International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Colorectal cancer (CRC) ranks third among the most prevalent diseases and is the second deadliest malignant disease globally (1,2). In 2020, there were ~1.9 million newly reported cases of colorectal cancer (CRC) globally, with >900,000 deaths attributed to CRC (3). Key factors contributing to its high mortality include challenges in early diagnosis, tumor heterogeneity and the ineffectiveness of late-stage treatments (4–6). Tumor heterogeneity encompasses both cellular heterogeneity within the tumor and heterogeneity of the tumor microenvironment (7,8). A growing body of research indicates that the heterogeneity of CRC is among the most notable in malignant tumors and exerts a profound influence on the prognosis and treatment of the disease (5,9,10). Moreover, previous studies leveraging bioinformatics analysis for molecular classification and prognostic prediction of CRC have emerged, indicating its considerable clinical promise (11–13). However, due to its heterogeneity, CRC progression is a multifactorial, multistage and dynamic process, which imposes certain limitations on existing models. Consequently, it is imperative to explore novel molecular subtypes and models of CRC for prognostic evaluation.
Tissue inhibitor of matrix metalloproteinase 1 (TIMP1), a member of the TIMP gene family, has been reported to inhibit matrix metalloproteinase (MMP) activity (14). The TIMP1 gene is situated on human chromosome Xp11.1-p11.4 and its mRNA codes for a protein composed of 184 amino acids, exhibiting a molecular mass of 29–34 kDa (15). Accumulating evidence indicates that it interacts with several molecules to regulate biological processes such as cell proliferation, apoptosis and differentiation, and it is implicated in the pathological mechanisms of several diseases, particularly cancer (16,17). A total of >35 independent clinical studies have reported that high TIMP1 expression is positively associated with poor prognosis across several cancer types, including lung, brain, prostate, breast and colon cancer (14). Moreover, the levels of TIMP1 are elevated in advanced-stage tumors (14). Tian et al (18) reported that pancreatic cancer cell-derived TIMP1 activates CD63/PI3K/AKT signaling to promote perineural invasion by stimulating Schwann cells. Moreover, TIMP1 loss has been reported to alter the senescence-associated secretory phenotype of senescent tumor cells and enhance prostate cancer metastasis via MMP activation (19). However, the role of TIMP1 in CRC remains poorly understood. Therefore, the present study aimed to explore the clinical and prognostic significance of TIMP1 in CRC.
TCGA database (http://cancergenome.nih.gov/) was the primary resource for acquiring CRC mRNA expression data and clinical information. The present study analyzed multi-omics patient data, which were systematically organized across multiple databases, including the original TCGA datasets (http://cancergenome.nih.gov/). Subsequent databases, such as the University of Alabama at Birmingham CANcer data analysis Portal (UALCAN) (20,21) and Gene Expression Profiling Interactive Analysis (GEPIA) (22), provided comprehensive insights into several aspects. The patient records from each database were specifically utilized for diverse analyses.
The UALCAN database (https://ualcan.path.uab.edu/) (20,21) was used to analyze TIMP1 mRNA expression levels in pan-cancer and in colon cancer samples (primary tumor, n=286; normal, n=41). GEPIA2 (http://gepia2.cancer-pku.cn/#general) (21) was used to assess TIMP1 expression in TCGA tumors by matching TCGA normal and Genotype-Tissue Expression project data for CRC (tumor, n=275; normal, n=349). The GSE24550 dataset was used to analyze TIMP1 mRNA expression levels in CRC tissues (G2, n=77) and normal tissues (G1, n=13) (23). In addition, the Clinical Proteomic Tumor Analysis Consortium (CPTAC) database within UALCAN was used to evaluate TIMP1 protein expression in CRC (normal, n=100; primary tumor, n=97).
STAR-counts data and the corresponding clinical information for CRC tumors were downloaded from TCGA database (24). The data in transcripts per million (TPM) format were extracted and normalized using log2(TPM+1) transformation. Following filtering of the samples with both RNA sequencing (RNAseq) and clinical data, CRC samples with high TIMP1 expression (top 25%, n=156) and low TIMP1 expression (top 25%, n=157) were selected for analysis (Table I). Statistical analysis was performed using R software (version v4.0.3; The R Foundation). P<0.05 was considered to indicate a statistically significant difference.
