1
|
Akli A, Takam Kamga P, Julie C, Capron C, Costantini A, Dumenil C, Dumoulin J, Giraud V, Parent F, Seferian A, Guettier C, Glorion M, Longchampt E, Emile JF, Giroux-Leprieur É. Role of the HGF/c-MET pathway in resistance to immune checkpoint inhibitors in advanced non-small cell lung cancer. Cancer Immunol Immunother 2025; 74:58. [PMID: 39751636 PMCID: PMC11698708 DOI: 10.1007/s00262-024-03882-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/03/2024] [Indexed: 01/04/2025]
Abstract
Most of advanced non-small cell lung cancer (NSCLC) patients will experience tumor progression with immunotherapy (IO). Preliminary data suggested an association between high plasma HGF levels and poor response to IO in advanced NSCLC. Our study aimed to evaluate further the role of the HGF/MET pathway in resistance to IO in advanced NSCLC. We included retrospectively 82 consecutive NSCLC patients from two academic hospitals. Among them, 49 patients received ICIs alone or in combination with chemotherapy (CT), while 33 patients received chemotherapy alone as the control group. We analyzed plasma HGF levels by ELISA and expression of PD-L1, MET/phospho-MET, and CD8+ T-Cell infiltration on lung tumor tissue by immunohistochemistry. We investigated the contribution of HGF/MET to IO response by culturing peripheral blood mononuclear cells (PBMC) with or without pembrolizumab, with recombinant HGF, or cocultured with NSCLC patients-derived explants. Additionally, c-MET inhibitors were used to evaluate the contribution of MET activation in NSCLC-mediated immunosuppression. High HGF levels were associated with high progression rate with IO (p = 0.0092), but not with CT. ELISA analysis of supernatants collected from cultured NSCLC cells showed that HGF was produced by tumor cells. Furthermore, when activated PBMCs were cultured in the presence of recombinant HGF or on NSCLC monolayer, the proliferation of CD3+CD8+ lymphocytes was inhibited, even in the presence of pembrolizumab. The addition of HGF/MET inhibitors restored lymphocyte activation and induced IFNγ production. In conclusion, inhibiting the HGF/MET signaling pathway could be a promising approach to enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Assya Akli
- Université Paris-Saclay, UVSQ, EA 4340 BECCOH, Boulogne-Billancourt, France
| | - Paul Takam Kamga
- Université Paris-Saclay, UVSQ, EA 4340 BECCOH, Boulogne-Billancourt, France
| | - Catherine Julie
- Université Paris-Saclay, UVSQ, EA 4340 BECCOH, Boulogne-Billancourt, France
- Department of Pathology, APHP-Ambroise Paré Hospital, Boulogne-Billancourt, France
| | - Claude Capron
- Université Paris-Saclay, UVSQ, EA 4340 BECCOH, Boulogne-Billancourt, France
- Department of Hematology and Immunology, APHP-Ambroise Paré Hospital, Boulogne-Billancourt, France
| | - Adrien Costantini
- Department of Respiratory Diseases and Thoracic Oncology, APHP - Hopital Ambroise Pare, 9 Avenue Charles de Gaulle, 92100, Boulogne-Billancourt, France
| | - Coraline Dumenil
- Department of Respiratory Diseases and Thoracic Oncology, APHP - Hopital Ambroise Pare, 9 Avenue Charles de Gaulle, 92100, Boulogne-Billancourt, France
| | - Jennifer Dumoulin
- Department of Respiratory Diseases and Thoracic Oncology, APHP - Hopital Ambroise Pare, 9 Avenue Charles de Gaulle, 92100, Boulogne-Billancourt, France
| | - Violaine Giraud
- Department of Respiratory Diseases and Thoracic Oncology, APHP - Hopital Ambroise Pare, 9 Avenue Charles de Gaulle, 92100, Boulogne-Billancourt, France
| | - Florence Parent
- Department of Pathology, APHP-Kremlin Bicetre Hospital, Kremlin Bicetre, France
| | - Andrei Seferian
- Department of Pathology, APHP-Kremlin Bicetre Hospital, Kremlin Bicetre, France
| | - Catherine Guettier
- Department of Pathology, APHP-Kremlin Bicetre Hospital, Kremlin Bicetre, France
| | - Mathieu Glorion
- Department of Thoracic Surgery, Foch Hospital, Suresnes, France
| | | | - Jean-François Emile
- Université Paris-Saclay, UVSQ, EA 4340 BECCOH, Boulogne-Billancourt, France
- Department of Pathology, APHP-Ambroise Paré Hospital, Boulogne-Billancourt, France
| | - Étienne Giroux-Leprieur
- Université Paris-Saclay, UVSQ, EA 4340 BECCOH, Boulogne-Billancourt, France.
- Department of Respiratory Diseases and Thoracic Oncology, APHP - Hopital Ambroise Pare, 9 Avenue Charles de Gaulle, 92100, Boulogne-Billancourt, France.
| |
Collapse
|
2
|
Korbecki J, Bosiacki M, Szatkowska I, Kupnicka P, Chlubek D, Baranowska-Bosiacka I. The Clinical Significance and Involvement in Molecular Cancer Processes of Chemokine CXCL1 in Selected Tumors. Int J Mol Sci 2024; 25:4365. [PMID: 38673949 PMCID: PMC11050300 DOI: 10.3390/ijms25084365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Chemokines play a key role in cancer processes, with CXCL1 being a well-studied example. Due to the lack of a complete summary of CXCL1's role in cancer in the literature, in this study, we examine the significance of CXCL1 in various cancers such as bladder, glioblastoma, hemangioendothelioma, leukemias, Kaposi's sarcoma, lung, osteosarcoma, renal, and skin cancers (malignant melanoma, basal cell carcinoma, and squamous cell carcinoma), along with thyroid cancer. We focus on understanding how CXCL1 is involved in the cancer processes of these specific types of tumors. We look at how CXCL1 affects cancer cells, including their proliferation, migration, EMT, and metastasis. We also explore how CXCL1 influences other cells connected to tumors, like promoting angiogenesis, recruiting neutrophils, and affecting immune cell functions. Additionally, we discuss the clinical aspects by exploring how CXCL1 levels relate to cancer staging, lymph node metastasis, patient outcomes, chemoresistance, and radioresistance.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28, 65-046 Zielona Góra, Poland
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| | - Iwona Szatkowska
- Department of Ruminants Science, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology, Klemensa Janickiego 29 St., 71-270 Szczecin, Poland;
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| |
Collapse
|
3
|
Bersano J, Lashuk K, Edinger A, Schueler J. A Subset of Colon Cancer Cell Lines Displays a Cytokine Profile Linked to Angiogenesis, EMT and Invasion Which Is Modulated by the Culture Conditions In Vitro. Cells 2023; 12:2539. [PMID: 37947617 PMCID: PMC10648033 DOI: 10.3390/cells12212539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Colorectal cancer (CRC) is one of the deadliest cancers worldwide. The dysregulation of secretory pathways is a crucial driver of CRC progression, since it modulates cell proliferation, angiogenesis and survival. This study explores the changes in the CRC cytokine profile depending on the culture conditions and the presence of fibroblasts and macrophages as cellular components of the tumor microenvironment in 2D and in 3D formed spheroids. Upon analysis of 45 different cytokines, chemokines and growth factors, 20 CRC cell lines were categorized into high and low secretors. In the high secretor group cytokines related to angiogenesis, EMT and invasion were significantly upregulated. LIF and HFG were identified as the best discriminator between both groups. Independent of this grouping, the addition of normal as well as cancer-associated fibroblasts had a similar impact on the cytokine profile by increasing the total amount of secreted cytokines in most of the investigated cell lines. In contrast, the differentiation and polarization of macrophages was modulated differently by normal vs. cancer-associated fibroblasts. In summary, we identified two groups of CRC cell lines that differ in their cytokine profile. The dependance of this profile was analyzed in detail-not only from the tumor cell line but as well from the culture condition in vitro. Key cytokines that discriminate the two groups were identified and their importance as promising biomarker candidates for CRC discussed.
Collapse
Affiliation(s)
| | | | | | - Julia Schueler
- Charles River Discovery Research Services Germany GmbH, Am Flughafen 12–14, 79108 Freiburg, Germany; (J.B.); (K.L.); (A.E.)
| |
Collapse
|
4
|
Gurudatt NG, Gwak H, Hyun KA, Jeong SE, Lee K, Park S, Chung MJ, Kim SE, Jo JH, Jung HI. Electrochemical detection and analysis of tumor-derived extracellular vesicles to evaluate malignancy of pancreatic cystic neoplasm using integrated microfluidic device. Biosens Bioelectron 2023; 226:115124. [PMID: 36758487 DOI: 10.1016/j.bios.2023.115124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/03/2023] [Indexed: 02/07/2023]
Abstract
Tumor-derived extracellular vesicles (tdEVs) are one of the most promising biomarkers for liquid biopsy-based cancer diagnostics, owing to the expression of specific membrane proteins of their cellular origin. The investigation of epithelial-to-mesenchymal transition (EMT) in cancer using tdEVs is an alternative way of evaluating the risk of malignancy transformation. An ultra-sensitive selection and detection methodology is an essential step in developing a tdEVs-based cancer diagnostic device. In this study, we developed an indium-tin-oxide (ITO) sensor integrated microfluidic device consisting of two main parts: 1) a multi-orifice flow-fractionation (MOFF) channel for extraction of pure EVs by removing blood cellular debris, and 2) an ITO sensor coupled with a geometrically activated surface interaction (GASI) channel for enrichment and quantification of tdEV. The microfluidic channel and the ITO sensors are assembled with a 3D printed magnetic housing to prevent sample leakage and to easily attach/detach the sensors to/from the microfluidic channel. The tdEVs were successfully captured on the specific antibody modified ITO surfaces in the integrated microfluidic channel. The integrated sensors showed an excellent linear response between 103 and 109 tdEVs/mL. Simultaneous evaluation of the epithelial and mesenchymal markers on the tdEV surfaces successfully revealed the EMT index of the corresponding pancreatic cancer cells. Our ITO sensor integrated microfluidic device showed excellent detection in the clinically relevant tdEVs-concentration range for patients with pancreatic cystic neoplasms. Hence, this system is expected to open a new avenue for liquid biopsy-based cancer prognostics and diagnostics.
