Next Article in Journal
Assessing the Prognosis of Patients with Myelodysplastic Syndromes (MDS)
Next Article in Special Issue
Precision Medicine in Metastatic Colorectal Cancer: Targeting ERBB2 (HER-2) Oncogene
Previous Article in Journal
Familial Risks for Liver, Gallbladder and Bile Duct Cancers and for Their Risk Factors in Sweden, a Low-Incidence Country
Previous Article in Special Issue
Colorectal Cancer: The Contribution of CXCL12 and Its Receptors CXCR4 and CXCR7
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

High Dual Expression of the Biomarkers CD44v6/α2β1 and CD44v6/PD-L1 Indicate Early Recurrence after Colorectal Hepatic Metastasectomy

1
Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University, Marchioninistraße 15, 81377 Munich, Germany
2
German Cancer Consortium (DKTK), Partner Site Munich, Pettenkoferstraße 8a, 80336 Munich, Germany
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(8), 1939; https://doi.org/10.3390/cancers14081939
Submission received: 9 March 2022 / Revised: 28 March 2022 / Accepted: 7 April 2022 / Published: 12 April 2022

Abstract

:

Simple Summary

Distant metastasis in colorectal cancer still correlates with poor prognosis, emphasizing the high need for new diagnostic and therapeutic strategies. In the present study, liver and lung metastases revealed profound differences in the expression pattern of metastasis-driving protein biomarkers. This suggests the adaption of the therapy to the biology of the metastatic organ site. High expression of the cell adhesion molecule CD44v6 and high dual expression of CD44v6, combined with the cell adhesion molecules integrin α2β1, as well as the checkpoint inhibitor molecule PD-L1, correlated significantly with early recurrence after hepatectomy, in a substantial number of liver metastatic patients. These findings suggest the need for the implementation of biological risk factors into clinical risk scores, aiming to make the prognosis of the individual patient more precise. Further, dual expression of protein biomarkers that are druggable, such as CD44v6/α2β1 and CD44v6/PD-L1, can identify high-risk patients for targeted therapy that might provide a survival benefit.

Abstract

Considering the biology of CRC, distant metastases might support the identification of high-risk patients for early recurrence and targeted therapy. Expression of a panel of druggable, metastasis-related biomarkers was immunohistochemically analyzed in 53 liver (LM) and 15 lung metastases (LuM) and correlated with survival. Differential expression between LM and LuM was observed for the growth factor receptors IGF1R (LuM 92.3% vs. LM 75.8%, p = 0.013), EGFR (LuM 68% vs. LM 41.5%, p = 0.004), the cell adhesion molecules CD44v6 (LuM 55.7% vs. LM 34.9%, p = 0.019) and α2β1 (LuM 88.3% vs. LM 58.5%, p = 0.001) and the check point molecule PD-L1 (LuM 6.1% vs. LM 3.3%, p = 0.005). Contrary, expression of HGFR, Hsp90, Muc1, Her2/neu, ERα and PR was comparable in LuM and LM. In the LM cohort (n = 52), a high CD44v6 expression was identified as an independent factor of poor prognosis (PFS: HR 2.37, 95% CI 1.18–4.78, p = 0.016). High co-expression of CD44v6/α2β1 (HR 4.14, 95% CI 1.65–10.38, p = 0.002) and CD44v6/PD-L1 (HR 2.88, 95% CI 1.21–6.85, p = 0.017) indicated early recurrence after hepatectomy, in a substantial number of patients (CD44v6/α2β1: 11 (21.15%) patients; CD44v6/PD-L1: 12 (23.1%) patients). Dual expression of druggable protein biomarkers may refine prognostic prediction and stratify high-risk patients for new therapeutic concepts, depending on the metastatic location.

Graphical Abstract

1. Introduction

According to international guidelines [1,2,3], metastasectomy currently offers the best chance for long-term survival for selected colorectal cancer patients. Additional standard chemotherapy for patients with resectable liver metastases resulted in the prolongation of disease-free survival (DFS) and progression-free survival (PFS) but revealed no significant improvement in overall survival (OS) [4,5]. In patients with resectable pulmonary metastases, the outcome of peri-operative chemotherapy is inconclusive [6,7]. However, despite curative-intent metastasectomy, more than half of the patients suffer recurrence [8,9]. This highlights the urgent need for the implementation of new strategies to identify high-risk patients suitable for personalized therapy, aiming to improve treatment outcome and survival [10].
Colorectal cancer preferentially metastasizes to the liver, followed by the lung and the peritoneum and, more rarely, in bone, ovary and the brain [11,12,13]. The metastatic pattern depends on the sidedness of the primary colorectal tumor. Elucidating the underlying mechanisms of the metastatic organotropism, profound molecular differences were observed between right-sided and left-sided CRC cancers. Similarly, the tumor microenvironment seems to have a deep impact on the metastatic site [14]. Indeed, for primary metastatic colorectal cancer, a growing body of molecular data is available, resulting in the continuous development of targeted therapies and improvement in survival [15,16].
Comparative analysis of primary CRC and corresponding metastatic sites revealed maintenance of the main driver mutations in both liver and lung metastases, some of which are approved for CRC therapy, such as RAS, BRAF and MSI [17,18,19]. In contrast, genomic [20,21,22], transcriptomic [23] and proteomic [24] profiling identified molecular differences between primary tumor, liver and lung metastases that might have potential therapeutic implications for specific metastatic sites. Moreover, distant metastases in different organs revealed discordant responses to standard chemotherapy [25], all together, supporting the concept of inter- and intratumor heterogeneity, which is one of the key factors in tumor progression, therapeutic resistance, and poor patient outcome.
In the present study, a panel of protein biomarkers was selected, which drive the complex metastatic process of primary colorectal cancer and lead to poor prognosis. In contrast, little information is available on the expression pattern of these prognostic factors in liver and lung metastases. The protein biomarker panel encompassed the growth factor receptors epidermal growth factor receptor (EGF-R) and hepatocyte growth factor receptor (HGF-R) [26], human epidermal growth factor receptor (Her2/neu) [27], insulin-like growth factor 1 receptor (IGF-1R) [28], estrogen receptor alpha (Erα) [29] and progesterone receptor (PR) [30], the cell adhesion molecules CD44v6 [31], Muc1 [32] and integrin α2β1 [33], the chaperone heat shock protein 90 (Hsp90) [34], and the immune checkpoint molecule programmed death ligand 1 (PD-L1) [35]. Interestingly, the protein biomarkers selected are drug targets, for which drugs are already approved or for which clinical trials are ongoing, in primary colorectal cancer or other cancer types. This could open up new options for second and further line treatments in colorectal cancer.
The present study aimed (1) to identify the phenotypic heterogeneity in tumor biology between colorectal liver and lung metastases and (2) to stratify patients with a high risk for early recurrence after hepatic metastasectomy.

2. Materials and Methods

2.1. Patient Cohort

The patient cohort consists of 68 patients with metastatic colorectal cancer, receiving metastasectomy with curative intent at the Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University, Munich, Germany. A liver metastasis (LM, n = 53) or a lung metastasis (LuM, n = 15) was analyzed from each patient. Double-coded tissues and the corresponding data used in this study were provided by the Biobank of the Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany. This Biobank operates under the administration of the Human Tissue and Cell Research (HTCR) Foundation. The framework of HTCR Foundation, which includes obtaining written informed consent from all donors, has been approved by the ethics commission of the Faculty of Medicine at the LMU (approval number 025-12) as well as the Bavarian State Medical Association (approval number 11142) in Germany. All liver metastases were diagnosed as the first relapse of the individual patient. Lung metastases represented first (n = 3), second (n = 8) and later stage relapse (n = 4). Survival analysis was performed for 52 patients diagnosed with liver metastases. One patient was lost to follow up. Follow-up period of the patient cohort was from December 2010 until February 2018.

