Next Article in Journal
Intratumoural Cytochrome P450 Expression in Breast Cancer: Impact on Standard of Care Treatment and New Efforts to Develop Tumour-Selective Therapies
Next Article in Special Issue
Patient-Specific Variables Determine the Extent of Cellular Senescence Biomarkers in Ovarian Tumors In Vivo
Previous Article in Journal
Iron Oxide-Based Magneto-Optical Nanocomposites for In Vivo Biomedical Applications
Previous Article in Special Issue
BECN1 and BRCA1 Deficiency Sensitizes Ovarian Cancer to Platinum Therapy and Confers Better Prognosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Integrin α2β1 Represents a Prognostic and Predictive Biomarker in Primary Ovarian Cancer

1
Department of General, Visceral and Transplant Surgery, University Hospital, Ludwig-Maximilians-University Munich, Marchioninistraße 15, 81377 Munich, Germany
2
Gynecology and Obstetrics Clinic, Klinikum Dritter Orden, Menzinger Straße 44, 80638 Munich, Germany
3
Department of Obstetrics and Gynecology, Klinikum Rechts der Isar, Technical University Munich, Ismaninger Straße 22, 81675 Munich, Germany
4
Department of Obstetrics and Gynecology, Starnberg Hospital, Oßwaldstraße 1, 82319 Starnberg, Germany
5
Department of Obstetrics and Gynecology, Munich Clinic Harlaching, Sanatoriumsplatz 2, 81545 Munich, Germany
6
Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University Munich, Marchioninistraße 15, 81377 Munich, Germany
7
German Cancer Consortium (DKTK), Partner Site Munich, Pettenkoferstraße 8a, 80336 Munich, Germany
*
Author to whom correspondence should be addressed.
Biomedicines 2021, 9(3), 289; https://doi.org/10.3390/biomedicines9030289
Submission received: 24 January 2021 / Revised: 23 February 2021 / Accepted: 3 March 2021 / Published: 12 March 2021

Abstract

:
Currently, the same first-line chemotherapy is administered to almost all patients suffering from primary ovarian cancer. The high recurrence rate emphasizes the need for precise drug treatment in primary ovarian cancer. Being crucial in ovarian cancer progression and chemotherapeutic resistance, integrins became promising therapeutic targets. To evaluate its prognostic and predictive value, in the present study, the expression of integrin α2β1 was analyzed immunohistochemically and correlated with the survival data and other therapy-relevant biomarkers. The significant correlation of a high α2β1-expression with the estrogen receptor alpha (ERα; p = 0.035) and epithelial growth factor receptor (EGFR; p = 0.027) was observed. In addition, high α2β1-expression was significantly associated with a low number of tumor-infiltrating immune cells (CD3 intratumoral, p = 0.017; CD3 stromal, p = 0.035; PD-1 intratumoral, p = 0.002; PD-1 stromal, p = 0.049) and the lack of PD-L1 expression (p = 0.005). In Kaplan–Meier survival analysis, patients with a high expression of integrin α2β1 revealed a significant shorter progression-free survival (PFS, p = 0.035) and platinum-free interval (PFI, p = 0.034). In the multivariate Cox regression analysis, integrin α2β1 was confirmed as an independent prognostic factor for both PFS (p = 0.021) and PFI (p = 0.020). Dual expression of integrin α2β1 and the hepatocyte growth factor receptor (HGFR; PFS/PFI, p = 0.004) and CD44v6 (PFS, p = 0.000; PFI, p = 0.001; overall survival [OS], p = 0.025) impaired survival. Integrin α2β1 was established as a prognostic and predictive marker in primary ovarian cancer with the potential to stratify patients for chemotherapy and immunotherapy, and to design new targeted treatment strategies.

Graphical Abstract

1. Introduction

Several clinicopathological factors, such as advanced tumor stage and residual tumor after surgery, have been established as strong prognostic factors in primary ovarian cancer [1]. In addition, a few tumor biological characteristics have been identified as prognostic markers. Examples are distinct gene signatures [2] or a high number of T-cells [3]. Although recently new promising candidates were detected [4], predictive markers are rare in primary ovarian cancer. Two targeted therapy approaches are recommended under current guidelines, namely vascular endothelial growth factor (VEGF) inhibition and poly (ADP-ribose) polymerase (PARP) inhibition, which are both administered in addition to the standard chemotherapy [5,6,7]. For VEGF inhibition, no predictive biomarker is available to select appropriate patients for anti-angiogenic therapy. Similarly, BRCA mutation or HRD status, which so far represent a prerequisite for some of the PARP inhibition treatments, need to be re-evaluated [8]. Thus, robust biomarkers for precise prognosis and treatment response are urgently required in primary ovarian cancer. This importance is emphasized by the fact that, despite standard therapy combining radical surgery and adjuvant platinum-based chemotherapy, 70–80% of the patients suffer from relapse [9].
Integrins are transmembrane cell adhesion molecules, which mediate cell–cell and cell–extracellular matrix (ECM) interaction. Currently, 18 α-subunits and 8 β-subunits are identified, forming a variety of integrin heterodimers [10]. Due to their ability of inside-out and outside-in signaling, they are known to be involved in migration, invasion, and metastasis promoting tumor progression in several cancer types [11,12,13]. Considering the mechanism of ovarian cancer metastasis by spreading in the peritoneal fluid and attaching to the omental and peritoneal tissue [14,15,16], integrins seem to be a promising therapeutic target in ovarian cancer.
While there is already some information about ovarian cancer and other β1-heterodimers, such as integrins α4β1 and α5β1 [17], less information is available about integrin α2β1. The main ligand of integrin α2β1 is collagen type I, but binding to other collagen types, laminins, and other ECM-proteins is also possible [18,19]. Expression of integrin α2β1 is not only observed on the epithelial cells, but also on the endothelial cells, platelets, white blood cells, and fibroblasts [20,21].
Previous studies indicate a role of integrin α2β1 in chemotherapy resistance [22,23], which constitutes a special interest for ovarian cancer. In the present study, the expression of integrin α2β1 in primary ovarian cancer and its prognostic and predictive role will be evaluated.