Collapse
Affiliation(s)
- N G Gurudatt
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hogyeong Gwak
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kyung-A Hyun
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Se-Eun Jeong
- Korea Electronics Technology Institute (KETI), 25, Saenari-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13509, Republic of Korea
| | - Kyungyeon Lee
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sunyoung Park
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seong-Eun Kim
- Korea Electronics Technology Institute (KETI), 25, Saenari-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13509, Republic of Korea.
| | - Jung Hyun Jo
- Division of Gastroenterology, Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Hyo-Il Jung
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; The DABOM Inc., Seoul, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
5
|
Xue B, Schüler J, Harrod CM, Lashuk K, Bomya Z, Hribar KC. A Novel Hydrogel-Based 3D In Vitro Tumor Panel of 30 PDX Models Incorporates Tumor, Stromal and Immune Cell Compartments of the TME for the Screening of Oncology and Immuno-Therapies. Cells 2023; 12:1145. [PMID: 37190054 PMCID: PMC10137152 DOI: 10.3390/cells12081145] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Human-relevant systems that mimic the 3D tumor microenvironment (TME), particularly the complex mechanisms of immuno-modulation in the tumor stroma, in a reproducible and scalable format are of high interest for the drug discovery industry. Here, we describe a novel 3D in vitro tumor panel comprising 30 distinct PDX models covering a range of histotypes and molecular subtypes and cocultured with fibroblasts and PBMCs in planar (flat) extracellular matrix hydrogels to reflect the three compartments of the TME-tumor, stroma, and immune cells. The panel was constructed in a 96-well plate format and assayed tumor size, tumor killing, and T-cell infiltration using high-content image analysis after 4 days of treatment. We screened the panel first against the chemotherapy drug Cisplatin to demonstrate feasibility and robustness, and subsequently assayed immuno-oncology agents Solitomab (CD3/EpCAM bispecific T-cell engager) and the immune checkpoint inhibitors (ICIs) Atezolizumab (anti-PDL1), Nivolumab (anti-PD1) and Ipilimumab (anti-CTLA4). Solitomab displayed a strong response across many PDX models in terms of tumor reduction and killing, allowing for its subsequent use as a positive control for ICIs. Interestingly, Atezolizumab and Nivolumab demonstrated a mild response compared to Ipilimumab in a subset of models from the panel. We later determined that PBMC spatial proximity in the assay setup was important for the PD1 inhibitor, hypothesizing that both duration and concentration of antigen exposure may be critical. The described 30-model panel represents a significant advancement toward screening in vitro models of the tumor microenvironment that include tumor, fibroblast, and immune cell populations in an extracellular matrix hydrogel, with robust and standardized high content image analysis in a planar hydrogel. The platform is aimed at rapidly screening various combinations and novel agents and forming a critical conduit to the clinic, thus accelerating drug discovery for the next generation of therapeutics.
Collapse
Affiliation(s)
- Bin Xue
- Cypre, Inc., South San Francisco, CA 94080, USA
| | - Julia Schüler
- Charles River Discovery Research Services Germany GmbH, 79108 Freiburg, Germany
| | | | - Kanstantsin Lashuk
- Charles River Discovery Research Services Germany GmbH, 79108 Freiburg, Germany
| | - Zoji Bomya
- Cypre, Inc., South San Francisco, CA 94080, USA
| | | |
Collapse
|
6
|
Peterfi L, Yusenko MV, Kovacs G, Beothe T. FAPα and αSMA mark different subsets of fibroblasts in normal kidney and conventional renal cell carcinoma. Neoplasia 2022; 35:100854. [PMID: 36516488 PMCID: PMC9755362 DOI: 10.1016/j.neo.2022.100854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/08/2022] [Indexed: 12/14/2022] Open
Abstract
Several studies suggested a correlation between cancer associated fibroblasts (CAF) and cancer progression, but data on conventional renal cell carcinoma (cRCC) is still lacking. We aimed to analyse the impact of αSMA positive myo-CAF and FAPα expressing i-CAF on postoperative relapse of cRCC. We applied immunohistochemistry on tissue-multiarray (TMA) containing 736 consecutively operated cRCC without metastasis at the time of diagnosis. We analysed the correlation between the amount and pattern of αSMA and FAPα expressing CAFs and tumour cells and postoperative tumour relapse. Stromal fibroblasts of each cRCC displayed αSMA immunreaction but only 142 of the 736 tumours showed positive FAPα staining. There was no correlation between the amount of αSMA and or FAPα positive CAFs and tumour progression. However, tumours with large tourtous vessels with strong αSMA positive immunreaction have more then two times higher risk of postoperative tumour relapse (RR=2.198, p = 0.005). Patients with cRCC (57) showing cytoplasmic αSMA staining of tumour cells had a nearly two times higher risk for postoperative progression (RR=1.776, p = 0.014). There is no significant correlation between the density of αSMA or FAPα positive CAFs and postoperative relapse of cRCCs, therefore CAFs in cRCC are not suitable targets for therapy. Further limitation of anti-CAF therapy of cRCC that stromal cells of normal kidney are positive with αSMA antibody.
Collapse
Affiliation(s)
- Lehel Peterfi
- Department of Urology, Medical School, University of Pecs, Hungary
| | - Maria V. Yusenko
- Institute of Biochemistry, University of Muenster, Muenster, Germany
| | - Gyula Kovacs
- Department of Urology, Medical School, University of Pecs, Hungary,Medical Faculty, Ruprecht-Karls-University, Heidelberg, Germany,Correspondending author at: Department of Urology, Munkacsy M utca 2, 7621 Pecs, Hungary.
| | - Tamas Beothe
- Department of Urology, Peterfy Sandor Hospital, Budapest, Hungary
| |
Collapse
|
7
|
Habibi A, Bakhshi N, Moradi shoili Z, Amirmozafari N. Iron Oxide Nanoparticles Conjugated to Thiosemicarbazone Reduce the Survival of Cancer Cells by Increasing the Gene Expression of MicroRNA let-7c in Lung Cancer A549 Cells. ARCHIVES OF IRANIAN MEDICINE 2022; 25:807-816. [PMID: 37543908 PMCID: PMC10685841 DOI: 10.34172/aim.2022.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/07/2021] [Indexed: 08/08/2023]
Abstract
BACKGROUND Cancer cells have a higher demand for iron to grow and proliferate. A new complex of iron nanoparticles and thiosemicarbazones was synthesized. Confirmation tests included UV-visible, scanning electron microscopy (SEM), energy dispersive X-ray analysis (EDX), Fourier transform infrared (FTIR), X-ray diffraction (XRD) and zeta potential. METHODS MTT assay, flow cytometry and qRT-PCR were used to investigate anti-proliferative effect, amount of apoptosis and the effect of Fe3 O4 @Glu/BTSC on changes in gene expression of microRNA let-7c (let-7c), respectively. The specifications of Fe3 O4 @ Glu/BTSC were confirmed at 5 nm. RESULTS Fe3O4@Glu/BTSC was more effective than BTSC and Fe3 O4 on A549 cells (IC50=166.77 µg/mL) but its effect on healthy cells was smaller (CC50=189.15 µg/mL). The drug selectivity index (SI) was calculated to be 1.13. The initial apoptosis rate was 46.33% for Fe3 O4 @Glu/BTSC, 28.27% for BTSC and 26.02% for Fe3 O4 . BTSC and BTSC@Fe3 O4 inhibited the cell cycle progression in the Sub-G1 and S phases. let-7c expression was 6.9 times higher in treated cells compared to the control group. The expression rate was 2.2 with BTSC compared to the control group and 1.6 times for Fe3 O4. CONCLUSION Fe3 O4 @Glu/BTSC has proper anti-proliferative effects against lung cancer cells by increasing the expression of let-7c and inhibiting the cell cycle with the apoptosis activation pathway.
Collapse
Affiliation(s)
- Alireza Habibi
- Departman of Basic Sciences, Faculty of Science, Imam Hossein University, Tehran, Iran
| | - Nesa Bakhshi
- Departman of Molecular Cell Biology, Faculty of Science, Islamic Azad University of Lahijan, Lahijan, Iran
| | | | - Nour Amirmozafari
- Departman of Microbiology, School of Medicine, University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Schueler J, Borenstein J, Buti L, Dong M, Masmoudi F, Hribar K, Anderson E, Sommergruber W. How to build a tumor: An industry perspective. Drug Discov Today 2022; 27:103329. [PMID: 35908685 PMCID: PMC9585375 DOI: 10.1016/j.drudis.2022.07.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/23/2022] [Accepted: 07/25/2022] [Indexed: 12/15/2022]
Abstract
During the past 15 years, a plethora of innovative 3D in vitro systems has been developed. They offer the possibility of identifying crucial cellular and molecular contributors to the disease by permitting manipulation of each in isolation. However, improvements are needed particularly with respect to the predictivity and validity of those models. The major challenge now is to identify which assay and readout combination(s) best suits the current scientific question(s). A deep understanding of the different platforms along with their pros and cons is a prerequisite to make this decision. This review aims to give an overview of the most prominent systems with a focus on applications, translational relevance and adoption drivers from an industry perspective.