2.2. Immunohistochemistry and Evaluation of Biomarker Expression

Fresh tumor samples including adjacent benign reference tissue were collected according to international biobanking standards. After surgery the tumor samples were immediately snap frozen in liquid nitrogen. Serial cryosections (5 µm) were performed and air dried over night at room temperature. Sections were either fixed in acetone, or for the ERα und PR staining in formalin solution (10%). Immunohistochemistry was performed using the standard avidin-biotin-peroxidase complex method [36,37,38]. Briefly, unspecific Fc receptors were blocked with 10% AB-serum in D-PBS, pH 7.4 for 20 min. Endogenous biotin was blocked using the Avidin-/Biotin-blocking Kit for 15 min. The primary antibodies (Table 1) were incubated for one hour. Some antibodies were detected with the secondary biotinylated antibody (111-065-114; wc 7.0 µg/mL; JacksonImmunoResearch, West Grove, PA, USA for anti-rabbit and 315-065-048; wc 0.75 µg/mL; JacksonImmunoResearch for anti-mouse) for 30 min, followed by the peroxidase-conjugated streptavidin (016-030-084; wc 1.0 µg/mL; affymetrix eBiosciences, Santa Clara, CA, USA) for another 30 min. Other primary antibodies were detected with the amplification Kit ZytoChem Plus (HRP060; Zytomed Systems, Bargteheide, Germany) according to the instructions of the manufacturer (marked in Table 1 with Kit: +). For visualization of the antigen–antibody reaction all slides were developed in a 3-Amino-9-ethylcarbazole solution containing 35% hydrogen peroxide (AEC staining) for eight minutes in darkness. Counterstaining was performed with Mayer’s hemalum solution. All incubation steps were performed in a humid chamber at room temperature. Specificity of the staining was controlled by the corresponding isotype controls (Table 1). Cancer cells were visualized by EpCAM and pan-cytokeratin expression.
For the evaluation of biomarker expression, the size of the measurement field was standardized using a normalized grid at 100× magnification (Olympus microscope BX50, Olympus, Hamburg, Germany). The biomarker-positive tumor area was determined in relation to the total tumor area. The percentage of biomarker-positive tumor cells was expressed by semiquantitative estimation in 10% increments. Staining results were evaluated by two independent observers (FW, BM). External monitoring was performed by local pathologists (Institute of Pathology, LMU Munich, Munich, Germany, T. Kirchner) and for Her2/neu expression by J. Rüschoff (Institute of Pathology Nordhessen, Kassel, Germany, Rüschoff) [39].
For some biomarkers standardized cut-off values are given, namely ERα and PR [40], Her2/neu [39,41], Muc1 [42,43], and PD-L1 [36,44]. In the absence of standardized cut-offs for other biomarkers, cut-offs were assessed using the biphasic distribution, which was statistically defined using the mean antigen expression in liver or lung metastases. Biomarker expression below the calculated cut-off was defined as low expression, and biomarker expression above the calculated cut-off was defined as high expression. The same cut-off values were used for single biomarker analysis and the evaluation of dual biomarker expression. In addition to the tumor tissue, antigen expression was evaluated on the adjacent benign liver and lung tissues.

2.3. Statistical Analysis

All statistical analyses were performed with IBM SPSS v. 23. Mean biomarker ex-pression between liver and lung metastases was compared using the Mann–Whitney U-test. The prognostic impact of single and dual biomarker expression was evaluated using Kaplan–Meier analysis (log rank test, ‘pairwise over strata’) and multivariate Cox regression analysis (biomarker expression used as ‘categorical covariate’, ‘First’ as reference category). OS was defined as the time from metastasectomy until the last follow-up or death of the patient. PFS was defined as the time from metastasectomy until the next progression. A p-value of ≤0.05 was considered as significant.

3. Results

3.1. Patient Characteristics

In the present study, 53 liver metastases and 15 lung metastases surgically resected from colorectal cancer patients were analyzed. Men were more frequently affected than women (LM: ratio 1.79:1; LuM: ratio 4:1). Most (66.04%) liver metastases were detected at primary diagnosis (synchronous), whereas all lung metastases were documented at a later time (metachronous). Liver and lung metastases were diagnosed as single organ metastases. However, at the organ site, tumor disease was frequently extensive (number of nodules within the metastatic organ >1; LM: 64.15%, LuM: 53.33%; multilobular involvement; LM: 56.6%, LuM: 66.67%). Still, most patients were resected with curative intent (R0; LM: 73.58%, LuM: 80%). Further, 32 of 53 (60.38%) patients diagnosed with liver metastases received first-line chemotherapy (5-FU as single agent: 34.38%, oxaliplatin-based: 43.75%, irinotecan-based: 15.63%, others: 6.25%) and 23 of 53 (43.40%) received neoadjuvant chemotherapy before liver metastasectomy. Of these, 10 of 15 (66.67%) patients were treated with front line chemotherapy (5-FU as single agent: 10%, oxaliplatin-based: 80%, others: 10%) and 8 of 15 (53.33%) patients received neoadjuvant chemotherapy, right before surgery of the lung metastasis studied. Complete treatment records were not available for all patients with lung metastases.
Patient characteristics are summarized in detail in Table 2.
Survival analysis was performed in the patient cohort with liver metastases but was omitted in patients with lung metastases because of small sample size. Patients diagnosed with multiple (>1) LM had a significantly shorter PFS compared to patients diagnosed with a single liver metastasis (multiple metastases, PFS: 6.5 months; single metastasis, PFS: 10 months; log-rank, p = 0.014). Patients with synchronous LM relapsed much faster compared to patients with metachronous LM (synchronous, PFS: 7 months; metachronous, PFS: 16 months; log rank, p = 0.001). None of the patient characteristics revealed an impact on OS.

3.2. Differential Biomarker Expression in Colorectal Liver and Lung Metastases

Liver and lung metastases were comparatively analyzed with a panel of metastasis-related protein biomarkers. A differential expression pattern between liver and lung metastases was observed for the growth factor receptors IGF-1R (LuM 92.3% vs. LM 75.8%, p = 0.013) and EGF-R (LuM 68% vs. LM 41.5%, p = 0.004), showing a significantly higher fraction of positive cancer cells in the lung metastases, respectively. Similar results were obtained for the cell adhesion molecules CD44v6 (LuM 55.7% vs. LM 34.9%, p = 0.019) and integrin α2β1 (LuM 88.3% vs. LM 58.5%, p = 0.001), as well as for the check point molecule PD-L1 (LuM 6.1% vs. LM 3.3%, p = 0.005). In contrast, no significant difference was observed for the growth factor receptor HGF-R and the chaperon molecule Hsp90, both showing a high fraction of positive cancer cells in almost all distant metastases. Conversely, all but one metastatic lesion were found negative for the hormone receptors ERα and PR. One individual liver metastasis demonstrated 30% ERα positive cancer cells. Moreover, in colorectal liver and lung metastases, a minor fraction of the cancer cells were found positive for the cell adhesion molecule Muc1 and growth factor receptor Her2/neu. In fact, only one liver metastasis (60% Her2/neu positive cancer cells) qualified for anti-Her2/neu therapy. The number of biomarker-positive lesions and the means of biomarker expression are given in Table 3. The distribution of biomarker expression is shown for liver and lung metastases (Figure 1).
Biomarker analysis showed most of the benign liver tissues positive for HGF-R, EGF-R, and Hsp90. IGF-1R and PD-L1 were detected in a fraction of benign liver samples (IGF-1R: 11 out of 52, 21.2%; PD-L1: 10 out of 52, 19.2%). Interestingly, benign liver tissue was negative for Muc1, CD44v6 and the integrin α2β1. In contrast, all biomarkers tested were detected on benign lung tissue, although the integrin α2β1 (10 out of 15, 66.6%) and Muc1 (8 out of 15, 53%) were observed on a reduced number of adjacent lung tissues. Data obtained in benign tissue samples are summarized in Table S1. Figure 2 demonstrates the significantly different staining patterns by each biomarker of liver and lung metastases.