Table I.Tissue inhibitor of matrix metalloproteinase 1 expression and clinical features in 313 samples from patients with colorectal cancer. |
The GEPIA2 database was used for the analysis of the association between TIMP1 expression, overall survival and disease-free survival in patients with CRC. In addition, the UALCAN database was used for the analysis of the association between TIMP1 expression and overall survival. CRC samples with high TIMP1 expression (median, n=309) and low TIMP1 expression (median, n=310) from TCGA database were used for further analysis. For survival analysis, the log-rank test was employed to compare survival differences between the two groups in the Kaplan-Meier analysis, obtaining the P-value, hazard ratio (HR) and 95% confidence interval (CI) using the log-rank test and univariate Cox regression. To perform Cox regression analysis, univariate and multivariate Cox proportional hazards regression analyses were performed. The ‘forestplot’ package was used to visually present the P-value, HR and 95% CI for each variable using a forest plot. Subsequently, based on the multivariate Cox proportional hazard model results, a Nomogram was constructed using the ‘rms’ package to predict the total recurrence rate over X years. All statistical analyses were performed using R software (v4.0.3).
To reliably evaluate immune scores in CRC, ‘immunedeconv’ was used, which is an R package (v4.1.3) that combines two advanced algorithms, including Tumor IMune Estimation Resource (TIMER) and Estimate the Proportion of Immune and Cancer (EPIC), applied to TCGA CRC data (n=620) (25,26). Each algorithm has been benchmarked for its unique strengths. For result analysis and visualization, the R package ‘ggClusterNet’ was employed. In addition, Spearman's correlation analysis was performed to assess the relationship between TIMP1 and immune cells (CD4+ and CD8+ T cells) in CRC using TCGA data (n=620).
RNAseq data and clinical information of CRC (n=620) were obtained from TCGA database. Spearman's correlation analysis was used to assess the correlation between TIMP1 and TMB/MSI (25,26).
CRC RNAseq data and the clinical information (n=620) were retrieved from TCGA database. Following compilation of the genes from the relevant pathways, the ‘GSVA’ package was used in R software with the parameter method ‘ssgsea’ used to perform single-sample gene set enrichment analysis (ssGSEA). Finally, the correlation between gene expression and pathway scores was assessed using Spearman's correlation analysis (27–29).
A colon cancer tissue microarray (TMA) was purchased from Hunan Aifang Biotechnology Co., Ltd., and the relevant follow-up data were included. The TMA comprised 78 pairs of colon cancer and normal tissues. The patient characteristics are summarized in Table SI. The present study was approved by the Medical Ethics Committee of Soochow University's First Affiliated Hospital (approval no. 2021-327).
IHC staining was performed as follows (30): Tissue samples were subjected to fixation utilizing a 4% paraformaldehyde solution (Beyotime Institute of Biotechnology) maintained at an ambient temperature of 25°C for a duration of 24 h. Subsequently, the specimens were processed for paraffin embedding. Sections, each with a thickness of 5 µm, excised from the paraffin-embedded tissue blocks, underwent deparaffinization and rehydration procedures. Following antigen retrieval conducted with a 10 mM sodium citrate buffer (pH 6.0; Beyotime Institute of Biotechnology), the sections were treated with a 3% hydrogen peroxide solution at room temperature for 10 min to inhibit endogenous peroxidase activity and to mitigate non-specific protein interactions. The sections were then incubated with anti-TIMP1 antibodies (1:300 dilution; cat. no. 16644-1-AP; Proteintech Group, Inc.) overnight at 4°C, succeeded by incubation with a biotinylated goat anti-rabbit secondary antibody working solution (1:500 dilution; cat. no. SA1020; Boster Biological Technology Co. Ltd.) at 37°C for 30 min. Immunodetection was subsequently executed employing the Dako EnVision detection system (Agilent Technologies, Inc.). The prepared slides were examined and images were captured under a fluorescence microscope (Leica Microsystems). The IHC score was determined by multiplying the staining intensity (negative, 0; mild, 1; moderate, 2; and strong, 3) by the percentage of the stained area (0%, 0; ≤25%, 1; 25–50%, 2; 50–75%, 3; and >75%, 4).