Collapse
Affiliation(s)
- Julia Schueler
- Charles River Discovery Research Services Germany GmbH, Freiburg, Germany,Corresponding author.
| | | | | | - Meng Dong
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tuebingen, Stuttgart, Germany
| | | | | | | | | |
Collapse
|
9
|
Oswald E, Bug D, Grote A, Lashuk K, Bouteldja N, Lenhard D, Löhr A, Behnke A, Knauff V, Edinger A, Klingner K, Gaedicke S, Niedermann G, Merhof D, Feuerhake F, Schueler J. Immune cell infiltration pattern in non-small cell lung cancer PDX models is a model immanent feature and correlates with a distinct molecular and phenotypic make-up. J Immunother Cancer 2022; 10:jitc-2021-004412. [PMID: 35483746 PMCID: PMC9052060 DOI: 10.1136/jitc-2021-004412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND The field of cancer immunology is rapidly moving towards innovative therapeutic strategies, resulting in the need for robust and predictive preclinical platforms reflecting the immunological response to cancer. Well characterized preclinical models are essential for the development of predictive biomarkers in the oncology as well as the immune-oncology space. In the current study, gold standard preclinical models are being refined and combined with novel image analysis tools to meet those requirements. METHODS A panel of 14 non-small cell lung cancer patient-derived xenograft models (NSCLC PDX) was propagated in humanized NOD/Shi-scid/IL-2Rnull mice. The models were comprehensively characterized for relevant phenotypic and molecular features, including flow cytometry, immunohistochemistry, histology, whole exome sequencing and cytokine secretion. RESULTS Models reflecting hot (>5% tumor-infiltrating lymphocytes/TILs) as opposed to cold tumors (<5% TILs) significantly differed regarding their cytokine profiles, molecular genetic aberrations, stroma content, and programmed cell death ligand-1 status. Treatment experiments including anti cytotoxic T-lymphocyte-associated protein 4, anti-programmed cell death 1 or the combination thereof across all 14 models in the single mouse trial format showed distinctive tumor growth response and spatial immune cell patterns as monitored by computerized analysis of digitized whole-slide images. Image analysis provided for the first time qualitative evaluation of the extent to which PDX models retain the histological features from their original human donors. CONCLUSIONS Deep phenotyping of PDX models in a humanized setting by combinations of computational pathology, immunohistochemistry, flow cytometry and proteomics enables the exhaustive analysis of innovative preclinical models and paves the way towards the development of translational biomarkers for immuno-oncology drugs.
Collapse
Affiliation(s)
- Eva Oswald
- Charles River Discovery Research Services Gemany GmbH, Charles River Laboratories Inc, Freiburg, Germany
| | - Daniel Bug
- Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
| | - Anne Grote
- Department of Pathology, Hannover Medical School, Hannover, Germany
| | - Kanstantsin Lashuk
- Charles River Discovery Research Services Gemany GmbH, Charles River Laboratories Inc, Freiburg, Germany
| | - Nassim Bouteldja
- Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
| | - Dorothee Lenhard
- Charles River Discovery Research Services Gemany GmbH, Charles River Laboratories Inc, Freiburg, Germany
| | - Anne Löhr
- Charles River Discovery Research Services Gemany GmbH, Charles River Laboratories Inc, Freiburg, Germany
| | - Anke Behnke
- Charles River Discovery Research Services Gemany GmbH, Charles River Laboratories Inc, Freiburg, Germany
| | - Volker Knauff
- Charles River Discovery Research Services Gemany GmbH, Charles River Laboratories Inc, Freiburg, Germany
| | - Anna Edinger
- Charles River Discovery Research Services Gemany GmbH, Charles River Laboratories Inc, Freiburg, Germany
| | - Kerstin Klingner
- Charles River Discovery Research Services Gemany GmbH, Charles River Laboratories Inc, Freiburg, Germany
| | - Simone Gaedicke
- Department of Radiation Oncology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Gabriele Niedermann
- Department of Radiation Oncology, Medical Center-University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Heidelberg, Germany
| | - Dorit Merhof
- Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
| | | | - Julia Schueler
- Charles River Discovery Research Services Gemany GmbH, Charles River Laboratories Inc, Freiburg, Germany
| |
Collapse
|
10
|
Valencia K, Sainz C, Bértolo C, de Biurrun G, Agorreta J, Azpilikueta A, Larrayoz MJ, Bosco G, Zandueta C, Redrado M, Redín E, Exposito F, Serrano D, Echepare M, Ajona D, Melero I, Pio R, Thomas R, Calvo A, Montuenga LM. Two alternative cell line models for the study of multiorganic metastasis and immunotherapy in Lung Squamous Cell Carcinoma. Dis Model Mech 2021; 15:273637. [PMID: 34870316 PMCID: PMC8822220 DOI: 10.1242/dmm.049137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/29/2021] [Indexed: 11/20/2022] Open
Abstract
There is a paucity of adequate mouse models and cell lines available to study lung squamous cell carcinoma (LUSC). We have generated and characterized two models of phenotypically different transplantable LUSC cell lines (UN-SCC679 and UN-SCC680) derived from an N-nitroso-tris-chloroethylurea (NTCU) chemically-induced mouse model in A/J mice. Furthermore, we genetically characterized and compared both LUSC cell lines by performing whole exome and RNA sequencing. These experiments revealed similar genetic and transcriptomic patterns that may correspond to the classical LUSC human subtype. In addition, we compared the immune landscape generated by both tumor cells lines in vivo and assessed their response to immune checkpoint inhibition. The differences between the two cell lines are a good model for the remarkable heterogeneity of human squamous cell carcinoma. Study of the metastatic potential of these models revealed that both cell lines represent the human LUSC organotropism to the brain, bones, liver and adrenal glands. In summary, we have generated a very valuable cell line tools for LUSC research that recapitulates the complexity of the human disease.
Collapse
Affiliation(s)
- Karmele Valencia
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Cristina Sainz
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Cristina Bértolo
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Gabriel de Biurrun
- Department of Environmental Biology School of Sciences, University of Navarra, Pamplona, Spain
| | - Jackeline Agorreta
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Health Sciences, Biochemistry Area, Public University of Navarra, Pamplona, Spain
| | - Arantza Azpilikueta
- Program of Immunology and Immunotherapy, CIMA-University of Navarra, Pamplona, Spain
| | - Marta J Larrayoz
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Graziella Bosco
- Department of Translational Genomics, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Carolina Zandueta
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Miriam Redrado
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Esther Redín
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Francisco Exposito
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Diego Serrano
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Mirari Echepare
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Daniel Ajona
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Ignacio Melero
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, CIMA-University of Navarra, Pamplona, Spain.,Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Ruben Pio
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Roman Thomas
- Department of Translational Genomics, Medical Faculty, University of Cologne, 50931 Cologne, Germany.,Department of Pathology, University Hospital Cologne, 50937 Cologne, Germany.,German Cancer Research Center, German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Alfonso Calvo
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Luis M Montuenga
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| |
Collapse
|
11
|
Gwak H, Park S, Kim J, Lee JD, Kim IS, Kim SI, Hyun KA, Jung HI. Microfluidic chip for rapid and selective isolation of tumor-derived extracellular vesicles for early diagnosis and metastatic risk evaluation of breast cancer. Biosens Bioelectron 2021; 192:113495. [PMID: 34273737 DOI: 10.1016/j.bios.2021.113495] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
The epithelial-to-mesenchymal transition (EMT) index in cancer is a complementary approach for estimating metastatic risk. Considering the demand for evaluating metastatic risk based on liquid biopsies, tumor-derived extracellular vesicles (EVs) can be exploited to generate the EMT index. For the generation of EVs-based EMT index, it is essential to selectively isolate each epithelial cell and mesenchymal cell-derived EVs. This study proposes a novel microfluidic chip for selectively separating two types of EVs in an efficient and timely manner. The microfluidic chip is fully integrated with a micromixer for the creation of efficient collision between EVs and specific antibody-coated microbeads (7 and 15 μm in diameter) and a hydrodynamic particle separator for the stratification of EVs bound microbeads according to the sizes of microbeads. Using this chip, over 90% of EVs expressing the epithelial marker (epithelial cell adhesion molecule, EpCAM) and the mesenchymal marker (CD49f) can be selectively isolated within 6.7 min per 100 μL of sample volume. The clinical relevance of EMT is investigated using plasma samples from 20 breast cancer patients and 10 age-matched controls. The EMT index produced from the microfluidic chip is in a good agreement with the conventional tissue-based EMT index and is significantly high in patients with aggressive breast cancer subtypes, compared with healthy controls. In addition, the patients with high scores on the EMT index (≥5) shows recurrence within 5 years after adjuvant treatment. Predicting EMT-index-based metastatic risk using our microfluidic chip can be beneficial for cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Hogyeong Gwak
- Department of Mechanical Engineering, Yonsei University, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sunyoung Park
- Department of Mechanical Engineering, Yonsei University, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Junmoo Kim
- Department of Mechanical Engineering, Yonsei University, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jeong Dong Lee
- Department of Surgery, College of Medicine, Yonsei University, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - In-Soo Kim
- EUDIPIA Inc., Cheongju-si, Chungcheongbuk-do, 28160, Republic of Korea
| | - Seung-Il Kim
- Department of Surgery, College of Medicine, Yonsei University, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Kyung-A Hyun
- Department of Mechanical Engineering, Yonsei University, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Hyo-Il Jung
- Department of Mechanical Engineering, Yonsei University, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
12
|
Stadler M, Pudelko K, Biermeier A, Walterskirchen N, Gaigneaux A, Weindorfer C, Harrer N, Klett H, Hengstschläger M, Schüler J, Sommergruber W, Oehler R, Bergmann M, Letellier E, Dolznig H. Stromal fibroblasts shape the myeloid phenotype in normal colon and colorectal cancer and induce CD163 and CCL2 expression in macrophages. Cancer Lett 2021; 520:184-200. [PMID: 34256095 DOI: 10.1016/j.canlet.2021.07.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/30/2022]
Abstract
Colorectal cancer (CRC) accounts for about 10% of cancer deaths worldwide. Colon carcinogenesis is critically influenced by the tumor microenvironment. Cancer associated fibroblasts (CAFs) and tumor associated macrophages (TAMs) represent the major components of the tumor microenvironment. TAMs promote tumor progression, angiogenesis and tissue remodeling. However, the impact of the molecular crosstalk of tumor cells (TCs) with CAFs and macrophages on monocyte recruitment and their phenotypic conversion is not known in detail so far. In a 3D human organotypic CRC model, we show that CAFs and normal colonic fibroblasts are critically involved in monocyte recruitment and for the establishment of a macrophage phenotype, characterized by high CD163 expression. This is in line with the steady recruitment and differentiation of monocytes to immunosuppressive macrophages in the normal colon. Cytokine profiling revealed that CAFs produce M-CSF, and IL6, IL8, HGF and CCL2 secretion was specifically induced by CAFs in co-cultures with macrophages. Moreover, macrophage/CAF/TCs co-cultures increased TC invasion. We demonstrate that CAFs and macrophages are the major producers of CCL2 and, upon co-culture, increase their CCL2 production twofold and 40-fold, respectively. CAFs and macrophages expressing high CCL2 were also found in vivo in CRC, strongly supporting our findings. CCL2, CCR2, CSF1R and CD163 expression in macrophages was dependent on active MCSFR signaling as shown by M-CSFR inhibition. These results indicate that colon fibroblasts and not TCs are the major cellular component, recruiting and dictating the fate of infiltrated monocytes towards a specific macrophage population, characterized by high CD163 expression and CCL2 production.