3.3. Prognostic Impact of Biomarker Expression in Colorectal Liver Metastases

The prognostic impact of the biomarkers was analyzed in patients with liver metastases. CD44v6, but none of the other biomarkers tested, was identified as an indicator for early recurrence. Liver metastases with a high fraction (>30%, n = 22) of CD44v6+ tumor cells significantly correlated with a shorter (median 7.0 months) PFS compared to LM with a low CD44v6 expression (≤30% CD44v6+ cells, n = 30; median 15.5 months; log rank p = 0.01). Recurrent liver metastases with a high proportion of CD44v6+ cancer cells showed more frequent multi-organ metastases (6 out of 19, 31.58%), compared to liver metastases with a low proportion of CD44v6+ cancer cells (3 out of 22, 13.65%). Almost all multi-organ metastases involved liver and lung, regardless of the extent of CD44v6 expression. Cox regression analysis confirmed the independent prognostic impact of CD44v6 on PFS (Table 4). No significant correlation was found between CD44v6 expression in LM and OS.

3.4. CD44v6-Related Dual Biomarker Expression in Colorectal Liver Metastases

Co-expression analysis was performed on CD44v6 and the metastasis-related biomarkers. Univariate analysis identified three pairs of highly expressed biomarkers associated with short PFS. Patients with liver metastases with strong expression of CD44v6 and integrin α2β1 showed a shorter mean PFS (3 months) compared to the group with only high expression of CD44v6 (7 months) (Table 5, Figure 3). Multivariate Cox regression analysis identified the combination of a high CD44v6 and a high integrin α2β1 expression (HR: 4.135, 95% CI: 1.648–10.375, p = 0.002) and the combination of a high CD44v6 and a high PD-L1 expression (HR: 2.882, 95% CI: 1.213–6.848, p = 0.017), as independent prognostic factors for short progression-free survival (Table 6). High co-expression was detected in a substantial number of patients; i.e., CD44v6 high (>30% positive tumors cells) combined with integrin α2β1 high (>80% positive tumor cells) in 11 out of 52 (21.15%) patients, CD44v6 high combined with Hsp90 high (>70% positive tumor cells) in 14 out of 52 (26.92%) patients and CD44v6 high combined with PD-L1 high (>1% positive cells) in 12 out of 52 (23.1%) patients.

4. Discussion

It is well published that primary colorectal cancer differs in its biology, depending on sidedness [45]. This also includes treatment-relevant characteristics, such as the RAS [46,47], MSI [48] and BRAF status [49]. In the present study, biomarker heterogeneity was identified between colorectal liver and lung metastases, namely for the cell adhesion molecules α2β1, CD44v6, the growth factor receptors IGF-1R, EGF-R and the immune checkpoint biomarker PD-L1. These site-specific differences in biomarker expression might reflect the complex multifactorial interactions between disseminated cancer cells and the target organ microenvironment [50]. Cancer cells with a unique tumor biology are homing to metastatic niches with a microenvironment promoting colonization, survival, and proliferation [51,52]. Liver and lung metastases reveal biological differences; for example, in the cellular composition of the microenvironment [36,52,53,54], the ECM signature [52,55,56] and the secretome profile [52,57]. Quantitative differences in protein biomarker expression were found between liver and lung metastases, showing a significantly higher proportion of IGF-1R-, EGR-R-, CD44v6-, α2β1-, and PD-L1-positive cancer cells in the lung. This observation confirms published data, showing a higher frequency of genetic drivers, such as KRAS alterations and MET amplification in lung metastases [20,58]. At the same time, lung metastases exhibit an increased immunosuppressive microenvironment and prometastatic inflammation [36,59]. These findings suggest distinct colonization mechanisms, involving both specific cancer cells with a higher propensity to metastasize to the lung and a lung-specific environment that facilitates metastasis of specific cancer cells. Targeting metastasis-relevant biomarker expression will open up new therapeutic opportunities, adjusted to specific metastatic localizations. This is in deep contrast to the current guideline, which recommends the concept of treating distant metastasis with the same therapy, independent from the metastatic organ site.
The protein biomarker expression pattern in liver metastases was tested for prognostic relevance. A high (>30%) fraction of CD44v6+ liver metastatic cells was identified as an independent prognostic factor mediating short progression-free survival. This finding supports CD44v6 as a metastatic driver. Multiple underlying molecular mechanisms have been described for CD44v6-mediated progression in colorectal cancer. Examples are interactions with the extracellular matrix components osteopontin and hyaluronic acid and the binding of different cytokines, such as HGF, EGF and VEGF [31,60]. Co-expression analysis identified two new independent risk factors associated with poor prognosis of CRC patients with liver metastases. Most interesting, high dual expression of CD44v6 and integrin α2β1 represents an indicator of early recurrence, defined as tumor relapse within six months after liver resection for colorectal metastases [61,62]. Direct and extracellular matrix-mediated molecular crosstalk between CD44v6 and various integrins, including α2β1, was found to promote cancer cell proliferation and invasion, tumor angiogenesis and chemoresistance, all involved in a considerable shortening of progression-free survival compared to the single CD44v6 expression [63,64,65]. In addition, dual expression of CD44v6 and PD-L1, indicating the crosstalk between tumor cells and the tumor microenvironment, significantly correlated with short survival. The subset of CD44v6+ colorectal cancers simultaneously expressing PD-L1 might represent stem-like properties and contributes to immune evasion mediating poor prognosis [66,67]. Similarly, co-mutations in RAS, TP53 and SMAD4, as well as in APC and PIK3CA, resulted in a worse outcome after hepatectomy compared to single mutations [19]. Therefore, our findings support the strategy of combining prognostic protein biomarkers to render the prediction of outcome more precise [68,69]. Further, these new factors might be included in clinical risk scores, similar as reported for the KRAS status in the GAME score [70] and the KRAS/NRAS/BRAF status in the CERR score [71], which resulted in the refinement to predict recurrence after resection of CRC liver metastases. In contrast to some of the most investigated therapeutic biomarkers, namely BRAF, MSI-high, and Her2/neu, all detected in a very small patient cohort [19,20], dual expression of the druggable targets CD44v6/α2β1 and CD44v6/PD-L1 was identified in about 20% of the liver metastatic patients.
In addition, these novel findings might have an impact on the development of new therapeutic strategies for liver metastatic CRC patients. Currently, new anti-CD44v6 treatment strategies, such as half antibodies conjugated nanoparticles [72], peptides (NCT03009214) and CD44v6-specific CAR gene-engineered T cells (NCT04427449, [73]) are under investigation and might also become a treatment option for CRC patients with CD44v6-positive liver metastases. Combination of two biomarkers might help to stratify patients more precisely for targeted therapy compared to single biomarker expression. For example, Shek et al., 2021, reported that only a subgroup of PD-L1-positive mCRCs responded to checkpoint inhibitor therapy [74]. In addition, dual expression of druggable biomarkers will further promote the promising concept of multiple target inhibition, aiming to improve treatment outcome and reduce the risk of drug resistance. Recently, the combination of the BRAF inhibitor Encorafenib with the EGF-R inhibitor Cetuximab has been reported as the new standard for the treatment of metastatic BRAF-mutated colorectal cancer [75]. Currently, a number of clinical trials are ongoing in advanced colorectal cancer, simultaneously inhibiting different targets. This includes combination therapy of the EGF-R inhibitor Panitumumab with the multi-kinase inhibitor Cabozantinib [76]. Further, anti-PD-L1 checkpoint inhibitors have been combined with targeted therapies, aiming to improve the response to immunotherapy [77]. In the present study, dual expression of PD-L1 and CD44v6 was found to correlate with poor prognosis and might represent a new therapeutic option for combination therapy. The second interesting pair of therapeutic targets identified in the present study was the co-expression of CD44v6 and the integrin α2β1. Both cell adhesion molecules were found to mediate chemoresistance [65,78]. Simultaneous inhibition of both targets might result in the circumvention of chemoresistance and represent a new anti-metastatic strategy of targeted therapy. Consideration of metastasis-driving protein biomarkers that predict early recurrence after hepatectomy might play a critical role in the clinical management of patients diagnosed with liver metastases [79]. The findings in the present study need to be confirmed in a larger, prospective trial.

