Next Article in Journal
Orchestrating Treatment Modalities in Metastatic Pancreatic Neuroendocrine Tumors—Need for a Conductor
Previous Article in Journal
Efficient Radiomics-Based Classification of Multi-Parametric MR Images to Identify Volumetric Habitats and Signatures in Glioblastoma: A Machine Learning Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Chick Chorioallantoic Membrane (CAM) Assays as a Model of Patient-Derived Xenografts from Circulating Cancer Stem Cells (cCSCs) in Breast Cancer Patients

1
Department of Research and Development, Transfusion Center Bayreuth, 95448 Bayreuth, Germany
2
Department of Organic Chemistry, University of Bayreuth, 95440 Bayreuth, Germany
3
Department of Cardiac Surgery, Clinic of Bayreuth, 95455 Bayreuth, Germany
4
Department of Clinical and Experimental Pathology, Wroclaw Medical University, 50-556 Wroclaw, Poland
5
Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, 43-309 Bielsko-Biała, Poland
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(6), 1476; https://doi.org/10.3390/cancers14061476
Submission received: 8 February 2022 / Revised: 7 March 2022 / Accepted: 8 March 2022 / Published: 14 March 2022

Abstract

:

Simple Summary

Circulating cancer cells—and in particular their very rare subpopulation, circulating cancer stem cells (cCSCs)—are responsible for recurrence and metastasis. In this study, we present a novel process in which patient-derived xenograft (PDX) can be harvested on chorioallantoic membrane (CAM) from circulating cancer stem cells. In our opinion, the CAM-based PDX model using circulating cancer stem cells can provide a fast, low-cost, easy-to-use, and efficient preclinical platform for drug screening, therapy optimization, and biomarker discovery.

Abstract

Background: cCSCs are a small subset of circulating tumor cells with cancer stem cell features: resistance to cancer treatments and the capacity for generating metastases. PDX are an appreciated tool in oncology, providing biologically meaningful models of many cancer types, and potential platforms for the development of precision oncology approaches. Commonly, mouse models are used for the in vivo assessment of potential new therapeutic targets in cancers. However, animal models are costly and time consuming. An attractive alternative to such animal experiments is the chicken chorioallantoic membrane assay. Methods: In this study, primary cultures from cCSCs were established using the sphere-forming assay. Subsequently, tumorspheres were transplanted onto the CAM membrane of fertilized chicken eggs to form secondary microtumors. Results: We have developed an innovative in vitro platform for cultivation of cCSCs from peripheral blood of cancer patients. The number of tumorspheres increased significantly with tumor progression and aggressiveness of primary tumor. The number of tumorspheres was positively correlated with Ki-67, Her2 status, and grade score in primary breast tumors. The grafting of tumorspheres onto the CAM was successful and positively correlated with aggressiveness and proliferation capacity of the primary tumor. These tumors pathologically closely resembled the primary tumor. Conclusions: The number of tumorspheres cultured from peripheral blood and the success rate of establishing PDX directly reflect the aggressiveness and proliferation capacity of the primary tumor. A CAM-based PDX model using cCSC provides a fast, low-cost, easy to handle, and powerful preclinical platform for drug screening, therapy optimization, and biomarker discovery.

1. Introduction

Breast cancer is the most common neoplasms among women, and it continues to be associated with a high mortality rate despite the increasing number of early diagnoses and improvement of the initial tumor cure rate [1]. Distant metastases remain the leading cause of death in breast cancer patients and they arise from hematogenous spread of circulating tumor cells (CTCs) from solid primary or metastatic tumor lesions [2,3,4]. Recent studies have provided strong support for the cancer stem cell hypothesis, which suggests that many cancers, including breast cancer, are driven by a subpopulation of cells that exhibit stem cell properties. These cells may mediate metastasis and, because of their resistance to radiotherapy and chemotherapy, contribute to disease recurrence and progression in cancer patients [5,6]. As a consequence, eradicating this subpopulation would be critical in order to achieve patient’s cure [7,8].
Interestingly, some experimental findings have highlighted that a small subpopulation of CTCs displays CSCs features (tumor initiating capability), so that they can be considered as circulating cancer stem cells [9,10,11]. A number of non-invasive diagnostic assays are being developed in order to either diagnose patients with metastatic disease and/or to screen patients for relapse after treatment. A new diagnostic tool, developed in recent years by our team, involves measuring extremely rare circulating cancer stem cells in patients’ blood to detect early-stage metastases or cancer recurrence in treated patients. This functional assay is based on the unique property of cancer stem cells to survive and grow in suspension culture [12]. cCSCs were able to form tumorspheres in vitro, confirming that this subpopulation of CTCs represent tumor initiating cells which can potentially grow into metastatic tumors in vivo. To assess tumorigenicity, breast cancer stem cells can be xenotransplanted into the subcutaneous area under the mammary fat pad of immunocompromised female mice [5]. Although they are associated with ethical and financial limitations, this assay is the ‘gold standard’ for assessing CSC tumorigenicity.
In contrast, the chick embryo chorioallantoic membrane (CAM) is a low cost, reproducible, and reliable model used to investigate several functional features of tumor biology—such as the ability of cells to be tumorigenic, to invade, and to metastasize into the embryo [13]. CAM is a highly vascularized extraembryonic membrane, thus providing rich nutrient conditions for the growth of human tumor cells. Cancer cells can be implanted easily, non-invasively, and in an immunodeficient environment because the chick embryo has an immature immune system in the early stages of development when the cells are implanted. Within days, tumor formation occurs and, in the case of aggressive tumors, metastasizing cells can colonize the embryo’s organs via hematogenous metastasis. In human medicine, this model is widely used in preclinical oncology research to study the growth of many types of tumors, including breast cancer [14]. Moreover, it has also been used as a platform to analyze the values of anticancer drugs [15] and radiation therapy [16]. A recent important advance of the CAM assay is that patient tumor samples can be used to transplant on the CAM, enabling the establishment of a patient-derived CAM tumor [17].
In this manuscript, we describe, for the first time, the use of tumorspheres cultured from circulating cancer stem cells of breast cancer patients to establish patient-derived xenografts on the CAM membrane. These results allowed us to propose that the chick CAM model can be a new tool to evaluate breast CSC properties in vivo with the potential to be used in a personalized medicine context.