Collapse
Affiliation(s)
- Mira Stadler
- Institute of Medical Genetics, Medical University of Vienna, Währinger Straße 10, A-1090 Vienna, Austria
| | - Karoline Pudelko
- Institute of Medical Genetics, Medical University of Vienna, Währinger Straße 10, A-1090 Vienna, Austria
| | - Alexander Biermeier
- Institute of Medical Genetics, Medical University of Vienna, Währinger Straße 10, A-1090 Vienna, Austria
| | - Natalie Walterskirchen
- Department of Surgery, Medical University of Vienna, Währinger Gürtel 20, A-1090 Vienna, Austria
| | - Anthoula Gaigneaux
- Department of Life Sciences and Medicine, University of Luxembourg, 6 Avenue du Swing, L-4367, Campus Belval, Luxembourg
| | - Claudia Weindorfer
- Institute of Medical Genetics, Medical University of Vienna, Währinger Straße 10, A-1090 Vienna, Austria
| | - Nathalie Harrer
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria, Dr. Boehringer-Gasse 5-11, A-1130 Vienna, Austria
| | - Hagen Klett
- Charles River Research Services Germany GmbH, Am Flughafen 12-14, 79108 Freiburg, Germany
| | - Markus Hengstschläger
- Institute of Medical Genetics, Medical University of Vienna, Währinger Straße 10, A-1090 Vienna, Austria
| | - Julia Schüler
- Charles River Research Services Germany GmbH, Am Flughafen 12-14, 79108 Freiburg, Germany
| | - Wolfgang Sommergruber
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria, Dr. Boehringer-Gasse 5-11, A-1130 Vienna, Austria
| | - Rudolf Oehler
- Department of Surgery, Medical University of Vienna, Währinger Gürtel 20, A-1090 Vienna, Austria
| | - Michael Bergmann
- Department of Surgery, Medical University of Vienna, Währinger Gürtel 20, A-1090 Vienna, Austria
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, University of Luxembourg, 6 Avenue du Swing, L-4367, Campus Belval, Luxembourg
| | - Helmut Dolznig
- Institute of Medical Genetics, Medical University of Vienna, Währinger Straße 10, A-1090 Vienna, Austria.
| |
Collapse
|
13
|
Lin C, Zhu X, Jin Q, Sui A, Li J, Shen L. Effects of Holothurian Glycosaminoglycan on the Sensitivity of Lung Cancer to Chemotherapy. Integr Cancer Ther 2021; 19:1534735420911430. [PMID: 32202167 PMCID: PMC7092648 DOI: 10.1177/1534735420911430] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Sea cucumber is a kind of food. Holothurian glycosaminoglycan (hGAG) is extracted from the body wall of the sea cucumber. Administration of hGAG and cisplatin (DDP) together to treat lung cancer was investigated. Lung adenocarcinoma A549 cells were cultured and divided into 4 groups: control group, hGAG 100 µg/mL group, DDP 3 µg/mL group, and hGAG 100 µg/mL + DDP 3 µg/mL group. Cell inhibition and apoptosis was evaluated by CCK8 and Hoechst33258 staining. Cell cycle was tested by Annexin V-FITC/PI (propidium iodide) double-staining and flow cytometry. The expression of mRNA and protein of Bcl-2, Bax, caspase-3, and survivin were detected by reverse transcriptase-polymerase chain reaction and Western blot, respectively. The results showed that hGAG combined with DDP enhanced the inhibitory effect of DDP on A549 lung cells through apoptosis pathway. The mechanism of apoptosis may be related to the reduction of Bcl-2 and survivin, as well as the ascension of Bax and caspase-3. hGAG could promote A549 cell cycle arrest in G1 and G2 phase and improve the DDP chemotherapy effects on A549 cells.
Collapse
Affiliation(s)
- Cunzhi Lin
- Department of Pulmonary Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xinhong Zhu
- Internal Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Qing Jin
- Department of Intensive Care Unit, The 903rd Hospital of People's Liberation Army, Hangzhou, Zhejiang, China
| | - Aihua Sui
- Department of Respiratory & Critical Care Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jinfeng Li
- Department of Respiratory & Critical Care Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Liyan Shen
- Department of Respiratory & Critical Care Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
14
|
Jin Q, Lin C, Zhu X, Cao Y, Guo C, Wang L. 125I seeds irradiation inhibits tumor growth and induces apoptosis by Ki-67, P21, survivin, livin and caspase-9 expression in lung carcinoma xenografts. Radiat Oncol 2020; 15:238. [PMID: 33059701 PMCID: PMC7559445 DOI: 10.1186/s13014-020-01682-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 10/06/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Lung cancer is a fatal disease and a serious health problem worldwide. Patients are usually diagnosed at an advanced stage, and the effectiveness of chemotherapy for such patients is very limited. Iodine 125 seed (125I) irradiation can be used as an important adjuvant treatment for lung carcinoma. The purpose of this study was to examine the role of irradiation by 125I seeds in human lung cancer xenograft model and to determine the underlying mechanisms involved, with a focus on apoptosis. METHODS 40 mice with A549 lung adenocarcinoma xenografts were randomly divided into 4 groups: control group (n = 10), sham seed (0 mCi) implant group (n = 10), 125I seed (0.6 mCi) implant group (n = 10) and 125I seed (0.8 mCi) implant group (n = 10), respectively. The body weight and tumor volume, were recorded every 4 days until the end of the study. Apoptotic cells were checked by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and activities of caspase-3 and caspase-8 enzyme were tested. Expression of P21, survivin, livin, caspase-9 and proliferating cell nuclear antigen (Ki-67) was detected with immunohistochemical staining. RESULTS The results of TUNEL staining assays showed that 125I seed irradiation suppresses the growth of lung cancer xenografts in nude mice and induced apoptosis. The activity of caspase-3 and caspase-8 was significantly higher. The expression levels Ki67, survivin and livin were substantially downregulated, while P21 and caspase-9 protein expression were significantly increased following 125I seed irradiation. This study revealed that 125I seed irradiation could significantly change apoptosis-related protein in human lung cancer xenografts. CONCLUSIONS Overall, our study demonstrates that radiation exposure by 125I seeds could be a new treatment option for lung cancer.
