PODXL Might Be a New Prognostic Biomarker in Various Cancers: A Meta-analysis and Sequential Verification with TCGA Datasets

DOI: https://doi.org/10.21203/rs.3.rs-25453/v1

Abstract

Background Several studies have investigated the associations between the podocalyxin-like protein (PODXL) expression quantity or locations and cancers survival, but the results were far from conclusive. Therefore, we proceeded a meta-analysis on PODXL in various human cancers to find its prognostic value and followed confirmation using the TCGA datasets.

Methods We performed a systematic search, and 18 citations, including 5705 patients were pooled in meta-analysis. The results were verified with TCGA datasets.

Results Total eligible studies comprised 5705 patients with 10 types of cancer. And the result indicated that PODXL high-expression or membrane-expression were significantly related to poor overall survival (OS). However, subgroup analysis showed a significant association between high expressed PODXL and poor OS in the colorectal cancer, pancreatic cancer, urothelial bladder cancer, renal cell carcinoma and glioblastoma multiforme. Then, we validated the inference using TCGA datasets, and the consistent results were demonstrated in patients with pancreatic cancer, glioblastoma multiforme, gastric cancer, esophageal cancer and lung adenocarcinoma.

Conclusion The result of meta-analysis showed that high expressed PODXL was significantly linked with poor OS in pancreatic cancer and glioblastoma multiforme, but not in gastric cancer, esophageal cancer or lung adenocarcinoma. And the membrane expression of PODXL might also associate with poor OS. PODXL may act as tumor promotor and may serve as a potential target for antitumor therapy. 

Background

Nowadays, noncommunicable diseases (NCDs) account for the majority of global deaths, and cancer predicts to be the leading cause. According to the latest global cancer statistics, 18.1 million new cancer diagnoses and 9.6 million deaths are expected in 2018 [1].

Podocalyxin-like protein (PODXL) is a highly glycosylated type I transmembrane protein associated with CD34 [24]. PODXL expression has been reported in the cytoplasm of some tumor cells, in some cases protruding toward the cell membrane, but not in the nucleus [5]. PODXL is encoded on chromosome 7q32-q33, and highly expressed by glomerular podocytes, vascular endothelium, hematopoietic cells and breast epithelial cells[68], which involved in many physiologic processes, such as hematopoiesis [9], leucocyte-endothelial cell interaction [10], regulating vascular permeability [11]and neural development [12].

The clinical significance of PODXL in the progression of various cancers has been studied, and it was found as a stem cell marker in the testicular cancer at the first time [3]. The later findings proved that, PODXL associates with advanced tumor phenotype in some cancers, including breast cancer [13, 1], colorectal cancer [5, 1416], esophageal cancer [17], gastric cancer [18, 19, 17], glioblastoma multiforme [20], lung adenocarcinoma [21], oral squamous cell carcinoma [4, 22], ovarian cancer [23], pancreatic cancer [2427], prostate cancer [28, 29], renal cell carcinoma [30], urothelial bladder cancer [31], and so on. We performed a meta-analysis at pooling data, in order to estimate the potential prognostic value of PODXL. Finally, the sequential verification were performed with the Cancer Genome Atlas (TCGA, http://cancergenome.nih.gov ) datasets.

Methods

Publication Search

Our meta-analysis followed the guidance of the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) [32]. We performed a systematic search of the PubMed, Web of Science, Embase and Cochrane Library database from January 1, 2000 to October 31, 2018, using both MeSH search for keywords and full text. Our search terms were: (“cancer” OR “tumor” OR “neoplasm” OR “carcinoma”) AND (“Podocalyxin like protein” OR “Podocalyxin” OR “PODXL”) AND (“prognosis” OR “prognostic” OR “outcome”). Additionally, the references and other related researches were reviewed to find more potential articles.

Inclusion and Exclusion Criteria

The eligible articles selection process was done by two authors (Siying He and Menglan Li).The inclusion criteria were as followed: (1) involved the correlation between the expression of PODXL and survival data of cancer patients; (2) provided the relevant clinicopathological parameters; (3) the number of patients involved in the studies should be more than 50.

The exclusion criteria were as followed: (1) studies that not based on human; (2) insufficient Hazard ratios (HRs) or other data; (3) repetitive patients; (4) reviews, case reports or a meta-analysis.

Data Collection and Quality Detection

Two researchers evaluated and collected data from these eligible articles with a predefined standard independently. The following information was recorded: (1) first author’s name; (2) publication year; (3) countries; (4) types of cancers; (5) number of patients; (6) detection methods; (7) cut-off criteria; (8) clinical parameters; (9) data about overall survival (OS), disease-free survival (DFS) or cancer-specific survival (CSS). The Engauge Digitizer 4.1 software was used to extract data from Kaplan-Meier (K-M) plot, when there was no HRs and its 95% confidence inter(CI) offered directly [33]. In addition, the included studies should be evaluated with the Newcastle-Ottawa Scale (NOS) [34].

Data Collection and Analysis in TCGA

Data for the expression of PODXL and clinicopathological parameters in TCGA were recorded from the Gene Expression Profiling Interactive Analysis (GEPIA, http://gepia.cancer-pku.cn ) [35] and the UALCAN (http://ualcan.path.uab.edu ) [36]. There were 31 types of cancer, including 9040 subjects which had both PODXL expression and cancer survival data. In order to make the K-M survival analysis and generated overall survival plots, the expression levels of PODXL were divided into low/median and high expression group according to the TPM value. The difference between two groups was conducted by Log-rank test.