2. Material and Methods

2.1. Study Population

The patient population of this study consists of 75 women diagnosed with breast cancer in different stages of disease. Blood samples were drawn into normal blood count tubes with ethylenediaminetetraacetic acid (EDTA) as an anticoagulant and processed within 72 h of collection in accordance with a recently published protocol [3,12,18]. The study was conducted in accordance with the Declaration of Helsinki and was approved by the ethics committee of University Bayreuth (O 1305/1-GB).

2.2. Sphere-Forming Assays

Circulating cancer stem cells were cultured together with leukocytes in medium composed of RPMI-1640 supplemented with L-glutamine, HEPES, penicillin/streptomycin, and growth factors such as EGF, insulin, and hydrocortisone at a density of 2 × 105 cells/mL. Cells were incubated at 37 °C in a humidified 5% CO2 atmosphere, and the fresh culture medium was added once a week until cells started to form non adherent aggregates. The formation of spheres was confirmed under an inverted microscope (Zeiss, Jena, Germany) and after 14 days collected by gentle centrifugation for further experiments. Tumorspheres cultured from cCSCs were characterized using typical combination of markers for breast cancer stem cells. EpCAM positive tumorspheres were additionally stained for CD24 (clone ML5, mouse anti-human, BD Bioscience, Franklin Lakes, NJ, USA) and CD44 (clone 515, mouse anti-human, BD Bioscience, Franklin Lakes, NJ, USA) PE-conjugated antibody. To confirm tumor stem cell properties of tumorspheres, we used an aldehyde dehydrogenase isoform 1 (ALDH1) enzymatic assay, which quantifies the ALDH1 activity of tumorspheres. We used an ALDEFLUOR assay kit (Stem Cell TechnologiesTM, Vancouver, BC, Canada) according to the manufacturer’s protocol. Tumorspheres expressing high levels of ALDH1 becoming brightly fluorescent could be easily distinguished from single cells because they develop a solid spherical formation with a diameter >50 µm.

2.3. CAM Xenografts Model from Circulating Cancer Stem Cells

In 10 cases of breast cancer patients, CAM xenograft experiments were performed. Briefly, fertilized chicken eggs were maintained in a humidified egg incubator (Grumbach, Asslar, Germany) at 37 °C. Protocols were approved by the Ethics Committee of the University Bayreuth. A small window was made in the shell on day 4 of chick embryo development under aseptic conditions. The window was resealed with adhesive tape and eggs were returned to the incubator until day 8 of chick embryo development. At day 8 the tumorspheres cultured from peripheral blood were mixed (1:1) with standard Matrigel (BD Biosciences) basement membrane matrix to prevent cell dispersion, in a total volume of 20 µL and directly seeded on the membrane of CAM through the small window created earlier (Figure 1).
After completion of the study protocol, the embryo was sacrificed by decapitation. The CAM bearing the tumor was then excised with sterilized surgical scissors, fixed with 4% paraformaldehyde for 24 h, and embedded in paraffin.

2.4. Histopathology

Paraffin blocks were produced for each CAM tumor. The histological sections were deparaffinized, hydrated in xylene and graded alcohol series and then stained with H&E according to standard protocol. Histological images (40×, 100×, 200×, and 400×) were acquired by light microscope (Olympus BX43, Tokio, Japan) and digitized using a RGB video camera (Olympus DP 20, Tokio, Japan).

2.5. Graphs and Statistical Analyses

All statistical analyses were performed using SigmaPlot Version 13.0 (released 3 December 2017, Systat Software Inc., San Jose, CA, USA). A paired or unpaired Student’s t-test was used for continuous variables, as appropriate. One-way ANOVA was performed to calculate the differences among multiple groups. A two-sided p-value less than 0.05 was considered statistically significant.

3. Results

3.1. Clinicopathological Characteristics of Breast Cancer Patients

Clinicopathological information is summarized in Table 1. Twenty-six patients (34.7%) had T1, 33 (44%) T2, 9 (12%) T3, and 7 (9.3%) T4. Thirty-five patients (46.7%) had lymph node involvement. Sixteen patients (40%) had ER-positive and Her2-negative breast cancer, 16 (21%) had Her2-positive disease, of which 5 were also ER-positive, and 13 (17%) had triple-negative breast cancer (TNBC). The majority of breast cancer patients had non metastatic disease (52; 69.3%), while 30% of patients had metastatic breast cancer.