2. Materials and Methods

2.1. Study Population

Forty-eight patients diagnosed with a primary, chemonaive ovarian, fallopian tube, or peritoneal cancer from the SpheroID-Study were included. Patients suffering from another neoplasia within the last five years were excluded. Patients were recruited between September 2012 and January 2015 from five ovarian cancer centers, namely University Hospital, LMU Munich (n = 16), Klinikum Dritter Orden (n = 15), Klinikum rechts der Isar, Technical University Munich (n = 7), Munich clinic Harlaching (n = 5), and Starnberg Hospital (n = 5). Standardized surgical resection and pathological analysis was conducted by the recruiting hospital. Patient-, tumor- and treatment-related data for correlations were given in the routine reports and delivered in a pseudonymized form. Survival analysis was performed after the completion of chemotherapy. Seven patients with no chemotherapy or a reduced number of treatment cycles (≤ 2) had to be excluded. Progression-free survival (PFS) was defined as the time from surgical treatment to relapse or progression. Platinum-free interval (PFI) was defined as the time from end of the chemotherapy to relapse or progression. Overall survival (OS) was defined as the time from surgical treatment to death. Data from patients who did not die and had no relapse or progression were censored at the date of their last visit.

2.2. Immunohistochemistry

After surgical removal, tumor samples were snap frozen in liquid nitrogen. Serial cryosections (5 µm) were performed. The samples were stained immunohistochemically using the avidin–biotin–peroxidase method [24]. Tissue sections were fixed either in acetone for 8 min or, for the antigens ERα and PgR, in formalin for 3 min and afterwards in a citrate buffer for 7 min at 90 °C. Blocking of unspecific Fc receptors was performed with 10% AB Serum (Biotest, Dreieich, Germany) in either PBS (acetone fixation) or in a TRIS–HCl buffer (formalin fixation) for 20 min. Endogenous biotin was blocked with a two-step avidin–biotin blocking kit (Vector Laboratories, Burlingame, CA, USA) according to the manufacturer’s instructions for 20 min. Primary antibodies were applied for one hour. Details about primary and secondary antibodies and working concentrations, including the appropriate positive and negative controls, are given in Table 1. Secondary biotinylated antibodies and peroxidase conjugated streptavidin (Dianova, Hamburg, Germany) were incubated for 30 min each.

2.3. Evaluation of Biomarker Expression

Sections were evaluated semiquantitatively using a light microscope (Figure S1). The percentage of positively stained carcinoma cells was evaluated for each antigen. Tumors were defined as hormone receptor-positive if ≥1% of the cancer cells revealed a nuclear staining of ER or PR [25]. Her2/neu expression was scored according to breast cancer [26] and gastric cancer [27] guidelines. Due to the lack of further references, the other biomarkers’ expression was estimated as a percentage of positive cancer cells in 10% steps. Validation was conducted by a second observer (FS). In the absence of standardized cut-offs for other biomarkers, cut-offs were evaluated according to the biphasic distribution or the group size (see Table 1). Quantitative evaluation of CD3, CD8, and PD-1, and semiquantitative evaluation of PD-L1 was performed according to Dotzer et al. [24].

2.4. Statistical Analysis

Clinicopathological factors were grouped by clinical relevance. Integrin expression was correlated with clinicopathological factors, other biomarkers’ expression, and immune infiltrate using the Fisher’s exact two-tailed test. Univariate analysis was performed by calculating cumulative survival probabilities with the Kaplan–Meier method and comparing them with a log-rank test. A Cox regression model was used for the multivariate analysis of survival. p-values < 0.05 were considered to be statistically significant. All statistical analyses were performed using IBM SPSS Statistics 23 (Armonk, NY, USA).

3. Results

3.1. Patient Characteristic

The clinicopathological data are shown in Table 2. Forty-eight patients were included in this study. The mean age at time of diagnosis was 62 years. Most patients suffered from high-grade, serous ovarian carcinoma in an advanced FIGO (Fédération Internationale de Gynécologie et d’Obstétrique) stage with the presence of ascites. Complete surgical resection without macroscopic residual tumor was achieved in 72.9% of all patients. In total, 83.4% of the patients received chemotherapy based on carboplatin and paclitaxel. The median OS was 42 months, the median PFS was 22 months, and the median PFI was 17 months.
Survival data are summarized in Table 2. The presence of distant metastases (FIGO IV) was related to a shorter OS (p = 0.015) and tended to predict a shorter PFS (p = 0.081) and PFI (p = 0.068). Furthermore, patients with a macroscopic residual tumor after surgery showed a significant shorter OS (p = 0.041), PFS (p = 0.008) and PFI (p = 0.01).

3.2. Prognostic and Predictive Impact of Integrin α2β1

High integrin α2β1 expression in primary ovarian cancer was found to be associated with an unfavorable prognosis. Patients with a high expression of integrin α2β1 showed a median PFS of 16 months, which was significantly shorter compared to patients with low α2β1 expression (PFS 29 months, p = 0.035). In addition, high expression of integrin α2β1 predicted a shorter PFI (11 months) in contrast to patients with a low α2β1-expressing primary tumor (25 months, p = 0.034). Most importantly, a high expression of integrin α2β1 in primary ovarian cancer was found to be an independent prognostic factor for a shorter PFS (HR 2.46, CI 95% 1.14–5.29, p = 0.021) and a shorter PFI (HR 2.44, CI 95% 1.14–5.26, p = 0.022). No impact of the extent of α2β1 expression on OS was observed (Table 3). In addition, no significant correlation between the expression of integrin α2β1 and clinicopathological factors could be found.

3.3. Correlation of Integrin α2β1 with Other Biomarkers

In almost all patients (17 out of 18, 94.4%), a high expression of integrin α2β1 significantly correlated with a high expression of ERα (p = 0.035). Furthermore, a high expression of integrin α2β1 could be found more frequently in patients with a high expression of EGFR (7 out of 10, 70%) compared to patients with a low expression of EGFR (11 out of 38, 28.9%, p = 0.027, Table 4).

