or Carboplatin | IHC, RT-PCR | 30/74 = 40.5% in cancer | Significant association of HMGA2(+) expression with shorter OS 5-year OS rate: 78% vs. 35% (P = 0.02) |
Wu et al. (2015) (US - China) | Cross-sectional | 278 FFPE (Training), 150 FFPE (Validation) | Training: serous 80%, mucinous 20% Validation: serous 72%, mucinous 28% |
IHC | Training (P < 0.001): serous 130/222 = 58.6%; mucinous 15/56 = 26.8% Validation (P = 0.001): serous 56/108 = 54.6%; mucinous 10/42 = 23.8% |
|
Gastric cancer | ||||||
Motoyama et al. (2008) (Japan) | Cohort | 110 frozen, FFPE | 110 gastric carcinoma | IHC, RT-PCR | 83/110 = 75.4% | OS HR = 2.00 (1.32–3.15) |
Kong et al. (2014) (China) | Cohort | 212 FFPE | 158 cancers, 30 peritumoral tissues, 24 normal gastric tissues | IHC, RT-PCR | 68/1 18 = 43.0% | Multivariate OS: HMGA2(+) only HR = 0.98 (0.34–2.33); HMGA2(+)/Oct4(+) HR = 2.89 (1.02–5.14) |
Lee et al. (2015) (South Korea) | Cohort | 170 FFPE | 170 gastric cancer | IHC, RT-PCR | 39/170 = 22.9% | 5-year OS rate: 54.2% vs. 43.6% (P = 0.028) |
Jun et al. (2015) (South Korea) | Retrospective cohort | 169 FFPE | 110 gastric cancer; 29 adenoma; 30 non-cancerous gastric tissues | IHC | 72/1 10 = 65.5%, (P < 0.001) | RFS HR = 3.20 (1.50–6.79) |
Colorectal cancer | ||||||
Huang et al. (2009) (China) | Cross-sectional | 62 | 6-A colorectal; 2-S colon; 3-D colon; 20 rectum | RT-PCR | 47/62 = 76% | |
Wang et al. (2011) (USA - China) | Cohort | 280 FFPE | 89 (training, USA), 191 (validation, China); 66/191 had adjuvant chemotherapy | IHC, RT-PCR | Training: 32/89 = 35.9; validation: 70/191 = 36.6% | OS stages I + II vs. III + IV in training: HR = 2.38 (1.30–4.34), validation: HR = 2.14 (1.21–3.79) |
Helmke et al. (2012) (Germany) | Cross-sectional | 38 FFPE | 38 colon cancer expressing HGMA2 | IHC, RT-PCR | 19/38 = 50% | |
Rizzi et al. (2013) (Italy) | Retrospective cohort | 103 FFPE | 103 colorectal cancer | IHC | 90/103 = 87.4% | No OS difference in HMGA2(+) vs. (−) (P = 0.59) |
Liver cancer | ||||||
Wu et al. (2012) (China) | Cohort | 23 pairs = 46 FFPE=107 |
23 HCC | IHC, RT-PCR | 51/107 = 47.7%; HGMA2 high in tumor vs. normal (38.7 vs. 8.4, P < 0.01) | OS HR = 1.97 (1.17–3.33) |
Lee et al. (2013) (USA-China) | Cross-sectional | 86 FFPE | 15 FL-HCC; 15 hepatoblastomas; 34 HCC; 22 hepatic adenomas | IHC | 15/15 = 100% | |
Lee et al. (2014) (USA) | Retrospective cohort | 68 FFPE | 14 - stage I; 21 - stage II; 7 - stage III; 11 - stage IV | IHC | 18/55 = 33% | OS HR = 2.20 (1.12–4.33) |
Breast cancer | ||||||
Rogalla et al. (1998) (Germany) | Cross-sectional | 57 | 44 breast cancer, 13 normal adjacent tissue | RT-PCR | 20/44 = 45.45% | |
Jones et al. (2008) (UK) | Cross-sectional | 23 | 12 benign phylloides tumors, 11 borderline malignant phylloides tumors | RT-PCR | Microarray: 4-fold change RT-PCR: 6-fold change |
|
Lung cancer | ||||||
Sarhadi et al. (2006) (Finland) | Cohort | 152 FFPE | 152 mainly small cell lung carcinoma | IHC, RT-PCR | Overall: 130/144 = 90% SCC: 60/62 = 96.8%; HGMA2 high in tumor vs. normal | Significant association of HGMA2 expression in AC with poor survival (P = 0.05) |
Meyer et al. (2007) (Germany) | Cross-sectional | 68 (34 pairs) | 17 - AC, 17 - SCC | IHC, RT-PCR | 10–50% AC; 80% SCC AC: mean 158.41-fold (1.02–911.02) SCC: mean 336.26-fold (4.34–2,503.68) |
|
Oral cancer | ||||||
Miyazawa et al. (2004) (USA-Japan) | Retrospective cohort | 42 FFPE | 42 primary oral squamous cell carcinomas | IHC, RT-PCR | 31/42 = 73.8%; expression level: 163.4 ± 90.4 (P < 0.05) | DFS HR = 3.48 (1.40–8.69) |
Chang et al. (2015) (Taiwan) | Cohort | 215 FFPE | 215 oral squamous cell carcinoma | IHC | HGMA2 high in tumor vs. normal (P < 0.001) | 5-year OS: 75.6% vs. 57.7% (P = 0.007) 5-year DSS: 78.1% vs. 59.1% (P = 0.006) 5-year DFS: 72.7% vs. 53.1% (P = 0.002) |
RT-PCR | 48 ± 75 vs. 1 ± 1.5 copy/105 GAPDH copy, P < 0.001 | |||||
Nasopharyngeal cancer | ||||||
Liu et al. (2015) (China) | Cohort | 145 FFPE | 116 NPC, 29 non-cancerous NP tissues | IHC | 62/116 = 52.6% | OS HR = 1.72 (1.02–2.91) |
Xia et al. (2015) (China) | Retrospective cohort | 144 FFPE | 124 NPC, 20 non-tumoral nasopharynx | IHC | 54/124 = 43.55% (P < 0.001) | OS HR = 2.68 (1.18–6.08) |
Pancreatic cancer | ||||||
Piscuoglio et al. (2012) (Switzerland) | Retrospective cohort | 210 FFPE | 210 PDAC, 40 PanIN-3; 40 Normal control | IHC | 197/210 = 93.8% (PAD) 37/40 = 92.5% (PanIN-3) |
OS was not significantly different between HMGA2(+) and (−) |
Melanoma | ||||||
Raskin et al. (2013) (USA) | Retrospective cohort | 127 frozen (training), 330 FFPE (validation) | 67 primary melanoma, 20 melanoma metastases, 40 normal skin | Expression microarray, IHC, RT-PCR | Primary melanoma: 26/46 = 53.1% | Training: OS HR = 6.30 (1.80–22.30), DMFS HR = 6.40 (1.4–29.7) |
Melanoma metastasis: 10/12 = 83,3% | Validation: OS HR = 1.72 (1.09–2.73) | |||||
Bladder cancer | ||||||
Yang et al. (2011) (USA) | Retrospective cohort | 148 FFPE | 148 urothelial bladder cancer | IHC, RT-PCR | 77/148 = 52%; HGMA2 expression: 121 ± 31.13 (cancer) vs. 1.74 ± 0.42 (normal), P < 0.001 | RFS HR = 3.83 (2.19–6.71), PFS HR = 3.47 (1.43–8.45) |
Bile duct carcinoma | ||||||
Zakharov et al. (2013) (USA) | Cross-sectional | 48 FFPE | 22 adenocarcinoma, 12 adenoma, 14 reactive atypia | IHC | 41/48 = 86% | |
Gallbladder cancer | ||||||
Zou et al. (2012) (China) | Retrospective cohort | 204 FFPE | 108 AC, 45 adjacent tissue, 15 polyps, 35 chronic cholecystitis | IHC | Adenocarcinoma: 64/108 = 59.3% (P < 0.01) | OS HR = 3.02 (1.58–5.78) |
Glioma | ||||||
Liu et al. (2014) (China - Japan) | Retrospective cohort | 85 FFPE | 78 gliomas | IHC, RT-PCR | 67.9% (53/78) | HMGA2 at a higher level had a significantly shorter progression-free survival time (11.2 months vs. 18.8 months; P = 0.02) |
Esophageal cancer | ||||||
Liu et al. (2014) (China) | Cross-sectional | 226 FFPE | 113 esophageal squamous cell carcinomas | IHC, RT-PCR | 98/113 = 86.7% |
AC, adenocarcinoma; AFP, serum alpha-fetoprotein; AJCC, American Joint Committee on Cancer; ATC, anaplastic thyroid carcinoma; AUC, area-under-curve; CCOC, clear cell ovarian carcinoma; chemo, chemotherapy; DFS, disease-free survival; DMFS, distant metastases-free survival; DSS, disease-specific survival; EOC, endometroid ovarian carcinoma; FA, follicular adenoma; FFPE, formalin-fixed paraffin-embedded; FIGO, International Federation of Gynecology & Obstetrics; FL-HCC, fibrolamellar hepatocellular carcinoma; FNA, fine-needle aspiration; FTC, follicular carcinoma; FVPTC, follicular variant of papillary thyroid carcinomas; GAPDH, glyceraldehyde 3-phosphate dehydronase; GBM, glioblastoma multiforme; HA, Hürthle cell adenoma; HC, Hurthle cell carcinoma; HCC, hepatocellular carcinoma; HG-PSC, high-grade papillary serous carcinoma; HMGA2, high mobility group A2 protein; HR, hazard ratio; IHC, immunohistochemistry; MMMT, malignant mixed Mullerian tumor; MOC, mucinous ovarian carcinoma; NPC, nasopharyngeal carcinoma; PAD, pancreatic adenocarcinoma; PanIN-3, pancreatic intraepithelial neoplasia, grade 3; PDAC, pancreatic ductal adenocarcinoma; PFS, progression-free survival; PTC, papillary thyroid carcinomas; radio, radiotherapy; RFS, recurrence-free survival; RT-PCR, real-time PCR; SBT, serous borderline tumor; SCC, squamous cell carcinoma; SCC, squamous cell carcinoma; Sen, sensitivity; Spe, specificity.