Statistical Analysis

Our meta-analysis was based on the Stata12.0 software (Stata Corporation, College Station, TX, United States). The prognostic value of PODXL on OS, DFS and CSS was calculated by pooled HRs with 95% CIs. On the other hand, odds ratios (ORs) with corresponding 95% CIs were used to assess the relation between PODXL and clinicopathological features. Chi square-based Cochran Q test and I2 test were used to determine the heterogeneity among these eligible articles. I2 > 50% or P-value < 0.05 was considered as significant heterogeneity, and a random-effect model would be adopted; otherwise, a fix-effect model would be chose. The sources of heterogeneity could be dissect with subgroup analysis. In addition, the sensitivity and publication bias were performed. P < 0.05 was considered statistically significant with two-sided.

Results

Search results and research characteristics

In total, 436 records were identified and 87 duplicates were excluded. 39 articles remained after scanning the titles and abstracts, and among the 39 studies, 7 were excluded for not for human, 9 were excluded for insufficient HRs or other data, 3 were excluded because the included patients were repetitive in other studies, and 1 meta-analysis was excluded, and the flow diagram was shown in Fig. 1. Finally, 18 eligible studies were include in this meta-analysis. [5, 37, 1416, 2427, 18, 17, 38, 13, 30, 23, 20, 31, 21, 1, 19]. These eligible researches contained 5705 patients, involved 10 types of cancers, including the breast cancer (n = 2), renal cell carcinoma (n = 1), colorectal cancer (n = 4), ovarian cancer (n = 1), glioblastoma multiforme (n = 1), urothelial bladder cancer (n = 2), pancreatic adenocarcinoma (n = 4), esophageal cancer (n = 1), gastric cancer (n = 3) and lung adenocarcinoma (n = 1). In these studies, PODXL expression levels were evaluated by immunohistochemistry (IHC). The characteristics of the eligible articles were listed in Table 1.

Table 1

Characteristics of eligible studies in this meta-analysis.

Author

Year

Country

No. of Patient

Tumor type

Method

Cut-off

Outcome

Analysis

Antibody

NOS

Somasiri

2004

Canada

272

Breast cancer

IHC

IHC ≥ 50%

CSS

K-M Curve

M

7

Hsu

2010

Taiwan

303

Renal cell carcinoma

IHC

IHC score ≥ 1

OS, CSS, MFS

Multivariate

P

8

Larsson

2011

Sweden

626

Colorectal cancer

IHC

IHC score ≥ 3

OS, CSS

Multivariate

P

8

Cipollone

2012

Canada

479

Ovarian cancer

IHC

IHC score ≥ 1

DFS

K-M Curve

M

8

Larsson

2012

Sweden

607

Colorectal cancer

IHC

IHC score ≥ 3

OS, DFS, TTR

Multivariate

P

9

Binder

2013

America

181

Glioblastoma multiforme

IHC

NA

OS

Multivariate

NA

7

Boman

2013

Sweden

100

Urothelial bladder cancer

IHC

IHC score ≥ 3

OS

Multivariate

M/P

7

Boman

2013

Sweden

343

Urothelial bladder cancer

IHC

IHC score ≥ 3

OS, CSS, PFS

Multivariate

M/P

8

Forse

2013

Canada

698

Breast cancer

IHC

IHC score ≥ 3

DFS

Multivariate

P

9

Kaprio

2014

Finland

840

Colorectal cancer

IHC

IHC score ≥ 3

CSS

K-M Curve

M/P

9

Heby

2015

Sweden

175

Pancreatic and periampullary adenocarcinoma

IHC

IHC score ≥ 2

OS, DFS

Multivariate

P

7

Laitinen

2015

Finland

337

Gastric cancer

IHC

IHC score ≥ 1

CSS

Multivariate

M/P

8

Saukkonen

2015

Finland

189

Pancreatic ductal adenocarcinoma

IHC

IHC score ≥ 3

CSS

Multivariate

M/P

7

Borg

2016

Sweden

106

Esophageal cancer

IHC

IHC score ≥ 1

OS, TTR

K-M Curve

P

7

Borg

2016

Sweden

65

Gastric cancer

IHC

IHC score ≥ 1

OS, TTR

K-M Curve

p

7

Chijiiwa

2016

Japan

70

Pancreatic cancer

IHC

IHC score ≥ 4

OS, DFS

K-M Curve

M

7

Taniuchi

2016

Japan

102

Pancreatic cancer

IHC

IHC score ≥ 3

OS

Multivariate

P

7

Kusumoto

2017

Japan

114

Lung adenocarcinoma

IHC

IHC score ≥ 1

OS, DFS, CSS

K-M Curve

NA

8

Yuan

2018

China

87

Colorectal cancer

IHC

IHC score ≥ 3

OS

Multivariate

M

7

Zhang

2018

China

54

Gastric cancer

IHC

IHC score ≥ 1

OS, DFS

Multivariate

NA

7

IHC, Immunohistochemistry; NA, Not Available; OS, Overall Survival; DFS, Disease-free Survival; CSS, Cancer-specific Survival; NOS, Newcastle-Ottawa Scale

Meta-analysis of PODXL expression levels and locations on OS/ DFS/ CSS

A total of 11 eligible studies, including 13 cohorts and 2272 patients, were recruited to evaluate the expression level of PODXL on OS. The pooled HR and 95% CI indicated that high-expressed PODXL was significantly related to poor OS in patients with various cancers (HR = 2.33, 95% CI = 1.76–3.09, P < 0.0001) with a significant heterogeneity across these studies (I2 = 63.4%, P = 0.001) (Fig. 2a). In addition, there were 6 studies performed the relationships between PODXL expression levels and DFS, and 8 studies investigated the associations between PODXL expression levels and CSS respectively. Heterogeneity test indicated both the DFS (I2 = 73.4%, P = 0.002) and CSS (I2 = 70.0%, P = 0.002) should be analyzed using the random-effect model. Finally, the results indicated the association between the high expressed PODXL and the shorter DFS (HR = 1.76, 95% CI = 1.20–2.58, P = 0.004) or the shorter CSS (HR = 2.84, 95% CI = 1.85–4.38, P < 0.0001) (Fig. 2b-2c). On the other hand, among these eligible 18 papers, 5 studies involved the expression locations of PODXL and the prognosis of cancers, and only 2 studies, including 4 cohorts, showed the association between membrane expressed PODXL and poor OS (HR = 2.98, 95% CI = 1.29–6.90, P = 0.011), also by using the random-effect model (I2 = 84.7%, P < 0.0001) (Fig. 2d).