3.4. Prognostic and Predictive Impact of Integrin α2β1 Combined with Other Biomarkers

The dual expression of integrin α2β1 and various growth factor receptors revealed an impact on PFS and PFI (Table 5). Patients with a high expression of integrin α2β1 and a positive Her-2/neu status showed a shorter PFS (p = 0.043) and PFI (p = 0.037) than patients with a low expression of integrin α2β1, Her-2/neu, or both. Combined high expression of integrin α2β1 and IGF1R correlated significantly with a shorter PFS (p = 0.045) and PFI (p = 0.043). Most interestingly, a high expression of integrin α2β1 and HGFR was related to a shorter PFS (p = 0.004) and PFI (p = 0.004) and impaired prognosis in comparison to integrin α2β1 as single biomarker.
Likewise, a high expression of both integrin α2β1 and CD44v6 was found to be a strong factor in a poor prognosis that correlated with a shorter PFS (p = 0.000), PFI (p = 0.001) and a reduced OS (p = 0.025, Table 5).

3.5. Correlation of Integrin α2β1 and Immune Infiltrate

In patients with a high expression of integrin α2β1, low numbers of stromal and intratumoral CD3+ cells were found (14 out of 18, 77.8%, p = 0.035 and p = 0.017, Table 6). Furthermore, most tumors with a high expression of integrin α2β1 showed a low density of stromal (16 out of 18, 88.9%, p = 0.049) and intratumoral (17 out of 18, 94.4%, p = 0.002) PD-1+ cells. PD-L1 positivity was found more often in tumors with a low expression of integrin α2β1 (23 out of 30, 76.7%) compared to samples with a high expression (6 out of 18, 33.3%; p = 0.005). No correlations for CD8+ cells have been found.

4. Discussion

In the present study, integrin α2β1 was identified as a potential new prognostic and predictive marker in primary ovarian cancer.
A high expression of integrin α2β1 was identified as a marker for a poor prognosis with equal strength, as reported for the established clinical factors: FIGO stage and macroscopic residual tumor after surgical resection. The positive correlation between a high expression of the integrin β1 chain and short survival is documented for various tumor entities [28,29,30]. In particular, integrin α5β1 is already known to be an unfavorable prognostic factor for ovarian cancer [31], but also for cervical, gastric, and non-small-cell lung cancer [32,33,34].
Integrin α2β1 is involved in many steps of cancer progression. Binding to components of the extracellular matrix (ECM), integrin α2β1 mediates tumor cell invasion and metastasis [35,36,37]. This step is promoted by crosstalk with growth factor receptors [38,39]. Interestingly, in the present study, a combined expression of integrin α2β1 with ERα and EGFR was observed. Furthermore, the signaling of integrin α2β1 can induce chemoresistance. This mechanism was observed for chemotherapies containing paclitaxel [23,40], gemcitabine [41], and etoposide [42].
Early relapse and resistance to platinum-based chemotherapy are key problems in the treatment of ovarian cancer [43]. Therefore, the predictive value for the treatment response of integrin α2β1 was analyzed in the present study. Patients with a high expression of integrin α2β1 were observed to have a shorter median PFI. In particular, β1 integrins are already known to promote platinum resistance in ovarian cancer. The mechanisms of this effect are still unclear. Intracellular signaling initiated by binding to the ECM seems to be fundamental for cell adhesion-mediated drug resistance (CAM-DR) [44,45]. One of the main ECM molecules involved in this concept is collagen type I [46], which is the central binding partner of integrin α2β1 [18]. These molecular interactions suggest that the heterodimer α2β1 contributes to CAM-DR. Therefore, targeting integrin α2β1 represents a promising strategy for overcoming platinum resistance in primary ovarian cancer.
In addition, a high expression of integrin α2β1 was observed in patients with a low density of stromal and intratumoral CD3+ as well as PD-1+ cells. Inversely, more than 75% of patients with a low expression of integrin α2β1 showed PD-L1 positivity, which represents an established predictive biomarker for immunotherapy [47]. Several integrins are related to an immunosuppressive tumor microenvironment [48,49]. For example, αv-integrins are major activators of latent TGF-β, which is involved in immunotherapy resistance [50]. The present data suggest that integrin α2β1 might play a similar role. Recently, immunotherapy became a promising approach in ovarian cancer [51,52], and phase III studies with checkpoint inhibitors in combination with platinum-based chemotherapy are already ongoing (NCT03038100, NCT03740165, NCT03737643). Low expression of integrin α2β1, therefore, could be a potential predictive marker for immunotherapy in ovarian cancer. Taken together, integrin α2β1 represents a stratification marker for patients receiving platinum-based chemotherapy and immunotherapy.
Inhibition of integrin α2β1 should be considered as a targeted therapy in ovarian cancer. Several molecules and antibodies have been developed and evaluated for integrin α2β1 inhibition in other entities.
Anti-tumoral activity was shown in prostate cancer in vivo using the monoclonal antibody GBR-500 [53]. E-7820 is a sulphonamide derivative that inhibits the expression of α2-mRNA. In Phase I studies, treatment was associated with a stable disease in a variety of malignancies [54,55]. Phase II studies are ongoing to evaluate the combination with chemotherapy in colon carcinoma (NCT01347645, NCT01133990, NCT00309179). Another β1-antibody could improve the efficiency of platinum-based chemotherapy in non-small-cell lung cancer [56]. However, despite these promising approaches, the complex biology of heterodimers with promiscuous ligands, allosteric activation, and multiple intracellular signaling pathways might hinder successful treatment strategies [13,57,58].
Furthermore, the results of this study also indicate the potential efficiency of dual inhibition. Patients with a combined high expression of integrin α2β1 and HGFR or CD44v6 showed a very short median PFS and PFI, indicating a worse prognosis and platinum resistance.
Dual targeting has become a promising strategy in ovarian cancer. Its efficiency was proven in tumor spheroid and mouse models [59,60]; thus, various phase I studies are ongoing (NCT03895788, NCT03695380, NCT04315233). In future studies, dual inhibition including integrin α2β1-antagonists should be considered for patients with an appropriate biomarker profile.
The main limitation of this study is the small cohort, though it is representative and comparable to cohorts of other clinical trials. The promising role of integrin α2β1 as a new prognostic and predictive biomarker in primary ovarian cancer needs to be confirmed by an enlarged study.