5. Conclusions

A differential expression pattern of the druggable protein biomarkers α2β1, CD44v6, IGF-1R, EGF-R and PD-L1 was identified between colorectal liver and lung metastases. High expression of CD44v6, CD44v6/α2β1, and CD44v6/PD-L1 correlated significantly with early recurrence after hepatic metastasectomy. Dual biomarker expression may render the prognostic prediction more precise and stratify high-risk patients for new therapeutic concepts, depending on the metastatic organ site.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers14081939/s1, Table S1: Positivity and Distribution of Biomarkers in Benign Tissue of Liver and Lung Metastases.

Author Contributions

Conceptualization, B.M.; methodology, K.D. and F.W.; validation, B.M. and K.D.; investigation, F.W. and B.M.; resources, B.M.; data curation, F.W. and M.P.; writing—original draft preparation, F.W. and B.M.; writing—review and editing, K.D., J.W. and M.P.; visualization, F.W.; supervision, B.M.; project administration, B.M.; funding acquisition, B.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the German Federal Ministry of Education and Research, Leading Edge Cluster m4 (B.M.) under Grant FKZ 16EX1021N.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Biobank of the Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany. This Biobank operates under the administration of the Human Tissue and Cell Research (HTCR) Foundation. The framework of the HTCR Foundation has been approved by the ethics commission of the Faculty of Medicine at the LMU (approval number 025-12), as well as the Bavarian State Medical Association (approval number 11142) in Germany.

Informed Consent Statement

Informed consent from all patients was obtained by the HTCR Foundation.

Data Availability Statement

Data corresponding to the analyzed tissues were delivered in anonymized form by the HTCR Foundation.