Subgroup analysis for OS

In order to find the source of heterogeneity, the subgroup analysis of OS was performed, and all of the 2272 patients were classified based on cancer types, analysis types, antibody types, ethnicities and sample sizes (Table 2). Single study which assessed the relationship between the expression and OS in renal cell carcinoma, glioblastoma multiforme, esophageal cancers and lung adenocarcinoma were defined as “other cancers” in the other cancers subgroup. Subgroup analysis showed that, high expressed PODXL were linked with poor OS in colorectal cancer (HR = 1.79, 95% CI = 1.35–2.37, P < 0.0001), pancreatic cancer (HR = 2.98, 95% CI = 1.95–4.55, P < 0.0001), urothelial bladder cancer (HR = 2.14, 95% CI = 1.48–3.10) and other cancers (HR = 2.60, 95% CI = 1.45–4.66, P = 0.001), but not in patients with the gastric cancer (HR = 2.76, 95% CI = 0.45–15.84, P = 0.256). In conclusion, high expressed level of PODXL was associated with poor OS in 6 types of cancers.

Table 2

Subgroup analysis of pooled HR for OS

Categories

No. of studies

No. of patients

Pooled HR (95%CI)

Heterogeneity

Fix/Random

P-value

I2 (%)

P-value

1. OS

13

2272

2.33 (1.76, 3.09)

0

63.4

0.001

2. Cancer type

8

         

1) Colorectal cancer

3

834

1.79 (1.35, 2.37)

0

0

0.499

2) Pancreatic cancer

2

172

2.98 (1.95, 4.55)

0

0

0.391

3) Gastric cancer

2

119

2.76 (0.48, 15.84)

0.256

59.9

0.114

4) Urothelial bladder cancer

2

443

2.14 (1.48, 3.10)

0

0

0.880

5) Other cancers

4

704

2.60 (1.45, 4.66)

0.001

83.3

0

3. Analysis

           

1) K-M curve

4

355

1.85 (1.17, 2.95)

0.009

0

0.89

2) Multivariate

9

2017

2.59 (1.77, 3.80)

0

74.7

0

4. Antibody type

           

1) Monoclonal antibody

2

157

2.25 (1.36, 3.73)

0.002

0

0.975

2) Polyclonal antibody

6

1672

2.55 (1.45, 4.50)

0.001

0

81.6

3) M + P

2

443

2.14 (1.48, 3.10)

0

0

0.880

5. Ethnicity

           

1) European

6

1361

1.84 (1.47, 2.30)

0

0

0.834

2) Asian

6

730

3.49 (2.02, 6.02)

0

64.5

0.015

3) North American

1

181

1.67 (1.23, 2.29)

 

-

-

6. Sample size

           

1) < 150

8

628

2.46 (1.81, 3.33)

0

0

0.536

4) ≥ 150

5

1644

2.36 (1.53, 3.65)

0

81.3

0

OS, overall survival; HR, hazard ratio.

And regarding the analysis type, we also found that the high expression of PODXL was significantly associated with the much shorter OS, when the studies were assessed with K-M curve. In the subgroups based on ethnicities, antibody types and sample sizes, we also found that, the relation between high expression level of PODXL and poor OS, except for patients from Asia or the sample size ≥ 150.

PODXL overexpression and relative clinical parameters

In order to obtain more clinical values of PODXL, we investigated the associations between PODXL expression levels and clinical parameters in several cancers (Table 3). From these results, we found that the expression level of PODXL was related with the TNM stage (HR = 1.63, 95% CI = 1.19–2.23, P = 0.002, fixed-effects), tumor grade (HR = 4.29, 95% CI = 1.84–9.99, P = 0.001, random-effects), differentiation (HR = 2.84, 95% CI = 1.82–4.42, P < 0.0001, fixed-effects), distant metastasis (HR = 5.46, 95% CI = 2.55–11.66, P < 0.0001, fixed-effects), lymph node metastasis (HR = 1.51, 95% CI = 1.03–2.22, P = 0.034, fixed-effects), neural invasion (HR = 2.43, 95% CI = 1.02–5.79, P = 0 .45, fixed-effects) and vascular invasion (HR = 2.27, 95% CI = 1.56–3.30, P < 0.0001, fixed-effects) significantly. Whereas, there was no significant correlations between PODXL expression and age (HR = 0.88, 95% CI = 0.71–1.10, P = 0.269, fixed-effects), gender (HR = 1.04, 95% CI = 0.82–1.32, P = 0.749, fix-effects) and tumor size (HR = 0.90, 95% CI = 0.61–1.34, P = 0.614, fixed-effects). As a result, these correlations indicated that the high expressed PODXL was associated with the advanced biological behavior in various cancers.

Table 3

Clinicopathological features of the enrolled studies with high expressed PODXL in patients with cancer.