3.2. Detection of Circulating Cancer Stem Cells

One of our aims was to detect circulating cancer stem cells using tumorsphere-forming assay. To determine the self-renewal and growth potentials of cCSCs, we maintained the suspension cultures. We were able to generate tumorspheres from peripheral blood in 58 (77.35%) cases. Tumorspheres cultured from cCSCs were very compact and the size increased with time, reaching a diameter of 50–100 µm after 21 days (Figure 2a). Tumorspheres were positive for EpCAM and CD 44, and negative or very low positive for CD24 (Figure 2b). The EpCAM and CD44 staining was very heterogeneous in the individual cells within individual tumorspheres. Most of the tumorspheres showed distinct fluorescence for ALDH1 (Figure 2c) typical for breast cancer stem cells.

3.3. Tumorspheres Count and Its Association with Clinicopathological Features

We correlated tumorsphere counts with clinicopathological features. The number of tumorspheres was significantly associated with presence and amount of distant metastasis and also aggressiveness of primary tumor. We found that patients with distant metastasis had statistically significantly more tumorspheres compared to patients without spreading to distant organs (median 32.5 vs. 10.0; p < 0.01) (Figure 3a). Furthermore, the highest level of tumorspheres was observed in the patient group with multiple metastases (median 61; p < 0.001) (Figure 3b). Additionally, our results indicate that the number of tumorspheres correlated significantly with the aggressiveness of the primary tumor. Patients with Her2-neu positive primary tumor had significantly higher levels of tumorspheres compared to patients with negative Her2-neu histology (median 35 vs. 10; p < 0.05) (Figure 3c). Additionally, the number of tumorspheres was positively correlated with the proliferation index Ki-67 and histological grade in primary breast tumor. By definition, tumors exhibiting a Ki-67 index of 15% or more were considered to be connected with worse prognosis. Based on this cut-off criterion, 18 out of the 75 patients were shown to have primary tumors displaying highly proliferative potential. Patients with Ki-67 > 15% had significantly higher numbers of tumorspheres compared to those with Ki-67 < 15% (median 34 vs. 8; p < 0.01) (Figure 3d). With respect to differentiation status, we observed that patients with G3 primary tumors had significantly more tumorspheres compared to patients with G1 (median 32.5 vs. 0; p < 0.01) (Figure 4). In contrast, age, tumor size, lymph node and ER/PR status were not significantly associated with the number of tumorspheres.

3.4. Establishing a Breast Cancer Model from Tumorspheres Using the Chicken Egg CAM System

To establish a breast cancer chicken egg model, we used tumorsphere cultured from peripheral blood of breast cancer patients (n = 10). We were able to reproducibly generate tumors on CAM from 5 out of 10 breast cancer cases tested. Each patient was characterized separately in Table 2. In the CAM, tumors derived from tumorspheres were able to invade the CAM and to be vascularized (Figure 5a). Histological examination revealed that the CAM tumors contained extensive neoplastic tissue with a well-organized, preserved, and homogeneous structure, clearly distinguishable from the surrounding stroma and membrane border which resembled the morphology of the patient’s primary breast tumor (Figure 5b). The success rate of tumor development on CAM membrane depended on the number of spheres and the aggressiveness of the primary tumor. Patients with a large number of tumorspheres and high Ki-67 index of primary tumor were more likely to develop tumors on the CAM membrane (Figure 6a,b). The estrogen receptor status did not have any influence on tumorsphere formation and CAM-tumor development.