Acknowledgments

We thank the staff members of the Biobank, especially Maresa Demmel, Nadine Gese, Ute Bossmanns and Beatrice Rauter for tissue sample/data organization, Michael Pohr for technical support and Robin v. Holzschuher for providing language help.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Benson, A.B.; Venook, A.P.; Al-Hawary, M.M.; Arain, M.A.; Chen, Y.J.; Ciombor, K.K.; Cohen, S.; Cooper, H.S.; Deming, D.; Farkas, L.; et al. Colon Cancer, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2021, 19, 329–359. [Google Scholar] [CrossRef] [PubMed]
  2. 2020 Exceptional Surveillance of Colorectal Cancer (NICE Guideline NG151); Copyright © NICE 2020; National Institute for Health and Care Excellence: London, UK, 2020.
  3. Van Cutsem, E.; Cervantes, A.; Adam, R.; Sobrero, A.; Van Krieken, J.H.; Aderka, D.; Aranda Aguilar, E.; Bardelli, A.; Benson, A.; Bodoky, G.; et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2016, 27, 1386–1422. [Google Scholar] [CrossRef] [PubMed]
  4. Nordlinger, B.; Sorbye, H.; Glimelius, B.; Poston, G.J.; Schlag, P.M.; Rougier, P.; Bechstein, W.O.; Primrose, J.N.; Walpole, E.T.; Finch-Jones, M.; et al. Perioperative FOLFOX4 chemotherapy and surgery versus surgery alone for resectable liver metastases from colorectal cancer (EORTC 40983): Long-term results of a randomised, controlled, phase 3 trial. Lancet Oncol. 2013, 14, 1208–1215. [Google Scholar] [CrossRef]
  5. Kanemitsu, Y.; Shimizu, Y.; Mizusawa, J.; Inaba, Y.; Hamaguchi, T.; Shida, D.; Ohue, M.; Komori, K.; Shiomi, A.; Shiozawa, M.; et al. Hepatectomy Followed by mFOLFOX6 Versus Hepatectomy Alone for Liver-Only Metastatic Colorectal Cancer (JCOG0603): A Phase II or III Randomized Controlled Trial. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2021, 39, 3789–3799. [Google Scholar] [CrossRef] [PubMed]
  6. Guerrera, F.; Falcoz, P.E.; Renaud, S.; Massard, G. Does perioperative chemotherapy improve survival in patients with resectable lung metastases of colorectal cancer? Interact. Cardiovasc. Thorac. Surg. 2017, 24, 789–791. [Google Scholar] [CrossRef] [Green Version]
  7. Li, Y.; Qin, Y. Peri-operative chemotherapy for resectable colorectal lung metastasis: A systematic review and meta-analysis. J. Cancer. Res. Clin. Oncol. 2020, 146, 545–553. [Google Scholar] [CrossRef] [Green Version]
  8. Martin, J.; Petrillo, A.; Smyth, E.C.; Shaida, N.; Khwaja, S.; Cheow, H.K.; Duckworth, A.; Heister, P.; Praseedom, R.; Jah, A.; et al. Colorectal liver metastases: Current management and future perspectives. World J. Clin. Oncol. 2020, 11, 761–808. [Google Scholar] [CrossRef]
  9. Lang, H.; Baumgart, J.; Roth, W.; Moehler, M.; Kloth, M. Cancer gene related characterization of patterns and point of recurrence after resection of colorectal liver metastases. Ann. Transl. Med. 2021, 9, 1372. [Google Scholar] [CrossRef]
  10. Filip, S.; Vymetalkova, V.; Petera, J.; Vodickova, L.; Kubecek, O.; John, S.; Cecka, F.; Krupova, M.; Manethova, M.; Cervena, K.; et al. Distant Metastasis in Colorectal Cancer Patients-Do We Have New Predicting Clinicopathological and Molecular Biomarkers? A Comprehensive Review. Int. J. Mol. Sci. 2020, 21, 5255. [Google Scholar] [CrossRef]
  11. Riihimaki, M.; Hemminki, A.; Sundquist, J.; Hemminki, K. Patterns of metastasis in colon and rectal cancer. Sci. Rep. 2016, 6, 29765. [Google Scholar] [CrossRef] [Green Version]
  12. Gao, Y.; Bado, I.; Wang, H.; Zhang, W.; Rosen, J.M.; Zhang, X.H. Metastasis Organotropism: Redefining the Congenial Soil. Dev. Cell 2019, 49, 375–391. [Google Scholar] [CrossRef] [PubMed]
  13. Pretzsch, E.; Bösch, F.; Neumann, J.; Ganschow, P.; Bazhin, A.; Guba, M.; Werner, J.; Angele, M. Mechanisms of Metastasis in Colorectal Cancer and Metastatic Organotropism: Hematogenous versus Peritoneal Spread. J. Oncol. 2019, 2019, 7407190. [Google Scholar] [CrossRef] [PubMed]
  14. Chandra, R.; Karalis, J.D.; Liu, C.; Murimwa, G.Z.; Voth Park, J.; Heid, C.A.; Reznik, S.I.; Huang, E.; Minna, J.D.; Brekken, R.A. The Colorectal Cancer Tumor Microenvironment and Its Impact on Liver and Lung Metastasis. Cancers 2021, 13, 6206. [Google Scholar] [CrossRef] [PubMed]
  15. Dekker, E.; Tanis, P.J.; Vleugels, J.L.A.; Kasi, P.M.; Wallace, M.B. Colorectal cancer. Lancet 2019, 394, 1467–1480. [Google Scholar] [CrossRef]
  16. Imyanitov, E.; Kuligina, E. Molecular testing for colorectal cancer: Clinical applications. World J. Gastrointest. Oncol. 2021, 13, 1288–1301. [Google Scholar] [CrossRef]
  17. Bhullar, D.S.; Barriuso, J.; Mullamitha, S.; Saunders, M.P.; O’Dwyer, S.T.; Aziz, O. Biomarker concordance between primary colorectal cancer and its metastases. eBioMedicine 2019, 40, 363–374. [Google Scholar] [CrossRef] [Green Version]
  18. Testa, U.; Castelli, G.; Pelosi, E. Genetic Alterations of Metastatic Colorectal Cancer. Biomedicines 2020, 8, 414. [Google Scholar] [CrossRef]
  19. Diener, M.K.; Fichtner-Feigl, S. Biomarkers in colorectal liver metastases: Rising complexity and unknown clinical significance? Ann. Gastroenterol. Surg. 2021, 5, 477–483. [Google Scholar] [CrossRef]
  20. Wang, Z.; Zheng, X.; Wang, X.; Chen, Y.; Li, Z.; Yu, J.; Yang, W.; Mao, B.; Zhang, H.; Li, J.; et al. Genetic differences between lung metastases and liver metastases from left-sided microsatellite stable colorectal cancer: Next generation sequencing and clinical implications. Ann. Transl. Med. 2021, 9, 967. [Google Scholar] [CrossRef]
  21. Jiang, B.; Mu, Q.; Qiu, F.; Li, X.; Xu, W.; Yu, J.; Fu, W.; Cao, Y.; Wang, J. Machine learning of genomic features in organotropic metastases stratifies progression risk of primary tumors. Nat. Commun. 2021, 12, 6692. [Google Scholar] [CrossRef]
  22. Puccini, A.; Seeber, A.; Xiu, J.; Goldberg, R.M.; Soldato, D.; Grothey, A.; Shields, A.F.; Salem, M.E.; Battaglin, F.; Berger, M.D.; et al. Molecular differences between lymph nodes and distant metastases compared with primaries in colorectal cancer patients. NPJ Precis. Oncol. 2021, 5, 95. [Google Scholar] [CrossRef] [PubMed]
  23. Eide, P.W.; Moosavi, S.H.; Eilertsen, I.A.; Brunsell, T.H.; Langerud, J.; Berg, K.C.G.; Røsok, B.I.; Bjørnbeth, B.A.; Nesbakken, A.; Lothe, R.A.; et al. Metastatic heterogeneity of the consensus molecular subtypes of colorectal cancer. NPJ Genom. Med. 2021, 6, 59. [Google Scholar] [CrossRef] [PubMed]
  24. Fahrner, M.; Bronsert, P.; Fichtner-Feigl, S.; Jud, A.; Schilling, O. Proteome biology of primary colorectal carcinoma and corresponding liver metastases. Neoplasia 2021, 23, 1240–1251. [Google Scholar] [CrossRef]
  25. Vigano, L.; Corleone, P.; Darwish, S.S.; Turri, N.; Famularo, S.; Viggiani, L.; Rimassa, L.; Del Fabbro, D.; Di Tommaso, L.; Torzilli, G. Hepatic and Extrahepatic Colorectal Metastases Have Discordant Responses to Systemic Therapy. Pathology Data from Patients Undergoing Simultaneous Resection of Multiple Tumor Sites. Cancers 2021, 13, 464. [Google Scholar] [CrossRef] [PubMed]
  26. Xie, Y.H.; Chen, Y.X.; Fang, J.Y. Comprehensive review of targeted therapy for colorectal cancer. Signal. Transduct. Target. 2020, 5, 22. [Google Scholar] [CrossRef] [PubMed]
  27. Siena, S.; Sartore-Bianchi, A.; Marsoni, S.; Hurwitz, H.I.; McCall, S.J.; Penault-Llorca, F.; Srock, S.; Bardelli, A.; Trusolino, L. Targeting the human epidermal growth factor receptor 2 (HER2) oncogene in colorectal cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2018, 29, 1108–1119. [Google Scholar] [CrossRef] [PubMed]