Clinicopathological parameters

Studies

No. of patients

Risk of high PODXL

OR (95% CI)

Significant

Z

P-value

Heterogeneity

I2 (%)

P-value

Model

Age (< 65 vs ≥ 65)

10

2905

0.88 (0.71, 1.10)

1.11

0.269

42.6

0.084

Fixed effects

Gender (male vs female)

11

3081

1.04 (0.82, 1.32)

0.32

0.749

0

0.835

Fixed effects

Tumor size (< 5 cm vs ≥ 5 cm)

5

1334

0.90 (0.61, 1.34)

0.50

0.614

0

0.703

Fixed effects

TNM stage (III-IV vs I-II)

12

2417

1.63 (1.19, 2.23)

3.04

0.002

13.1

0.319

Fixed effects

Tumor grade (3–4 vs 1–2)

6

2268

4.29 (1.84, 9.99)

3.38

0.001

78.6

0

Random effects

Tumor differentiation

(moderate/well vs poor)

6

1429

2.84 (1.82, 4.42)

4.62

0

0

0.559

Fixed effects

Distant metastasis

(positive vs Negative)

3

475

5.46 (2.55, 11.66)

4.38

0

44.5

0.165

Fixed effects

Lymph node metastasis

(positive vs negative)

6

1574

1.51 (1.03, 2.22)

2.11

0.034

0

0.614

Fixed effects

Neural invasion

(positive vs negative)

3

264

2.43 (1.02, 5.79)

2.00

0.045

0

1.000

Fixed effects

Vascular invasion

(positive or negative)

6

1240

2.27 (1.56, 3.30)

4.29

0

2.1

0.403

Fixed effects

Table 4

The difference of PODXL expression in cancers and corresponding normal tissues in TCGA datasets.

Types of cancer

TCGA dataset

No. of cancer tissues

No. of normal tissues

Log2(FC)

P value

Adenoid cystic carcinoma

ACC

77

128

-1.068

1.10e-10

Breast invasion carcinoma

BRCA

1085

291

-0.514

4.01e-16

Cervical squamous cell carcinoma

CESC

306

13

-0.590

0.191

Esophagus cancer

ESCA

182

286

1.391

5.97e-22

Glioblastoma multiforme

GBM

163

207

0.866

1.12e-10

Head and neck squamous cell carcinoma

HNSCC

519

44

0.656

0.123

Kidney chromophobe

KICH

66

53

-2.863

1.53e-10

Kidney renal clear cell carcinoma

KIRC

523

100

-0.732

1.85e-9

Kidney renal papillary cell carcinoma

KIRP

286

60

-4.247

1.31e-43

Acute myeloid leukemia

LAML

173

70

1.210

3.26e-2

Liver hepatocellular carcinoma

LIHC

369

160

1.508

8.20e-32

Lung adenocarcinoma

LUAD

483

347

-2.064

6.27e-122

Lung squamous cell carcinoma

LUSC

486

338

-2.832

3.86e-153

Ovarian serous cystadenocarcinoma

OVSC

426

88

1.449

3.96e-14

Pancreatic adenocarcinoma

PAAD

179

171

0.492

4.05e-5

Prostate carcinoma

PRAD

492

152

-0.479

0.044

Rectum adenocarcinoma

READ

92

318

0.598

8.17e-8

Skin cutaneous melanoma

SKCM

461

558

-0.636

3.83e-6

Stomach adenocarcinoma

STAD

408

211

1.597

1.64e-49

Testicular germ cell tumor

TGCT

137

165

2.750

3.93e-30

Thyroid carcinoma

THCA

512

337

-0.796

6.95e-22

Uterine corpus endometrial carcinoma

UCEC

174

91

-0.797

2.97e-5

Uterine carcinosarcoma

UCS

58

78

-2.075

3.79e-12

Table 5

The difference of overall survival in cancer patients with high PODXL expression vs low/median expression.

Cancer type

No. of cancer tissues

P value

High

Low/Median

Total

ACC

20

59

79

0.37

BLCA

102

304

406

0.34

BRCA

272

809

1081

0.4

CESC

73

218

291

0.77

CHOL

9

27

36

0.57

COAD

69

210

279

0.32

ESCA

46

138

184

0.16

GBM

39

113

152

0.041

HNSCC

130

389

519

0.3

KICH

15

49

64

0.35

KIRC

134

397

531

< 0.0001

KIRP

72

215

287

0.0037

LAML

43

120

163

0.64

LIHC

93

272

365

0.82

LUAD

125

377

502

0.37

LUSC

126

368

494

0.33

DLBC

12

35

47

0.21

MESO

22

63

85

0.23

OVSC

76

227

303

0.95

PAAD

45

132

177

0.013

PCPG

45

134

179

0.13

PRAD

125

372

497

0.92

READ

42

123

165

0.44

SARC

65

194

259

0.12

SKCM

115

344

459

0.22

TGCT

34

100

134

0.29

THYM

30

89

119

0.78

THCA

127

377

504

0.87

UCS

15

41

56

0.58

UCEC

136

407

543

0.006

UVM

20

60

80

0.36

ACC, adrenocortical carcinoma; BLCA, bladder urothelial carcinoma; BRCA, breast invasion carcinoma; CESE, cervical squamous cell carcinoma; CHOL, cholangiocarcinoma; COAD, colon adenocarcinoma; ESCA, esophageal carcinoma; GBM, glioblastoma multiforme; HNSCC, head and neck squamous cell carcinoma; KICH, kidney chromophobe; KIRC, kidney renal clear cell carcinoma; KIRP, kidney renal papillary cell carcinoma; LAML, acute myeloid leukemia; LIHC, liver hepatocellular carcinoma; LUAD, lung adenocarcinoma; LUSC, lung squamous cell carcinoma; DLBC, lymphoid neoplasm diffuse large B-cell lymphoma; MESO, mesothelioma; OVSC, ovarian serous cystadenocarcinoma; PAAD, pancreatic adenocarcinoma; PCPG, pheochromocytoma and paraganglioma; PRAD, prostate adenocarcinoma; READ, rectum adenocarcinoma; SARC, sarcoma; SKCM, skin cutaneous melanoma; STAD, stomach adenocarcinoma; TGCT, testicular germ cell tumors; THYM, thymoma; THCA, thyroid carcinoma; UCS, uterine carcinosarcoma; UCEC, uterine corpus endometrial carcinoma; UVM, uveal melanoma.