4. Discussion

Cancers exist in an extraordinary variety of types and subtypes, making each cancer individually unique. Tumors are heterogeneous and many cancer cell populations with different features are present. Among these tumor populations, cells with properties of stemness, commonly called CSCs, have been described and associated with more aggressive phenotypes [19]. There is strong evidence suggesting that cancer cells with stem properties selectively resist current cancer therapies, indicating the important role that CSCs play in tumor evolution, relapse, and metastasis [20,21]. Interestingly, some experimental findings have highlighted that a small population of CTCs also displays cancer stem cell characteristics (tumor initiating capability) [12,22,23]. Sphere-forming assays are well-described culture methods that have been used for stem cell isolation, identification, and enrichment from different tissues [24,25,26,27]. Starting materials for these cultures have been commercial cell lines, surgical resection specimens, and also peripheral blood with a subset of circulating cancer stem cells [11,12]. The role of cCSCs was investigated in several studies and the results indicated that cCSCs are linked to an unfavorable prognosis in various cancers [28,29,30,31]. In this report, we confirmed our previous findings that circulating cancer stem cells are able to generate tumorspheres and the number of tumorspheres was associated with presence of metastasis [12]. Patients with metastatic disease had more tumorspheres compared to the patients without metastases. Furthermore, patients with multiple metastases had the highest number of tumorspheres. These observations suggest that the number of spheres may reflect the aggressiveness of the tumor and provide additional information about the progression of the disease. We also observed that sphere-formation rates were positively correlated with Her2-neu, Ki-67, and grading of primary tumor. Patients with Her2-neu positive tumor had more tumorspheres in comparison to patients with Her2-neu negative status. Our results are consistent with those of Korkaya et al. [32] who showed that overexpression of Her2-neu increases the population of mammary stem cells as well as their ability to form spheres. Ginestier et al. [33] showed that, in human breast cancers, there was a correlation between Her2-neu amplification and cancer stem cells frequency as assessed by expression of the breast CSC marker ALDH-1.
Ki-67 is universally expressed in proliferating cells, and it is a predictive and prognostic marker for clinical practice in breast cancer patients [34]. The level of Ki-67 determines the categorization of low risk of recurrence and high risk of recurrence. Clinically, the most widely used cut-off for low-risk recurrence is a Ki-67 index below 15% [35]. In our study, the generation of tumorspheres was associated with Ki-67 status in the primary tumor. Patients with Ki-67 >15% had more tumorspheres compared to patients with Ki-67 <15%. Cidado´s et al. [36] demonstrated that Ki-67 was required for maintenance of cancer stem cells but not cell proliferation. However, additional studies are needed to clarify the exact mechanism of how Ki-67 regulates CSC properties.
The histologic grade also is a prognostic factor for breast cancer, regardless of tumor size and the number of involved axillary lymph nodes [37]. In our population, patients with poorly differentiated primary tumor (G3) had significantly more tumorspheres as compared to patients with well differentiated tumor (G1). To our knowledge, there are no data in the literature with respect to the relationship between the number of cancer stem cells and the histologic grade in breast cancer. However, Mohanta et al. [38] showed that patients with poorly differentiated squamous cell carcinoma of the oral cavity had more CSCs, which correlated with poor prognosis in clinical settings.
The CAM assay is a frequently applied model to study cancer cell invasion and metastasis [13]. In our approach we sought to investigate the functional properties of tumorspheres. The CAM model has many advantages. It is cost effective, allows large scale screening and is an easily reproducible in vivo model [39,40]. We show here, for the first time, that tumorspheres cultured from peripheral blood in patients with breast cancer are able to induce tumors on CAM upon application. The efficacy of inoculation using surgical specimens varies widely, ranging from about 45% [41] to much higher rates of 70–80% [42,43,44] depending on the type of tumor. Baccelli et al. [45] identified a population of tumor cells circulating in blood from breast cancer patients that initiates metastasis in a xenograft assay, albeit at a much lower frequency in obtaining xenografts. As shown in the Table 2, five patients (50%) have successfully grown tumors on CAM and the success rate was associated with the number of tumorspheres. Moreover, we suppose that the inoculation rate is also dependent upon Ki-67 status and histologic grade of primary tumor, because only patients with high Ki-67 and low histologic grade have developed tumors on CAM membrane. Histopathological analysis was the key endpoint assay of our experiment, which confirmed that tumors grown on CAM had the original morphological profile of the patient’s tumor. Given the high efficiency of patient derived tumor on CAM in a short 8-day period, it holds great promise as an in vivo platform to pursue pilot drug screening on an individual patient’s tumor. Our model has an advantage over others because the biological material used for CAM-assay is easily and repeatedly available without high risk to the patient. A limitation of our study is the small number of patients who underwent CAM experiment, but further clinical validation for a large-size sample set remains to be done in future work.
Personalized medicine aims to provide cancer patients with tailored treatments, so it is necessary to use patient-derived tumor models to characterize possible treatment options. Currently, tumor organoids [46] or patient-derived xenograft mouse systems [47] are used for this purpose. Based on our success in growing patient derived tumors from tumor cells circulating in peripheral blood in the CAM assay, our model may provide an interesting alternative for investigating the individual tumor biology and a suitable platform for the testing of different drugs in personalized oncology.

5. Conclusions

The number of tumorspheres cultured from peripheral blood of cancer patients and the success rate of establishing patient derived xenografts on CAM directly reflect the aggressiveness and proliferation capacity of the primary tumor. The CAM-based PDX model using circulating cancer stem cells could provide an alternative step in translational cancer research to assess experimental and novel strategies for both diagnostic and therapeutic purposes.

Author Contributions

Conceptualization, M.P. (Monika Pizon) and K.P.; Methodology, M.P. (Monika Pizon), D.S. and R.S.; Software, D.S.; Validation, M.P. (Monika Pizon) and D.S.; Formal analysis, D.S. and M.P. (Marek Pizon); Investigation, D.S. and M.P. (Monika Pizon); Resources, K.P. and U.P.; Data curation, M.P. (Monika Pizon), D.S. and M.W.; Writing—original draft preparation, M.P. (Monika Pizon); Writing—review and editing, K.P., M.W., U.P., R.B. and M.P. (Marek Pizon); Visualization, D.S.; Supervision, K.P.; Project administration, R.S.; Funding acquisition, U.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and was approved by the ethics committee of University Bayreuth (O 1305/1-GB).