5. Conclusions

In the present study, integrin α2β1 was identified as a prognostic and predictive marker in primary ovarian cancer. High expression of integrin α2β1 correlated with a short PFS. Prognosis was even worse in integrin α2β1-positive tumors co-expressing HGFR or CD44v6. This finding might lead to new biomarker-directed treatment strategies in primary ovarian cancer. In addition, the high expression of integrin α2β1 correlated with a short PFI, supporting the hypothesis that integrins mediate platinum resistance. Thus, a high expression of integrin α2β1 might represent a stratification marker for personalized treatment.

Supplementary Materials

The following are available online at https://www.mdpi.com/2227-9059/9/3/289/s1, Figure S1: Immunohistochemical stainings.

Author Contributions

Conceptualization, B.M.; formal analysis, K.D. and B.M.; funding acquisition, B.M.; investigation, K.D.; methodology, K.D., F.S. and B.M.; project administration, B.M.; resources, F.E.v.K., C.E.B., S.A., S.F., B.C., A.B. and S.M.; validation, F.S., B.C. and B.M.; visualization, K.D.; writing—original draft, K.D. and B.M.; writing—review and editing, F.S., C.E.B., S.F., J.W. and S.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) to B.M., Grant FKZ 16EX1021N.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the institutional review board at 07th August 2012 of the Ludwig-Maximilians-Universität, Munich, Germany (No.278/04).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to protection of detailed patient-related data.

Acknowledgments

The authors appreciate Maja Schorkowitz for acquisition of follow-up data and Anton Stolp, Frank Arnold and Michael Pohr for technical support.