Sensitivity analysis and publication bias

We performed sensitivity analysis to determine whether an individual study could affected the overall result. Results of association studies between PODXL expression and OS and CSS demonstrated that single study had no influence on the result of meta-analysis (Fig. 3). Funnel plots and Begg’s test were performed and the results showed no publication bias existed in studies on associations between PODXL overexpression and OS (P = 0.502), DFS (P = 0.133) and CSS (P = 0.266). And no publication bias existed in our meta-analysis on associations between PODXL membrane expression and OS (P = 1.000) as well (Fig. 4).

The expression Data of PODXL Extracted from TCGA Datasets

The differences of PODXL expression level between various tumor tissues and corresponding normal tissues were obtained with GEPIA, which was a common web-based tool that can provide a quick and customizable survey of function based on TCGA and GTEx data [35]. PODXL was detected in 23 types of cancers, and the result that the PODXL expression was significantly much higher than the corresponding normal tissues was found in 9 types of cancers, including the esophagus cancer, glioblastoma multiforme, acute myeloid leukemia, liver hepatocellular carcinoma, ovarian serous cystadenocarcinoma, pancreatic adenocarcinoma, rectum adenocarcinoma, stomach adenocarcinoma, testicular germ cell tumor.

Validation of prognostic correlation by TCGA datasets

To validate the clinical prognosis indication value of PODXL, we explored TCGA datasets by using UALCAN, which was an interactive online tool that could analyze the expression data of genes in TCGA [36]. And among the 31 types of cancers, 9040 patients, the significant association between high expressed PODXL and poor OS was found in 3 types of cancers, including the glioblastoma multiforme, kidney renal papillary cell carcinoma and pancreatic adenocarcinoma. But there were adverse results in kidney renal clear cell carcinoma and uterine corpus endometrial carcinoma, which showed a significant correlation between the low expressed PODXL and poor OS (Fig. 5).

A joint result of our meta-analysis and TCGA datasets validation identified the correlation between the expression level of PODXL and the glioblastoma multiforme, pancreatic adenocarcinoma, esophagus cancer, gastric cancer and lung adenocarcinoma.

Discussion

Recently, increasing evidences have suggested that PODXL was involved in multiple links in several processes of tumor development, such as cell adhesion and morphology [39], lymphatic metastasis [40], tumor cells motility and invasiveness [26], tumor angiogenesis [41] and prognosis. Recent researches indicated that the expression level and location of PODXL could be a new biomarker to assess the prognosis of various types of cancers. However, a single study is limited by insufficient data and single experimental model, so that a meta-analysis of pooling studies is necessary to explore the potential clinical value of PODXL.

So far as we know, the meta-analysis was the first one to analyze and validate the prognostic value of PODXL in various cancers from both the published studies and sequential verification using TCGA datasets. Among these published studies, there were 10 types of cancers, including 5705 patients. Our meta-analysis not only indicated that high expressed PODXL was associated with poor OS, DFS or CSS in patients with cancers, but also showed that membrane expression was correlated with poor OS as well. Clinicopathological features analysis showed that the overexpressed PODXL was linked with poor stage and differentiation, and high incidences of metastasis and invasion in cancers, which indicated that there might be a significant association between PODXL expression level and advanced features of cancer. Subgroup analysis showed that the association between overexpressed PODXL and poor OS in patients with cancers, was only significative in the glioblastoma multiforme, pancreatic cancer, renal cell carcinoma, colorectal cancer and urothelial bladder cancer, but not in the esophageal cancer, gastric cancer and lung adenocarcinoma. Then we used GEPIA and UALCAN to explore TCGA datasets, to compare the expression difference of PODXL among tumor tissues and correlated normal tissues, and the survival curves. Consistent results of meta-analysis and TCGA datasets validation were found in 5 types of cancers.

Among the eligible 18 studies, there were only 2 researches mentioned the expression location of PODXL and prognosis of cancers, containing 4 cohorts. The studies showed a significant association between membrane expression of PODXL and poor OS, but the sensitivity analysis showed that this result is not credible. On the premise of appropriate number of included studies, samples that may introduce heterogeneity are moved, but the sensitivity is still high, so this result can only be used as a descriptive hypothesis, and need more included studies.

Although, PODXL has been found to be highly expressed in various malignancies and was related to a more aggressive phenotype and poor prognosis, the exact mechanisms of which role did PODXL play in tumorigenesis remains unclear [42]. Some latest studies showed that PODXL promoted the gelsolin-actin interaction in cell protrusions to enhance the motility and invasiveness [26], and some showed that the PODXL-ezrin signaling axis could rearrange the dynamic cytoskeleton for transendothelial migration [43]. PODXL also took part in the NF-kB, PI3K/AKT, Hippo and MAPK/ERK signaling pathway, and facilitated tumor progression by increasing cell proliferation, migration and invasion as well as suppressing apoptosis [21, 44, 45].