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.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Mohme, M.; Riethdorf, S.; Pantel, K. Circulating and Disseminated Tumour Cells—Mechanisms of Immune Surveillance and Escape. Nat. Rev. Clin. Oncol. 2017, 14, 155–167. [Google Scholar] [CrossRef] [PubMed]
  3. Schott, D.S.; Pizon, M.; Pachmann, U.; Pachmann, K.; Schobert, R.; Wittig, A.; Mäurer, M. Influence of Adjuvant Radiotherapy on Circulating Epithelial Tumor Cells and Circulating Cancer Stem Cells in Primary Non-Metastatic Breast Cancer. Transl. Oncol. 2021, 14, 101009. [Google Scholar] [CrossRef] [PubMed]
  4. Zhang, H.; Lin, X.; Huang, Y.; Wang, M.; Cen, C.; Tang, S.; Dique, M.R.; Cai, L.; Luis, M.A.; Smollar, J.; et al. Detection Methods and Clinical Applications of Circulating Tumor Cells in Breast Cancer. Front. Oncol. 2021, 11, 652253. [Google Scholar] [CrossRef] [PubMed]
  5. Liu, H.; Patel, M.R.; Prescher, J.A.; Patsialou, A.; Qian, D.; Lin, J.; Wen, S.; Chang, Y.-F.; Bachmann, M.H.; Shimono, Y.; et al. Cancer Stem Cells from Human Breast Tumors Are Involved in Spontaneous Metastases in Orthotopic Mouse Models. Proc. Natl. Acad. Sci. USA 2010, 107, 18115–18120. [Google Scholar] [CrossRef] [Green Version]
  6. Dittmer, J. Breast Cancer Stem Cells: Features, Key Drivers and Treatment Options. Semin. Cancer Biol. 2018, 53, 59–74. [Google Scholar] [CrossRef]
  7. Klonisch, T.; Wiechec, E.; Hombach-Klonisch, S.; Ande, S.R.; Wesselborg, S.; Schulze-Osthoff, K.; Los, M. Cancer Stem Cell Markers in Common Cancers—Therapeutic Implications. Trends Mol. Med. 2008, 14, 450–460. [Google Scholar] [CrossRef] [Green Version]
  8. Saeg, F.; Anbalagan, M. Breast Cancer Stem Cells and the Challenges of Eradication: A Review of Novel Therapies. Stem Cell Investig. 2018, 5, 39. [Google Scholar] [CrossRef]
  9. Krawczyk, N.; Meier-Stiegen, F.; Banys, M.; Neubauer, H.; Ruckhaeberle, E.; Fehm, T. Expression of Stem Cell and Epithelial-Mesenchymal Transition Markers in Circulating Tumor Cells of Breast Cancer Patients. BioMed Res. Int. 2014, 2014, 415721. [Google Scholar] [CrossRef]
  10. Kantara, C.; O’Connell, M.R.; Luthra, G.; Gajjar, A.; Sarkar, S.; Ullrich, R.L.; Singh, P. Methods for Detecting Circulating Cancer Stem Cells (CCSCs) as a Novel Approach for Diagnosis of Colon Cancer Relapse/Metastasis. Lab. Investig. 2015, 95, 100–112. [Google Scholar] [CrossRef] [Green Version]
  11. Pizon, M.; Zimon, D.; Carl, S.; Pachmann, U.; Pachmann, K.; Camara, O. Heterogeneity of Circulating Epithelial Tumour Cells from Individual Patients with Respect to Expression Profiles and Clonal Growth (Sphere Formation) in Breast Cancer. Ecancermedicalscience 2013, 7, 343. [Google Scholar] [PubMed]
  12. Pizon, M.; Schott, D.; Pachmann, U.; Pachmann, K. The Number of Tumorspheres Cultured from Peripheral Blood Is a Predictor for Presence of Metastasis in Patients with Breast Cancer. Oncotarget 2016, 7, 48143–48154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Ribatti, D. The Chick Embryo Chorioallantoic Membrane (CAM). A Multifaceted Experimental Model. Mech. Dev. 2016, 141, 70–77. [Google Scholar] [CrossRef] [PubMed]
  14. Nowak-Sliwinska, P.; Segura, T.; Iruela-Arispe, M.L. The Chicken Chorioallantoic Membrane Model in Biology, Medicine and Bioengineering. Angiogenesis 2014, 17, 779–804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Swadi, R.; Mather, G.; Pizer, B.L.; Losty, P.D.; See, V.; Moss, D. Optimising the Chick Chorioallantoic Membrane Xenograft Model of Neuroblastoma for Drug Delivery. BMC Cancer 2018, 18, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Dünker, N.; Jendrossek, V. Implementation of the Chick Chorioallantoic Membrane (CAM) Model in Radiation Biology and Experimental Radiation Oncology Research. Cancers 2019, 11, 1499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Chu, P.-Y.; Koh, A.P.-F.; Antony, J.; Huang, R.Y.-J. Applications of the Chick Chorioallantoic Membrane as an Alternative Model for Cancer Studies. Cells Tissues Organs 2021, 1–16. [Google Scholar] [CrossRef]
  18. Pachmann, K.; Clement, J.H.; Schneider, C.-P.; Willen, B.; Camara, O.; Pachmann, U.; Höffken, K. Standardized Quantification of Circulating Peripheral Tumor Cells from Lung and Breast Cancer. Clin. Chem. Lab. Med. 2005, 43, 617–627. [Google Scholar] [CrossRef]
  19. Batlle, E.; Clevers, H. Cancer Stem Cells Revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef]
  20. Dean, M.; Fojo, T.; Bates, S. Tumour Stem Cells and Drug Resistance. Nat. Rev. Cancer 2005, 5, 275–284. [Google Scholar] [CrossRef]
  21. Jordan, C.T.; Guzman, M.L.; Noble, M. Cancer Stem Cells. N. Engl. J. Med. 2006, 355, 1253–1261. [Google Scholar] [CrossRef] [PubMed]