Collapse
Affiliation(s)
- Qing Jin
- Department of Critical Care Medicine, The 903th Hospital of PLA Joint Logistics Support Force, Zhejiang Province, Hangzhou, 310013, China
| | - Cunzhi Lin
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Xinhong Zhu
- Department of Internal Medicine, Qingdao Municipal Hospital, Qingdao, 266071, Shandong Province, China
| | - Yiwei Cao
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Caihong Guo
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Lijun Wang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China.
| |
Collapse
|
15
|
Ancel J, Dewolf M, Deslée G, Nawrocky-Raby B, Dalstein V, Gilles C, Polette M. Clinical Impact of the Epithelial-Mesenchymal Transition in Lung Cancer as a Biomarker Assisting in Therapeutic Decisions. Cells Tissues Organs 2020; 211:91-109. [PMID: 32750701 DOI: 10.1159/000510103] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/11/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is one of the most common solid cancers and represents the leading cause of cancer death worldwide. Over the last decade, research on the epithelial-mesenchymal transition (EMT) in lung cancer has gained increasing attention. Here, we review clinical and histological features of non-small-cell lung cancer associated with EMT. We then aimed to establish potential clinical implications of EMT in current therapeutic options, including surgery, radiation, targeted therapy against oncogenic drivers, and immunotherapy.
Collapse
Affiliation(s)
- Julien Ancel
- Inserm, Université de Reims Champagne Ardenne, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France.,Service de Pneumologie, Hôpital Maison Blanche, CHU de Reims, Reims, France
| | - Maxime Dewolf
- Service de Pneumologie, Hôpital Maison Blanche, CHU de Reims, Reims, France
| | - Gaëtan Deslée
- Inserm, Université de Reims Champagne Ardenne, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France.,Service de Pneumologie, Hôpital Maison Blanche, CHU de Reims, Reims, France
| | - Béatrice Nawrocky-Raby
- Inserm, Université de Reims Champagne Ardenne, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France
| | - Véronique Dalstein
- Inserm, Université de Reims Champagne Ardenne, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France.,Laboratoire de Pathologie, Hôpital Maison Blanche, CHU de Reims, Reims, France
| | - Christine Gilles
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium,
| | - Myriam Polette
- Inserm, Université de Reims Champagne Ardenne, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France.,Laboratoire de Pathologie, Hôpital Maison Blanche, CHU de Reims, Reims, France
| |
Collapse
|
16
|
Chen T, Zhao Z, Chen B, Wang Y, Yang F, Wang C, Dong Q, Liu Y, Liang H, Zhao W, Qi L, Xu Y, Gu Y. An individualized transcriptional signature to predict the epithelial-mesenchymal transition based on relative expression ordering. Aging (Albany NY) 2020; 12:13172-13186. [PMID: 32639951 PMCID: PMC7377874 DOI: 10.18632/aging.103407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/25/2020] [Indexed: 12/14/2022]
Abstract
The epithelial-mesenchymal transition (EMT) process is involved in cancer cell metastasis and immune system activation. Hence, identification of gene expression signatures capable of predicting the EMT status of cancer cells is essential for development of therapeutic strategies. However, quantitative identification of EMT markers is limited by batch effects, the platform used, or normalization methods. We hypothesized that a set of EMT-related relative expression orderings are highly stable in epithelial samples yet are reversed in mesenchymal samples. To test this hypothesis, we analyzed transcriptome data for ovarian cancer cohorts from publicly available databases, to develop a qualitative 16-gene pair signature (16-GPS) that effectively distinguishes the mesenchymal from epithelial phenotype. Our method was superior to previous quantitative methods in terms of classification accuracy and applicability to individualized patients without requiring data normalization. Patients with mesenchymal-like ovarian cancer showed poorer overall survival compared to patients with epithelial-like ovarian cancer. Additionally, EMT score was positively correlated with expression of immune checkpoint genes and metastasis. We, therefore, established a robust EMT 16-GPS that is independent of detection platform, batch effects and individual variations, and which represents a qualitative signature for investigating the EMT and providing insights into immunotherapy for ovarian cancer patients.
Collapse
Affiliation(s)
- Tingting Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zhangxiang Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Bo Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yuquan Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Fan Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Chengyu Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Qi Dong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yaoyao Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Haihai Liang
- College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wenyuan Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lishuang Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yunyan Gu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Abugomaa A, Elbadawy M, Yamanaka M, Goto Y, Hayashi K, Mori T, Uchide T, Azakami D, Fukushima R, Yoshida T, Shibutani M, Yamashita R, Kobayashi M, Yamawaki H, Shinohara Y, Kaneda M, Usui T, Sasaki K. Establishment of 2.5D organoid culture model using 3D bladder cancer organoid culture. Sci Rep 2020; 10:9393. [PMID: 32523078 PMCID: PMC7287130 DOI: 10.1038/s41598-020-66229-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 05/18/2020] [Indexed: 02/07/2023] Open
Abstract
Three-dimensional (3D) organoid culture holds great promises in cancer precision medicine. However, Matrigel and stem cell-stimulating supplements are necessary for culturing 3D organoid cells. It costs a lot of money and consumes more time and effort compared with 2D cultured cells. Therefore, the establishment of cheaper and Matrigel-free organoid culture that can maintain the characteristics of a part of 3D organoids is demanded. In the previous study, we established a dog bladder cancer (BC) 3D organoid culture system by using their urine samples. Here, we successfully isolated cells named "2.5D organoid" from multiple strains of dog BC 3D organoids using 2.5 organoid media. The cell proliferation speed of 2.5D organoids was faster than parental 3D organoid cells. The expression pattern of stem cell markers was close to 3D organoids. Injection of 2.5D organoid cells into immunodeficient mice formed tumors and showed the histopathological characteristics of urothelial carcinoma similar to the injection of dog BC 3D organoids. The 2.5D organoids had a similar sensitivity profile for anti-cancer drug treatment to their parental 3D organoids. These data suggest that our established 2.5D organoid culture method might become a reasonable and useful tool instead of 3D organoids in dog BC research and therapy.
Collapse
Affiliation(s)
- Amira Abugomaa
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
- Faculty of Veterinary Medicine, Mansoura University, 35516, Mansoura, Egypt
| | - Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, 13736, Moshtohor, Toukh, Egypt
| | - Megumi Yamanaka
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Yuta Goto
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Kimika Hayashi
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Takashi Mori
- Laboratory of Veterinary Clinical Oncology, Faculty of Applied Biological Sciences, Gifu University, 1-1, Yanagido, Gifu, Gifu, 501-1193, Japan
- Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University (G-CHAIN), 1-1, Yanagido, Gifu, Gifu, 501-1193, Japan
| | - Tsuyoshi Uchide
- Department of Veterinary Surgery, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Daigo Azakami
- Department of Veterinary Clinical Oncology, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Ryuji Fukushima
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Risako Yamashita
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Mio Kobayashi
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, 35-1 Higashi 23 ban-cho, Towada, Aomori, 034-8628, Japan
| | - Yuta Shinohara
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
- Pet Health & Food Division, Iskara Industry CO., LTD, 1-14-2, Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan.
| | - Kazuaki Sasaki
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan
| |
Collapse
|
18
|
Wernitznig D, Kiakos K, Del Favero G, Harrer N, Machat H, Osswald A, Jakupec MA, Wernitznig A, Sommergruber W, Keppler BK. First-in-class ruthenium anticancer drug (KP1339/IT-139) induces an immunogenic cell death signature in colorectal spheroids in vitro. Metallomics 2019; 11:1044-1048. [DOI: 10.1039/c9mt00051h] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
ICD enhances antigenicity from dying cancer cells, which leads to antitumor immunity. We show for the first time that a ruthenium-complex induces the ICD signature in a 3D model.