PODXL was expected to be a novel therapeutic and monitoring biomarker in certain cancers, because the high expressed PODXL might be a potential indicator of poor prognosis of cancers. ATF3 could activate PODXL transcription, which suggested that ATF3 pathway might be beneficial for anticancer therapy [46]. High expression of miR-509-3-5p and miR-5100 inhibited the invasion and metastasis of gastric cancers and pancreatic cancers by directly targeting PODXL, functioning as a tumor suppressor [27, 40]. A core fucose-deficient monoclonal antibody (mAb) of PODXL might be a new antibody-based therapy method against PODXL high-expressed oral squamous cell carcinoma [47]. And patients with gastric or esophageal adenocarcinoma would have a much better prognosis after treating with neoadjuvant ± adjuvant fluoropyrimidine– and oxaliplantin-based chemotherapy, if the expression level of PODXL is high [48].

However, there are still some limitations. First of all, many unavoidable reasons, such as different types of cancers, the analysis methods, ethnicities and sample sizes might attribute to the heterogeneity. Secondly, we extracted the data of HRs and 95% CIs from the K-M plots when it could not be obtained from the paper directly, and this process might decrease the accuracy of results. Thirdly, the sensitivity analysis only showed that individual study had no influence on the association study between the high expressed PODXL and poor OS or CSS, that is to say, the results of the association between the membrane expressed PODXL and poor OS in cancers can only be seen as a descriptive hypothesis, might be induced by the insufficient studies or the small sample size. Fourthly, our meta-analysis seemed have no publication bias, but as the chance of negative results being published is very small, more studies are needed to verify the results of our meta-analysis.

Conclusion

PODXL is a significant clinical indicator for tumor prognosis and detection, the expression level and location in tumor tissues, and even the serum concentration of which could be associated significantly with tumor progression [49]. Our meta-analysis showed that PODXL plays a significant role in cancer progression, and high-expressed PODXL could be linked to aggressive biological phenotype and poor prognosis. Specifically, the high expressed PODXL was correlated with poor prognosis significantly in the glioblastoma multiforme and pancreatic cancer, but not in the esophageal adenocarcinoma, gastric cancer and lung adenocarcinoma.

Abbreviations

CI: confidence inter

CSS: cancer-specific survival

DFS: disease-free survival

GEPIA: Gene Expression Profiling Interactive Analysis

HR: Hazard ratio

IHC: immunohistochemistry

K-M: Kaplan-Meier

mAb: monoclonal antibody

NCD: noncommunicable disease

NOS: Newcastle-Ottawa Scale

OR: odds ratio

OS: overall survival

PRISMA: Preferred Reporting Items for Systematic Reviews and Meta-Analysis

PODXL: podocalyxin-like protein

TCGA: the Cancer Genome Atlas

Declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Availability of data and materials

All data generated or analyzed during this study are included in this article and referenced articles are listed in the References section.

Competing interests

The authors declare that they have no competing interests.

Funding

Not applicable.

Authors’ Contributions

MY and SH conceived the study. SH and ML searched the databases and extracted the data. WD and SH analyzed the data. SH and WD wrote the draft of the paper. FZ and MY reviewed the manuscript. All authors have read and approved the manuscript.

Acknowledgements

Not applicable.