  28. Oliveres, H.; Pesántez, D.; Maurel, J. Lessons to Learn for Adequate Targeted Therapy Development in Metastatic Colorectal Cancer Patients. Int. J. Mol. Sci. 2021, 22, 5019. [Google Scholar] [CrossRef]
  29. Liang, R.; Lin, Y.; Yuan, C.L.; Liu, Z.H.; Li, Y.Q.; Luo, X.L.; Ye, J.Z.; Ye, H.H. High expression of estrogen-related receptor α is significantly associated with poor prognosis in patients with colorectal cancer. Oncol. Lett. 2018, 15, 5933–5939. [Google Scholar] [CrossRef] [Green Version]
  30. Ye, S.B.; Cheng, Y.K.; Zhang, L.; Wang, X.P.; Wang, L.; Lan, P. Prognostic value of estrogen receptor-α and progesterone receptor in curatively resected colorectal cancer: A retrospective analysis with independent validations. BMC Cancer 2019, 19, 933. [Google Scholar] [CrossRef] [Green Version]
  31. Ma, L.; Dong, L.; Chang, P. CD44v6 engages in colorectal cancer progression. Cell Death Dis. 2019, 10, 30. [Google Scholar] [CrossRef]
  32. Li, C.; Zuo, D.; Liu, T.; Yin, L.; Li, C.; Wang, L. Prognostic and Clinicopathological Significance of MUC Family Members in Colorectal Cancer: A Systematic Review and Meta-Analysis. Gastroenterol Res. Pr. 2019, 2019, 2391670. [Google Scholar] [CrossRef] [PubMed]
  33. Hou, S.; Wang, J.; Li, W.; Hao, X.; Hang, Q. Roles of Integrins in Gastrointestinal Cancer Metastasis. Front Mol. Biosci. 2021, 8, 708779. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, S.; Guo, S.; Li, Z.; Li, D.; Zhan, Q. High expression of HSP90 is associated with poor prognosis in patients with colorectal cancer. Peer J. 2019, 7, e7946. [Google Scholar] [CrossRef]
  35. Oliveira, A.F.; Bretes, L.; Furtado, I. Review of PD-1/PD-L1 Inhibitors in Metastatic dMMR/MSI-H Colorectal Cancer. Front Oncol. 2019, 9, 396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Schlueter, F.; Doetzer, K.; Pruefer, M.; Bazhin, A.V.; Werner, J.; Mayer, B. Integrating Routine Clinical Factors to Stratify Colorectal Cancer Patients with Liver and Lung Metastases for Immune Therapy. J. Cancer Sci. Clin. Ther. 2021, 5, 49–62. [Google Scholar] [CrossRef]
  37. Dotzer, K.; Schluter, F.; Schoenberg, M.B.; Bazhin, A.V.; von Koch, F.E.; Schnelzer, A.; Anthuber, S.; Grab, D.; Czogalla, B.; Burges, A.; et al. Immune Heterogeneity Between Primary Tumors and Corresponding Metastatic Lesions and Response to Platinum Therapy in Primary Ovarian Cancer. Cancers 2019, 11, 1250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Mayer, B.; Lorenz, C.; Babic, R.; Jauch, K.W.; Schildberg, F.W.; Funke, I.; Johnson, J.P. Expression of leukocyte cell adhesion molecules on gastric carcinomas: Possible involvement of LFA-3 expression in the development of distant metastases. Int. J. Cancer 1995, 64, 415–423. [Google Scholar] [CrossRef]
  39. Rüschoff, J.; Dietel, M.; Baretton, G.; Arbogast, S.; Walch, A.; Monges, G.; Chenard, M.P.; Penault-Llorca, F.; Nagelmeier, I.; Schlake, W.; et al. HER2 diagnostics in gastric cancer-guideline validation and development of standardized immunohistochemical testing. Virchows Arch. Int. J. Pathol. 2010, 457, 299–307. [Google Scholar] [CrossRef] [Green Version]
  40. Sieh, W.; Kobel, M.; Longacre, T.A.; Bowtell, D.D.; deFazio, A.; Goodman, M.T.; Hogdall, E.; Deen, S.; Wentzensen, N.; Moysich, K.B.; et al. Hormone-receptor expression and ovarian cancer survival: An Ovarian Tumor Tissue Analysis consortium study. Lancet Oncol. 2013, 14, 853–862. [Google Scholar] [CrossRef] [Green Version]
  41. Wolff, A.C.; Hammond, M.E.H.; Allison, K.H.; Harvey, B.E.; Mangu, P.B.; Bartlett, J.M.; Bilous, M.; Ellis, I.O.; Fitzgibbons, P.; Hanna, W. Human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline focused update. Arch. Pathol. Lab. Med. 2018, 142, 1364–1382. [Google Scholar] [CrossRef] [Green Version]
  42. Nagai, S.; Takenaka, K.; Sonobe, M.; Ogawa, E.; Wada, H.; Tanaka, F. A novel classification of MUC1 expression is correlated with tumor differentiation and postoperative prognosis in non-small cell lung cancer. J. Thorac. Oncol. 2006, 1, 46–51. [Google Scholar]
  43. Zeng, Y.; Zhang, Q.; Zhang, Y.; Lu, M.; Liu, Y.; Zheng, T.; Feng, S.; Hao, M.; Shi, H. MUC1 Predicts Colorectal Cancer Metastasis: A Systematic Review and Meta-Analysis of Case Controlled Studies. PLoS ONE 2015, 10, e0138049. [Google Scholar] [CrossRef] [PubMed]
  44. Korehisa, S.; Oki, E.; Iimori, M.; Nakaji, Y.; Shimokawa, M.; Saeki, H.; Okano, S.; Oda, Y.; Maehara, Y. Clinical significance of programmed cell death-ligand 1 expression and the immune microenvironment at the invasive front of colorectal cancers with high microsatellite instability. Int. J. Cancer 2018, 142, 822–832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Baran, B.; Mert Ozupek, N.; Yerli Tetik, N.; Acar, E.; Bekcioglu, O.; Baskin, Y. Difference Between Left-Sided and Right-Sided Colorectal Cancer: A Focused Review of Literature. Gastroenterol. Res. 2018, 11, 264–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Zihui Yong, Z.; Ching, G.T.H.; Ching, M.T.C. Metastatic Profile of Colorectal Cancer: Interplay Between Primary Tumor Location and KRAS Status. J. Surg. Res. 2020, 246, 325–334. [Google Scholar] [CrossRef] [PubMed]
  47. Xue, X.; Li, X.; Pan, Z.; Zhao, L.; Ding, Y. Comparison of clinicopathological features and KRAS gene mutation of left-sided and right-sided colon cancers. Int. J. Clin. Exp. Pathol. 2017, 10, 11353–11359. [Google Scholar] [PubMed]
  48. Takahashi, Y.; Sugai, T.; Habano, W.; Ishida, K.; Eizuka, M.; Otsuka, K.; Sasaki, A.; Takayuki, M.; Morikawa, T.; Unno, M.; et al. Molecular differences in the microsatellite stable phenotype between left-sided and right-sided colorectal cancer. Int. J. Cancer 2016, 139, 2493–2501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Brulé, S.Y.; Jonker, D.J.; Karapetis, C.S.; O’Callaghan, C.J.; Moore, M.J.; Wong, R.; Tebbutt, N.C.; Underhill, C.; Yip, D.; Zalcberg, J.R.; et al. Location of colon cancer (right-sided versus left-sided) as a prognostic factor and a predictor of benefit from cetuximab in NCIC CO.17. Eur. J. Cancer 2015, 51, 1405–1414. [Google Scholar] [CrossRef] [PubMed]
  50. Welch, D.R.; Hurst, D.R. Defining the Hallmarks of Metastasis. Cancer Res. 2019, 79, 3011–3027. [Google Scholar] [CrossRef]
  51. Dmello, R.S.; To, S.Q.; Chand, A.L. Therapeutic Targeting of the Tumour Microenvironment in Metastatic Colorectal Cancer. Int. J. Mol. Sci. 2021, 22, 2067. [Google Scholar] [CrossRef]
  52. Altorki, N.K.; Markowitz, G.J.; Gao, D.; Port, J.L.; Saxena, A.; Stiles, B.; McGraw, T.; Mittal, V. The lung microenvironment: An important regulator of tumour growth and metastasis. Nat. Rev. Cancer 2019, 19, 9–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kong, J.; Tian, H.; Zhang, F.; Zhang, Z.; Li, J.; Liu, X.; Li, X.; Liu, J.; Li, X.; Jin, D.; et al. Extracellular vesicles of carcinoma-associated fibroblasts creates a pre-metastatic niche in the lung through activating fibroblasts. Mol. Cancer 2019, 18, 175. [Google Scholar] [CrossRef] [PubMed]
  54. Gonzalez-Zubeldia, I.; Dotor, J.; Redrado, M.; Bleau, A.M.; Manrique, I.; de Aberasturi, A.L.; Villalba, M.; Calvo, A. Co-migration of colon cancer cells and CAFs induced by TGFβ₁ enhances liver metastasis. Cell Tissue Res. 2015, 359, 829–839. [Google Scholar] [CrossRef] [PubMed]
  55. Huang, J.; Pan, C.; Hu, H.; Zheng, S.; Ding, L. Osteopontin-enhanced hepatic metastasis of colorectal cancer cells. PLoS ONE 2012, 7, e47901. [Google Scholar] [CrossRef] [Green Version]