  22. Theodoropoulos, P.A.; Polioudaki, H.; Agelaki, S.; Kallergi, G.; Saridaki, Z.; Mavroudis, D.; Georgoulias, V. Circulating Tumor Cells with a Putative Stem Cell Phenotype in Peripheral Blood of Patients with Breast Cancer. Cancer Lett. 2010, 288, 99–106. [Google Scholar] [CrossRef] [PubMed]
  23. Scatena, R.; Bottoni, P.; Giardina, B. Circulating Tumour Cells and Cancer Stem Cells: A Role for Proteomics in Defining the Interrelationships between Function, Phenotype and Differentiation with Potential Clinical Applications. Biochim. Biophys. Acta (BBA)-Rev. Cancer 2013, 1835, 129–143. [Google Scholar] [CrossRef] [PubMed]
  24. Al-Hajj, M.; Wicha, M.S.; Benito-Hernandez, A.; Morrison, S.J.; Clarke, M.F. Prospective Identification of Tumorigenic Breast Cancer Cells. Proc. Natl. Acad. Sci. USA 2003, 100, 3983–3988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ricci-Vitiani, L.; Lombardi, D.G.; Pilozzi, E.; Biffoni, M.; Todaro, M.; Peschle, C.; De Maria, R. Identification and Expansion of Human Colon-Cancer-Initiating Cells. Nature 2007, 445, 111–115. [Google Scholar] [CrossRef] [PubMed]
  26. Singh, S.K.; Clarke, I.D.; Terasaki, M.; Bonn, V.E.; Hawkins, C.; Squire, J.; Dirks, P.B. Identification of a Cancer Stem Cell in Human Brain Tumors. Cancer Res. 2003, 63, 5821–5828. [Google Scholar] [PubMed]
  27. Collins, A.T.; Berry, P.A.; Hyde, C.; Stower, M.J.; Maitland, N.J. Prospective Identification of Tumorigenic Prostate Cancer Stem Cells. Cancer Res. 2005, 65, 10946–10951. [Google Scholar] [CrossRef] [Green Version]
  28. Iinuma, H.; Watanabe, T.; Mimori, K.; Adachi, M.; Hayashi, N.; Tamura, J.; Matsuda, K.; Fukushima, R.; Okinaga, K.; Sasako, M.; et al. Clinical Significance of Circulating Tumor Cells, Including Cancer Stem-Like Cells, in Peripheral Blood for Recurrence and Prognosis in Patients with Dukes’ Stage B and C Colorectal Cancer. J. Clin. Oncol. 2011, 29, 1547–1555. [Google Scholar] [CrossRef]
  29. Toyoshima, K.; Hayashi, A.; Kashiwagi, M.; Hayashi, N.; Iwatsuki, M.; Ishimoto, T.; Baba, Y.; Baba, H.; Ohta, Y. Analysis of Circulating Tumor Cells Derived from Advanced Gastric Cancer: Analysis of CTC Derived from Gastric Cancer. Int. J. Cancer 2015, 137, 991–998. [Google Scholar] [CrossRef]
  30. Pirozzi, G.; Tirino, V.; Camerlingo, R.; La Rocca, A.; Martucci, N.; Scognamiglio, G.; Franco, R.; Cantile, M.; Normanno, N.; Rocco, G. Prognostic Value of Cancer Stem Cells, Epithelial-Mesenchymal Transition and Circulating Tumor Cells in Lung Cancer. Oncol. Rep. 2013, 29, 1763–1768. [Google Scholar] [CrossRef] [Green Version]
  31. Chang, P.-H.; Wu, M.-H.; Liu, S.-Y.; Wang, H.-M.; Huang, W.-K.; Liao, C.-T.; Yen, T.-C.; Ng, S.-H.; Chen, J.-S.; Lin, Y.-C.; et al. The Prognostic Roles of Pretreatment Circulating Tumor Cells, Circulating Cancer Stem-Like Cells, and Programmed Cell Death-1 Expression on Peripheral Lymphocytes in Patients with Initially Unresectable, Recurrent or Metastatic Head and Neck Cancer: An Exploratory Study of Three Biomarkers in One-Time Blood Drawing. Cancers 2019, 11, 540. [Google Scholar] [CrossRef] [Green Version]
  32. Korkaya, H.; Paulson, A.; Iovino, F.; Wicha, M.S. Her2 Regulates the Mammary Stem/Progenitor Cell Population Driving Tumorigenesis and Invasion. Oncogene 2008, 27, 6120–6130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ginestier, C.; Hur, M.H.; Charafe-Jauffret, E.; Monville, F.; Dutcher, J.; Brown, M.; Jacquemier, J.; Viens, P.; Kleer, C.G.; Liu, S.; et al. ALDH1 Is a Marker of Normal and Malignant Human Mammary Stem Cells and a Predictor of Poor Clinical Outcome. Cell Stem Cell 2007, 1, 555–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Fasching, P.A.; Heusinger, K.; Haeberle, L.; Niklos, M.; Hein, A.; Bayer, C.M.; Rauh, C.; Schulz-Wendtland, R.; Bani, M.R.; Schrauder, M.; et al. Ki67, Chemotherapy Response, and Prognosis in Breast Cancer Patients Receiving Neoadjuvant Treatment. BMC Cancer 2011, 11, 486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Harbeck, N.; Penault-Llorca, F.; Cortes, J.; Gnant, M.; Houssami, N.; Poortmans, P.; Ruddy, K.; Tsang, J.; Cardoso, F. Breast Cancer. Nat. Rev. Dis. Primers 2019, 5, 66. [Google Scholar] [CrossRef] [PubMed]