Conflicts of Interest

All but one author declare no conflict of interest. Sven Mahner: Research support, advisory board, honoraria and travel expenses from AbbVie, AstraZeneca, Clovis, Eisai, GlaxoSmithKline, Medac, MSD, Novartis, Olympus, PharmaMar, Roche, Sensor Kinesis, Teva and Tesaro. This support was not related to work in the present study. The funder 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. du Bois, A.; Reuss, A.; Pujade-Lauraine, E.; Harter, P.; Ray-Coquard, I.; Pfisterer, J. Role of surgical outcome as prognostic factor in advanced epithelial ovarian cancer: A combined exploratory analysis of 3 prospectively randomized phase 3 multicenter trials: By the Arbeitsgemeinschaft Gynaekologische Onkologie Studiengruppe Ovarialkarzinom (AGO-OVAR) and the Groupe d’Investigateurs Nationaux Pour les Etudes des Cancers de l’Ovaire (GINECO). Cancer 2009, 115, 1234–1244. [Google Scholar] [CrossRef]
  2. Waldron, L.; Haibe-Kains, B.; Culhane, A.C.; Riester, M.; Ding, J.; Wang, X.V.; Ahmadifar, M.; Tyekucheva, S.; Bernau, C.; Risch, T.; et al. Comparative meta-analysis of prognostic gene signatures for late-stage ovarian cancer. J. Natl. Cancer Inst. 2014, 106. [Google Scholar] [CrossRef] [Green Version]
  3. Li, J.; Wang, J.; Chen, R.; Bai, Y.; Lu, X. The prognostic value of tumor-infiltrating T lymphocytes in ovarian cancer. Oncotarget 2017, 8, 15621–15631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Plotti, F.; Terranova, C.; Guzzo, F.; De Cicco Nardone, C.; Luvero, D.; Bartolone, M.; Dionisi, C.; Benvenuto, D.; Fabris, S.; Ciccozzi, M.; et al. Role of BRCA Mutation and HE4 in Predicting Chemotherapy Response in Ovarian Cancer: A Retrospective Pilot Study. Biomedicines 2021, 9, 55. [Google Scholar] [CrossRef]
  5. Leitlinienprogramm Onkologie (Deutsche Krebsgesellschaft, D.K., AWMF). S3-Leitlinie Diagnostik, Therapie und Nachsorge Maligner Ovarialtumoren, Langversion 4.0. Available online: https://www.leitlinienprogramm-onkologie.de/leitlinien/ovarialkarzinom/ (accessed on 24 January 2021).
  6. National Institute for Health and Care Excellence. Olaparib for Maintenance Treatment of BRCA Mutation-Positive Advanced Ovarian, Fallopian Tube or Peritoneal Cancer after Response to First-Line Platinum-Based Chemotherapy. Available online: https://www.nice.org.uk/guidance/ta598 (accessed on 24 January 2021).
  7. Armstrong, D.K.; Alvarez, R.D.; Bakkum-Gamez, J.N.; Barroilhet, L.; Behbakht, K.; Berchuck, A.; Berek, J.S.; Chen, L.M.; Cristea, M.; DeRosa, M.; et al. NCCN Guidelines Insights: Ovarian Cancer, Version 1.2019. J. Natl. Compr. Cancer Netw. 2019, 17, 896–909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Colombo, N.; Sessa, C.; du Bois, A.; Ledermann, J.; McCluggage, W.G.; McNeish, I.; Morice, P.; Pignata, S.; Ray-Coquard, I.; Vergote, I.; et al. ESMO-ESGO consensus conference recommendations on ovarian cancer: Pathology and molecular biology, early and advanced stages, borderline tumours and recurrent disease. Ann. Oncol. 2019, 30, 672–705. [Google Scholar] [CrossRef] [Green Version]
  9. Pignata, S.; Cecere, S.C.; Du Bois, A.; Harter, P.; Heitz, F. Treatment of recurrent ovarian cancer. Ann. Oncol. 2017, 28, viii51–viii56. [Google Scholar] [CrossRef] [PubMed]
  10. Hynes, R.O. Integrins: Bidirectional, allosteric signaling machines. Cell 2002, 110, 673–687. [Google Scholar] [CrossRef] [Green Version]
  11. Bianconi, D.; Unseld, M.; Prager, G.W. Integrins in the Spotlight of Cancer. Int. J. Mol. Sci. 2016, 17, 2037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Ganguly, K.K.; Pal, S.; Moulik, S.; Chatterjee, A. Integrins and metastasis. Cell Adh. Migr. 2013, 7, 251–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Alday-Parejo, B.; Stupp, R.; Rüegg, C. Are Integrins Still Practicable Targets for Anti-Cancer Therapy? Cancers 2019, 11, 978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lengyel, E. Ovarian cancer development and metastasis. Am. J. Pathol. 2010, 177, 1053–1064. [Google Scholar] [CrossRef]
  15. Aziz, M.; Agarwal, K.; Dasari, S.; Mitra, A.A.K. Productive Cross-Talk with the Microenvironment: A Critical Step in Ovarian Cancer Metastasis. Cancers 2019, 11, 1608. [Google Scholar] [CrossRef] [Green Version]
  16. Ghoneum, A.; Afify, H.; Salih, Z.; Kelly, M.; Said, N. Role of tumor microenvironment in ovarian cancer pathobiology. Oncotarget 2018, 9, 22832–22849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Kobayashi, M.; Sawada, K.; Kimura, T. Potential of Integrin Inhibitors for Treating Ovarian Cancer: A Literature Review. Cancers 2017, 9, 83. [Google Scholar] [CrossRef] [Green Version]
  18. Heino, J. The collagen receptor integrins have distinct ligand recognition and signaling functions. Matrix Biol. 2000, 19, 319–323. [Google Scholar] [CrossRef]
  19. Humphries, J.D.; Byron, A.; Humphries, M.J. Integrin ligands at a glance. J. Cell Sci. 2006, 119, 3901–3903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Van de Walle, G.R.; Vanhoorelbeke, K.; Majer, Z.; Illyés, E.; Baert, J.; Pareyn, I.; Deckmyn, H. Two functional active conformations of the integrin {alpha}2{beta}1, depending on activation condition and cell type. J. Biol. Chem. 2005, 280, 36873–36882. [Google Scholar] [CrossRef] [Green Version]
  21. Zutter, M.M.; Santoro, S.A. Widespread histologic distribution of the alpha 2 beta 1 integrin cell-surface collagen receptor. Am. J. Pathol. 1990, 137, 113–120. [Google Scholar]
  22. Naci, D.; Vuori, K.; Aoudjit, F. Alpha2beta1 integrin in cancer development and chemoresistance. Semin. Cancer Biol. 2015, 35, 145–153. [Google Scholar] [CrossRef]