Sources of materials were both FFPE and fine-needle aspiration, except two studies (Motoyama et al., 2008; Raskin et al., 2013) in which HMGA2 was also from frozen samples. The method to quantify HMGA2 gene expression was either IHC or RT-PCR and expression microarray was also used additionally in two studies (Arora et al., 2009; Raskin et al., 2013) (Table 1 and Supplemental Table 1, Supplemental digital content 1, http://links.lww.com/EJCP/A297).
Thyroid cancer
Diagnostic
Between 2008 and 2019, eleven cross-sectional studies (Belge et al., 2008; Chiappetta et al., 2008; Prasad et al., 2008; Arora et al., 2009; Lappinga et al., 2010; Jin et al., 2011; Prasad et al., 2012; Klemke et al., 2014; Nagar et al., 2014; Jin et al., 2015; Jang et al., 2015) investigated the gene expression of HMGA2 in thyroid cancer. The frequency of HMGA2 expression in tumor samples varied from 30.8% in a study by Jin et al. (2015) (in a histologic diagnosis of Hürthle cell carcinoma) to 100% in a study by Belge et al. (2008). Additionally, two studies (Arora et al., 2009; Klemke et al, 2014) reported fold change of HMGA2 in tumor sample in comparison with benign tumor. Accordingly, Arora et al. (2009) found that the expression level of HMGA2 was 3.56-fold higher in thyroid tumor than that in benign tumor (P = 0.02). Also, using result of frequency of HMGA2 gene expression (100% in thyroid tumor), Belge et al. (2008) found that the decision limit for the discrimination between benign and malignant tissues was 3.99 with a sensitivity of 95.9% and specificity of 93.9%; one of the best known single biomarker to distinguish between benign and malignant thyroid neoplasm.
Prognostic
In the current review, we did not find any such study for prognostic using HMGA2 in thyroid cancer.
Ovarian cancer
Diagnostic
Between 2009 and 2019 there were one case-only study (Mahajan et al., 2010), one cross-sectional study (Wu et al., 2015) and three cohort studies (Hetland et al., 2012; Califano et al., 2014; Kim et al., 2015) which investigated the expression of HMGA2 in ovarian cancer. The frequency of HMGA2 expression was found to be lowest in a study of mucinous ovarian carcinoma (6.7%) by Mahajan et al. (2010) and highest in a study by Hetland et al. (2012) (96.0%).
Prognostic
Findings on the prognostic value of HMGA2 to ovarian cancer is inconclusive. Accordingly, Hetland et al. (2012), in a study of 199 ovarian cancer patients, found null association between HMGA2 expression and PFS or OS in effusions (P = 0.5 and P = 0.9, respectively), primary tumors (P = 0.7 and P = 0.5, respectively) or metastatic samples (P = 0.1 and P = 0.5, respectively). However, a study from Italy by Califano et al. (2014), found null association between HMGA2 expression only and DFS (HR = 0.83, 95% CI 0.38–1.82); they did not find a significant association between combination/interaction between HMGA2-BMI (low vs. high score) and OS of ovarian cancer (HR = 3.17, 95% CI 1.25–8.03). Recently, Kim et al. (2015) reported that HMGA2 expression was associated with distant metastasis (P = 0.001), FIGO stage (P = 0.004), and lymph node (P = 0.008). The expression of HMGA2 was also correlated with OS of patients with high grade ovarian serous carcinomas (5-year OS rate: 78% vs. 35%, P = 0.02).
Gastric cancer
Diagnostic
From 2008 up to date, there are four cohort studies (Motoyama et al., 2008; Kong et al., 2014; Jun et al., 2015; Lee et al., 2015) investigating the association between expression of HMGA2 and risk of gastric cancer. The lowest frequency of HMGA2 gene expression was found in a study by Lee et al. (2015) of 170 FFPE samples (22.9%) and highest was in a study by Motoyama et al. (2008) of 110 frozen samples (75.4%)
Prognostic
Data from these four studies (Motoyama et al., 2008; Kong et al., 2014; Jun et al., 2015; Lee et al., 2015) showed consistently that HMGA2 had poor survival for gastric cancer patients. Accordingly, in a study of 110 frozen samples in Japan, Motoyama et al. (2008) shown that HMGA2 expression level was associated with reduced survival (OS HR = 2.00, 95% CI 1.32–3.15). In another study by Kong et al. (2014) of 158 gastric cancer and surrounding non-tumor tissues, they found that while there was no association between HMGA2 or Oct4 with poor survival of gastric cancer (HR = 0.99, 95% CI 0.34–2.33; and 1.00, 95% CI 0.41–2.86, respectively) the combination between these two proteins was a predictor of poor survival of gastric cancer (HR = 2.89, 95% CI 1.02–5.14). The other study by Lee et al. (2015) reported that patients with high-level expression have a significantly worse 5-year OS rate than those with low-level expression (43.6% vs. 54.2%; P = 0.028). Finally, Jun et al. (2015) found that high level of HMGA2 expression in gastric cancer patients were significantly associated with recurrence-free survival (RFS) (HR = 3.20, 95% CI 1.50–6.79).
Colorectal cancer
Diagnostic
Between 2009 and 2019, four studies (Huang et al., 2009; Wang et al., 2011; Helmke et al., 2012; Rizzi et al., 2013) investigated the expression of HMGA2 and colorectal cancer status of which two (Huang et al., 2009; Rizzi et al., 2013) were of cross-sectional study design and two (Wang et al., 2011; Helmke et al., 2012) were of cohort study design. The frequency of HMGA2 expression in colorectal cancer was found from 36 (Wang et al., 2011) to 87.4% (Rizzi et al., 2013).