  56. Naba, A.; Clauser, K.R.; Whittaker, C.A.; Carr, S.A.; Tanabe, K.K.; Hynes, R.O. Extracellular matrix signatures of human primary metastatic colon cancers and their metastases to liver. BMC Cancer 2014, 14, 518. [Google Scholar] [CrossRef] [Green Version]
  57. Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef]
  58. Passot, G.; Kim, B.J.; Glehen, O.; Mehran, R.J.; Kopetz, S.E.; Goere, D.; Overman, M.J.; Pocard, M.; Marchal, F.; Conrad, C.; et al. Impact of RAS Mutations in Metastatic Colorectal Cancer After Potentially Curative Resection: Does Site of Metastases Matter? Ann. Surg. Oncol. 2018, 25, 179–187. [Google Scholar] [CrossRef]
  59. García-Mulero, S.; Alonso, M.H.; Pardo, J.; Santos, C.; Sanjuan, X.; Salazar, R.; Moreno, V.; Piulats, J.M.; Sanz-Pamplona, R. Lung metastases share common immune features regardless of primary tumor origin. J. Immunother. Cancer 2020, 8, 491. [Google Scholar] [CrossRef]
  60. Wang, J.L.; Su, W.Y.; Lin, Y.W.; Xiong, H.; Chen, Y.X.; Xu, J.; Fang, J.Y. CD44v6 overexpression related to metastasis and poor prognosis of colorectal cancer: A meta-analysis. Oncotarget 2017, 8, 12866–12876. [Google Scholar] [CrossRef] [Green Version]
  61. Viganò, L.; Capussotti, L.; Lapointe, R.; Barroso, E.; Hubert, C.; Giuliante, F.; Ijzermans, J.N.; Mirza, D.F.; Elias, D.; Adam, R. Early recurrence after liver resection for colorectal metastases: Risk factors, prognosis, and treatment. A LiverMetSurvey-based study of 6025 patients. Ann. Surg. Oncol. 2014, 21, 1276–1286. [Google Scholar] [CrossRef]
  62. Dai, S.; Ye, Y.; Kong, X.; Li, J.; Ding, K. A predictive model for early recurrence of colorectal-cancer liver metastases based on clinical parameters. Gastroenterol. Rep. 2021, 9, 241–251. [Google Scholar] [CrossRef] [PubMed]
  63. Gao, C.; Guo, H.; Downey, L.; Marroquin, C.; Wei, J.; Kuo, P.C. Osteopontin-dependent CD44v6 expression and cell adhesion in HepG2 cells. Carcinogenesis 2003, 24, 1871–1878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Jung, T.; Gross, W.; Zöller, M. CD44v6 coordinates tumor matrix-triggered motility and apoptosis resistance. J. Biol. Chem. 2011, 286, 15862–15874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Naci, D.; Vuori, K.; Aoudjit, F. Alpha2beta1 integrin in cancer development and chemoresistance. Semin Cancer Biol. 2015, 35, 145–153. [Google Scholar] [CrossRef]
  66. Lee, Y.; Shin, J.H.; Longmire, M.; Wang, H.; Kohrt, H.E.; Chang, H.Y.; Sunwoo, J.B. CD44+ Cells in Head and Neck Squamous Cell Carcinoma Suppress T-Cell-Mediated Immunity by Selective Constitutive and Inducible Expression of PD-L1. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 3571–3581. [Google Scholar] [CrossRef] [Green Version]
  67. Lin, C.C.; Liao, T.T.; Yang, M.H. Immune Adaptation of Colorectal Cancer Stem Cells and Their Interaction With the Tumor Microenvironment. Front. Oncol. 2020, 10, 588542. [Google Scholar] [CrossRef]
  68. Chen, K.; Li, Z.; Jiang, P.; Zhang, X.; Zhang, Y.; Jiang, Y.; He, Y.; Li, X. Co-expression of CD133, CD44v6 and human tissue factor is associated with metastasis and poor prognosis in pancreatic carcinoma. Oncol. Rep. 2014, 32, 755–763. [Google Scholar] [CrossRef] [Green Version]
  69. Ueda, A.; Takasawa, A.; Akimoto, T.; Takasawa, K.; Aoyama, T.; Ino, Y.; Nojima, M.; Ono, Y.; Murata, M.; Osanai, M.; et al. Prognostic significance of the co-expression of EGFR and HER2 in adenocarcinoma of the uterine cervix. PLoS ONE 2017, 12, e0184123. [Google Scholar] [CrossRef]
  70. Margonis, G.A.; Sasaki, K.; Gholami, S.; Kim, Y.; Andreatos, N.; Rezaee, N.; Deshwar, A.; Buettner, S.; Allen, P.J.; Kingham, T.P.; et al. Genetic And Morphological Evaluation (GAME) score for patients with colorectal liver metastases. Br. J. Surg. 2018, 105, 1210–1220. [Google Scholar] [CrossRef]
  71. Chen, Y.; Chang, W.; Ren, L.; Chen, J.; Tang, W.; Liu, T.; Jian, M.; Liu, Y.; Wei, Y.; Xu, J. Comprehensive Evaluation of Relapse Risk (CERR) Score for Colorectal Liver Metastases: Development and Validation. Oncologist 2020, 25, e1031–e1041. [Google Scholar] [CrossRef] [Green Version]
  72. Lourenço, B.N.; Pereira, R.F.; Barrias, C.C.; Fischbach, C.; Oliveira, C.; Granja, P.L. Engineering Modular Half-Antibody Conjugated Nanoparticles for Targeting CD44v6-Expressing Cancer Cells. Nanomaterials 2021, 11, 295. [Google Scholar] [CrossRef] [PubMed]
  73. Porcellini, S.; Asperti, C.; Corna, S.; Cicoria, E.; Valtolina, V.; Stornaiuolo, A.; Valentinis, B.; Bordignon, C.; Traversari, C. CAR T Cells Redirected to CD44v6 Control Tumor Growth in Lung and Ovary Adenocarcinoma Bearing Mice. Front. Immunol. 2020, 11, 99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Shek, D.; Akhuba, L.; Carlino, M.S.; Nagrial, A.; Moujaber, T.; Read, S.A.; Gao, B.; Ahlenstiel, G. Immune-Checkpoint Inhibitors for Metastatic Colorectal Cancer: A Systematic Review of Clinical Outcomes. Cancers 2021, 13, 4345. [Google Scholar] [CrossRef] [PubMed]
  75. Tabernero, J.; Grothey, A.; Van Cutsem, E.; Yaeger, R.; Wasan, H.; Yoshino, T.; Desai, J.; Ciardiello, F.; Loupakis, F.; Hong, Y.S.; et al. Encorafenib Plus Cetuximab as a New Standard of Care for Previously Treated BRAF V600E-Mutant Metastatic Colorectal Cancer: Updated Survival Results and Subgroup Analyses from the BEACON Study. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2021, 39, 273–284. [Google Scholar] [CrossRef]
  76. Strickler, J.H.; Rushing, C.N.; Uronis, H.E.; Morse, M.A.; Niedzwiecki, D.; Blobe, G.C.; Moyer, A.N.; Bolch, E.; Webb, R.; Haley, S.; et al. Cabozantinib and Panitumumab for RAS Wild-Type Metastatic Colorectal Cancer. Oncologist 2021, 26, 465-e917. [Google Scholar] [CrossRef]
  77. Ooki, A.; Shinozaki, E.; Yamaguchi, K. Immunotherapy in Colorectal Cancer: Current and Future Strategies. J. Anus Rectum Colon 2021, 5, 11–24. [Google Scholar] [CrossRef]
  78. Pereira, C.; Ferreira, D.; Mendes, N.; Granja, P.L.; Almeida, G.M.; Oliveira, C. Expression of CD44v6-Containing Isoforms Influences Cisplatin Response in Gastric Cancer Cells. Cancers 2020, 12, 858. [Google Scholar] [CrossRef] [Green Version]
  79. Kamphues, C.; Beyer, K.; Margonis, G.A. Prognostic and Therapeutic Implications of Tumor Biology in Colorectal Liver Metastases. Cancers 2021, 14, 88. [Google Scholar] [CrossRef]
Figure 1. Biomarker Expression Pattern of Liver and Lung Metastases. horizontal bars, Means. Each dot represents a metastatic lesion; empty dots represent liver metastases (LM); filled dots represent lung metastases (LuM).
Figure 1. Biomarker Expression Pattern of Liver and Lung Metastases. horizontal bars, Means. Each dot represents a metastatic lesion; empty dots represent liver metastases (LM); filled dots represent lung metastases (LuM).
Cancers 14 01939 g001
Figure 2. Immunohistochemical Staining of Different Biomarkers. Differential biomarker expression between liver (1) and lung (2) metastases demonstrated by immuno-histochemistry. (A), CD44v6; (B), α2β1; (C), PD-L1; (D), IGF-1R; (E), EGFR; Tu, tumor tissue; BT, Benign tissue.
Figure 2. Immunohistochemical Staining of Different Biomarkers. Differential biomarker expression between liver (1) and lung (2) metastases demonstrated by immuno-histochemistry. (A), CD44v6; (B), α2β1; (C), PD-L1; (D), IGF-1R; (E), EGFR; Tu, tumor tissue; BT, Benign tissue.
Cancers 14 01939 g002
Figure 3. Kaplan–Meier Curves of CD44v6-Related Biomarker Expression in Colorectal Liver Metastases. Blue lines, low/low expression; green lines, high/high expression; red lines, high/low and low/high expression; log-rank p-values are given; cut-off values defining high- and low-level expression for the individual biomarker are given in Table 3.