  23. Zheng, W.; Ge, D.; Meng, G. Reversing microtubule-directed chemotherapeutic drug resistance by co-delivering alpha2beta1 inhibitor and paclitaxel with nanoparticles in ovarian cancer. Cell Biol. Int. 2020, 44, 610–620. [Google Scholar] [CrossRef] [PubMed]
  24. 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]
  25. 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]
  26. Wolff, A.C.; Hammond, M.E.H.; Allison, K.H.; Harvey, B.E.; Mangu, P.B.; Bartlett, J.M.S.; Bilous, M.; Ellis, I.O.; Fitzgibbons, P.; Hanna, W.; et al. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. J. Clin. Oncol. 2018, 36, 2105–2122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Ruschoff, 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. 2010, 457, 299–307. [Google Scholar] [CrossRef] [Green Version]
  28. Yao, E.S.; Zhang, H.; Chen, Y.Y.; Lee, B.; Chew, K.; Moore, D.; Park, C. Increased beta1 integrin is associated with decreased survival in invasive breast cancer. Cancer Res. 2007, 67, 659–664. [Google Scholar] [CrossRef] [Green Version]
  29. Liu, Q.Z.; Gao, X.H.; Chang, W.J.; Gong, H.F.; Fu, C.G.; Zhang, W.; Cao, G.W. Expression of ITGB1 predicts prognosis in colorectal cancer: A large prospective study based on tissue microarray. Int. J. Clin. Exp. Pathol. 2015, 8, 12802–12810. [Google Scholar]
  30. Sun, Q.; Zhou, C.; Ma, R.; Guo, Q.; Huang, H.; Hao, J.; Liu, H.; Shi, R.; Liu, B. Prognostic value of increased integrin-beta 1 expression in solid cancers: A meta-analysis. OncoTargets Ther. 2018, 11, 1787–1799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Sawada, K.; Mitra, A.K.; Radjabi, A.R.; Bhaskar, V.; Kistner, E.O.; Tretiakova, M.; Jagadeeswaran, S.; Montag, A.; Becker, A.; Kenny, H.A.; et al. Loss of E-cadherin promotes ovarian cancer metastasis via alpha 5-integrin, which is a therapeutic target. Cancer Res. 2008, 68, 2329–2339. [Google Scholar] [CrossRef] [Green Version]
  32. Adachi, M.; Taki, T.; Higashiyama, M.; Kohno, N.; Inufusa, H.; Miyake, M. Significance of integrin alpha5 gene expression as a prognostic factor in node-negative non-small cell lung cancer. Clin. Cancer Res. 2000, 6, 96–101. [Google Scholar]
  33. Ren, J.; Xu, S.; Guo, D.; Zhang, J.; Liu, S. Increased expression of α5β1-integrin is a prognostic marker for patients with gastric cancer. Clin. Transl. Oncol. 2014, 16, 668–674. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, H.Y.; Chen, Z.; Wang, Z.H.; Wang, H.; Huang, L.M. Prognostic significance of α5β1-integrin expression in cervical cancer. Asian Pac. J. Cancer Prev. 2013, 14, 3891–3895. [Google Scholar] [CrossRef] [Green Version]
  35. Shield, K.; Riley, C.; Quinn, M.A.; Rice, G.E.; Ackland, M.L.; Ahmed, N. Alpha2beta1 integrin affects metastatic potential of ovarian carcinoma spheroids by supporting disaggregation and proteolysis. J. Carcinog. 2007, 6, 11. [Google Scholar] [CrossRef]
  36. Burnier, J.V.; Wang, N.; Michel, R.P.; Hassanain, M.; Li, S.; Lu, Y.; Metrakos, P.; Antecka, E.; Burnier, M.N.; Ponton, A.; et al. Type IV collagen-initiated signals provide survival and growth cues required for liver metastasis. Oncogene 2011, 30, 3766–3783. [Google Scholar] [CrossRef] [Green Version]
  37. Bartolomé, R.A.; Barderas, R.; Torres, S.; Fernandez-Aceñero, M.J.; Mendes, M.; García-Foncillas, J.; Lopez-Lucendo, M.; Casal, J.I. Cadherin-17 interacts with α2β1 integrin to regulate cell proliferation and adhesion in colorectal cancer cells causing liver metastasis. Oncogene 2014, 33, 1658–1669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Eliceiri, B.P. Integrin and growth factor receptor crosstalk. Circ. Res. 2001, 89, 1104–1110. [Google Scholar] [CrossRef] [Green Version]
  39. Hamidi, H.; Ivaska, J. Every step of the way: Integrins in cancer progression and metastasis. Nat. Rev. Cancer 2018, 18, 533–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Aoudjit, F.; Vuori, K. Integrin signaling inhibits paclitaxel-induced apoptosis in breast cancer cells. Oncogene 2001, 20, 4995–5004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Duxbury, M.S.; Ito, H.; Benoit, E.; Waseem, T.; Ashley, S.W.; Whang, E.E. RNA interference demonstrates a novel role for integrin-linked kinase as a determinant of pancreatic adenocarcinoma cell gemcitabine chemoresistance. Clin. Cancer Res. 2005, 11, 3433–3438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Sethi, T.; Rintoul, R.C.; Moore, S.M.; MacKinnon, A.C.; Salter, D.; Choo, C.; Chilvers, E.R.; Dransfield, I.; Donnelly, S.C.; Strieter, R.; et al. Extracellular matrix proteins protect small cell lung cancer cells against apoptosis: A mechanism for small cell lung cancer growth and drug resistance in vivo. Nat. Med. 1999, 5, 662–668. [Google Scholar] [CrossRef]
  43. Wilson, M.K.; Pujade-Lauraine, E.; Aoki, D.; Mirza, M.R.; Lorusso, D.; Oza, A.M.; du Bois, A.; Vergote, I.; Reuss, A.; Bacon, M.; et al. Fifth Ovarian Cancer Consensus Conference of the Gynecologic Cancer InterGroup: Recurrent disease. Ann. Oncol. 2017, 28, 727–732. [Google Scholar] [CrossRef] [PubMed]
  44. Scalici, J.M.; Harrer, C.; Allen, A.; Jazaeri, A.; Atkins, K.A.; McLachlan, K.R.; Slack-Davis, J.K. Inhibition of alpha4beta1 integrin increases ovarian cancer response to carboplatin. Gynecol. Oncol. 2014, 132, 455–461. [Google Scholar] [CrossRef] [Green Version]
  45. Wantoch von Rekowski, K.; König, P.; Henze, S.; Schlesinger, M.; Zawierucha, P.; Januchowski, R.; Bendas, G. The Impact of Integrin-Mediated Matrix Adhesion on Cisplatin Resistance of W1 Ovarian Cancer Cells. Biomolecules 2019, 9, 788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Baltes, F.; Pfeifer, V.; Silbermann, K.; Caspers, J.; Wantoch von Rekowski, K.; Schlesinger, M.; Bendas, G. β(1)-Integrin binding to collagen type 1 transmits breast cancer cells into chemoresistance by activating ABC efflux transporters. Biochim. Biophys. Acta Mol. Cell Res. 2020, 1867, 118663. [Google Scholar] [CrossRef] [PubMed]