Collapse
Affiliation(s)
- Debora Wernitznig
- Department of Inorganic Chemistry and Research Cluster ‘Translational Cancer Therapy Research’
- Faculty of Chemistry
- University of Vienna
- Vienna
- Austria
| | - Konstantinos Kiakos
- Department of Inorganic Chemistry and Research Cluster ‘Translational Cancer Therapy Research’
- Faculty of Chemistry
- University of Vienna
- Vienna
- Austria
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology
- Faculty of Chemistry
- University of Vienna
- Vienna
- Austria
| | | | | | | | - Michael A. Jakupec
- Department of Inorganic Chemistry and Research Cluster ‘Translational Cancer Therapy Research’
- Faculty of Chemistry
- University of Vienna
- Vienna
- Austria
| | | | | | - Bernhard K. Keppler
- Department of Inorganic Chemistry and Research Cluster ‘Translational Cancer Therapy Research’
- Faculty of Chemistry
- University of Vienna
- Vienna
- Austria
| |
Collapse
|
19
|
Osswald A, Hedrich V, Sommergruber W. 3D-3 Tumor Models in Drug Discovery for Analysis of Immune Cell Infiltration. Methods Mol Biol 2019; 1953:151-162. [PMID: 30912021 DOI: 10.1007/978-1-4939-9145-7_10] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The cross talk between tumor cells and other cells present in the tumor microenvironment such as stromal and immune cells highly influences the behavior and progression of disease. Understanding the underlying mechanisms of interaction is a prerequisite to develop new treatment strategies and to prevent or at least reduce therapy failure in the future. Specific reactivation of the patient's immune system is one of the major goals today. However, standard two-dimensional (2D) cell culture techniques lack the necessary complexity to address related questions. Novel three-dimensional (3D) in vitro models-embedded in a matrix or encapsulated in alginate-recapitulate the in vivo situation much better. Cross talk between different cell types can be studied starting from co-cultures. As cancer immune modulation is becoming a major research topic, 3D in vitro models represent an important tool to address immune regulatory/modulatory questions for T, NK, and other cells of the immune system. The 3D systems consisting of tumor cells, fibroblasts, and immune cells (3D-3) already proved as a reliable tool for us. For instance, we made use of those models to study the molecular mechanisms of the cross talk of non-small cell lung cancer (NSCLC) and fibroblasts, to unveil macrophage plasticity in the tumor microenvironment and to mirror drug responses in vivo. Generation of those 3D models and how to use them to study immune cell infiltration and activation will be described in the present book chapter.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Bioreactors
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Cells, Immobilized/drug effects
- Cells, Immobilized/immunology
- Cells, Immobilized/pathology
- Coculture Techniques/methods
- Drug Discovery/methods
- Drug Screening Assays, Antitumor/methods
- Fibroblasts/drug effects
- Fibroblasts/immunology
- Fibroblasts/pathology
- Humans
- Immunity, Cellular/drug effects
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/immunology
- Spheroids, Cellular/pathology
- Tumor Cells, Cultured
- Tumor Microenvironment/drug effects
Collapse
Affiliation(s)
| | - Viola Hedrich
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | |
Collapse
|
20
|
Bolt R, Foran B, Murdoch C, Lambert DW, Thomas S, Hunter KD. HPV-negative, but not HPV-positive, oropharyngeal carcinomas induce fibroblasts to support tumour invasion through micro-environmental release of HGF and IL-6. Carcinogenesis 2018; 39:170-179. [PMID: 29140428 DOI: 10.1093/carcin/bgx130] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 11/07/2017] [Indexed: 01/02/2023] Open
Abstract
Human papillomavirus (HPV) infection is causally related to a subset of oropharyngeal carcinomas (OPC) and is linked to a more favourable prognosis compared to HPV-negative OPC. The mechanisms underlying this effect on prognosis are not fully understood, but interactions with the tumour microenvironment may be pivotal. Here, we investigated the role of the tumour microenvironment in HPV-positive compared to HPV-negative cancer using 2D and 3D modelling of OPC interactions with stromal fibroblasts. HPV-negative, but not HPV-positive, OPC-derived cell lines induced a rapid fibroblast secretory response that supported 2D cancer cell migration and invasion in vitro. Array profiling of this HPV-negative induced fibroblast secretome identified hepatocyte growth factor (HGF) as the principal secreted factor that promoted cancer cell migration. The interaction between HPV-negative cell lines and fibroblasts in 2D was prevented using c-Met (HGF receptor) inhibitors, which further restricted both HPV-negative and positive cell invasion in 3D co-culture models. Furthermore, we discovered a synergistic relationship between HGF and IL-6 in the support of migration that relates JAK activation to HGF responsiveness in HPV-negative lines. In summary, our data show significant differences in the interactions between HPV-positive and HPV-negative OPC cells and stromal fibroblasts. In addition, we, provide in vitro evidence to support the clinical application of c-MET inhibitors in the control of early HPV-negative OPC.
Collapse
Affiliation(s)
- Robert Bolt
- School of Clinical Dentistry, University of Sheffield, Sheffield, South Yorkshire, UK
| | - Bernadette Foran
- Department of Oncology, Weston Park Hospital, Sheffield, South Yorkshire, UK
| | - Craig Murdoch
- School of Clinical Dentistry, University of Sheffield, Sheffield, South Yorkshire, UK
| | - Daniel W Lambert
- School of Clinical Dentistry, University of Sheffield, Sheffield, South Yorkshire, UK
| | - Sally Thomas
- Department of Biomedical Sciences, University of Sheffield, Sheffield, South Yorkshire, UK
| | - Keith D Hunter
- School of Clinical Dentistry, University of Sheffield, Sheffield, South Yorkshire, UK
| |
Collapse
|
21
|
Kanda P, Alarcon EI, Yeuchyk T, Parent S, de Kemp RA, Variola F, Courtman D, Stewart DJ, Davis DR. Deterministic Encapsulation of Human Cardiac Stem Cells in Variable Composition Nanoporous Gel Cocoons To Enhance Therapeutic Repair of Injured Myocardium. ACS NANO 2018; 12:4338-4350. [PMID: 29660269 DOI: 10.1021/acsnano.7b08881] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Although cocooning explant-derived cardiac stem cells (EDCs) in protective nanoporous gels (NPGs) prior to intramyocardial injection boosts long-term cell retention, the number of EDCs that finally engraft is trivial and unlikely to account for salutary effects on myocardial function and scar size. As such, we investigated the effect of varying the NPG content within capsules to alter the physical properties of cocoons without influencing cocoon dimensions. Increasing NPG concentration enhanced cell migration and viability while improving cell-mediated repair of injured myocardium. Given that the latter occurred with NPG content having no detectable effect on the long-term engraftment of transplanted cells, we found that changing the physical properties of cocoons prompted explant-derived cardiac stem cells to produce greater amounts of cytokines, nanovesicles, and microRNAs that boosted the generation of new blood vessels and new cardiomyocytes. Thus, by altering the physical properties of cocoons by varying NPG content, the paracrine signature of encapsulated cells can be enhanced to promote greater endogenous repair of injured myocardium.
Collapse
Affiliation(s)
- Pushpinder Kanda
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Canada K1Y4W7
| | - Emilio I Alarcon
- Division of Cardiac Surgery, Department of Surgery, University of Ottawa Heart Institute , University of Ottawa , Ottawa , Canada K1Y4W7
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Canada K1H8M5
| | - Tanya Yeuchyk
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Canada K1Y4W7
| | - Sandrine Parent
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Canada K1Y4W7
| | - Robert A de Kemp
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Canada K1Y4W7
| | - Fabio Variola
- Department of Mechanical Engineering , University of Ottawa , Ottawa , Canada K1N6N5
- Department of Cellular and Molecular Medicine , University of Ottawa , Ottawa , Canada K1H8M5
| | - David Courtman
- Regenerative Medicine Program , Ottawa Hospital Research Institute , Ottawa , Canada K1H8L6
| | - Duncan J Stewart
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Canada K1Y4W7
- Department of Cellular and Molecular Medicine , University of Ottawa , Ottawa , Canada K1H8M5
- Regenerative Medicine Program , Ottawa Hospital Research Institute , Ottawa , Canada K1H8L6
| | - Darryl R Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Canada K1Y4W7
- Department of Cellular and Molecular Medicine , University of Ottawa , Ottawa , Canada K1H8M5
| |
Collapse
|
22
|
Rebelo SP, Pinto C, Martins TR, Harrer N, Estrada MF, Loza-Alvarez P, Cabeçadas J, Alves PM, Gualda EJ, Sommergruber W, Brito C. 3D-3-culture: A tool to unveil macrophage plasticity in the tumour microenvironment. Biomaterials 2018; 163:185-197. [PMID: 29477032 DOI: 10.1016/j.biomaterials.2018.02.030] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/09/2018] [Accepted: 02/12/2018] [Indexed: 12/12/2022]
Abstract
The tumour microenvironment (TME) shapes disease progression and influences therapeutic response. Most aggressive solid tumours have high levels of myeloid cell infiltration, namely tumour associated macrophages (TAM). Recapitulation of the interaction between the different cellular players of the TME, along with the extracellular matrix (ECM), is critical for understanding the mechanisms underlying disease progression. This particularly holds true for prediction of therapeutic response(s) to standard therapies and interrogation of efficacy of TME-targeting agents. In this work, we explored a culture platform based on alginate microencapsulation and stirred culture systems to develop the 3D-3-culture, which entails the co-culture of tumour cell spheroids of non-small cell lung carcinoma (NSCLC), cancer associated fibroblasts (CAF) and monocytes. We demonstrate that the 3D-3-culture recreates an invasive and immunosuppressive TME, with accumulation of cytokines/chemokines (IL4, IL10, IL13, CCL22, CCL24, CXCL1), ECM elements (collagen type I, IV and fibronectin) and matrix metalloproteinases (MMP1/9), supporting cell migration and promoting cell-cell interactions within the alginate microcapsules. Importantly, we show that both the monocytic cell line THP-1 and peripheral blood-derived monocytes infiltrate the tumour tissue and transpolarize into an M2-like macrophage phenotype expressing CD68, CD163 and CD206, resembling the TAM phenotype in NSCLC. The 3D-3-culture was challenged with chemo- and immunotherapeutic agents and the response to therapy was assessed in each cellular component. Specifically, the macrophage phenotype was modulated upon treatment with the CSF1R inhibitor BLZ945, resulting in a decrease of the M2-like macrophages. In conclusion, the crosstalk between the ECM and tumour, stromal and immune cells in microencapsulated 3D-3-culture promotes the activation of monocytes into TAM, mimicking aggressive tumour stages. The 3D-3-culture constitutes a novel tool to study tumour-immune interaction and macrophage plasticity in response to external stimuli, such as chemotherapeutic and immunomodulatory drugs.