R software (version 4.0.3) and GraphPad Prism 8.0 (Dotmatics) were used for both data analysis and visualization. The data are presented as the mean ± SD from a minimum of three replicates. Differences between two groups were assessed using an unpaired t-test, Wilcoxon rank-sum test or paired t-test, as appropriate. Fisher's exact test or χ2 test were used to analyze the associations between the expression of TIMP1 and clinicopathological features. Spearman's correlation analysis was performed to assess the correlation between TIMP1 and immune cells (CD4+ and CD8+ T cells) in CRC using TCGA data (n=620). P<0.05 was considered to indicate a statistically significant difference.
Using the UALCAN database, TIMP1 mRNA expression was assessed across TCGA cancer tissues. The results indicated that, compared with that in normal tissues, its expression was markedly upregulated in several tumor types, including CRC. In 15/24 tumors, TIMP1 was overexpressed (Fig. 1A). Based on a database visualization website of UALCAN with TCGA, the mRNA expression levels of TIMP1 were also significantly higher in colon cancer samples compared with those in normal samples (Fig. 1B). The GSE24550 dataset was also evaluated, where the specimens of patients with colon cancer exhibited high TIMP1 expression compared with those of normal controls (Fig. 1C). Furthermore, TCGA data analysis of GEPIA confirmed this result (Fig. 1D). Moreover, CPTAC data revealed that the protein expression of TIMP1 exhibited significant upregulation specifically in the tumor tissue, in comparison with that in normal tissues (Fig. 1E).
To assess the association between TIMP1 expression levels in CRC tissues and the clinicopathological characteristics of patients with CRC, STAR-counts data and relevant clinical information were obtained from TCGA database. The results revealed that TIMP1 expression was significantly associated with survival (P=0019) and ethnicity (P=0.0404); however, no significant associations were noted between TIMP1 levels and age, sex, pathological (p) tumor (T)-node (N)-metastasis (M) stage, pT stage, pN stage or pM stage (Table I).
Patients with CRC and high TIMP1 expression exhibited shorter overall survival compared with those with low expression (Fig. 2A-C). Notably, TIMP1 expression was significantly associated with disease-free survival (Fig. 2D). Univariate Cox regression analysis identified TIMP1 expression, age, pT stage (T4 vs. T1), pN stage (N1/2 vs. N0), pM stage (M1/X vs. M0) and pTNM stage (III vs. I and IV vs. I) as significant predictors of overall survival in patients with CRC (Fig. 3A). The results of multivariate Cox regression analysis further confirmed that TIMP1 expression, age, pN stage (N1/2 vs. N0) and pTNM stage (III vs. I, IV vs. I) were independent prognostic factors for overall survival (Fig. 3B). Based on these factors, nomograms were constructed using TCGA datasets to calculate prognostic scores for patients with CRC, yielding a survival probability of 0.715 (Fig. 3C). The calibration curves for TCGA cohorts were closely aligned with the 45-degree diagonal line (Fig. 3D), indicating optimal concordance between predicted and actual survival probabilities.
Subsequently, the role of TIMP1 in immunity was used to evaluate its potential in CRC immunotherapy. Network connection diagrams and heatmaps illustrated the link between TIMP1 expression and immune scores, where red/blue intensity and ring size reflected correlation strength (Fig. 4A and B). Red lines indicate the positive correlations and green lines the negative. Both TIMER and EPIC scores revealed the positive correlation of TIMP1 with CD4+ and CD8+ T cells. Spearman's correlation analysis using TCGA CRC samples further confirmed this significant association (Fig. 4C).
Previous studies have reported that TMB and MSI are valuable biomarkers that provide insights into cancer prognosis and treatment decisions (31–33). Both TMB and MSI serve roles in cancer prognosis, treatment selection and immunotherapy development (34). The findings of the present study demonstrate a significant but weak positive correlation between TIMP1 expression and TMB and MSI in CRC (Fig. 4D). This suggests that TIMP1 can potentially serve as a biomarker to identify patients who may benefit from immune checkpoint inhibitors, particularly in tumors where high TMB and MSI predict a favorable response to immunotherapy.