Prognostic
In a study of 280 FFPE samples, Wang et al. (2011) reported an association between HMGA2 overexpression with poor survival (Training set: HR-OS/OS = 2.38, 95% CI 1.30–4.34; Validation set: HR-OS = 2.14, 95% CI 1.21–3.79). They also shown a significant association between HMGA2 overexpression and distant metastasis (training set: OR = 3.53, 95% CI 1.37–9.70; validation set: OR = 6.38, 95% CI 1.47–43.95). In another study of 103 colorectal cancer cases in Italy, Rizzi et al. (2013) found that the increased HMGA2 expression was strongly associated with an increase in tumor invasiveness, measured through both budding and vascular invasion (P < 0.0001).
Liver cancer
Diagnostic
From 2012 to date, we found three studies (Wu et al., 2012; Lee et al., 2013; Lee et al., 2014) that investigated the expression of HMGA2 and liver cancer. The frequency of HMGA2 expression in liver cancer was found to be as low as 33% in a study by Lee et al. (2013) and as high as 100% in a study by Lee et al. (2014). In the other study, Wu et al. (2012) also reported that HMGA2 expression level was higher in tumor than non-tumor tissues (mean ± SD: 38.70 ± 10.41 vs. 8.41 ± 4.06, respectively; P < 0.01) and that HMGA2 was expressed in 48% of liver cancer tumors.
Prognostic
HMGA2 overexpression in liver cancer patients had consistently poor survival outcome. Accordingly, Wu et al. (2012) shown that HMGA2 expression was associated with decreased OS (OS-HR = 1.97, 95% CI 1.17–3.33). Similarly, Lee et al. (2013), in a study of 15 hepatoblastoma, a rare but most common type of hepatocellular carcinoma-HCC in children with 71 other HCC types samples, reported that patients with HMGA2 was 2.20 times higher risk of death than those without HMGA2 (HR = 2.20, 95% CI 1.12–4.33).
Breast cancer
Diagnostic
Between 1998 and up to date, there are two cross-sectional studies (Rogalla et al., 1997; Jones et al., 2008) examining the relationship between HMGA2 expression and breast cancer status. Accordingly, Rogalla et al. (1997) reported that HMGA2 over-expressed in 45.45% of breast tumors while Jones et al. (2008) reported that HMGA2 expression was 4.2-fold change in microarray test and six-fold change in RT-PCR test (P = 0.003).
Prognostic
We did not find any studies on prognosis for HMGA2 in breast cancer in this review.
Lung cancer
Diagnostic
We found two studies, one cross-sectional study (Sarhadi et al., 2006) and one cohort study (Meyer et al., 2007) between 2006 and up to date, examining the expression of HMGA2 and lung cancer status. The overexpression of HMGA2 was particularly high in squamous cell carcinoma (SCC) sub-type (i.e. 96.8% in a study by Sarhadi et al. (2006) and 80% in a study by Meyer et al. (2007)). Also, Meyer et al. (2007) reported that the HMGA2 expression levels were increased up to 911-fold (mean: 158.41, range: 1.02–911.02, P < 0.0001) for adenocarcinoma and up to 2504-fold for SCC (mean: 336.26, range: 4.34–2.503.68, P < 0.0001).
Prognostic
Only a study by Sarhadi et al. (2006) reported the survival data in which they shown that there was a significant association between HMGA2 positive and poor survival in adenocarcinoma patients (P = 0.05).
Oral cancer
Diagnostic
From 2004 to date, we found two cohort studies (Miyazawa et al., 2004; Chang et al., 2015) that investigated the overexpression levels of HMGA2 and oral cancer status. Accordingly, Miyazawa et al. (2004) reported that HMGA2 was detected in 73.8% carcinomas but none in normal oral tissues. They also found that oral carcinoma tissues expressed the HMGA2 gene at levels 84.4–315.2-fold greater than that of normal tissues (mean ± SD: 163.4 ± 90.4; P < 0.05). Similarly, Chang et al. (2015) reported that HMGA2 levels was significantly expressed in oral SCC specimens compared with adjacent normal tissues (mean ± SD: 48 ± 75 vs. 1 ± 1.5 copy/105 GAPDH-glyceraldehyde 3-phosphate dehydronase copy, P < 0.001)
Prognostic
Both cohort studies shown that oral cancer patient with HMGA2 had poorer survival than patient without HMGA2 gene (Miyazawa et al., 2004; Chang et al., 2015). Accordingly, Miyazawa et al. (2004) reported that HMGA2 was significantly associated with poor survival (DFS HR = 3.48, 95% CI 1.39–8.69) while Chang et al. (2015) indicated that the 5-year OS, disease-specific survival (DSS), and DFS rates for patient subgroups stratified by the absence or presence of HMGA2 expression were 75.6% vs. 57.7% (P = 0.007), 78% vs. 59.1% (P = 0.006), and 72.7% vs. 53.1% (P = 0.002), respectively. In multivariable analysis (adjusted for age, sex, overall stage, perineural invasion), HMGA2 expression is independent predictor of OS, DSS, and DFS (P = 0.028, 0.025, and 0.015, respectively) (Chang et al., 2015).
Nasopharyngeal cancer
Diagnostic
Recently, two cohort studies (Liu et al., 2015; Xia et al., 2015) have examined the relation between HMGA2 expression and nasopharyngeal cancer status. The levels of HMGA2 expression ranged from 43.5 (Liu et al., 2015) to 52.6% (Xia et al., 2015).
Prognostic
Both cohort studies (Liu et al., 2015; Xia et al., 2015) provided consistent evidence that HMGA2 positive is a predictor of poor survival for patients with nasopharyngeal cancer. Accordingly, Liu et al. (2015), in a cohort study of 145 samples has shown that the OS HR for a patient of nasopharyngeal cancer with HMGA2 positive was 1.72 (95% CI 1.02–2.91) compared with a patient without HMGA2 gene. At the same time, Xia et al. (2015) found even higher estimate on the HMGA2 expression in relation to with poor survival (OS-HR = 2.68, 95% CI 1.18–6.08).
Pancreatic cancer
Diagnostic
In a cohort study of 210 ductal pancreatic adenocarcinomas (PAD) in Switzerland, Piscuoglio et al. (2012) found that HMGA2 was overexpressed in 94% of PAD tissues and 92% of pancreatic intraepithelial neoplasia-grade 3 (PanIN-3). They also reported that the mean ± SD for the percentage of cells showing HMGA2 protein expression were found to 0.2 ± 0.9 in normal tissue, as compared with 16.3 ± 28.4 in carcinomas (P < 0.001). HMGA2 protein expression were significantly higher in ductal PAD (mean ± SD: 16.3 ± 28.4) than in PanIN cases (2.7 ± 13.5) (P < 0.001). Similar observation was found between PanIN vs. normal tissue (2.7 ± 13.5 vs. 0.2 ± 0.9, P < 0.001).
Prognostic
In the same cohort study, Piscuoglio et al. (2012) did not find a difference in median survival between HMGA2positive vs. HMGA2-negative tissues (P = 0.20).
Melanoma
Diagnostic
In 2013, from a cohort study of 127 frozen samples (training set) and 330 FFPE samples (validation set), Raskin et al. (2013) showed that the frequency of HMGA2 overexpression was 53.1 and 83.3% in primary melanoma and melanoma metastasis tissues, respectively. They also reported that HMGA2 expression is significantly upregulated in primary melanoma and metastases (P = 9 × 10−5) compared with normal tissues.
Prognostic
In the same cohort study, Raskin et al. (2013) also reported that in the training set HMGA2 is independently associated with DFS (HR = 6.3, 95% CI 1.8–22.3), OS (stratified log-rank P = 0.008), and DMFS (HR = 6.4, 95% CI 1.4–29.7) after adjusting for American Joint Committee on Cancer (AJCC) stage and age at. The validation set also confirmed that HMGA2 overexpression was significantly associated with reduced OS of melanoma patients, after adjustment for AJCC stage and age at diagnosis (HR = 1.72, 95% CI 1.09–2.73).
Bladder cancer
Diagnostic
Yang et al. (2011), in a cohort study of 148 bladder cancer and 30 specimens of adjacent normal bladder tissues, reported that HMGA2 was overexpressed in 52% of tumor samples and that the expression levels of HMGA2 were significant higher in tumor tissues than adjacent normal tissues (mean ± SD: 121.84 ± 31.13 vs. 1.74 ± 0.42, respectively; P < 0.001).