  36. Cidado, J.; Wong, H.Y.; Rosen, D.M.; Cimino-Mathews, A.; Garay, J.P.; Fessler, A.G.; Rasheed, Z.A.; Hicks, J.; Cochran, R.L.; Croessmann, S.; et al. Ki-67 Is Required for Maintenance of Cancer Stem Cells but Not Cell Proliferation. Oncotarget 2016, 7, 6281–6293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Schwartz, A.M.; Henson, D.E.; Chen, D.; Rajamarthandan, S. Histologic Grade Remains a Prognostic Factor for Breast Cancer Regardless of the Number of Positive Lymph Nodes and Tumor Size: A Study of 161,708 Cases of Breast Cancer from the SEER Program. Arch. Pathol. Lab. Med. 2014, 138, 1048–1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Mohanta, S.; Siddappa, G.; Valiyaveedan, S.G.; Dodda Thimmasandra Ramanjanappa, R.; Das, D.; Pandian, R.; Khora, S.S.; Kuriakose, M.A.; Suresh, A. Cancer Stem Cell Markers in Patterning Differentiation and in Prognosis of Oral Squamous Cell Carcinoma. Tumour Biol. 2017, 39, 101042831770365. [Google Scholar] [CrossRef] [Green Version]
  39. Tufan, A.; Satiroglu-Tufan, N. The Chick Embryo Chorioallantoic Membrane as a Model System for the Study of Tumor Angiogenesis, Invasion and Development of Anti-Angiogenic Agents. Curr. Cancer Drug Targets 2005, 5, 249–266. [Google Scholar] [CrossRef]
  40. Cimpean, A.M.; Ribatti, D.; Raica, M. The Chick Embryo Chorioallantoic Membrane as a Model to Study Tumor Metastasis. Angiogenesis 2008, 11, 311–319. [Google Scholar] [CrossRef]
  41. Sommers, S.C.; Sullivan, B.A.; Warren, S. Heterotransplantation of Human Cancer. III. Chorioallantoic Membranes of Embryonated Eggs. Cancer Res. 1952, 12, 915–917. [Google Scholar] [PubMed]
  42. Kunz, P.; Schenker, A.; Sähr, H.; Lehner, B.; Fellenberg, J. Optimization of the Chicken Chorioallantoic Membrane Assay as Reliable in Vivo Model for the Analysis of Osteosarcoma. PLoS ONE 2019, 14, e0215312. [Google Scholar] [CrossRef] [PubMed]
  43. Rovithi, M.; Avan, A.; Funel, N.; Leon, L.G.; Gomez, V.E.; Wurdinger, T.; Griffioen, A.W.; Verheul, H.M.W.; Giovannetti, E. Development of Bioluminescent Chick Chorioallantoic Membrane (CAM) Models for Primary Pancreatic Cancer Cells: A Platform for Drug Testing. Sci. Rep. 2017, 7, 44686. [Google Scholar] [CrossRef] [PubMed]
  44. Uloza, V.; Kuzminiene, A.; Palubinskiene, J. Model of Human Recurrent Respiratory Papilloma on Chicken Embryo Chorioallantoic Membrane for Tumor Angiogenesis Research. Histol. Histopathol. 2017, 699–710. [Google Scholar] [CrossRef]
  45. Baccelli, I.; Schneeweiss, A.; Riethdorf, S.; Stenzinger, A.; Schillert, A.; Vogel, V.; Klein, C.; Saini, M.; Bäuerle, T.; Wallwiener, M.; et al. Identification of a Population of Blood Circulating Tumor Cells from Breast Cancer Patients That Initiates Metastasis in a Xenograft Assay. Nat. Biotechnol. 2013, 31, 539–544. [Google Scholar] [CrossRef] [PubMed]
  46. Weeber, F.; Ooft, S.N.; Dijkstra, K.K.; Voest, E.E. Tumor Organoids as a Pre-Clinical Cancer Model for Drug Discovery. Cell Chem. Biol. 2017, 24, 1092–1100. [Google Scholar] [CrossRef] [PubMed]
  47. Jung, J.; Seol, H.S.; Chang, S. The Generation and Application of Patient-Derived Xenograft Model for Cancer Research. Cancer Res. Treat. 2018, 50, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. In vivo workflow: A timeline of chorioallantoic member (CAM). Fertilized eggs are incubated for 4 days, at which time a shell window is opened. At embryonic development day 8, tumorspheres are inoculated on top of the CAM. At day 16, the eggs are sacrificed, and the tumor growth is examined.
Figure 1. In vivo workflow: A timeline of chorioallantoic member (CAM). Fertilized eggs are incubated for 4 days, at which time a shell window is opened. At embryonic development day 8, tumorspheres are inoculated on top of the CAM. At day 16, the eggs are sacrificed, and the tumor growth is examined.
Cancers 14 01476 g001
Figure 2. (a) Exemplary pictures of tumorspheres in light microscope. (b) Fluorescence staining of tumorspheres with cancer stem cell makers typical for breast cancer. (c) Expression of aldehyde dehydrogenase 1 (ALDH-1) in tumorspheres. Scale bar = 40 µm; magnification ×200.
Figure 2. (a) Exemplary pictures of tumorspheres in light microscope. (b) Fluorescence staining of tumorspheres with cancer stem cell makers typical for breast cancer. (c) Expression of aldehyde dehydrogenase 1 (ALDH-1) in tumorspheres. Scale bar = 40 µm; magnification ×200.
Cancers 14 01476 g002
Figure 3. (a) Box plot analysis of the number of tumorspheres in patients without and with metastatic disease. (b) Box plot analysis showing the number of tumorspheres in patients with single and multiple metastases. (c) Box plot analysis of the number of tumorspheres in patients negative and positive for Her2-neu in primary tumor. (d) Box plot analysis showing the number of tumorspheres in patients with Ki-67 < 15% and >15% in primary tumor. The horizontal line in box-plot depicts median value. Dots specify outliers.