The ssGSEA algorithm was applied to compute the enrichment fraction of each sample for specific pathways, thereby exploring the sample-pathway relationship (27). The calculated fractions intuitively reflected the interaction and association between samples and pathways. The results indicated that in CRC, TIMP1 may be positively correlated with epithelial-mesenchymal transition (EMT) markers, extracellular matrix (ECM)-related genes, collagen formation, angiogenesis, apoptosis, ferroptosis, the upregulation of the expression levels of genes caused by reactive oxygen species (ROS), tumor inflammation and the TGF-β signaling pathway (Fig. 5). Overall, these findings indicate that TIMP1 may be associated with multiple pathways and may regulate the progression of CRC.
The IHC assay indicated that the expression levels of TIMP1 were significantly higher in colon cancer tissues than those in adjacent normal tissues (Fig. 6A and B). Notably, individuals with high TIMP1 expression (IHC score >4) exhibited a significantly reduced survival rate compared with those with low TIMP1 expression (IHC score ≤4) in colon cancer (Fig. 6C).
Most patients with CRC are diagnosed at an advanced stage, which contributes to the poor prognosis commonly noted in this patient population (35,36). However, previous research indicates that an early diagnosis of CRC can markedly improve patient outcomes (37). As such, the identification of effective biomarkers for early CRC detection is crucial, as they can serve a pivotal role in enhancing the prognosis of patients with CRC.
TIMP1 belongs to the TIMP family, which includes other members such as TIMP2, TIMP3 and TIMP4 (14). Research has indicated that TIMP1 expression is upregulated across multiple tumor types and this upregulation is associated with poor prognosis and reduced survival in patients with cancer (16). For example, TIMP1 expression has been associated with gastric cancer differentiation and poor prognosis in patients with gastric cancer (38). Similarly, high TIMP1 expression in lung cancer tumors is associated with an unfavorable prognosis (39). Macedo et al (40) also reported that elevated TIMP1 levels in patients with colon and gastric cancer are associated with poor prognosis. These reports are consistent with the findings of the present study, which revealed that both TIMP1 mRNA and protein levels were markedly elevated in CRC tissues, and positively associated with worse overall and disease-free survival in patients with CRC. Univariate and multivariate Cox regression analyses further established high TIMP1 expression as an independent prognostic indicator for overall survival in CRC. These results indicate that TIMP1 expression is likely a predictor of CRC prognosis.
Previous studies have established immunity as a prognostic indicator for cancer progression (41–43). Emerging mechanistic studies identify TIMP1 as a critical tumor immune modulator (44,45). In thyroid cancer, TIMP1 promotes cancer cell progression by inducing macrophage phenotypic polarization via the PI3K/AKT signaling pathway (44). In glioblastoma, the expression of TIMP1, induced by Sp1, is markedly elevated in tumor-infiltrating lymphocytes and is associated with cancer progression (45). Moreover, TIMP1 expression in gliomas is associated with tumor immune infiltration and immune checkpoint-related gene expression (46). Wang et al (47) reported that intracellular TIMP-1-CD63 signaling directs immune escape and metastasis evolution in KRAS-mutated pancreatic cancer cells. In the present study, network connection diagrams and heatmaps visualized the association between TIMP1 expression and immune scores. Notably, TIMP1 indicated a significant but weak correlation with CD4+ and CD8+ T cells in CRC. Overall, these findings suggest that TIMP1 may modulate the CRC tumor immune environment by influencing CD4+ and CD8+ T cell infiltration and function.
Immunotherapy, particularly the use of immune checkpoint inhibitors (ICIs), has transformed cancer treatment by leveraging the capacity of the immune system to combat tumors (48). However, in contrast to cancer types, such as non-small-cell lung cancer and melanoma, which are known to be sensitive to immunotherapy, there is a lack of reliable predictive biomarkers in multiple cancer types (49). TMB and MSI serve as genomic biomarkers to identify patients likely to benefit from ICIs, and ICIs have shown promise in colon cancer treatment, notably in patients with high TMB and MSI (34). The present study indicated a significant association between TIMP1 expression and TMB and MSI in CRC. This suggests that TIMP1 may be a potential predictive biomarker for colon cancer immunotherapy.