References

  1. Somasiri A, Nielsen JS, Makretsov N, McCoy ML, Prentice L, Gilks CB et al. Overexpression of the anti-adhesin podocalyxin is an independent predictor of breast cancer progression. Cancer Res. 2004;64(15):5068-73. doi:10.1158/0008-5472.can-04-0240.
  2. McNagny KM, Pettersson I, Rossi F, Flamme I, Shevchenko A, Mann M et al. Thrombomucin, a novel cell surface protein that defines thrombocytes and multipotent hematopoietic progenitors. J Cell Biol. 1997;138(6):1395-407.
  3. Schopperle WM, Kershaw DB, DeWolf WC. Human embryonal carcinoma tumor antigen, Gp200/GCTM-2, is podocalyxin. Biochemical and biophysical research communications. 2003;300(2):285-90.
  4. Itai S, Yamada S, Kaneko MK, Sano M, Nakamura T, Yanaka M et al. Podocalyxin is crucial for the growth of oral squamous cell carcinoma cell line HSC-2. Biochem Biophys Rep. 2018;15:93-6. doi:10.1016/j.bbrep.2018.07.008.
  5. Larsson A, Johansson ME, Wangefjord S, Gaber A, Nodin B, Kucharzewska P et al. Overexpression of podocalyxin-like protein is an independent factor of poor prognosis in colorectal cancer. Br J Cancer. 2011;105(5):666-72. doi:10.1038/bjc.2011.295.
  6. Kershaw DB, Beck SG, Wharram BL, Wiggins JE, Goyal M, Thomas PE et al. Molecular cloning and characterization of human podocalyxin-like protein. Orthologous relationship to rabbit PCLP1 and rat podocalyxin. J Biol Chem. 1997;272(25):15708-14.
  7. Li P, Karaczyn AA, McGlauflin R, Favreau-Lessard AJ, Jachimowicz E, Vary CP et al. Novel roles for podocalyxin in regulating stress myelopoiesis, Rap1a, and neutrophil migration. Experimental hematology. 2017;50:77-83.e6. doi:10.1016/j.exphem.2017.04.001.
  8. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394-424. doi:10.3322/caac.21492.
  9. Kerosuo L, Juvonen E, Alitalo R, Gylling M, Kerjaschki D, Miettinen A. Podocalyxin in human haematopoietic cells. British journal of haematology. 2004;124(6):809-18.
  10. Porras G, Ayuso MS, Gonzalez-Manchon C. Leukocyte-endothelial cell interaction is enhanced in podocalyxin-deficient mice. The international journal of biochemistry & cell biology. 2018;99:72-9. doi:10.1016/j.biocel.2018.03.018.
  11. Horrillo A, Porras G, Ayuso MS, Gonzalez-Manchon C. Loss of endothelial barrier integrity in mice with conditional ablation of podocalyxin (Podxl) in endothelial cells. European journal of cell biology. 2016;95(8):265-76. doi:10.1016/j.ejcb.2016.04.006.
  12. Vitureira N, Andres R, Perez-Martinez E, Martinez A, Bribian A, Blasi J et al. Podocalyxin is a novel polysialylated neural adhesion protein with multiple roles in neural development and synapse formation. PLoS One. 2010;5(8):e12003. doi:10.1371/journal.pone.0012003.
  13. Forse CL, Yilmaz YE, Pinnaduwage D, O'Malley FP, Mulligan AM, Bull SB et al. Elevated expression of podocalyxin is associated with lymphatic invasion, basal-like phenotype, and clinical outcome in axillary lymph node-negative breast cancer. Breast Cancer Res Treat. 2013;137(3):709-19. doi:10.1007/s10549-012-2392-y.
  14. Larsson A, Fridberg M, Gaber A, Nodin B, Leveen P, Jonsson G et al. Validation of podocalyxin-like protein as a biomarker of poor prognosis in colorectal cancer. BMC Cancer. 2012;12:282.
  15. Kaprio T, Hagstrom J, Fermer C, Mustonen H, Bockelman C, Nilsson O et al. A comparative study of two PODXL antibodies in 840 colorectal cancer patients. BMC Cancer. 2014;14:494.
  16. Yuan D, Chen H, Wang S, Liu F, Cheng Y, Fang J. Identification of LEA, a podocalyxin-like glycoprotein, as a predictor for the progression of colorectal cancer. Cancer Med. 2018;7(10):5155-66. doi:10.1002/cam4.1765.
  17. Borg D, Hedner C, Nodin B, Larsson A, Johnsson A, Eberhard J et al. Expression of podocalyxin-like protein is an independent prognostic biomarker in resected esophageal and gastric adenocarcinoma. BMC Clin Pathol. 2016;16:13. doi:10.1186/s12907-016-0034-8.
  18. Laitinen A, Bockelman C, Hagstrom J, Kokkola A, Fermer C, Nilsson O et al. Podocalyxin as a Prognostic Marker in Gastric Cancer. PLoS One. 2015;10(12):e0145079. doi:10.1371/journal.pone.0145079.
  19. Zhang J, Zhu Z, Wu H, Yu Z, Rong Z, Luo Z et al. PODXL, negatively regulated by KLF4, promotes the EMT and metastasis and serves as a novel prognostic indicator of gastric cancer. Gastric Cancer. 2019;22(1):48-59. doi:10.1007/s10120-018-0833-y.
  20. Binder ZA, Siu IM, Eberhart CG, Ap Rhys C, Bai RY, Staedtke V et al. Podocalyxin-like protein is expressed in glioblastoma multiforme stem-like cells and is associated with poor outcome. PLoS One. 2013;8(10):e75945. doi:10.1371/journal.pone.0075945.
  21. Kusumoto H, Shintani Y, Kanzaki R, Kawamura T, Funaki S, Minami M et al. Podocalyxin influences malignant potential by controlling epithelial-mesenchymal transition in lung adenocarcinoma. Cancer Sci. 2017;108(3):528-35. doi:10.1111/cas.13142.
  22. Lin CW, Sun MS, Wu HC. Podocalyxin-like 1 is associated with tumor aggressiveness and metastatic gene expression in human oral squamous cell carcinoma. Int J Oncol. 2014;45(2):710-8. doi:10.3892/ijo.2014.2427.
  23. Cipollone JA, Graves ML, Kobel M, Kalloger SE, Poon T, Gilks CB et al. The anti-adhesive mucin podocalyxin may help initiate the transperitoneal metastasis of high grade serous ovarian carcinoma. Clin Exp Metastasis. 2012;29:239-52. doi:10.1007/s10585-011-9446-0).
  24. Saukkonen K, Hagstrom J, Mustonen H, Juuti A, Nordling S, Fermer C et al. Podocalyxin Is a Marker of Poor Prognosis in Pancreatic Ductal Adenocarcinoma. PLoS One. 2015;10(6):e0129012. doi:10.1371/journal.pone.0129012.
  25. Heby M, Elebro J, Nodin B, Jirstrom K, Eberhard J. Prognostic and predictive significance of podocalyxin-like protein expression in pancreatic and periampullary adenocarcinoma. BMC Clin Pathol. 2015;15:10. doi:10.1186/s12907-015-0009-1.