Collapse
Affiliation(s)
- Sofia P Rebelo
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Catarina Pinto
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Tatiana R Martins
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Nathalie Harrer
- Boehringer Ingelheim RCV GmbH & Co KG, Department of Lead Discovery, 1121, Vienna, Austria
| | - Marta F Estrada
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Pablo Loza-Alvarez
- ICFO - Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, 08860, Castelldefels, Barcelona, Spain
| | - José Cabeçadas
- Serviço de Anatomia Patológica, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Emilio J Gualda
- ICFO - Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, 08860, Castelldefels, Barcelona, Spain
| | - Wolfgang Sommergruber
- Boehringer Ingelheim RCV GmbH & Co KG, Department of Lead Discovery, 1121, Vienna, Austria
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal.
| |
Collapse
|
23
|
Chen SC, Chen FW, Hsu YL, Kuo PL. Systematic Analysis of Transcriptomic Profile of Renal Cell Carcinoma under Long-Term Hypoxia Using Next-Generation Sequencing and Bioinformatics. Int J Mol Sci 2017; 18:ijms18122657. [PMID: 29215599 PMCID: PMC5751259 DOI: 10.3390/ijms18122657] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/25/2017] [Accepted: 12/04/2017] [Indexed: 01/28/2023] Open
Abstract
Patients with clear cell renal cell carcinoma (ccRCC) are often diagnosed with both von Hippel-Lindau (VHL) mutations and the constitutive activation of hypoxia-inducible factor-dependent signaling. In this study, we investigated the effects of long-term hypoxia in 786-O, a VHL-defective renal cell carcinoma cell line, to identify potential genes and microRNAs associated with tumor malignancy. The transcriptomic profiles of 786-O under normoxia, short-term hypoxia and long-term hypoxia were analyzed using next-generation sequencing. The results showed that long-term hypoxia promoted the ability of colony formation and transwell migration compared to normoxia. In addition, the differentially expressed genes induced by long-term hypoxia were involved in various biological processes including cell proliferation, the tumor necrosis factor signaling pathway, basal cell carcinoma and cancer pathways. The upregulated (L1CAM and FBN1) and downregulated (AUTS2, MAPT, AGT and USH1C) genes in 786-O under long-term hypoxia were also observed in clinical ccRCC samples along with malignant grade. The expressions of these genes were significantly correlated with survival outcomes in patients with renal cancer. We also found that long-term hypoxia in 786-O resulted in decreased expressions of hsa-mir-100 and hsa-mir-378 and this effect was also observed in samples of metastatic ccRCC compared to samples of non-metastatic ccRCC. These findings may provide a new direction for the study of potential molecular mechanisms associated with the progression of ccRCC.
Collapse
Affiliation(s)
- Szu-Chia Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan.
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Feng-Wei Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| |
Collapse
|
24
|
Protocols and characterization data for 2D, 3D, and slice-based tumor models from the PREDECT project. Sci Data 2017; 4:170170. [PMID: 29160867 PMCID: PMC5697359 DOI: 10.1038/sdata.2017.170] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 10/18/2017] [Indexed: 12/22/2022] Open
Abstract
Two-dimensional (2D) culture of cancer cells in vitro does not recapitulate the three-dimensional (3D) architecture, heterogeneity and complexity of human tumors. More representative models are required that better reflect key aspects of tumor biology. These are essential studies of cancer biology and immunology as well as for target validation and drug discovery. The Innovative Medicines Initiative (IMI) consortium PREDECT (www.predect.eu) characterized in vitro models of three solid tumor types with the goal to capture elements of tumor complexity and heterogeneity. 2D culture and 3D mono- and stromal co-cultures of increasing complexity, and precision-cut tumor slice models were established. Robust protocols for the generation of these platforms are described. Tissue microarrays were prepared from all the models, permitting immunohistochemical analysis of individual cells, capturing heterogeneity. 3D cultures were also characterized using image analysis. Detailed step-by-step protocols, exemplary datasets from the 2D, 3D, and slice models, and refined analytical methods were established and are presented.
Collapse
|
25
|
Organotypic three-dimensional cancer cell cultures mirror drug responses in vivo: lessons learned from the inhibition of EGFR signaling. Oncotarget 2017; 8:107423-107440. [PMID: 29296175 PMCID: PMC5746077 DOI: 10.18632/oncotarget.22475] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/27/2017] [Indexed: 01/07/2023] Open
Abstract
Complex three-dimensional (3D) in vitro models that recapitulate human tumor biology are essential to understand the pathophysiology of the disease and to aid in the discovery of novel anti-cancer therapies. 3D organotypic cultures exhibit intercellular communication, nutrient and oxygen gradients, and cell polarity that is lacking in two-dimensional (2D) monolayer cultures. In the present study, we demonstrate that 2D and 3D cancer models exhibit different drug sensitivities towards both targeted inhibitors of EGFR signaling and broad acting cytotoxic agents. Changes in the kinase activities of ErbB family members and differential expression of apoptosis- and survival-associated genes before and after drug treatment may account for the differential drug sensitivities. Importantly, EGFR oncoprotein addiction was evident only in the 3D cultures mirroring the effect of EGFR inhibition in the clinic. Furthermore, targeted drug efficacy was strongly increased when incorporating cancer-associated fibroblasts into the 3D cultures. Taken together, we provide conclusive evidence that complex 3D cultures are more predictive of the clinical outcome than their 2D counterparts. In the future, 3D cultures will be instrumental for understanding the mode of action of drugs, identifying genotype-drug response relationships and developing patient-specific and personalized cancer treatments.
Collapse
|
26
|
Avnet S, Di Pompo G, Chano T, Errani C, Ibrahim-Hashim A, Gillies RJ, Donati DM, Baldini N. Cancer-associated mesenchymal stroma fosters the stemness of osteosarcoma cells in response to intratumoral acidosis via NF-κB activation. Int J Cancer 2017; 140:1331-1345. [PMID: 27888521 DOI: 10.1002/ijc.30540] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/09/2016] [Indexed: 12/12/2022]
Abstract
The role of mesenchymal stem cells (MSC) in osteosarcoma (OS), the most common primary tumor of bone, has not been extensively elucidated. We have recently shown that OS is characterized by interstitial acidosis, a microenvironmental condition that is similar to a wound setting, in which mesenchymal reactive cells are activated to release mitogenic and chemotactic factors. We therefore intended to test the hypothesis that, in OS, acid-activated MSC influence tumor cell behavior. Conditioned media or co-culture with normal MSC previously incubated with short-term acidosis (pH 6.8 for 10 hr, H+ -MSC) enhanced OS clonogenicity and invasion. This effect was mediated by NF-κB pathway activation. In fact, deep-sequencing analysis, confirmed by Real-Time PCR and ELISA, demonstrated that H+ -MSC differentially induced a tissue remodeling phenotype with increased expression of RelA, RelB and NF-κB1, and downstream, of CSF2/GM-CSF, CSF3/G-CSF and BMP2 colony-promoting factors, and of chemokines (CCL5, CXCL5 and CXCL1), and cytokines (IL6 and IL8), with an increased expression of CXCR4. An increased expression of IL6 and IL8 were found only in normal stromal cells, but not in OS cells, and this was confirmed in tumor-associated stromal cells isolated from OS tissue. Finally, H+ -MSC conditioned medium differentially promoted OS stemness (sarcosphere number, stem-associated gene expression), and chemoresistance also via IL6 secretion. Our data support the hypothesis that the acidic OS microenvironment is a key factor for MSC activation, in turn promoting the secretion of paracrine factors that influence tumor behavior, a mechanism that holds the potential for future therapeutic interventions aimed to target OS.
Collapse
Affiliation(s)
- Sofia Avnet
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Gemma Di Pompo
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Tokuhiro Chano
- Department of Clinical Laboratory Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Costantino Errani
- Orthopaedic Oncology Surgical Unit, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Arig Ibrahim-Hashim
- Department of Imaging Research, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Robert J Gillies
- Department of Imaging Research, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Davide Maria Donati
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,Orthopaedic Oncology Surgical Unit, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Nicola Baldini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
27
|
Wu H, Zhou J, Mei S, Wu D, Mu Z, Chen B, Xie Y, Ye Y, Liu J. Circulating exosomal microRNA-96 promotes cell proliferation, migration and drug resistance by targeting LMO7. J Cell Mol Med 2017; 21:1228-1236. [PMID: 28026121 PMCID: PMC5431139 DOI: 10.1111/jcmm.13056] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 10/03/2016] [Indexed: 12/11/2022] Open
Abstract
Detection and treatment of lung cancer still remain a clinical challenge. This study aims to validate exosomal microRNA-96 (miR-96) as a serum biomarker for lung cancer and understand the underlying mechanism in lung cancer progression. MiR-96 expressions in normal and lung cancer patients were characterized by qPCR analysis. Changes in cell viability, migration and cisplatin resistance were monitored after incubation with isolated miR-96-containing exosomes, anti-miR-96 and anti-miR negative control (anti-miR-NC) transfections. Dual-luciferase reporter assay was used to study interaction between miR-96 and LIM-domain only protein 7 (LMO7). Changes induced by miR-96 transfection and LMO7 overexpression were also evaluated. MiR-96 expression was positively correlated with high-grade and metastatic lung cancers. While anti-miR-96 transfection exhibited a tumour-suppressing function, exosomes isolated from H1299 enhanced cell viability, migration and cisplatin resistance. Potential miR-96 binding sites were found within the 3'-UTR of wild-type LMO7 gene, but not of mutant LMO7 gene. LMO7 expression was inversely correlated with lung cancer grades, and LMO7 overexpression reversed promoting effect of miR-96. We have identified exosomal miR-96 as a serum biomarker of malignant lung cancer. MiR-96 promotes lung cancer progression by targeting LMO7. The miR-96-LMO7 axis may be a therapeutic target for lung cancer patients, and new diagnostic or therapeutic strategies could be developed by targeting the miR-96-LMO7 axis.