  47. Gibney, G.T.; Weiner, L.M.; Atkins, M.B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016, 17, e542–e551. [Google Scholar] [CrossRef] [Green Version]
  48. Schmid, M.C.; Avraamides, C.J.; Dippold, H.C.; Franco, I.; Foubert, P.; Ellies, L.G.; Acevedo, L.M.; Manglicmot, J.R.; Song, X.; Wrasidlo, W.; et al. Receptor tyrosine kinases and TLR/IL1Rs unexpectedly activate myeloid cell PI3kγ, a single convergent point promoting tumor inflammation and progression. Cancer Cell 2011, 19, 715–727. [Google Scholar] [CrossRef] [Green Version]
  49. Wu, A.; Zhang, S.; Liu, J.; Huang, Y.; Deng, W.; Shu, G.; Yin, G. Integrated Analysis of Prognostic and Immune Associated Integrin Family in Ovarian Cancer. Front. Genet. 2020, 11, 705. [Google Scholar] [CrossRef]
  50. Brown, N.F.; Marshall, J.F. Integrin-Mediated TGFbeta Activation Modulates the Tumour Microenvironment. Cancers 2019, 11, 1221. [Google Scholar] [CrossRef] [Green Version]
  51. Kandalaft, L.E.; Odunsi, K.; Coukos, G. Immunotherapy in Ovarian Cancer: Are We There Yet? J. Clin. Oncol. 2019, 37, 2460–2471. [Google Scholar] [CrossRef]
  52. Wang, W.; Liu, J.R.; Zou, W. Immunotherapy in Ovarian Cancer. Surg. Oncol. Clin. N. Am. 2019, 28, 447–464. [Google Scholar] [CrossRef]
  53. Attinger, A.; Hou, S.; Pesenti, E.A.; Mancini, L.; Mottl, H. Abstract LB-294: GBR 500, a monoclonal VLA-2 antibody inhibits tumor and metastasis growth but not extravasation in a prostate cancer animal model. In Proceedings of the AACR 102nd Annual Meeting 2011, Orlando, FL, USA, 2–6 April 2011. [Google Scholar]
  54. Milojkovic Kerklaan, B.; Slater, S.; Flynn, M.; Greystoke, A.; Witteveen, P.O.; Megui-Roelvink, M.; de Vos, F.; Dean, E.; Reyderman, L.; Ottesen, L.; et al. A phase I, dose escalation, pharmacodynamic, pharmacokinetic, and food-effect study of alpha2 integrin inhibitor E7820 in patients with advanced solid tumors. Investig. New Drugs 2016, 34, 329–337. [Google Scholar] [CrossRef] [PubMed]
  55. Mita, M.; Kelly, K.R.; Mita, A.; Ricart, A.D.; Romero, O.; Tolcher, A.; Hook, L.; Okereke, C.; Krivelevich, I.; Rossignol, D.P.; et al. Phase I study of E7820, an oral inhibitor of integrin alpha-2 expression with antiangiogenic properties, in patients with advanced malignancies. Clin. Cancer Res. 2011, 17, 193–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Kim, M.Y.; Cho, W.D.; Hong, K.P.; Choi da, B.; Hong, J.W.; Kim, S.; Moon, Y.R.; Son, S.M.; Lee, O.J.; Lee, H.C.; et al. Novel monoclonal antibody against beta 1 integrin enhances cisplatin efficacy in human lung adenocarcinoma cells. J. Biomed. Res. 2016, 30, 217–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Raab-Westphal, S.; Marshall, J.F.; Goodman, S.L. Integrins as Therapeutic Targets: Successes and Cancers. Cancers 2017, 9, 110. [Google Scholar] [CrossRef]
  58. Millard, M.; Odde, S.; Neamati, N. Integrin targeted therapeutics. Theranostics 2011, 1, 154–188. [Google Scholar] [CrossRef]
  59. Taylan, E.; Zayou, F.; Murali, R.; Karlan, B.Y.; Pandol, S.J.; Edderkaoui, M.; Orsulic, S. Dual targeting of GSK3B and HDACs reduces tumor growth and improves survival in an ovarian cancer mouse model. Gynecol. Oncol. 2020, 159, 277–284. [Google Scholar] [CrossRef] [PubMed]
  60. Camblin, A.J.; Tan, G.; Curley, M.D.; Yannatos, I.; Iadevaia, S.; Rimkunas, V.; Mino-Kenudson, M.; Bloom, T.; Schoeberl, B.; Drummond, D.C.; et al. Dual targeting of IGF-1R and ErbB3 as a potential therapeutic regimen for ovarian cancer. Sci. Rep. 2019, 9, 16832. [Google Scholar] [CrossRef] [Green Version]
Table 1. Biomarkers and antibodies.
Table 1. Biomarkers and antibodies.
AntigenCloneSpeciesFixationUse of Kitwc (μg/mL)SupplierCut-Off for Positivity
Primary antibodies
Integrin α2β1BHA2.1mAcetone-2.50Millipore, Burlington, MA, USA≥20%
ERα1D5mFormalin+2.50Dako, Santa Clara, CA, USA≥1%
PRPgR 636mFormalin+2.50Dako, Santa Clara, CA, USA≥1%
HER-2/neu4B5rAcetone-1.50Ventana, Roche, Basel, CH≥10% (Intensity 2+/3+)
EGFRH11mAcetone-2.94Dako, Santa Clara, CA, USA≥50%
HGFRSP44rAcetone-2.12Spring Bioscience, Pleasanton, CA, USA≥50%
IGF1R23-41mAcetone+4.00invitrogen, Carlsbad, CA, USA≥80%
MUC-1Ma552mAcetone-0.50Monosan, Uden, NL≥70%
CD44v6VFF-18mAcetone-1.00affymetrix eBioscience, Santa Clara, CA, USA≥10%
Integrin αVβ3LM609mAcetone-5.00Millipore, Burlington, MA, USA≥20%
CD3UCHT1mAcetone-1.25BD Biosciences, Franklin Lakes, NJ, USA
CD8C8/144BmAcetone+3.00Dako, Santa Clara, CA, USA
PD-1MIH4mAcetone+10.00affymetrix eBioscience, Santa Clara, CA, USA
PD-L1MIH1mAcetone+10.00affymetrix eBioscience, Santa Clara, CA, USA≥1%
Positive controls
Epithelial AntigenBer-EP4mAcetone-2.50Dako, Santa Clara, CA, USA
CD452B11 + PD7/26mAcetone-4.50Dako, Santa Clara, CA, USA
Isotype controls
MOPC 21MOPC 21m -5.00Sigma-Aldrich, St. Louis, MO, USA
MOPC 21m +4.00Sigma-Aldrich, St. Louis, MO, USA
MOPC 21m +10.00Sigma-Aldrich, St. Louis, MO, USA
DA1EDA1Er -2.12Cell Signaling, Danvers, MA, USA
Biotin conjugated secondary antibodies
111-065-114g anti r 7.00Jackson Immunoresearch, West Grove, PA, USA
315-065-048r anti m 0.75Jackson Immunoresearch, West Grove, PA, USA
Legend: wc: working concentration, m: mouse, r: rabbit, g: goat, all used antibodies’ isotype was IgG1. ERα: estrogen receptor α, PR: progesterone receptor, Her-2/neu: human epidermal growth factor receptor 2, EGFR: epidermal growth factor receptor, HGFR: hepatocyte growth factor receptor, IGF1R: insulin-like growth factor 1, MUC-1: mucin-1, PD-1: programmed cell death protein 1, PD-L1: programmed cell death-ligand 1.