Prognostic
Consistent with findings from other cancers, Yang et al. (2011) reported that HMGA2 expression was associated with poor survival. The HR of RFS and PFS were 3.83 (95% CI 2.19–6.71) and 3.47 (95% CI 1.43–8.45), respectively.
Bile duct carcinoma
Diagnostic
In a cross-sectional study of 48 FFPE samples of bile duct carcinoma in the USA, Zakharov et al. (2013) reported that the frequency of HMGA2 overexpression in tumor samples was 86%.
Prognostic
We found no study in the survival or recurrence of bile duct carcinoma in the current systematic review.
Gallbladder cancer
Diagnostic
In a cohort study of 204 FFPE samples, Zou et al. (2012) found that the percentage of HMGA2 overexpression in gallbladder cancer was 59% compared with only 23% in cancer adjacent tissues (P < 0.01), 20% in polyps (P < 0.01) and 14% in chronic cholecystitis (P < 0.01)
Prognostic
In the same study (Zou et al., 2012), it was found that gallbladder cancer patients with HMGA2 positive had poorer survival than patients without HMGA2 (OS-HR = 3.02, 95% CI 1.58–5.78).
Glioblastoma
Diagnostic
In a recent cohort study of 85 FFPE samples of glioblastoma, Liu et al. (2014) found that 68% of cancer tumor tissues had HMGA2 overexpression.
Prognostic
In this same cohort study, Liu et al. (2014) also found that patients with tumors expressing HMGA2 at a higher level had a significantly shorter PFS time (11.2 months vs. 18.8 months; P = 0.02).
Esophageal cancer
Diagnostic
Liu et al. (2014), in a study of 123 esophageal SCC (OSCC) and 123 normal adjacent tissue (NAT), found that the expression of HMGA2 was significantly more frequent in OSCC (98 of 113, 86.7%) than in NAT (50 of 113, 44.2%, P < 0.0001).
Prognostic
No data for prognostic purpose (i.e. survival) is currently available for review or further analysis.
Meta-analysis
To provide a better perspective on the frequency/levels of HMGA2 gene expression in cancer tumor samples (for diagnostic purpose) and the survival and recurrence among HMGA2 positive patients, compared with those without HMGA2, we performed a meta-analysis with articles that had relevant data and provided data after contacting to corresponding authors.
Overall, 37 over 42 articles had data on frequency (or percentage) of HMGA2 overexpression in tumor samples. The pooled percentage of HMGA2 gene expression in tumor samples was 65.14%. Six out of 42 articles had data on the levels of HMGA2 expression in tumor samples. The pooled levels of HMGA2 Gene Expression in tumor samples was 113.08-fold changes (Table 2).
Table 2.
Pooled percentage and levels of HMGA2 gene expression (fold change) in cancer specimens
Sample Size | % | Weight | Adjusted % | |
---|---|---|---|---|
| ||||
Belge et al. (2008) | 64 | 100 | 0.012493 | 0.01249268 |
Chiappetta et al. (2008) | 37 | 91.90 | 0.007222 | 0.006637322 |
16 | 81.20 | 0.003123 | 0.002536014 | |
4 | 100.00 | 0.000781 | 0.000780793 | |
45 | 75.50 | 0.008784 | 0.006631856 | |
21 | 4.00 | 0.004099 | 0.000163966 | |
12 | 91.60 | 0.002342 | 0.002145618 | |
Prasad et al. (2008) | 30 | 87 | 0.005856 | 0.005094671 |
16 | 81 | 0.003123 | 0.002529768 | |
14 | 79 | 0.002733 | 0.002158891 | |
Lappinga et al. (2010) | 13 | 85 | 0.002538 | 0.002156939 |
9 | 33 | 0.001757 | 0.000579738 | |
22 | 77 | 0.004294 | 0.003306656 | |
Jin et al. (2011) | 170 | 71.6 | 0.033184 | 0.023759516 |
170 | 79.8 | 0.033184 | 0.026480578 | |
226 | 79.8 | 0.044115 | 0.035203592 | |
226 | 88.6 | 0.044115 | 0.039085692 | |
Prasad et al. (2012) | 17 | 35.0 | 0.003318 | 0.001161429 |
20 | 60.0 | 0.003904 | 0.002342378 | |
36 | 72.0 | 0.007027 | 0.005059535 | |
Jin et al. (2015) | 24 | 95.8 | 0.004685 | 0.004487995 |
10 | 80.0 | 0.001952 | 0.001561585 | |
17 | 88.2 | 0.003318 | 0.002926801 | |
14 | 30.8 | 0.002733 | 0.000841694 | |
Jang et al. (2015) | 72 | 41.7 | 0.014054 | 0.005860629 |
79 | 55.7 | 0.015421 | 0.008589303 | |
Mahajan et al. (2010) | 30 | 64.3 | 0.005856 | 0.003765372 |
15 | 64.3 | 0.002928 | 0.001882686 | |
10 | 10.0 | 0.001952 | 0.000195198 | |
30 | 7.1 | 0.005856 | 0.000415772 | |
15 | 6.7 | 0.002928 | 0.000196174 | |
15 | 23.1 | 0.002928 | 0.000676362 | |
Hetland et al. (2012) | 199 | 94.5 | 0.038844 | 0.036707984 |
50 | 96.0 | 0.00976 | 0.00936951 | |
50 | 90.0 | 0.00976 | 0.008783916 | |
Califano et al. (2014) | 117 | 53.0 | 0.022838 | 0.012104236 |
Kim et al. (2015) | 74 | 40.5 | 0.014445 | 0.005850088 |
Wu et al. (2015) | 222 | 58.6 | 0.043334 | 0.025393715 |
56 | 26.8 | 0.010931 | 0.002929533 | |
108 | 54.6 | 0.021081 | 0.011510443 | |
42 | 23.8 | 0.008198 | 0.0019512 | |
Motoyama et al. (2008) | 110 | 75.4 | 0.021472 | 0.016189733 |
Kong et al. (2014) | 158 | 43.0 | 0.030841 | 0.013261761 |
Lee et al. (2015) | 170 | 22.9 | 0.033184 | 0.007599063 |
Jun et al. (2015) | 110 | 65.50 | 0.021472 | 0.014064025 |
Huang et al. (2009) | 62 | 76.0 | 0.012102 | 0.009197736 |
Wang et al. (2011) | 89 | 35.9 | 0.017373 | 0.006236775 |
191 | 36.6 | 0.037283 | 0.01364552 | |
Helmke et al. (2012) | 38 | 50.0 | 0.007418 | 0.003708764 |
Rizzi et al. (2013) | 103 | 87.4 | 0.020105 | 0.017572126 |
Wu et al. (2012) | 107 | 47.7 | 0.020886 | 0.009962717 |
Lee et al. (2013) | 15 | 100.0 | 0.002928 | 0.002927972 |
Lee et al. (2014) | 55 | 33.0 | 0.010736 | 0.003542846 |
Rogalla et al. (1998) | 44 | 45.4 | 0.008589 | 0.003899278 |
Sarhadi et al. (2006) | 144 | 90.3 | 0.028109 | 0.025382003 |
62 | 96.8 | 0.012102 | 0.011715011 | |
Meyer et al. (2007) | 68 | 50.0 | 0.013273 | 0.006636736 |
68 | 80.0 | 0.013273 | 0.010618778 | |
Miyazawa et al. (2004) | 42 | 73.8 | 0.008198 | 0.006050361 |
Liu et al. (2015) | 116 | 52.6 | 0.022643 | 0.011910209 |
Xia et al. (2015) | 144 | 43.5 | 0.028109 | 0.012227211 |
Piscuoglio et al. (2012) | 210 | 93.8 | 0.040992 | 0.038450127 |
40 | 92.5 | 0.007808 | 0.007222331 | |
Raskin et al. (2013) | 46 | 57.0 | 0.008979 | 0.005118095 |
12 | 83.0 | 0.002342 | 0.001944173 | |
Yang et al. (2011) | 148 | 52.0 | 0.028889 | 0.015022448 |
Zakharov et al. (2013) | 48 | 86.0 | 0.00937 | 0.008057779 |
Zou et al. (2012) | 108 | 59.3 | 0.021081 | 0.012501269 |
Liu et al. (2014) | 85 | 67.9 | 0.016592 | 0.01126586 |
Liu et al. (2014) | 113 | 86.7 | 0.022057 | 0.019123756 |
Total | 5123 | 0.651362288 | ||
| ||||
Sample size | Fold change | Weight | Adjusted fold change | |
| ||||
Arora et al. (2009) | 90 | 3.56 | 0.188679 | 0.671698113 |
Klemke et al. (2014) | 37 | 23.9 | 0.077568 | 1.853878407 |
37 | 156.9 | 0.077568 | 12.17044025 | |
37 | 128.2 | 0.077568 | 9.944234801 | |
Jones et al. (2008) | 23 | 4 | 0.048218 | 0.192872117 |
23 | 6 | 0.048218 | 0.289308176 | |
Nagar et al. (2014) | 52 | 28 | 0.109015 | 3.052410901 |
Meyer et al. (2007) | 68 | 158.41 | 0.142558 | 22.58255765 |
68 | 336.26 | 0.142558 | 47.93643606 | |
Myazawa et al. (2004) | 42 | 163.4 | 0.08805 | 14.38742138 |
Total | 477 | 113.0812579 |
Nine studies had available data for OS meta-analysis. We found that cancer patients with HMGA2 positive was significantly reduced survival in comparison with cancer patients without HMGA2 gene (pooled-HR = 1.85, 95% CI 1.48–2.22) (Fig. 2a). There was a positive association between cancer patients with HMGA2 positive with cancer recurrence (in six studies), though this association did not reach significant level (pooled-HR = 1.44, 95% CI 0.80–2.07) (Fig. 3a). There was no publication bias in both meta-analysis of OS and recurrence of cancer (Figs. 2b and 3b).
Fig. 2.
(a) Overall survival of cancers with HMGA2 positive vs. HMGA2 negative (using immunohistochemistry testing method only). (b) Funnel plots of publication bias in the overall survival of cancers with HMGA2 positive vs. HMGA2 negative.
Fig. 3.
(a) Recurrence of Cancers with HMGA2 positive vs. HMGA2 negative (using immunohistochemistry testing method only). (b) Funnel plots of publication bias recurrence of cancers with HMGA2 positive vs. HMGA2 negative.
Discussion
In this review, we identified 42 studies published between 1998 and June 2019, with a total of 5123 tumor samples, that evaluated HMGA2 expression in 15 cancer types, including thyroid cancer, ovarian cancer, gastric cancer, colorectal cancer, liver cancer, breast cancer, lung cancer, oral cancer, nasopharyngeal cancer, pancreatic cancer, melanoma, bladder cancer, bile duct carcinoma, gallbladder cancer, glioma and esophageal cancer. In our meta-analysis, we found that HMGA2 was overexpressed in more than two-third of all 15 types of cancer and HMGA2 overexpression was significantly associated with reduced survival. There was also a trend towards association between HMGA2 expression and cancer recurrence, although not statistically significant.
The fact that the current systematic review demonstrated that HMGA2 was overexpressed (i.e. more than two-third) of 15 cancer types in 42 included showed that HMGA2 might be an important marker as an universal tumor marker for prognostic. To our knowledge, the current review is the most comprehensive systematic reviews on the role of HMGA2 as tumor marker for diagnostic and prognostic in different types of cancer. A recent review by Pallante et al. (2015) found HMGA2 overexpressed in seven types of cancer, including breast cancer (two studies), colorectal cancer (three studies), lung cancer (two studies), ovarian cancer (four studies), pancreatic cancer (one study), and testis cancer (one study).
The difference between our review and the review by Pallante et al. (2015) is that in addition to 13 studies that were already identified, we found 29 more studies in eight more cancer sites, a strong indication of emerging attention of the field on the role of HMGA2 in diagnostic and prognostic of cancer. The other difference is that in our current review, not only did we identify articles associated with overexpression of HMGA2 for diagnostic purpose, we also found those for prognostic purpose. Indeed, we found nine studies that had available data for OS meta-analysis and that found that cancer patients with HMGA2 positive was significantly reduced survival in comparison with cancer patients without HMGA2 gene. This suggests the significant value of HMGA2 as a universal tumor marker for both purposes (i.e. diagnostic and prognostic) in different types of cancer. With the number of cancer patients increasing (i.e. 1.8 million new cases in 2018 (American Cancer Society, 2018), that it is expected that 18 million Americans are living with a diagnosis of cancer by 2022 (Siegel et al., 2012), and a great attention to precision medicine as well as the application of liquid biopsy, the role of HMGA2 as a tumor marker in cancer cannot be overemphasized.
A recent meta-analysis Binabaj et al. (2019) on HMGA2 and OS and correlation with clinicopathological parameters. The major difference between ours and the study by Binabaj et al. (2019) is that they did not evaluate the diagnostic value of HMGA2. In our study, we calculated pooled percentage of HMGA2 gene expression and levels of fold change in cancer specimens compared to benign tumor samples. Identifying the pooled level of HMGA2 expression in tumors in addition to survival is of vital importance as since it is quite high, more than 64% among 15 cancer types, HMGA2 may be a universal biomarker in diagnostic to determine severity and inform treatment plans as well as predict survival.
One interesting point is that while there are close to a dozen articles on thyroid cancer, studies on the role of HMGAs as tumor marker for diagnostic and prognostic and common and fatal cancers in the USA are few, including four studies in colorectal cancer (Mahajan et al., 2010; Wang et al., 2011; Helmke et al., 2012; Rizzi et al., 2013), two studies in lung cancer (Sarhadi et al., 2006; Meyer et al., 2007), two studies in breast cancer (Rogalla et al., 1997; Jones et al., 2008), five studies in ovarian cancer (Mahajan et al., 2010; Hetland et al., 2012; Califano et al., 2014; Kim et al., 2015; Wu et al., 2015) or one study in pancreatic cancer (Piscuoglio et al., 2012). Further studies on HMGAs and those cancers are thus warranted.
Several mechanisms may underlie the oncogenicity of HMGA2, including activation of transcription factor E2F1 by binding of HMGA2 to pRB (Fedele et al., 2006), direct or indirect induction of cyclin A (Pagano et al., 1992), or negative regulation of nucleotide excision-repair genes (Borrmann et al., 2003). The chemokine CXCL1 overexpressed in melanoma and involved in melanoma progression is also regulated by HMGA2 (Nirodi et al. (2001)). TGF-beta mediates epithelial-mesenchymal transition by inducing HMGA2 via the SMAD pathway and HMGA2 also enhances the NF-kB complex formation (Noro et al., 2003). miRNA let-7 family negatively regulates HMGA2 expression (Peng et al., 2008) and loss of let-7 expression upregulates c-Myc, RAS, CDK4, integrin-β (Bittner et al., 2000), and HMGA2 (Johnson et al., 2005; Müller and Bosserhoff, 2008; Schultz et al., 2008). Activated MAPK pathway negatively regulates let-7 by inducing LIN28 expression through Myc transcription (Dangi-Garimella et al., 2009). It is worth noting that different miRNAs (i.e. let-7a, miR-15, miR-16, miR-26a, miR-34b, miR-196a2, miR-326, miR-432, miR-548c-3p, miR-570, and miR-603) have been identified to be associated with post-transitional repression of HMGA family, including HMGA2 (D’Angelo et al., 2012; Palmieri et al., 2012). Therefore, more studies are warranted of both HMGA expression and microRNA levels in relation to diagnosis and OS.
The usage of HMGA2 with other markers to enhance their diagnostic values have been explored previously. For example, in a study to determine the diagnostic accuracy of different markers for follicular neoplasm, Jang et al. (2015) found that the sensitivity, specificity and diagnostic accuracy of HMGA2 to follicular neoplasm were 49.0, 75.6, and 54.7%, respectively. However, when HMGA2 was used in combination with either Hector Battifora mesothelial 1 or cyclin D1, these values increased to 80.8, 75.6, and 79.7%, respectively (Jang et al, 2015).
One of the main challenges for the current review is that there are three main types of quantification methods for HMGA2 expression (i.e. microarray, IHC, and RT-PCR), the source of antibody, concentration and evaluation methods used in selected studies are different. For this reason, a sub-group analysis for heterogeneity is not possible. Results from these testing methods, however, showed the presence or absence of HMGA2 in tumor tissues in comparison with normal tissues. In 23 of total 42 eligible studies, RT-PCR was performed first and their results were confirmed by IHC while 15 other used only IHC, three studies used RT-PCR only and one study used microarray for HMGA2 quantification. For RT-PCR, the relative HMGA2 expression between tumor tissue compared with normal tissue was calculated using 2−ΔΔt method (Keedy et al., 2011). We calculated OS and recurrence of cancers between HMGA2 positive vs. negative using data from studies used IHC only (Fig. 2a and b). In those studies (that used IHC method), standardized protocol was deployed (Gilligan et al., 2010) such that the staining (HMGA2) was considered positive when localized to the nucleus and the score of 4 was applied: 0 = no staining; 1 = 1–5%; 2 = 2–25%; 3 = 26–75%, and 4 = 76–100% stained tumor cells and that specimens should contain at least 100 tumor cells.
Additionally, there may be differences in cutoff thresholds for HMGA2 expression, using RT-PCR, for different studies; thus increasing heterogeneity of results. In studies using only IHC method that were used for meta-analysis of OS and recurrence of cancers, however, we did not find the publication bias (Figs. 2b and 3b). Also, since most studies in current review were from clinical settings, where clinicopathological variables were available, some important confounding factors (i.e. smoking, alcohol consumption, dietary, etc.) might not be available. For this reason, residual confounding in multivariable analysis models were unavoidable. The other limitations in our review are few prospective cohort studies and lack of comparison groups.
Despite these limitations, our work is one of the most comprehensive systematic reviews on the role of HMGA2 as a universal tumor marker for diagnostic and prognostic across different types of cancer. When used in combination with other markers, its clinical accuracy might increase. We believe that when being used in clinical settings, this marker might help to monitor response to treatment regimens and to guide treatment decision in cancer patients.
Supplementary Material
Acknowledgements
We thank Allison M. Howard (University of South Florida-Shimberg Health Science Library) for performing excellent search for this systematic review.
This work is partially supported by the University of Pittsburgh Medical Center Hillman Cancer Center start-up grant (PI: H.N.L.).
Footnotes
Conflicts of interest
There are no conflicts of interest.
In summary, our systematic review included 42 studies that demonstrated that HMGA2 was overexpressed (i.e. more than 64%) in all 15 types cancers and HMGA2 overexpression was significantly associated with reduced survival. We also found a trend towards association between HMGA2 expression and cancer recurrence, an indication of promising tumor marker for prognostic predictive value. Since prior effort has shown that using HMGA2 in combination with other tumor marker would enhance test accuracy, we believe that HMGA2 would be a promising tumor pronostic marker in the era of precision medicine.
Supplemental Digital Content is available for this article. Direct URL citations appear in the printed text and are provided in the HTML and PDF versions of this article on the journal’s website (www.eurjcancerprev.com).
References
- Altman DG, McShane LM, Sauerbrei W, Taube SE (2012). Reporting Recommendations for Tumor Marker Prognostic Studies (REMARK): explanation and elaboration. Plos Med 9:e1001216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- American Cancer Society (2018). Cancer Facts & Figures 2018. Atlanta, Georgia: American Cancer Society.. [Google Scholar]
- Arora N, Scognamiglio T, Lubitz CC, Moo TA, Kato MA, Zhu B, et al. (2009). Identification of borderline thyroid tumors by gene expression array analysis. Cancer 115:5421–5431. [DOI] [PubMed] [Google Scholar]
- Belge G, Meyer A, Klemke M, Burchardt K, Stern C, Wosniok W, et al. (2008). Upregulation of HMGA2 in thyroid carcinomas: a novel molecular marker to distinguish between benign and malignant follicular neoplasias. Genes Chromosomes Cancer 47:56–63. [DOI] [PubMed] [Google Scholar]
- Bigbee W, Herberman R (2003). Tumor markers and immunodiagnosis. In: Bast RC, Kufe DW, Pollock RE, et al. , editors. Cancer Medicine. Hamilton, ON: BC: Decker, pp. 209–220. [Google Scholar]
- Binabaj MM, Soleimani A, Rahmani F, Avan A, Khazaei M, Fiuji H, et al. (2019). Prognostic value of high mobility group protein A2 (HMGA2) over-expression in cancer progression. Gene 706:131–139. [DOI] [PubMed] [Google Scholar]
- Bittner M, Meltzer P, Chen Y, Jiang Y, Seftor E, Hendrix M, et al. (2000). Molecular classification of cutaneous malignant melanoma by gene expression profiling. Nature 406:536–540. [DOI] [PubMed] [Google Scholar]
- Borrmann L, Schwanbeck R, Heyduk T, Seebeck B, Rogalla P, Bullerdiek J, et al. (2003). High mobility group A2 protein and its derivatives bind a specific region of the promoter of DNA repair gene ERCC1 and modulate its activity. Nucleic Acids Res 31:6841–6851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bossuyt PM, Reitsma JB, Bruns DE, Gatsonis CA, Glasziou PP, Irwig LM, et al. ; Standards for Reporting of Diagnostic Accuracy (2003). Towards complete and accurate reporting of studies of diagnostic accuracy: the STARD initiative. Standards for reporting of diagnostic accuracy. Clin Chem 49:1–6. [DOI] [PubMed] [Google Scholar]
- Breslow NE, Day NE (1987). Chapter 2: rates and rate standardization. In: Breslow NE, Day NE, editors. Statistical Methods in Cancer Research, Vol. II, The Design and Analysis of Cohort Studies. (IARC Scientific Publications No. 82). Lyon, France: International Agency for Research on Cancer-WHO. pp. 48–79. [Google Scholar]
- Califano D, Pignata S, Losito NS, Ottaiano A, Greggi S, De Simone V, et al. (2014). High HMGA2 expression and high body mass index negatively affect the prognosis of patients with ovarian cancer. J Cell Physiol 229:53–59. [DOI] [PubMed] [Google Scholar]
- Chang KP, Lin SJ, Liu SC, Yi JS, Chien KY, Chi LM, et al. (2015). Low-molecular-mass secretome profiling identifies HMGA2 and MIF as prognostic biomarkers for oral cavity squamous cell carcinoma. Sci Rep 5:11689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chiappetta G, Ferraro A, Vuttariello E, Monaco M, Galdiero F, De Simone V, et al. (2008). HMGA2 mRNA expression correlates with the malignant phenotype in human thyroid neoplasias. Eur J Cancer 44:1015–1021. [DOI] [PubMed] [Google Scholar]
- D’Angelo D, Palmieri D, Mussnich P, Roche M, Wierinckx A, Raverot G, et al. (2012). Altered microRNA expression profile in human pituitary GH adenomas: down-regulation of miRNA targeting HMGA1, HMGA2, and E2F1. J Clin Endocrinol Metab 97:E1128–E1138. [DOI] [PubMed] [Google Scholar]
- Dangi-Garimella S, Yun J, Eves EM, Newman M, Erkeland SJ, Hammond SM, et al. (2009). Raf kinase inhibitory protein suppresses a metastasis signalling cascade involving LIN28 and let-7. EMBO J 28:347–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DerSimonian R, Laird N (1986). Meta-analysis in clinical trials. Control Clin Trials 7:177–188. [DOI] [PubMed] [Google Scholar]
- Egger M, Davey Smith G, Schneider M, Minder C (1997). Bias in meta-analysis detected by a simple, graphical test. BMJ 315:629–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fedele M, Visone R, De Martino I, Troncone G, Palmieri D, Battista S, et al. (2006). HMGA2 induces pituitary tumorigenesis by enhancing E2F1 activity. Cancer Cell 9:459–471. [DOI] [PubMed] [Google Scholar]
- Fusco A, Fedele M (2007). Roles of HMGA proteins in cancer. Nat Rev Cancer 7:899–910. [DOI] [PubMed] [Google Scholar]
- Gallo V, Egger M, McCormack V, Farmer PB, Ioannidis JP, Kirsch-Volders M, et al. ; STROBE Statement (2011). STrengthening the reporting of observational studies in Epidemiology–Molecular Epidemiology (STROBE-ME): an extension of the STROBE statement. Plos Med 8:e1001117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giancotti V, Bandiera A, Buratti E, Fusco A, Marzari R, Coles B, Goodwin GH (1991). Comparison of multiple forms of the high mobility group I proteins in rodent and human cells. Identification of the human high mobility group I-C protein. Eur J Biochem 198:211–216. [DOI] [PubMed] [Google Scholar]
- Giancotti V, Pani B, D’Andrea P, Berlingieri MT, Di Fiore PP, Fusco A, et al. (1987). Elevated levels of a specific class of nuclear phosphoproteins in cells transformed with v-ras and v-mos oncogenes and by cotransfection with c-myc and polyoma middle T genes. EMBO J 6:1981–1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilligan TD, Seidenfeld J, Basch EM, Einhorn LH, Fancher T, Smith DC, et al. ; American Society of Clinical Oncology (2010). American Society of Clinical Oncology Clinical Practice Guideline on uses of serum tumor markers in adult males with germ cell tumors. J Clin Oncol 28:3388–3404. [DOI] [PubMed] [Google Scholar]
- Hammond ME, Hayes DF, Wolff AC, Mangu PB, Temin S (2010). American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Oncol Pract 6:195–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Helmke BM, Markowski DN, Meyer A, Bullerdiek J (2012). The expression of HMGA2 varies strongly among colon carcinomas. Anticancer Res 32:1589–1593. [PubMed] [Google Scholar]
- Hetland TE, Holth A, Kærn J, Flørenes VA, Tropé CG, Davidson B (2012). HMGA2 protein expression in ovarian serous carcinoma effusions, primary tumors, and solid metastases. Virchows Arch 460:505–513. [DOI] [PubMed] [Google Scholar]
- Huang ML, Chen CC, Chang LC (2009). Gene expressions of HMGI-C and HMGI(Y) are associated with stage and metastasis in colorectal cancer. Int J Colorectal Dis 24:1281–1286. [DOI] [PubMed] [Google Scholar]
- Jang MH, Jung KC, Min HS (2015). The Diagnostic Usefulness of HMGA2, Survivin, CEACAM6, and SFN/14-3-3 δ in Follicular Thyroid Carcinoma. J Pathol Transl Med 49:112–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin L, Lloyd RV, Henry MR, Erickson LA, Sebo TJ, Rumilla KM, et al. (2015). The diagnostic utility of combination of HMGA2 and IMP3 qRT-PCR testing in thyroid neoplasms. Appl Immunohistochem Mol Morphol 23:36–43. [DOI] [PubMed] [Google Scholar]
- Jin L, Lloyd RV, Nassar A, Lappinga PJ, Sebo TJ, Swartz K, et al. (2011). HMGA2 expression analysis in cytological and paraffin-embedded tissue specimens of thyroid tumors by relative quantitative RT-PCR. Diagn Mol Pathol 20:71–80. [DOI] [PubMed] [Google Scholar]
- Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, et al. (2005). RAS is regulated by the let-7 microRNA family. Cell 120:635–647. [DOI] [PubMed] [Google Scholar]
- Jones AM, Mitter R, Poulsom R, Gillett C, Hanby AM, Tomlinson IP, et al. ; Phyllodes Tumour Consortium (2008). mRNA expression profiling of phyllodes tumours of the breast: identification of genes important in the development of borderline and malignant phyllodes tumours. J Pathol 216:408–417. [DOI] [PubMed] [Google Scholar]
- Jun KH, Jung JH, Choi HJ, Shin EY, Chin HM (2015). HMGA1/HMGA2 protein expression and prognostic implications in gastric cancer. Int J Surg 24:39–44. [DOI] [PubMed] [Google Scholar]
- Keedy VL, Temin S, Somerfield MR, Beasley MB, Johnson DH, McShane LM, et al. (2011). American Society of Clinical Oncology provisional clinical opinion: epidermal growth factor receptor (EGFR) mutation testing for patients with advanced non-small-cell lung cancer considering first-line EGFR tyrosine kinase inhibitor therapy. J Clin Oncol 29:2121–2127. [DOI] [PubMed] [Google Scholar]
- Kim TH, Song JY, Park H, Jeong JY, Kwon AY, Heo JH, et al. (2015). miR-145, targeting high-mobility group A2, is a powerful predictor of patient outcome in ovarian carcinoma. Cancer Lett 356:937–945. [DOI] [PubMed] [Google Scholar]
- Klemke M, Müller MH, Wosniok W, Markowski DN, Nimzyk R, Helmke BM, Bullerdiek J (2014). Correlated expression of HMGA2 and PLAG1 in thyroid tumors, uterine leiomyomas and experimental models. PLoS One 9: e88126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong D, Su G, Zha L, Zhang H, Xiang J, Xu W, et al. (2014). Coexpression of HMGA2 and Oct4 predicts an unfavorable prognosis in human gastric cancer. Med Oncol 31:130. [DOI] [PubMed] [Google Scholar]
- Lappinga PJ, Kip NS, Jin L, Lloyd RV, Henry MR, Zhang J, et al. (2010). HMGA2 gene expression analysis performed on cytologic smears to distinguish benign from malignant thyroid nodules. Cancer Cytopathol 118:287–297. [DOI] [PubMed] [Google Scholar]
- Lee CT, Wu TT, Lohse CM, Zhang L (2014). High-mobility group AT-hook 2: an independent marker of poor prognosis in intrahepatic cholangiocarcinoma. Hum Pathol 45:2334–2340. [DOI] [PubMed] [Google Scholar]
- Lee CT, Zhang L, Mounajjed T, Wu TT (2013). High mobility group AT-hook 2 is overexpressed in hepatoblastoma. Hum Pathol 44:802–810. [DOI] [PubMed] [Google Scholar]
- Lee J, Ha S, Jung CK, Lee HH (2015). High-mobility-group A2 overexpression provokes a poor prognosis of gastric cancer through the epithelial-mesenchymal transition. Int J Oncol 46:2431–2438. [DOI] [PubMed] [Google Scholar]
- Liu B, Pang B, Hou X, Fan H, Liang N, Zheng S, et al. (2014). Expression of high-mobility group AT-hook protein 2 and its prognostic significance in malignant gliomas. Hum Pathol 45:1752–1758. [DOI] [PubMed] [Google Scholar]
- Liu Q, Liu T, Zheng S, Gao X, Lu M, Sheyhidin I, et al. (2014). HMGA2 is down-regulated by microRNA let-7 and associated with epithelial-mesenchymal transition in oesophageal squamous cell carcinomas of Kazakhs. Histopathology 65:408–417. [DOI] [PubMed] [Google Scholar]
- Liu Z, Wu K, Yang Z, Wu A (2015). High-mobility group A2 overexpression is an unfavorable prognostic biomarker for nasopharyngeal carcinoma patients. Mol Cell Biochem 409:155–162. [DOI] [PubMed] [Google Scholar]
- Locker GY, Hamilton S, Harris J, Jessup JM, Kemeny N, Macdonald JS, et al. ; ASCO (2006). ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Oncol 24:5313–5327. [DOI] [PubMed] [Google Scholar]
- Mahajan A, Liu Z, Gellert L, Zou X, Yang G, Lee P, et al. (2010). HMGA2: a biomarker significantly overexpressed in high-grade ovarian serous carcinoma. Mod Pathol 23:673–681. [DOI] [PubMed] [Google Scholar]
- Malek A, Bakhidze E, Noske A, Sers C, Aigner A, Schäfer R, Tchernitsa O (2008). HMGA2 gene is a promising target for ovarian cancer silencing therapy. Int J Cancer 123:348–356. [DOI] [PubMed] [Google Scholar]