Furthermore, previous studies have reported that TIMP-1 can regulate tumor cell behavior by inducing signaling pathways associated with cell growth, proliferation and survival (14,16). For example, Song et al (50) reported that inhibiting TIMP1 expression reduced proliferation and metastasis, whilst promoting apoptosis by targeting the FAK-PI3K/AKT and MAPK pathways. The use of short hairpin RNA to knockdown TIMP1 expression was associated with a notable reduction in cell proliferation and invasion in right-sided patient-derived organoids from both left- and right-sided CRCs. This effect was achieved by modulating the FAK/AKT signaling pathway (51). In the present study, the ssGSEA algorithm revealed a positive correlation between TIMP1 expression in CRC and multiple biological processes and pathways, including EMT markers, ECM-related genes, collagen formation, angiogenesis, apoptosis, ferroptosis, ROS-upregulated genes, tumor inflammation signatures and the TGF-β pathway. Whilst further comprehensive experimental validation is required to evaluate the pathways associated with TIMP1, the existing data strongly suggest that TIMP1 serves pivotal roles in driving CRC progression and metastasis.
Moreover, whilst the present study highlights the clinical significance of TIMP1, certain limitations are present. Firstly, specific analyses rely on retrospective public-database data; therefore, prospective studies are required to verify the clinical relevance of the findings. Given the complexity of CRC and the varied histological phenotypes, more detailed mechanistic and clinical research is essential to explore the roles of TIMP1 across CRC subtypes. In addition, the sample size of the present CRC cohort is limited. In future work, the validation cohort will be expanded by constructing a larger, multi-center TMA with balanced representation of major CRC subtypes (such as CMS, MSI-H and BRAF-mutant), on which subtype-stratified analyses will be performed to determine if the role of TIMP1 is universal or subtype-specific. Furthermore, comprehensive in vitro and in vivo experiments are required to clarify the mechanistic role of TIMP1 in tumor progression and its interaction with the TME. Despite these limitations, the present study guides future research, including clinical work and basic experiments, which will be part of the subsequent research focus.
In conclusion, high TIMP1 expression in CRC is associated with a poor prognosis for patients with CRC. TIMP1 may also modulate the CRC immune microenvironment and facilitate CRC progression and metastasis. The data suggest that TIMP1 represents a promising diagnostic biomarker and therapeutic target for CRC.
Not applicable.
The present study was supported by the Collaborative Custom Development of RNA Probes (grant no. P112213323).
The data generated in the present study may be requested from the corresponding author.
SX, DZ and TS contributed to the study conception and design. JG contributed to performing experiments, acquisition of data and interpretation of data. FC performed experiments. JG and TS wrote the original manuscript. DZ and SX revised the manuscript. JG and TS confirm the authenticity of all the raw data. All authors read and approved the final manuscript.
The present study was granted approval by the Institutional Review Board of the First Affiliated Hospital of Soochow University (Suzhou, China; approval no. 2021-327). Written informed consent was obtained from all participating patients.
Not applicable.
The authors declare that they have no competing interests.