Figure 3. Kaplan–Meier Curves of CD44v6-Related Biomarker Expression in Colorectal Liver Metastases. Blue lines, low/low expression; green lines, high/high expression; red lines, high/low and low/high expression; log-rank p-values are given; cut-off values defining high- and low-level expression for the individual biomarker are given in Table 3.
Cancers 14 01939 g003
Table 1. Antibody Panel for Immunophenotyping of Colorectal Liver and Lung Metastases.
Table 1. Antibody Panel for Immunophenotyping of Colorectal Liver and Lung Metastases.
BiomarkerAntibody/CloneSpeciesIsotypeWorking Concentration (µg/mL)KitSource
HGF-RSp44rabbitIgG12.12-Spring Bioscience/Biomol, Pleasanton, CA, USA
IGF1-R24–31mouseIgG14.0+Invitrogen, Carlsbad, CA, USA
EGR-RH11mouseIgG12.94-Dako, Santa Clara, CA, USA
Her2/neu4B5rabbitIgG11.5-Ventana, Roche, Basel, Switzerland
ErαID5mouseIgG12.5+Dako
PRPgR 636mouseIgG12.5+Dako
Muc1Ma55.2mouseIgG10.5-Monosan, Uden, The Netherlands
CD44v6VFF-18mouseIgG11.0-eBioscience Affymetrix
α2β1BHA2.1mouseIgG12.5-Millipore, Burlington, MA, USA
Hsp90AC88mouseIgG110.0+Abcam, Cambridge, UK
PD-L1MIH1mouseIgG110.0+Affymetrix
Positive controls
EpcamBer-EP4mouseIgG15.0-Dako
Pan CytokeratinKL-1mouseIgG10.32-Zytomed Systems
isotype controls
MOPC-21MOPC-21mouseIgG15.0-Sigma-Aldrich, St. Louis, MO, USA
MOPC-21MOPC-21mouseIgG14.0+Sigma-Aldrich
MOPC-21MOPC-21mouseIgG110.0+Sigma-Aldrich
Rabbit mAbDA1ErabbitIgG12.12-Cell Signaling, Danvers, MA, USA
Table 2. Patient Characteristics.
Table 2. Patient Characteristics.
ParametersLiver MetastasesLung Metastases
n%n%
patient related
sex
male3464.151280.00
female1935.85320.00
age (years)
median64 62
mean64 59
range30–89 37–74
metastasis related
grading
G1/G23981.251173.33
G3918.75426.67
missing5 0
number of metastases *
11935.85746.67
>13464.15853.33
diameter of the largest metastases (cm)
median3.5 1.8
mean4.29 2.25
range1.3–21.7 0.9–3.3
type of metastasis
synchronous3566.0400.00
metachronous1833.615100.00
R-status
R03973.581280.00
R11426.42320.00
distinction of metastasis
unilobular2343.4533.33
multilobular3056.61066.67
anatomical site
left sided713.21746.67
right sided1528.30853.33
both sided3158.49
neoadjuvant chemotherapy #
yes2343.40853.33
no3056.60746.67
therapy options
oxaliplatin-based1147.83112.5
irinotecan-based730.43562.5
others521.74225.0
n, number of patients; R-status, residual status after surgery; *, nodules within the metastatic organ; #, administered directly before metastasectomy.
Table 3. Positivity and Distribution of Biomarkers in Liver and Lung Metastases.
Table 3. Positivity and Distribution of Biomarkers in Liver and Lung Metastases.
BiomarkerNumber of Positive LesionsNumber of Positive Cancer Cells (%)Number of Positive Lesions above Cut-Offs
LiverLungMedian Mean LiverLung
n = 53%n = 15%LiverLungp-ValueLiverLungCut Off *n = 53%n = 15%
HGF-R5298.11510095950.16687.795.3 n.t.
IGF-1R5094.315100901000.01375.892.3>802954.71280
EGF-R4584.91510040700.00441.568.0>502547.21280
Her2/neu1935.8853.3010.5755.71.7>5011.900
ERα11.90000n.t.0.60≥1n.t.
PR000000n.t.00≥1n.t.
Muc12649.1960010.6146.85.9+/−2649.1960
CD44v64584.91510030600.01934.955.7>302343.41066.7
α2β14686.81510070900.00158.588.3>802037.71173.3
Hsp905196.21510075800.47568.773.9>702649.1960
PD-L12445.31386.7010.0056.13.25>12445.31173.3
n, number of patients; n.t., not tested; *, calculation of the cut-offs is given in the Materials and Methods Section.
Table 4. Multivariate Survival Analysis of CD44v6 Expression in Colorectal liver Metastases.
Table 4. Multivariate Survival Analysis of CD44v6 Expression in Colorectal liver Metastases.
VariableGroupsCox Regression
HRp-Value95% CI
age (median in years)>64/≤641.4240.3570.671–3.021
number of metastases *>1/≤11.2210.5720.610–2.454
type of metastasessynchronous/metachronous4.2060.0041.572–11.254
CD44v6 expression>30%/≤30%2.3690.0161.175–4.777
HR, Hazard ratio; p-value was calculated for progression free survival; CI, confidence interval; *, nodules within the metastatic organ.
Table 5. Univariate Survival Analysis of CD44v6-Related Dual Biomarker Expression in Colorectal Liver Metastases.
Table 5. Univariate Survival Analysis of CD44v6-Related Dual Biomarker Expression in Colorectal Liver Metastases.
CombinationNumber of Patients (n)Log Rank p-ValueMedian PFS (month)
CD44v6 high *220.017
CD44v6 low3015.5
CD44v6 high/IGF1-R high150.1427
CD44v6 high/IGF1-R low or CD44v6 low/IGF1-R high209
CD44v6 low/IGF1-R low1717
CD44v6 high/EGF-R high110.2176
CD44v6 high/EGF-R low or CD44v6 low/EGF-R high2411.5
CD44v6 low/EGF-R low179
CD44v6 high/Muc1 high110.5748
CD44v6 high/Muc1 low or CD44v6 low/Muc1 high2311
CD44v6 high/Muc1 low187.5
CD44v6 high/α2β1 high110.0023
CD44v6 high/α2β1 low or CD44v6 low/α2β1 high189
CD44v6 low/α2β1 low2324
CD44v6 high/Hsp90 high140.0227
CD44v6 high/Hsp90 low or CD44v6 low/Hsp90 high219
CD44v6 low/Hsp90 low1717
CD44v6 high/PD-L1 high120.0237
CD44v6 high/PD-L1 low or CD44v6 low/PD-L1 high2114
CD44v6 low/PD-L1 low1911
PFS, progression-free survival; cut-off values defining high and low for the individual biomarker are given in Table 3; *, calculation of the cut-offs is given in the Materials and Methods Section.
Table 6. Multivariate Survival Analysis of CD44v6-Related Dual Biomarker Expression in Colorectal Liver Metastases.
Table 6. Multivariate Survival Analysis of CD44v6-Related Dual Biomarker Expression in Colorectal Liver Metastases.
VariableGroupsCox Regression (PFS)
HRp-Value95% CI
age (median in years)>64/≤641.5610.2560.724–3.366
number of metastases *>1/≤11.3980.3580.684–2.855
type of metastasessynchronous/metachronous3.8130.0081.407–10.332
CD44v6/α2β1 expressionhigh/high vs. low/low4.1350.0021.648–10.375
high/low and low/high vs. low/low1.7840.1450.819–3.886
age (median in years)>64/≤641.1290.7730.496–2.568
number of metastases>1/≤11.3210.4600.632–2.762
type of metastasessynchronous/metachronous3.3450.0131.289–8.680
CD44v6/Hsp90 expressionhigh/high vs. low/low2.0390.0850.906–4.586
high/low and low/high vs. low/low1.4120.4430.585–3.404
age (median in years)>64/≤641.2900.4930.623–2.675
number of metastases>1/≤11.3410.4180.659–2.728
type of metastasessynchronous/metachronous4.1540.0041.584–10.893
CD44v6/PD-L1 expressionhigh/high vs. low/low2.8820.0171.213–6.848
high/low and low/high vs. low/low0.8720.7230.409–1.860
HR, Hazard ratio; PFS, progression free survival; CI, confidence interval; *, nodules within the metastatic organ; cut-off values defining high- and low-level expression for the individual biomarker are given in Table 3.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wrana, F.; Dötzer, K.; Prüfer, M.; Werner, J.; Mayer, B. High Dual Expression of the Biomarkers CD44v6/α2β1 and CD44v6/PD-L1 Indicate Early Recurrence after Colorectal Hepatic Metastasectomy. Cancers 2022, 14, 1939. https://doi.org/10.3390/cancers14081939

AMA Style

Wrana F, Dötzer K, Prüfer M, Werner J, Mayer B. High Dual Expression of the Biomarkers CD44v6/α2β1 and CD44v6/PD-L1 Indicate Early Recurrence after Colorectal Hepatic Metastasectomy. Cancers. 2022; 14(8):1939. https://doi.org/10.3390/cancers14081939

Chicago/Turabian Style

Wrana, Friederike, Katharina Dötzer, Martin Prüfer, Jens Werner, and Barbara Mayer. 2022. "High Dual Expression of the Biomarkers CD44v6/α2β1 and CD44v6/PD-L1 Indicate Early Recurrence after Colorectal Hepatic Metastasectomy" Cancers 14, no. 8: 1939. https://doi.org/10.3390/cancers14081939

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop