Organoid > Volume 1; 2021 > Article
Choi, Lim, Kim, Lee, Kwon, and Lee: A novel method for the isolation of CD45-positive and CD45-negative cells from malignant pleural effusion

Abstract

Background

Patient-derived malignant pleural effusion (MPE) samples can be used to identify a patient-specific drug combination since MPE samples are readily available and cost-effective tumor cell sources. However, the isolation of target cancer cells from MPE has been inefficient because MPE samples contain a complex mixture of immune cells, non-cancerous cells, and cancer cells. Hence, new methods need to be developed to effectively isolate target cancer cells from MPE samples that can be used for 3-dimensional (3D) cell culture. Patient-derived in vitro 3D tumor models are expected to facilitate more precise drug treatment.

Methods

MPE samples were obtained from Seoul St. Mary’s Hospital, The Catholic University of Korea with consent from patients previously diagnosed with lung adenocarcinoma. We isolated target cells from MPE samples using 2 different Percoll-gradient centrifugation methods.

Results

The use of 40% and 75% Percoll-gradient centrifugation led to a clearer separation of CD45-positive (CD45pos) and CD45-negative (CD45neg) cells than the traditional 44% and 67% Percoll-gradient centrifugation method.

Conclusion

Our findings strongly suggest that the 40% and 75% Percoll-gradient centrifugation method is more useful for the isolation of CD45pos or CD45neg cells than the previously described Percoll-gradient centrifugation method. Furthermore, our novel method was useful for the isolation of MPE-derived target cancer cells that can be used to construct in vitro patient-specific 3D tumor models.

Introduction

Patient-derived cells isolated from tumor biopsies can be used to track disease progression and screen for chemotherapy drug response, representing an ideal platform for precision medicine applications [1]. Patient-derived mouse xenografts (PDX) models have been shown to predict clinical outcomes more accurately than conventional cell line-derived xenograft models, particularly when therapeutic compounds are evaluated with clinically relevant doses [2-4]. However, the time taken for engraftment and low tumor growth rates do not allow a timely assessment of a patient's chemosensitivity for therapeutic decisions. Furthermore, patient-derived PDX models are usually obtained from biopsies or resected primary tumors, but these samples often have limited availability or cellular vitality and are not adequately preserved, which causes low engraftment rates [5-7]. As an alternative to PDX models, in vitro 3-dimensional (3D) cell culture models mimicking in vivo environments have been applied to drug toxicity testing [8] and have offered better precision in drug discovery [9]. In vitro patient-derived 3D cell culture models have been developed by using extracellular matrix-based organoid culture protocols and 3D co-culture methods [10]. Both PDX and 3D cell culture models require the isolation of target cells from a limited number of patient-derived tumor biopsies. Hence, the efficient preparation of substantial amounts of target cells from tumor tissue biopsies is clearly needed to construct patient-derived tumor models allowing the identification of patient-specific combination regimens.
Malignant pleural effusion (MPE) is defined as a significant accumulation of exudate in the pleural cavity containing malignant cells or tumor tissue. MPE is a serious medical condition causing shortness of breath, pain, cachexia, and decreased physical activity [11,12]. MPE, which is a common occurrence in many end-stage epithelial cancers [13], contains cancer cells, as well as large numbers of immune cells including various lymphocytes and myeloid subsets [14]. Several mechanisms are involved in the generation of MPE, including pleural metastasis with increased permeability, pleural metastasis with obstruction of the pleural lymphatic vessels, mediastinal lymph node involvement, chest tube obstruction, bronchial obstruction, and pericardial involvement. In addition, pleural effusion can also occur as a result of hypoproteinemia, obstructive pneumonia, or pulmonary embolism in patients with malignant tumors [15].
Because MPE samples (liquid biopsies) are readily available and contain various cell types exhibiting the heterogeneity of lung adenocarcinoma, MPE samples can be used as cost-effective tumor cell sources to generate patient-specific lung cancer models for future precision medicine applications. In this study, we optimized the Percoll-gradient centrifugation protocol for isolating CD45-positive (CD45pos) and CD45-negative (CD45neg) cells from MPE aspirate biopsies in lung adenocarcinoma patients, followed by establishing patient-derived 3D organoid culture. In addition, we compared the 3D cell culture viability of CD45neg cells isolated from MPE samples with non-isolated cells (CD45pos/neg).

Materials and Methods

Ethics statement: This study was approved by the Institutional Review Board (IRB) of the Catholic University of Korea Seoul St. Mary's hospital (No.KC18TNSI0033). Informed consent was obtained from all participants.

1. Cell isolation

MPE samples were obtained from Seoul St. Mary’s Hospital, The Catholic University of Korea with consent from patients previously diagnosed with lung adenocarcinoma. This study was approved by the Institutional Review Board (IRB) of The Catholic University of Korea (IRB no. KC18TNSI0033).
Each sample contained between 40 and 60 mL of fluid, yielding 1×107 cells for use in culture. Samples were transferred from the primary container to a 50-mL centrifuge tube, followed by centrifuging the tube at 440×g for 10 minutes at room temperature. The cell pellets were resuspended and washed 2 times with advanced dulbecco's modified eagle medium: nutrient mixture F-12 medium. A 90% Percoll solution was prepared by mixing the appropriate amount of Percoll (d=1.130 g/mL) with Hank’s balanced salt solution (HBSS, with phenol red), and then 40% and 75% Percoll solutions were prepared by mixing an appropriate amount of 90% Percoll (d=1.130 g/mL) and buffer 1 (10% fetal bovine serum [FBS]+100 μM ethylenediamine tetraacetic acid [EDTA] in phosphate-buffered saline [PBS]). The MPE samples were strained with a 70-m cell strainer and centrifuged at 440×g for 10 minutes at room temperature, and the cell pellets were rinsed with rRoswell Park Memorial Institute medium. After spin-down, each cell pellet was resuspended in a single-cell suspension with 5 mL of 75% Percoll solution and transferred to a 15-mL centrifuge tube. The 40% Percoll solution was gently added to the 75% Percoll solution and centrifuged at 780×g for 20 minutes at room temperature without interruption.

2. Cell culture

Target cells were isolated from MPE of lung adenocarcinoma for 3D cell cultures. The isolated cells (CD45neg or CD45pos/neg; 1×105 cells) were resuspended in 30 μL of 70% Matrigel (354230; Corning Life Sciences, Corning, NY, USA), and the Matrigel-cell mixture was spotted into each well of 4-well TCP plates (Thermo, Rochester, NY, USA), which were then incubated face down at 4°C for 15 minutes to allow cell aggregation at the bottom of the spots. The plates were then placed at 37°C in a humidity chamber to allow spot gelation. After gelation, fresh medium (CODRP_LOM; Medical & Bio Decision Co., Ltd., Suwon, Korea) was added to culture the cells isolated from MPE.

3. Flow cytometry analysis

Cells were incubated with an allophycocyanin (APC)-conjugated anti-CD3 antibody and fluorescein isothiocyanate (FITC)-conjugated anti-CD45 antibody in PBS containing 1% FBS for 30 minutes at 4°C. After washing, flow cytometry analysis of the fluorescent-labeled cells was performed to identify the population of CD45pos and CD45neg cells. Data acquisition and analysis were performed on a BD FACS Canto II (BD, Franklin Lakes, NJ, USA).

Results

1. The isolation of MPE-derived tumor cells from lung cancer patients

Tumor cells present in MPE samples from patients can be used for both in vivo PDX and in vitro 3D culture models to predict patients’ chemosensitivity. However, the isolation of cancer cells from pleural fluid has been difficult because the fluid contains normal epithelial cells, fibroblasts, immune cells, and cancer cells. To isolate cancer cells from the pleural effusions of lung cancer patients, we used different Percoll-gradient centrifugation methods, which have been used for isolating lymphocytes and other cells from tumor tissues [16-18]. Rapp et al. [17] reported that 44% and 67% Percoll solutions have been used to isolate intratumoral leukocytes from tumor tissues. Using the previous method [17], we tried to isolate CD45pos cells, including leukocytes, and CD45neg cells, including tumor cells, from MPE. For Percoll-gradient centrifugation, appropriate amounts of Percoll (d=1.130 g/mL) and PBS were mixed to prepare 44% and 67% Percoll solutions. The next steps were to add 9 mL of 44% Percoll solution over 6 mL of the 67% Percoll solution in a 50-mL centrifuge tube. Finally, the resuspended single-cell suspension obtained from the MPE was added onto the top layer and centrifuged at 800×g for 30 minutes at 4°C without interruption (Fig. 1A). After centrifugation, a mixture of both CD45neg and CD45pos cells was found in the Percoll top layer and middle layer (Fig. 2A and 3B).
To solve this problem, we modified the Percoll-gradient centrifugation method. We prepare a 90% Percoll solution by mixing the appropriate amount of Percoll (d=1.130 g/mL) with HBSS (with phenol red). For Percoll-gradient centrifugation, 40% and 75% Percoll solutions were prepared by mixing an appropriate amount of 90% Percoll (d=1.130 g/mL) and buffer 1 (10% FBS+100 μM EDTA in PBS). The MPE samples were strained with a 70-m cell strainer and centrifuged at 440×g for 10 minutes at room temperature, and the cell pellets were rinsed with RPMI medium. After spin-down, each cell pellet was resuspended in a single-cell suspension with 5 mL of 75% Percoll solution and transferred to a 15-mL centrifuge tube. Then, 40% Percoll solution was gently added to the 75% Percoll solution and centrifuged at 780×g for 20 minutes at room temperature without interruption (Fig. 1B). Surprisingly, we were able to observe a two clear layer at the interface of the 2 Percoll layers (Fig. 2B). In addition, >94% of the CD45pos cells were found in the Percoll middle layer, whereas >98% of the CD45neg cells were found on the top layer of 40% Percoll solution (Fig. 2B and 3C). No cells were found in the region between the top and middle layer of Percoll solution (data not shown).

2. Separation of both CD45pos and CD45neg cells from MPE

To measure the levels of CD45 expression in the cells in the top and middle layer of the Percoll solution, we performed fluorescence-activated cell sorting (FACS) analysis. The cells from each Percoll layer were stained with anti-CD45 antibody conjugated with FITC and anti-CD3 antibody conjugated with APC. The FACS data showed that the CD45pos and CD45neg cells were not separated using the 44% and 67% Percoll-gradient centrifugation method (Fig. 3B). However, the 40% and 75% Percoll-gradient centrifugation method led to a clear separation of CD45pos and CD45neg cells (Fig. 3C).

3. The effects of CD45pos cells on the viability of CD45neg cells in 3D cell culture

Next, we tested whether a cell mixture (CD45pos/neg) could affect the viability of CD45neg cells in a 3D cell culture condition. The cells collected from each top layer of 2 different Percoll concentration methods were embedded into Matrigel for 3D cell culture. After 5 days, the number of CD45neg cells obtained from 44% and 67% Percoll solution was significantly reduced (Fig. 4A). In contrast, CD45neg cells isolated from the top layer of the 40% and 75% Percoll gradients formed 3D aggregates (organoids) without cell death (Fig. 4B). These results indicate that the significant growth inhibition of the non-separated CD45pos/neg cell mixture could have resulted from immune cell cytotoxicity in CD45pos cells, since there was no growth inhibition of the CD45neg cell mixture in the absence of CD45pos cells.

Discussion

MPE is associated with a poor prognosis and shortened survival of patients with non-small cell lung cancer. The development of MPE involves various mechanisms such as inflammation, angiogenesis promotion, and vascular leakage. We believe that aggressive cancers accompanied by MPE require novel therapeutic strategies of precision medicine. Therefore, it is necessary to expand patient-derived cancer cells to perform a customized chemosensitivity assay for patient-specific care.
Patient-derived organoids (PDOs) have recently been developed as a promising tool for personalized medical decisions [19-21]. When compared to cell lines and animal models, PDOs can offer more representative disease models, providing a genetically manipulable system that can be propagated, stored, and transferred into complex 3D in vitro or murine in vivo models [22]. These PDOs allow human in vitro immunotherapy modeling through co-culture with native syngeneic, autologous tumor-reactive tumor-infiltrating lymphocytes (TILs). However, the co-culture of primary tumor cells with endogenous, syngeneic TILs has been particularly troublesome. Recently, Neal et al. [23] developed an air-liquid interface method for propagating PDOs containing native immune cells (macrophages, B cells, and natural killer cells) to facilitate personalized immunotherapy testing. Along with the previous study, we observed that growth inhibition of patient-derived cancer cells was observed in the presence of CD45pos cells. Therefore, to preserve and proliferate patient-derived tumor cells, it is critical to remove CD45pos cells from patient-derived tumor tissue samples.
The results from this study showed that CD45neg cells were successfully separated from a mixture of CD45pos and CD45neg cells in MPE samples using the 40% and 75% Percoll-gradient centrifugation method. Moreover, after separation, we were able to freshly store CD45pos cells, which can be used as patient-derived immune cells for application in the development of immunotherapies. In addition, 3D cell culture using isolated CD45neg cells can be used for chemosensitivity assays or immunotherapy using immune checkpoint modulators. In summary, 3D cell culture models using the CD45neg cells isolated from patient MPE samples might be a promising strategy for developing personalized medicine.

NOTES

Conflict of interest

No potential conflict of interest relevant to this article was reported.

Funding

This study was supported by a grant from the National Research Foundation of Korea, which was funded by the Ministry of Science (NRF-2019R1A2C1089258).

Author contributions

Conceptualization: JC, SHL; Data curation: JC, JUL, WK; Formal analysis: Funding acquisition: SHL; Methodology: JC; Project administration: SHL; Investigation: DWL, SJK; Resources: JUL, SHL; Software: WK; Supervision: JUL, DWL; Validation: SJK; Writing-review & editing: SJK, SHL.

Data availability

Please contact the corresponding author for data availability.

Fig. 1.
Overview of the experimental design. (A) A previously proposed experimental method and (B) the new method for isolating CD45-negative (CD45neg) or CD45-positive (CD45pos) cells from patient malignant pleural effusion (MPE) samples.
organoid-2021-1-e13f1.jpg
Fig. 2.
Photos of the resulting Percoll-gradient layers after centrifugation. The top and middle layers containing CD45-positive and CD45-negative cell fractions were obtained from (A) 44% and 67% Percoll solutions and (B) 40% and 75% Percoll solutions.
organoid-2021-1-e13f2.jpg
Fig. 3.
Flow cytometry analysis of CD45-positive (CD45pos) and CD45-negative (CD45neg) cells. (A) The cell mixture from malignant pleural effusion samples were distributed as CD45pos and CD45neg cells. (B) Cell population in the top (upper panel) or middle layer (lower panel) of a 44% and 67% Percoll gradient. (C) Cell population in the top (upper panel) or middle layer (lower panel) of a 40% and 75% Percoll gradient. SSC-A, side scatter-A; APC, allophycocyanin; FITC, fluorescein isothiocyanate.
organoid-2021-1-e13f3.jpg
Fig. 4.
Phase contrast images of 3-dimensional (3D) cell culture in Matrigel after Percoll-gradient centrifugation. (A) 3D cell culture of the cells (mixture of CD45pos/neg cells) obtained from the top layer with a 44% and 67% Percoll gradient. (B) 3D cell culture of the cells (mostly CD45neg cells) obtained from the top layer of a 40% and 75% Percoll gradient. (A, B) Scale bars, 50 m. CD45pos, CD45-positive; CD45neg, CD45-negative.
organoid-2021-1-e13f4.jpg

REFERENCES

1. Stock K, Estrada MF, Vidic S, Gjerde K, Rudisch A, Santo VE, et al. Capturing tumor complexity in vitro: comparative analysis of 2D and 3D tumor models for drug discovery. Sci Rep 2016;6:28951.
crossref pmid pmc pdf
2. Kopetz S, Lemos R, Powis G. The promise of patient-derived xenografts: the best laid plans of mice and men. Clin Cancer Res 2012;18:5160-2.
crossref pmid pmc pdf
3. Ricci F, Bizzaro F, Cesca M, Guffanti F, Ganzinelli M, Decio A, et al. Patient-derived ovarian tumor xenografts recapitulate human clinicopathology and genetic alterations. Cancer Res 2014;74:6980-90.
crossref pmid
4. Tentler JJ, Tan AC, Weekes CD, Jimeno A, Leong S, Pitts TM, et al. Patient-derived tumour xenografts as models for oncology drug development. Nat Rev Clin Oncol 2012;9:338-50.
crossref pmid pmc pdf
5. Fujii E, Kato A, Chen YJ, Matsubara K, Ohnishi Y, Suzuki M. The status of donor cancer tissues affects the fate of patient-derived colorectal cancer xenografts in NOG mice. Exp Anim 2015;64:181-90.
crossref pmid pmc
6. Ilie M, Nunes M, Blot L, Hofman V, Long-Mira E, Butori C, et al. Setting up a wide panel of patient-derived tumor xenografts of non-small cell lung cancer by improving the preanalytical steps. Cancer Med 2015;4:201-11.
crossref pmid pmc pdf
7. Stein AP, Saha S, Liu CZ, Hartig GK, Lambert PF, Kimple RJ. Influence of handling conditions on the establishment and propagation of head and neck cancer patient derived xenografts. PLoS One 2014;9:e100995.
crossref pmid pmc
8. Bell CC, Dankers A, Lauschke VM, Sison-Young R, Jenkins R, Rowe C, et al. Comparison of hepatic 2D sandwich cultures and 3D spheroids for long-term toxicity applications: a multicenter study. Toxicol Sci 2018;162:655-66.
crossref pmid pmc
9. Langhans SA. Three-dimensional in vitro cell culture models in drug discovery and drug repositioning. Front Pharmacol 2018;9:6.
crossref pmid pmc
10. Atat OE, Farzaneh Z, Pourhamzeh M, Taki F, Abi-Habib R, Vosough M, et al. 3D modeling in cancer studies. Hum Cell 2022;35:23-36.
crossref pmid pdf
11. Dhupar R, Okusanya OT, Eisenberg SH, Monaco SE, Ruffin AT, Liu D, et al. Characteristics of malignant pleural effusion resident CD8+ T cells from a heterogeneous collection of tumors. Int J Mol Sci 2020;21:6178.
crossref pmid pmc
12. Psallidas I, Kalomenidis I, Porcel JM, Robinson BW, Stathopoulos GT. Malignant pleural effusion: from bench to bedside. Eur Respir Rev 2016;25:189-98.
crossref pmid pmc
13. Mongardon N, Pinton-Gonnet C, Szekely B, Michel-Cherqui M, Dreyfus JF, Fischler M. Assessment of chronic pain after thoracotomy: a 1-year prevalence study. Clin J Pain 2011;27:677-81.
crossref pmid
14. Roberts ME, Neville E, Berrisford RG, Antunes G, Ali NJ; BTS Pleural Disease Guideline Group. Management of a malignant pleural effusion: British Thoracic Society Pleural Disease Guideline 2010. Thorax 2010;65 Suppl 2:ii32-40.
crossref pmid
15. Dixit R, Agarwal KC, Gokhroo A, Patil CB, Meena M, Shah NS, et al. Diagnosis and management options in malignant pleural effusions. Lung India 2017;34:160-6.
crossref pmid pmc
16. Massani M, Stecca T, Fabris L, Caratozzolo E, Ruffolo C, Furlanetto A, et al. Isolation and characterization of biliary epithelial and stromal cells from resected human cholangiocarcinoma: a novel in vitro model to study tumor-stroma interactions. Oncol Rep 2013;30:1143-8.
crossref pmid
17. Rapp M, Anz D, Schnurr M. Isolation of intratumoral leukocytes of TLR-stimulated tumor-bearing mice. Methods Mol Biol 2014;1169:175-9.
crossref pmid
18. Santiago L, Castro M, Pardo J, Arias M. Mouse model of colitis-associated colorectal cancer (CAC): isolation and characterization of mucosal-associated lymphoid cells. Methods Mol Biol 2019;1884:189-202.
crossref pmid
19. Kim M, Mun H, Sung CO, Cho EJ, Jeon HJ, Chun SM, et al. Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat Commun 2019;10:3991.
crossref pmid pmc pdf
20. Li Z, Qian Y, Li W, Liu L, Yu L, Liu X, et al. Human lung adenocarcinoma-derived organoid models for drug screening. iScience 2020;23:101411.
crossref pmid pmc
21. Shi R, Radulovich N, Ng C, Liu N, Notsuda H, Cabanero M, et al. Organoid cultures as preclinical models of non-small cell lung cancer. Clin Cancer Res 2020;26:1162-74.
crossref pmid pdf
22. Boj SF, Hwang CI, Baker LA, Chio II, Engle DD, Corbo V, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 2015;160:324-38.
crossref pmid pmc
23. Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, et al. Organoid modeling of the tumor immune microenvironment. Cell 2018;175:1972-88.
crossref pmid pmc


ABOUT
BROWSE ARTICLES
EDITORIAL POLICY
FOR CONTRIBUTORS
Editorial Office
Room 319, Hall 1 of Chonbuk National University Dental College, 20, Geonji-ro, Deokjin-gu, Jeonju 54907, Korea
Tel: +82-63-270-4024    E-mail: editor@j-organoid.org                

Copyright © 2024 by The Organoid Society.

Developed in M2PI

Close layer
prev next