Table 2. Patient characteristics.
Table 2. Patient characteristics.
n or Value%
Agemean/median62/66 years
range24–83 years
FIGO StageI or II00.0%
III3470.8%
IV1429.2%
pTpT2510.4%
pT34389.6%
pNpN0612.5%
pN13164.6%
Nx1122.9%
cMcM03470.8%
cM11429.2%
Primary Tumor SiteOvarian3981.3%
Fallopian Tube612.5%
Peritoneal36.3%
Histological SubtypeSerous4491.7%
Other48.4%
GradingG1/G224.2%
G34695.8%
Ascitesyes4083.3%
no816.7%
Macroscopic Residual Tumor after SurgeryNone3572.9%
<1 cm612.5%
>1 cm714.6%
First-Line-TreatmentC48.3%
C + P1531.3%
C + P + B2552.1%
None48.3%
Relapse after Chemotherapy<6 months24.2%
6–12 months1225.0%
>12 months2858.3%
none or non-sufficient chemotherapy612.5%
Legend: n: number of patients, Nx: no evaluation of lymph node status, C: carboplatin, P: paclitaxel, B: bevacizumab.
Table 3. Univariate and multivariate survival analysis of clinicopathological factors and integrin α2β1.
Table 3. Univariate and multivariate survival analysis of clinicopathological factors and integrin α2β1.
PFSPFIOS
nLog-RankMV Cox RegressionLog-RankMV Cox RegressionLog-Rank
MSpHR (CI 95%)pMSpHR (CI 95%)pMSp
Age ≤ 62 years19220.965 170.970 nr0.193
Age > 62 years2322 17 42
<pT3c7270.665 220.679 450.928
pT3c3522 17 42
pN05290.163 170.145 450.929
pN12822 22 42
cM02927 22 nr
cM113160.0812.06 (0.92–4.62)0.081110.0682.10 (0.94–4.69)0.072300.015
G1/G22140.579 80.610 300.843
G34022 17 42
Ascites absent6350.147 300.139 420.408
Ascites present3619 15 38
MR Tumor absent3027 22 45
MR Tumor present12130.0082.19 (1.03–4.68)0.04390.0102.10 (0.99-4.51)0.057260.041
Integrin α2β1 low2729 25 45
Integrin α2β1 high15160.0352.46 (1.14–5.29)0.021110.0342.45 (1.14-5.26)0.022300.155
Legend: n: number of patients, Cox regression: multivariate Cox regression, MS: median survival (in months) in Kaplan–Meier estimator, HR: hazard ratio, CI: confidence interval, MR Tumor: macroscopic residual tumor; nr: median survival not reached.
Table 4. Correlation between integrin α2β1 and other biomarkers.
Table 4. Correlation between integrin α2β1 and other biomarkers.
Integrin α2β1
n<20%≥20%p#
Growth Factor-ReceptorERα 48 0.035
<1% 101
≥1% 2017
PR 48 0.127
<1% 229
≥1% 89
Her-2/neu 48 1
negative 2213
positive 85
EGFR 48 0.027
<50% 2711
≥50% 37
HGFR 48 0.133
<50% 165
≥50% 1413
IGF1R 48 0.451
<80% 44
≥80% 2614
Cell-Adhesion-MoleculeMUC-1 48 0.765
<70% 147
≥70% 1611
CD44v6 48 0.103
<10% 2410
≥10% 68
Integrin αvβ3 48 0.19
<20% 2411
≥20% 67
Legend: n: number of patients, #: p-value calculated by Fisher’s exact two-tailed test.
Table 5. Univariate survival analysis of dual expression of integrin α2β1 and other biomarkers.
Table 5. Univariate survival analysis of dual expression of integrin α2β1 and other biomarkers.
PFSPFIOS
nMSp *MSp *MSp *
Integrin α2β1 high15160.035110.034300.155
Integrin α2β1 low27292545
Integrin α2β1 high/ERα high14160.078110.073300.287
Remaining combinations #28272242
Integrin α2β1 high/PR high8160.57411190.578270.526
Remaining combinations #34241942
Integrin α2β1 high/Her-2/neu +5210.043150.037360.698
Remaining combinations #37272242
Integrin α2β1 high/EGFR high6140.28980.290300.482
Remaining combinations #36221742
Integrin α2β1 high/HGFR high11150.004100.004270.054
Remaining combinations #31292545
Integrin α2β1 high/IGFR high11160.045110.043360.381
Remaining combinations #31272242
Integrin α2β1 high/MUC-1 high9140.06390.055270.257
Remaining combinations #33272242
Integrin α2β1 high/CD44v6 high6130.00090.001190.025
Remaining combinations #36272242
Integrin α2β1 high/Integrin αvβ3 high5350.322300.320nr0.162
Remaining combinations #37221742
Legend: n: number of patients, MS: median survival (in months) in Kaplan–Meier estimator, *: p-value calculated by log-rank test. # The remaining combinations represent tumor samples which were integrin α2β1 high/biomarker X low, integrin α2β1 low/biomarker X high, or integrin α2β1 low/biomarker X low. nr: median survival not reached.
Table 6. Correlations between integrin α2β1 and the immune infiltrate.
Table 6. Correlations between integrin α2β1 and the immune infiltrate.
Immune Infiltrate Integrin α2β1
n<20%≥20%p#
CD3 stromal 48 0.034
Low 1314
High 174
CD3 intratumoral 48 0.017
Low 1214
High 184
CD8 stromal 48 0.133
Low 1413
High 165
CD8 intratumoral 48 0.363
Low 1713
High 135
PD-1 stromal 48 0.049
Low 1816
High 122
PD-1 intratumoral 48 0.002
Low 1517
High 151
PD-L1 positivity 48 0.005
No 712
Yes 236
Legend: n: number of patients, #: p-value as calculated by Fisher’s exact two-tailed test.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dötzer, K.; Schlüter, F.; Koch, F.E.v.; Brambs, C.E.; Anthuber, S.; Frangini, S.; Czogalla, B.; Burges, A.; Werner, J.; Mahner, S.; et al. Integrin α2β1 Represents a Prognostic and Predictive Biomarker in Primary Ovarian Cancer. Biomedicines 2021, 9, 289. https://doi.org/10.3390/biomedicines9030289

AMA Style

Dötzer K, Schlüter F, Koch FEv, Brambs CE, Anthuber S, Frangini S, Czogalla B, Burges A, Werner J, Mahner S, et al. Integrin α2β1 Represents a Prognostic and Predictive Biomarker in Primary Ovarian Cancer. Biomedicines. 2021; 9(3):289. https://doi.org/10.3390/biomedicines9030289

Chicago/Turabian Style

Dötzer, Katharina, Friederike Schlüter, Franz Edler von Koch, Christine E. Brambs, Sabine Anthuber, Sergio Frangini, Bastian Czogalla, Alexander Burges, Jens Werner, Sven Mahner, and et al. 2021. "Integrin α2β1 Represents a Prognostic and Predictive Biomarker in Primary Ovarian Cancer" Biomedicines 9, no. 3: 289. https://doi.org/10.3390/biomedicines9030289

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