- Meyer B, Loeschke S, Schultze A, Weigel T, Sandkamp M, Goldmann T, et al. (2007). HMGA2 overexpression in non-small cell lung cancer. Mol Carcinog 46:503–511. [DOI] [PubMed] [Google Scholar]
- Miyazawa J, Mitoro A, Kawashiri S, Chada KK, Imai K (2004). Expression of mesenchyme-specific gene HMGA2 in squamous cell carcinomas of the oral cavity. Cancer Res 64:2024–2029. [DOI] [PubMed] [Google Scholar]
- Motoyama K, Inoue H, Nakamura Y, Uetake H, Sugihara K, Mori M (2008). Clinical significance of high mobility group A2 in human gastric cancer and its relationship to let-7 microRNA family. Clin Cancer Res 14:2334–2340. [DOI] [PubMed] [Google Scholar]
- Müller DW, Bosserhoff AK (2008). Integrin beta 3 expression is regulated by let-7a miRNA in malignant melanoma. Oncogene 27:6698–6706. [DOI] [PubMed] [Google Scholar]
- Nagar S, Ahmed S, Peeples C, Urban N, Boura J, Thibodeau B, et al. (2014). Evaluation of genetic biomarkers for distinguishing benign from malignant thyroid neoplasms. Am J Surg 207:596–601. [DOI] [PubMed] [Google Scholar]
- Nirodi C, NagDas S, Gygi SP, Olson G, Aebersold R, Richmond A (2001). A role for poly(ADP-ribose) polymerase in the transcriptional regulation of the melanoma growth stimulatory activity (CXCL1) gene expression. J Biol Chem 276:9366–9374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Noro B, Licheri B, Sgarra R, Rustighi A, Tessari MA, Chau KY, et al. (2003). Molecular dissection of the architectural transcription factor HMGA2. Biochemistry 42:4569–4577. [DOI] [PubMed] [Google Scholar]
- Pagano M, Pepperkok R, Verde F, Ansorge W, Draetta G (1992). Cyclin A is required at two points in the human cell cycle. EMBO J 11:961–971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pallante P, Sepe R, Puca F, Fusco A (2015). High mobility group a proteins as tumor markers. Front Med (Lausanne) 2:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palmieri D, D’Angelo D, Valentino T, De Martino I, Ferraro A, Wierinckx A, et al. (2012). Downregulation of HMGA-targeting microRNAs has a critical role in human pituitary tumorigenesis. Oncogene 31:3857–3865. [DOI] [PubMed] [Google Scholar]
- Peng Y, Laser J, Shi G, Mittal K, Melamed J, Lee P, et al. (2008). Antiproliferative effects by Let-7 repression of high-mobility group A2 in uterine leiomyoma. Mol Cancer Res 6:663–673. [DOI] [PubMed] [Google Scholar]
- Piscuoglio S, Zlobec I, Pallante P, Sepe R, Esposito F, Zimmermann A, et al. (2012). HMGA1 and HMGA2 protein expression correlates with advanced tumour grade and lymph node metastasis in pancreatic adenocarcinoma. Histopathology 60:397–404. [DOI] [PubMed] [Google Scholar]
- Prasad NB, Kowalski J, Tsai HL, Talbot K, Somervell H, Kouniavsky G, et al. (2012). Three-gene molecular diagnostic model for thyroid cancer. Thyroid 22:275–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prasad NB, Somervell H, Tufano RP, Dackiw AP, Marohn MR, Califano JA, et al. (2008). Identification of genes differentially expressed in benign versus malignant thyroid tumors. Clin Cancer Res 14:3327–3337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramakrishna N, Temin S, Chandarlapaty S, Crews JR, Davidson NE, Esteva FJ, et al. (2018). Recommendations on disease management for patients with advanced human epidermal growth factor receptor 2-positive breast cancer and brain metastases: ASCO clinical practice guideline update. J Clin Oncol 36:2804–2807. [DOI] [PubMed] [Google Scholar]
- Raskin L, Fullen DR, Giordano TJ, Thomas DG, Frohm ML, Cha KB, et al. (2013). Transcriptome profiling identifies HMGA2 as a biomarker of melanoma progression and prognosis. J Invest Dermatol 133:2585–2592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reeves R (2010). Nuclear functions of the HMG proteins. Biochim Biophys Acta 1799:3–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rizzi C, Cataldi P, Iop A, Isola M, Sgarra R, Manfioletti G, Giancotti V (2013). The expression of the high-mobility group A2 protein in colorectal cancer and surrounding fibroblasts is linked to tumor invasiveness. Hum Pathol 44:122–132. [DOI] [PubMed] [Google Scholar]
- Rogalla P, Drechsler K, Kazmierczak B, Rippe V, Bonk U, Bullerdiek J (1997). Expression of HMGI-C, a member of the high mobility group protein family, in a subset of breast cancers: relationship to histologic grade. Mol Carcinog 19:153–156. [DOI] [PubMed] [Google Scholar]
- Sarhadi VK, Wikman H, Salmenkivi K, Kuosma E, Sioris T, Salo J, et al. (2006). Increased expression of high mobility group A proteins in lung cancer. J Pathol 209:206–212. [DOI] [PubMed] [Google Scholar]
- Schultz J, Lorenz P, Gross G, Ibrahi S, Kunz M (2008). MicroRNA let-7b targets important cell cycle molecules in malignant melanoma cells and interferes with anchorage-independent growth. Cell Res 18:549–557. [DOI] [PubMed] [Google Scholar]
- Sepulveda AR, Hamilton SR, Allegra CJ, Grody W, Cushman-Vokoun AM, Funkhouser WK, et al. (2017). Molecular biomarkers for the evaluation of colorectal cancer: guideline from the American Society for Clinical Pathology, College of American Pathologists, Association for Molecular Pathology, and the American Society of Clinical Oncology. J Clin Oncol 35: 1453–1486. [DOI] [PubMed] [Google Scholar]
- Siegel R, DeSantis C, Virgo K, Stein K, Mariotto A, Smith T, et al. (2012). Cancer treatment and survivorship statistics, 2012. CA Cancer J Clin 62:220–241. [DOI] [PubMed] [Google Scholar]
- Sturgeon C, Diamandis E (2008). Laboratory Medicine Practice Guidelines: Use of Tumor Markers in Clinical Practice-Quality Requirements. Washington, DC: National Academic of Clinical Biochemistry. [Google Scholar]
- Wang X, Liu X, Li AY, Chen L, Lai L, Lin HH, et al. (2011). Overexpression of HMGA2 promotes metastasis and impacts survival of colorectal cancers. Clin Cancer Res 17:2570–2580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu J, Lai M, Shao C, Wang J, Wei JJ (2015). STC2 overexpression mediated by HMGA2 is a biomarker for aggressiveness of high-grade serous ovarian cancer. Oncol Rep 34:1494–1502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu L, Wang Z, Lu R, Jiang W (2012). Expression of high mobility group A2 is associated with poor survival in hepatocellular carcinoma. Pathol Oncol Res 18:983–987. [DOI] [PubMed] [Google Scholar]
- Xia YY, Yin L, Tian H, Guo WJ, Jiang N, Jiang XS, et al. (2015). HMGA2 is associated with epithelial-mesenchymal transition and can predict poor prognosis in nasopharyngeal carcinoma. Onco Targets Ther 8:169–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang GL, Zhang LH, Bo JJ, Hou KL, Cai X, Chen YY, et al. (2011). Overexpression of HMGA2 in bladder cancer and its association with clinicopathologic features and prognosis HMGA2 as a prognostic marker of bladder cancer. Eur J Surg Oncol 37:265–271. [DOI] [PubMed] [Google Scholar]
- Zakharov V, Ren B, Ryan C, Cao W (2013). Diagnostic value of HMGAs, p53 and β-catenin in discriminating adenocarcinoma from adenoma or reactive atypia in ampulla and common bile duct biopsies. Histopathology 62:778–787. [DOI] [PubMed] [Google Scholar]
- Zou Q, Xiong L, Yang Z, Lv F, Yang L, Miao X (2012). Expression levels of HMGA2 and CD9 and its clinicopathological significances in the benign and malignant lesions of the gallbladder. World J Surg Oncol 10:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.