|
Abedizadeh R, Majidi F, Khorasani HR, Abedi H and Sabour D: Colorectal cancer: A comprehensive review of carcinogenesis, diagnosis, and novel strategies for classified treatments. Cancer Metastasis Rev. 43:729–753. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Tao XY, Li QQ and Zeng Y: Clinical application of liquid biopsy in colorectal cancer: Detection, prediction, and treatment monitoring. Mol Cancer. 23:1452024. View Article : Google Scholar : PubMed/NCBI | |
|
Mousavi SE, Ilaghi M, Hamidi Rad R and Nejadghaderi SA: Epidemiology and socioeconomic correlates of colorectal cancer in Asia in 2020 and its projection to 2040. Sci Rep. 15:266392025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Wang Y, Zhang B, Li P and Zhao Y: Methods and biomarkers for early detection, prediction, and diagnosis of colorectal cancer. Biomed Pharmacother. 163:1147862023. View Article : Google Scholar : PubMed/NCBI | |
|
Mo S, Tang P, Luo W, Zhang L, Li Y, Hu X, Ma X, Chen Y, Bao Y, He X, et al: Patient-derived organoids from colorectal cancer with paired liver metastasis reveal tumor heterogeneity and predict response to chemotherapy. Adv Sci (Weinh). 9:e22040972022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang W and Wang S: Relationships between nutritional status and serum adipokine levels with chemotherapy efficacy in late-stage colorectal cancer patients. Int J Colorectal Dis. 40:252025. View Article : Google Scholar : PubMed/NCBI | |
|
Prasetyanti PR and Medema JP: Intra-tumor heterogeneity from a cancer stem cell perspective. Mol Cancer. 16:412017. View Article : Google Scholar : PubMed/NCBI | |
|
Yu X, Liu R, Gao W, Wang X and Zhang Y: Single-cell omics traces the heterogeneity of prostate cancer cells and the tumor microenvironment. Cell Mol Biol Lett. 28:382023. View Article : Google Scholar : PubMed/NCBI | |
|
Guo L, Wang Y, Yang W, Wang C, Guo T, Yang J, Shao Z, Cai G, Cai S, Zhang L, et al: Molecular profiling provides clinical insights into targeted and immunotherapies as well as colorectal cancer prognosis. Gastroenterology. 165:414–428.e7. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Singh H, Sahgal P, Kapner K, Corsello SM, Gupta H, Gujrathi R, Li YY, Cherniack AD, El Alam R, Kerfoot J, et al: RAS/RAF Comutation and ERBB2 copy number modulates HER2 heterogeneity and responsiveness to HER2-directed therapy in colorectal cancer. Clin Cancer Res. 30:1669–1684. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Jalali P, Aliyari S, Etesami M, Saeedi Niasar M, Taher S, Kavousi K, Nazemalhosseini Mojarad E and Salehi Z: GUCA2A dysregulation as a promising biomarker for accurate diagnosis and prognosis of colorectal cancer. Clin Exp Med. 24:2512024. View Article : Google Scholar : PubMed/NCBI | |
|
Lu S, Sun X, Tang H, Yu J, Wang B, Xiao R, Qu J, Sun F, Deng Z, Li C, et al: Colorectal cancer with low SLC35A3 is associated with immune infiltrates and poor prognosis. Sci Rep. 14:3292024. View Article : Google Scholar : PubMed/NCBI | |
|
Parisi E, Hidalgo I, Montal R, Pallise O, Tarragona J, Sorolla A, Novell A, Campbell K, Sorolla MA, Casali A and Salud A: PLA2G12A as a novel biomarker for colorectal cancer with prognostic relevance. Int J Mol Sci. 24:108892023. View Article : Google Scholar : PubMed/NCBI | |
|
Jackson HW, Defamie V, Waterhouse P and Khokha R: TIMPs: Versatile extracellular regulators in cancer. Nat Rev Cancer. 17:38–53. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Caterina NC, Windsor LJ, Bodden MK, Yermovsky AE, Taylor KB, Birkedal-Hansen H and Engler JA: Glycosylation and NH2-terminal domain mutants of the tissue inhibitor of metalloproteinases-1 (TIMP-1). Biochim Biophys Acta. 1388:21–34. 1388. View Article : Google Scholar : PubMed/NCBI | |
|
Justo BL and Jasiulionis MG: Characteristics of TIMP1, CD63, and β1-Integrin and the functional impact of their interaction in cancer. Int J Mol Sci. 22:93192021. View Article : Google Scholar : PubMed/NCBI | |
|