  26. Taniuchi K, Furihata M, Naganuma S, Dabanaka K, Hanazaki K, Saibara T. Podocalyxin-like protein, linked to poor prognosis of pancreatic cancers, promotes cell invasion by binding to gelsolin. Cancer Sci. 2016;107(10):1430-42. doi:10.1111/cas.13018.
  27. Chijiiwa Y, Moriyama T, Ohuchida K, Nabae T, Ohtsuka T, Miyasaka Y et al. Overexpression of microRNA-5100 decreases the aggressive phenotype of pancreatic cancer cells by targeting PODXL. Int J Oncol. 2016;48(4):1688-700. doi:10.3892/ijo.2016.3389.
  28. Casey G, Neville PJ, Liu X, Plummer SJ, Cicek MS, Krumroy LM et al. Podocalyxin variants and risk of prostate cancer and tumor aggressiveness. Human molecular genetics. 2006;15(5):735-41. doi:10.1093/hmg/ddi487.
  29. Heath EI, Heilbrun LK, Smith D, Schopperle WM, Ju Y, Bolton S et al. Overexpression of the Pluripotent Stem Cell Marker Podocalyxin in Prostate Cancer. Anticancer research. 2018;38(11):6361-6. doi:10.21873/anticanres.12994.
  30. Hsu YH, Lin WL, Hou YT, Pu YS, Shun CT, Chen CL et al. Podocalyxin EBP50 ezrin molecular complex enhances the metastatic potential of renal cell carcinoma through recruiting Rac1 guanine nucleotide exchange factor ARHGEF7. Am J Pathol. 2010;176(6):3050-61. doi:10.2353/ajpath.2010.090539.
  31. Boman K, Larsson AH, Segersten U, Kuteeva E, Johannesson H, Nodin B et al. Membranous expression of podocalyxin-like protein is an independent factor of poor prognosis in urothelial bladder cancer. Br J Cancer. 2013;108(11):2321-8. doi:10.1038/bjc.2013.215.
  32. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6(7):e1000097. doi:10.1371/journal.pmed.1000097.
  33. Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR. Practical methods for incorporating summary time-to-event data into meta-analysis. Trials. 2007;8:16. doi:10.1186/1745-6215-8-16.
  34. Zeng X, Zhang Y, Kwong JS, Zhang C, Li S, Sun F et al. The methodological quality assessment tools for preclinical and clinical studies, systematic review and meta-analysis, and clinical practice guideline: a systematic review. Journal of evidence-based medicine. 2015;8(1):2-10. doi:10.1111/jebm.12141.
  35. Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45(W1):W98-W102. doi:10.1093/nar/gkx247.
  36. Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi B et al. UALCAN: A Portal for Facilitating Tumor Subgroup Gene Expression and Survival Analyses. Neoplasia. 2017;19(8):649-58. doi:10.1016/j.neo.2017.05.002.
  37. A S, S NJ, N M, L MM, L P, B GC et al. Overexpression of the anti-adhesin podocalyxin is an independent predictor of breast cancer progression. Cancer Res. 2004;64:5068-73.
  38. Zhang J, Zhu Z, Wu H, Yu Z, Rong Z, Luo Z et al. PODXL, negatively regulated by KLF4, promotes the EMT and metastasis and serves as a novel prognostic indicator of gastric cancer. Gastric Cancer. 2018. doi:10.1007/s10120-018-0833-y.
  39. Nielsen JS, Graves ML, Chelliah S, Vogl AW, Roskelley CD, McNagny KM. The CD34-related molecule podocalyxin is a potent inducer of microvillus formation. PLoS One. 2007;2(2):e237. doi:10.1371/journal.pone.0000237.
  40. Zhang J, Zhu Z, Sheng J, Yu Z, Yao B, Huang K et al. miR-509-3-5P inhibits the invasion and lymphatic metastasis by targeting PODXL and serves as a novel prognostic indicator for gastric cancer. Oncotarget. 2017;8(21):34867-83. doi:10.18632/oncotarget.16802.
  41. Amo L, Tamayo-Orbegozo E, Maruri N, Eguizabal C, Zenarruzabeitia O, Rinon M et al. Involvement of platelet-tumor cell interaction in immune evasion. Potential role of podocalyxin-like protein 1. Frontiers in oncology. 2014;4:245. doi:10.3389/fonc.2014.00245.
  42. Larsson AH, Lehn S, Wangefjord S, Karnevi E, Kuteeva E, Sundström M et al. Significant association and synergistic adverse prognostic effect of podocalyxin-like protein and epidermal growth factor receptor expression in colorectal cancer. Journal of translational medicine. 2016;14(1):128. doi:10.1186/s12967-016-0882-0.
  43. Fröse J, Chen MB, Hebron KE, Reinhardt F, Hajal C, Zijlstra A et al. Epithelial-Mesenchymal Transition Induces Podocalyxin to Promote Extravasation via Ezrin Signaling. Cell reports. 2018;24(4):962-72. doi:10.1016/j.celrep.2018.06.092.
  44. Lee WY, Kuo CC, Lin BX, Cheng CH, Chen KC, Lin CW. Podocalyxin-Like Protein 1 Regulates TAZ Signaling and Stemness Properties in Colon Cancer. Int J Mol Sci. 2017;18(10). doi:10.3390/ijms18102047.
  45. Zhi Q, Chen H, Liu F, Han Y, Wan D, Xu Z et al. PODXL promotes gastric cancer progression through interacting with RUFY1 protein. Cancer Sci. 2018. doi:10.1111/cas.13864.
  46. Buganim Y, Madar S, Rais Y, Pomeraniec L, Harel E, Solomon H et al. Transcriptional activity of ATF3 in the stromal compartment of tumors promotes cancer progression. Carcinogenesis. 2011;32(12):1749-57. doi:10.1093/carcin/bgr203.
  47. Itai S, Ohishi T, Kaneko MK, Yamada S, Abe S, Nakamura T et al. Anti-podocalyxin antibody exerts antitumor effects via antibody-dependent cellular cytotoxicity in mouse xenograft models of oral squamous cell carcinoma. Oncotarget. 2018;9(32):22480-97. doi:10.18632/oncotarget.25132.
  48. Borg D, Larsson AH, Hedner C, Nodin B, Johnsson A, Jirstrom K. Podocalyxin-like protein as a predictive biomarker for benefit of neoadjuvant chemotherapy in resectable gastric and esophageal adenocarcinoma. Journal of translational medicine. 2018;16(1):290. doi:10.1186/s12967-018-1668-3.
  49. Taniuchi K, Tsuboi M, Sakaguchi M, Saibara T. Measurement of serum PODXL concentration for detection of pancreatic cancer. Onco Targets Ther. 2018;11:1433-45. doi:10.2147/ott.s155367.