Figure 3. (a) Box plot analysis of the number of tumorspheres in patients without and with metastatic disease. (b) Box plot analysis showing the number of tumorspheres in patients with single and multiple metastases. (c) Box plot analysis of the number of tumorspheres in patients negative and positive for Her2-neu in primary tumor. (d) Box plot analysis showing the number of tumorspheres in patients with Ki-67 < 15% and >15% in primary tumor. The horizontal line in box-plot depicts median value. Dots specify outliers.
Cancers 14 01476 g003
Figure 4. Box plot analysis of the number of tumorspheres according to the histologic tumor grade (G1—well-differentiated tumor; G2—moderately differentiated tumor; G3—poorly differentiated tumor). The horizontal line in box-plot depicts median value. Dots specify outliers.
Figure 4. Box plot analysis of the number of tumorspheres according to the histologic tumor grade (G1—well-differentiated tumor; G2—moderately differentiated tumor; G3—poorly differentiated tumor). The horizontal line in box-plot depicts median value. Dots specify outliers.
Cancers 14 01476 g004
Figure 5. (a) Development of a microtumor from tumorspheres of a breast cancer patient on CAM membrane. Scale bar = 5 mm; magnification ×20. (b) H&E and pan-cytokeratin staining of a CAM xenograft developed from tumorspheres in light microscopy. Scale bar = 100 µm for H&E, scale bar = 200 µm for pan-cytokeratin; magnification ×200.
Figure 5. (a) Development of a microtumor from tumorspheres of a breast cancer patient on CAM membrane. Scale bar = 5 mm; magnification ×20. (b) H&E and pan-cytokeratin staining of a CAM xenograft developed from tumorspheres in light microscopy. Scale bar = 100 µm for H&E, scale bar = 200 µm for pan-cytokeratin; magnification ×200.
Cancers 14 01476 g005
Figure 6. (a) Box plot analysis of the number of tumorpheres cultured from peripheral blood in patients who developed a tumor or not on the CAM membrane. (b) Box plot showing the Ki-67 proliferation index of primary tumor in patients with breast cancer, with or without tumor on CAM membrane. The horizontal line in box-plot depicts median value.
Figure 6. (a) Box plot analysis of the number of tumorpheres cultured from peripheral blood in patients who developed a tumor or not on the CAM membrane. (b) Box plot showing the Ki-67 proliferation index of primary tumor in patients with breast cancer, with or without tumor on CAM membrane. The horizontal line in box-plot depicts median value.
Cancers 14 01476 g006
Table 1. Clinicopathological parameters of patients with breast cancer.
Table 1. Clinicopathological parameters of patients with breast cancer.
Clinicopathological
Characteristics
Number of
Patients
Median Number of Tumorspheresp-Value
Age
≤50 years2020p = 0.206
>50 years5510
Tumor size
T1267.5p = 0.318
T23312.5
T3925
T4730
Lymph node
positive3510p = 0.967
negative4010
Clinical stage
I135p < 0.01
II2310
III1610
IV2332.5
ER/PR status
positive6215p = 0.955
negative1320
Her2-neu
positive1635p < 0.05
negative5910
Grading
1140p < 0.01
2335
32832.5
Ki-67 Index
>15%2934p < 0.01
<15%468
Metastasis
positive2332.5p < 0.01
negative5210
Table 2. Characteristics of patients who underwent CAM experiment.
Table 2. Characteristics of patients who underwent CAM experiment.
PatientsGradingKi-67ER/PRHer2-neuMetastasisTumorspheresTumor on CAM
1G340%positivepositivepositive80present
2G375%positivenegativenegative100present
3G365%negativenegativenegative50present
4G240%positivepositivenegative55present
5G330%positivepositivenegative20present
6G215%positivepositivenegative55absent
7G320%positivenegativenegative45absent
8G25%positivenegativenegative5absent
9G320%positivenegativenegative15absent
10G210%positivenegativenegative0absent
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pizon, M.; Schott, D.; Pachmann, U.; Schobert, R.; Pizon, M.; Wozniak, M.; Bobinski, R.; Pachmann, K. Chick Chorioallantoic Membrane (CAM) Assays as a Model of Patient-Derived Xenografts from Circulating Cancer Stem Cells (cCSCs) in Breast Cancer Patients. Cancers 2022, 14, 1476. https://doi.org/10.3390/cancers14061476

AMA Style

Pizon M, Schott D, Pachmann U, Schobert R, Pizon M, Wozniak M, Bobinski R, Pachmann K. Chick Chorioallantoic Membrane (CAM) Assays as a Model of Patient-Derived Xenografts from Circulating Cancer Stem Cells (cCSCs) in Breast Cancer Patients. Cancers. 2022; 14(6):1476. https://doi.org/10.3390/cancers14061476

Chicago/Turabian Style

Pizon, Monika, Dorothea Schott, Ulrich Pachmann, Rainer Schobert, Marek Pizon, Marta Wozniak, Rafal Bobinski, and Katharina Pachmann. 2022. "Chick Chorioallantoic Membrane (CAM) Assays as a Model of Patient-Derived Xenografts from Circulating Cancer Stem Cells (cCSCs) in Breast Cancer Patients" Cancers 14, no. 6: 1476. https://doi.org/10.3390/cancers14061476

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