Eckfeld C, Haussler D, Schoeps B, Hermann CD and Kruger A: Functional disparities within the TIMP family in cancer: Hints from molecular divergence. Cancer Metastasis Rev. 38:469–481. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Tian Z, Ou G, Su M, Li R, Pan L, Lin X, Zou J, Chen S, Li Y, Huang K and Chen Y: TIMP1 derived from pancreatic cancer cells stimulates Schwann cells and promotes the occurrence of perineural invasion. Cancer Let. 546:2158632022. View Article : Google Scholar : PubMed/NCBI | |
|
Guccini I, Revandkar A, D'Ambrosio M, Colucci M, Pasquini E, Mosole S, Troiani M, Brina D, Sheibani-Tezerji R, Elia AR, et al: Senescence reprogramming by TIMP1 deficiency promotes prostate cancer metastasis. Cancer Cell. 39:68–82.e9. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chandrashekar DS, Karthikeyan SK, Korla PK, Patel H, Shovon AR, Athar M, Netto GJ, Qin ZS, Kumar S, Manne U, et al: UALCAN: An update to the integrated cancer data analysis platform. Neoplasia. 25:18–27. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi B and Varambally S: UALCAN: A portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia. 19:649–658. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Tang Z, Kang B, Li C, Chen T and Zhang Z: GEPIA2: An enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 47:W556–W560. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Sveen A, Agesen TH, Nesbakken A, Rognum TO, Lothe RA and Skotheim RI: Transcriptome instability in colorectal cancer identified by exon microarray analyses: Associations with splicing factor expression levels and patient survival. Genome Med. 3:322011. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Wang Y, Peng M and Yi L: UBASH3B is a novel prognostic biomarker and correlated with immune infiltrates in prostate cancer. Front Oncol. 9:15172019. View Article : Google Scholar : PubMed/NCBI | |
|
Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, Porta-Pardo E, Gao GF, Plaisier CL, Eddy JA, et al: The immune landscape of cancer. Immunity. 48:812–830.e14. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Bonneville R, Krook MA, Kautto EA, Miya J, Wing MR, Chen HZ, Reeser JW, Yu L and Roychowdhury S: Landscape of microsatellite instability across 39 cancer types. JCO Precis Oncol. 2017.PO.17.00073. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Wei J, Huang K, Chen Z, Hu M, Bai Y, Lin S and Du H: Characterization of glycolysis-associated molecules in the tumor microenvironment revealed by pan-cancer tissues and lung cancer single cell data. Cancers (Basel). 12:17882020. View Article : Google Scholar : PubMed/NCBI | |
|
Hanzelmann S, Castelo R and Guinney J: GSVA: Gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 14:72013. View Article : Google Scholar : PubMed/NCBI | |
|
Xiao Z, Dai Z and Locasale JW: Metabolic landscape of the tumor microenvironment at single cell resolution. Nat Commun. 10:37632019. View Article : Google Scholar : PubMed/NCBI | |
|
Sun L, Chen Y, Xia L, Wang J, Zhu J, Li J, Wang K, Shen K, Zhang D, Zhang G, et al: TRIM69 suppressed the anoikis resistance and metastasis of gastric cancer through ubiquitin-proteasome-mediated degradation of PRKCD. Oncogene. 42:3619–3632. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Kendre G, Murugesan K, Brummer T, Segatto O, Saborowski A and Vogel A: Charting co-mutation patterns associated with actionable drivers in intrahepatic cholangiocarcinoma. J Hepatol. 78:614–626. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Di Mauro A, Santorsola M, Savarese G, Sirica R, Ianniello M, Cossu AM, Ceccarelli A, Sabbatino F, Bocchetti M, Carratu AC, et al: High tumor mutational burden assessed through next-generation sequencing predicts favorable survival in microsatellite stable metastatic colon cancer patients. J Transl Med. 22:11072024. View Article : Google Scholar : PubMed/NCBI | |
|
Shi S, Wang Y, Wu J, Zha B, Li P, Liu Y, Yang Y, Kong J, Gao S, Cui H, et al: Predictive value of PD-L1 and TMB for short-term efficacy prognosis in non-small cell lung cancer and construction of prediction models. Front Oncol. 14:13422622024. View Article : Google Scholar : PubMed/NCBI | |
|
Hou W, Yi C and Zhu H: Predictive biomarkers of colon cancer immunotherapy: Present and future. Front Immunol. 13:10323142022. View Article : Google Scholar : PubMed/NCBI | |
|
Sasidharan Nair V, Saleh R, Taha RZ, Toor SM, Murshed K, Ahmed AA, Kurer MA, Abu Nada M, Al Ejeh F and Elkord E: Differential gene expression of tumor-infiltrating CD4+ T cells in advanced versus early stage colorectal cancer and identification of a gene signature of poor prognosis. Oncoimmunology. 9:18251782020. View Article : Google Scholar : PubMed/NCBI | |
|
Garborg K: Colorectal cancer screening. Surg Clin North Am. 95:979–989. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Tonini V and Zanni M: Why is early detection of colon cancer still not possible in 2023? World J Gastroenterol. 30:211–224. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng M, Wang P, Wang Y, Jia Z, Gao J, Tan X, Chen H and Zu G: Clinicopathological and prognostic significance of TIMP1 expression in gastric cancer: A systematic review and meta-analysis. Expert Rev Anticancer Ther. 24:1169–1176. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Dantas E, Murthy A, Ahmed T, Ahmed M, Ramsamooj S, Hurd MA, Lam T, Malbari M, Agrusa C, Elemento O, et al: TIMP1 is an early biomarker for detection and prognosis of lung cancer. Clin Transl Med. 13:e13912023. View Article : Google Scholar : PubMed/NCBI | |
|
Macedo FC, Cunha N, Pereira TC, Soares RF, Monteiro AR, Bonito N, Valido F and Sousa G: A prospective cohort study of TIMP1 as prognostic biomarker in gastric and colon cancer. Chin Clin Oncol. 11:432022. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng J, Peng L, Zhang S, Liao H, Hao J, Wu S and Shen H: Preoperative systemic immune-inflammation index as a prognostic indicator for patients with urothelial carcinoma. Front Immunol. 14:12750332023. View Article : Google Scholar : PubMed/NCBI | |
|
Xiong S, Dong L and Cheng L: Neutrophils in cancer carcinogenesis and metastasis. J Hematol Oncol. 14:1732021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu R, Lu J, Liu J, Liao Y, Guo Y, Shi P, Wang Z, Wang H and Lai J: Macrophages in prostate cancer: Dual roles in tumor progression and immune evasion. J Transl Med. 23:6152025. View Article : Google Scholar : PubMed/NCBI | |
|
Lin X, Zhao R, Bin Y, Huo R, Xue G and Wu J: TIMP1 promotes thyroid cancer cell progression through macrophage phenotypic polarization via the PI3K/AKT signaling pathway. Genomics. 116:1109142024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu L, Yang S, Lin K, Yu X, Meng J, Ma C, Wu Z, Hao Y, Chen N, Ge Q, et al: Sp1 induced gene TIMP1 is related to immune cell infiltration in glioblastoma. Sci Rep. 12:111812022. View Article : Google Scholar : PubMed/NCBI | |
|
Xu J, Wei C, Wang C, Li F, Wang Z, Xiong J, Zhou Y, Li S, Liu X, Yang G, et al: TIMP1/CHI3L1 facilitates glioma progression and immunosuppression via NF-kappaB activation. Biochim Biophys Acta Mol Basis Dis. 1870:1670412024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang CA, Hou YC, Hong YK, Tai YJ, Shen C, Hou PC, Fu JL, Wu CL, Cheng SM, Hwang DY, et al: Intercellular TIMP-1-CD63 signaling directs the evolution of immune escape and metastasis in KRAS-mutated pancreatic cancer cells. Mol Cancer. 24:252025. View Article : Google Scholar : PubMed/NCBI | |
|
Butterfield LH and Najjar YG: Immunotherapy combination approaches: Mechanisms, biomarkers and clinical observations. Nat Rev Immunol. 24:399–416. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Bai R, Lv Z, Xu D and Cui J: Predictive biomarkers for cancer immunotherapy with immune checkpoint inhibitors. Biomark Res. 8:342020. View Article : Google Scholar : PubMed/NCBI | |
|
Song G, Xu S, Zhang H, Wang Y, Xiao C, Jiang T, Wu L, Zhang T, Sun X, Zhong L, et al: TIMP1 is a prognostic marker for the progression and metastasis of colon cancer through FAK-PI3K/AKT and MAPK pathway. J Exp Clin Cancer Res. 35:1482016. View Article : Google Scholar : PubMed/NCBI | |
|
Ma B, Ueda H, Okamoto K, Bando M, Fujimoto S, Okada Y, Kawaguchi T, Wada H, Miyamoto H, Shimada M, et al: TIMP1 promotes cell proliferation and invasion capability of right-sided colon cancers via the FAK/Akt signaling pathway. Cancer Sci. 113:4244–4257. 2022. View Article : Google Scholar : PubMed/NCBI |