Collapse
Affiliation(s)
- Hao Wu
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
| | - Jingcheng Zhou
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
| | - Shanshan Mei
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
| | - Da Wu
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
| | - Zhimin Mu
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
| | - Baokun Chen
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
| | - Yuancai Xie
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
| | - Yiwang Ye
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
| | - Jixian Liu
- Department of Thoracic SurgeryPeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
| |
Collapse
|
28
|
Fang L, He J, Xia J, Dong L, Zhang X, Chai Y, Li Y, Niu M, Hang T, Li S. Resistance to epithelial growth factor receptor tyrosine kinase inhibitors in a patient with transformation from lung adenocarcinoma to small cell lung cancer: A case report. Oncol Lett 2017; 14:593-598. [PMID: 28693210 PMCID: PMC5494666 DOI: 10.3892/ol.2017.6229] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 02/23/2017] [Indexed: 01/04/2023] Open
Abstract
First-generation epithelial growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) have markedly improved the treatment of non-small cell lung cancer (non-SCLC) with EGFR-sensitive mutations. However, acquired resistance to these drugs was inevitable. The transformation of lung adenocarcinoma to SCLC following treatment with EGFR-TKIs is a rare phenomenon that contributes to resistance to EGFR-TKIs. The present case concerns a 74-year-old man previously diagnosed with and treated for pneumonia; however, this was later pathologically confirmed as lung adenocarcinoma by transbronchial lung biopsy. Deletion of exon 19 of EGFR was identified by next-generation sequencing technology. The patient improved markedly when treated with gefitinib, but relapsed after 1 year, with markedly increased serum levels of neuron-specific enolase (NSE). Transformation to SCLC was detected by endobronchial ultrasound transbronchial needle aspiration (EBUS-TBNA) re-biopsy, which was negative for the deletion of exon 19 of EGFR. The patient was positive for vimentin expression and refractory to etoposide and cisplatin chemotherapy, and succumbed to the disease 18 months after diagnosis. Transformation of the disease from adenocarcinoma to SCLC may have been due to cancer heterogeneity. Re-biopsy is therefore important in EGFR-TKI-resistant patients for genetic and histological re-evaluation. NSE serum levels may also be useful for detecting early SCLC transformation.
Collapse
Affiliation(s)
- Liying Fang
- Department of Respiratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Department of Respiratory Medicine, The Fourth People's Hospital of Shaanxi Xi'an, Shaanxi 710043, P.R. China
| | - Jian He
- Department of Respiratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jingwen Xia
- Department of Respiratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Liang Dong
- Department of Respiratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Xiujuan Zhang
- Department of Respiratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Yaqin Chai
- Department of Respiratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Department of Respiratory Medicine, Xi'an Aerospace General Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Ying Li
- Department of Respiratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Department of Respiratory Medicine, The Second People's Hospital of Shaanxi Xi'an, Shaanxi 710005, P.R. China
| | - Mengjie Niu
- Department of Respiratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Tianxing Hang
- Department of Respiratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Shengqing Li
- Department of Respiratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
29
|
Stromal-derived IGF2 promotes colon cancer progression via paracrine and autocrine mechanisms. Oncogene 2017; 36:5341-5355. [PMID: 28534511 DOI: 10.1038/onc.2017.116] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 02/25/2017] [Accepted: 03/23/2017] [Indexed: 12/31/2022]
Abstract
The insulin-like growth factor (IGF)2/IGF1 receptor (IGF1R) signaling axis has an important role in intestinal carcinogenesis and overexpression of IGF2 is an accepted risk factor for colorectal cancer (CRC) development. Genetic amplifications and loss of imprinting contribute to the upregulation of IGF2, but insufficiently explain the extent of IGF2 expression in a subset of patients. Here, we show that IGF2 was specifically induced in the tumor stroma of CRC and identified cancer-associated fibroblasts (CAFs) as the major source. Further, we provide functional evidence that stromal IGF2, via the paracrine IGF1R/insulin receptor axis, activated pro-survival AKT signaling in CRC cell lines. In addition to its effects on malignant cells, autocrine IGF2/IGF1R signaling in CAFs induced myofibroblast differentiation in terms of alpha-smooth muscle actin expression and contractility in floating collagen gels. This was further augmented in concert with transforming growth factor-β (TGFβ) signaling suggesting a cooperative mechanism. However, we demonstrated that IGF2 neither induced TGFβ/smooth muscle actin/mothers against decapentaplegic (SMAD) signaling nor synergized with TGFβ to hyperactivate this pathway in two dimensional and three dimensional cultures. IGF2-mediated physical matrix remodeling by CAFs, but not changes in extracellular matrix-modifying proteases or other secreted factors acting in a paracrine manner on/in cancer cells, facilitated subsequent tumor cell invasion in organotypic co-cultures. Consistently, colon cancer cells co-inoculated with CAFs expressing endogenous IGF2 in mouse xenograft models exhibited elevated invasiveness and dissemination capacity, as well as increased local tumor regrowth after primary tumor resection compared with conditions with IGF2-deficient CAFs. In line, expression of IGF2 correlated with elevated relapse rates and poor survival in CRC patients. In agreement with our results, high-level coexpression of IGF2 and TGFβ was predicting adverse outcome with higher accuracy than increased expression of the individual genes alone. Taken together, we demonstrate that stroma-induced IGF2 promotes colon cancer progression in a paracrine and autocrine manner and propose IGF2 as potential target for tumor stroma cotargeting strategies.
Collapse
|
30
|
ErbB Family Signalling: A Paradigm for Oncogene Addiction and Personalized Oncology. Cancers (Basel) 2017; 9:cancers9040033. [PMID: 28417948 PMCID: PMC5406708 DOI: 10.3390/cancers9040033] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 02/07/2023] Open
Abstract
ErbB family members represent important biomarkers and drug targets for modern precision therapy. They have gained considerable importance as paradigms for oncoprotein addiction and personalized medicine. This review summarizes the current understanding of ErbB proteins in cell signalling and cancer and describes the molecular rationale of prominent cases of ErbB oncoprotein addiction in different cancer types. In addition, we have highlighted experimental technologies for the development of innovative cancer cell models that accurately predicted clinical ErbB drug efficacies. In the future, such cancer models might facilitate the identification and validation of physiologically relevant novel forms of oncoprotein and non-oncoprotein addiction or synthetic lethality. The identification of genotype-drug response relationships will further advance personalized oncology and improve drug efficacy in the clinic. Finally, we review the most important drugs targeting ErbB family members that are under investigation in clinical trials or that made their way already into clinical routine. Taken together, the functional characterization of ErbB oncoproteins have significantly increased our knowledge on predictive biomarkers, oncoprotein addiction and patient stratification and treatment.
Collapse
|
31
|
Puré E, Lo A. Can Targeting Stroma Pave the Way to Enhanced Antitumor Immunity and Immunotherapy of Solid Tumors? Cancer Immunol Res 2016; 4:269-78. [PMID: 27036971 DOI: 10.1158/2326-6066.cir-16-0011] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Solid tumors are complex organ-like structures. The potential of normal neighboring cells to contribute to the initiation, progression, and metastasis of epithelial-derived carcinomas has long been appreciated. However, the role of host cells has proven complex. Through multiple local and systemic mechanisms, nontransformed host cells can promote transition from a tumor-resistant to tumor-permissive environment, drive neoplastic transformation of epithelial cells, promote tumor growth, progression, and metastasis, but also constrain tumorigenesis. This complexity reflects the spatially and temporally dynamic involvement of multiple cell types and processes, including the development and recruitment of inflammatory, immune, endothelial, and mesenchymal stromal cells, and the remodeling of extracellular matrix. Our mechanistic understanding, as well as our ability to translate advances in our understanding of these mechanisms for therapeutic benefit, is rapidly advancing. Further insights will depend on delineating pathways that mediate the communication networks between inflammatory and immune cells with tumor and mesenchymal stromal cells and extracellular matrix. Here, we discuss the diversity of mesenchymal stromal cell populations and how context can dictate either their promotion or constraint of tumorigenesis. We review evidence for plasticity that allows for reprogramming of stromal cells and how tumor immunogenicity and desmoplasia influence the balance of immune-independent and immune-dependent regulation of tumor growth. The pivotal roles of matrix and mesenchymal stromal cells in modulating inflammation, antitumor immunity, and the efficacy of immune-based therapies are discussed. These concepts have emerged from data obtained from tumors of multiple organs, but we focus mostly on studies of pancreatic ductal adenocarcinomas.
Collapse
Affiliation(s)
- Ellen Puré
- University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Albert Lo
- University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
32
|
Santo VE, Rebelo SP, Estrada MF, Alves PM, Boghaert E, Brito C. Drug screening in 3D in vitro tumor models: overcoming current pitfalls of efficacy read-outs. Biotechnol J 2016; 12. [PMID: 27966285 DOI: 10.1002/biot.201600505] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/24/2016] [Accepted: 11/10/2016] [Indexed: 12/13/2022]
Abstract
There is cumulating evidence that in vitro 3D tumor models with increased physiological relevance can improve the predictive value of pre-clinical research and ultimately contribute to achieve decisions earlier during the development of cancer-targeted therapies. Due to the role of tumor microenvironment in the response of tumor cells to therapeutics, the incorporation of different elements of the tumor niche on cell model design is expected to contribute to the establishment of more predictive in vitro tumor models. This review is focused on the several challenges and adjustments that the field of oncology research is facing to translate these advanced tumor cells models to drug discovery, taking advantage of the progress on culture technologies, imaging platforms, high throughput and automated systems. The choice of 3D cell model, the experimental design, choice of read-outs and interpretation of data obtained from 3D cell models are critical aspects when considering their implementation in drug discovery. In this review, we foresee some of these aspects and depict the potential directions of pre-clinical oncology drug discovery towards improved prediction of drug efficacy.
Collapse
Affiliation(s)
- Vítor E Santo
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Sofia P Rebelo
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Marta F Estrada
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | | | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
33
|
Åkerfelt M, Bayramoglu N, Robinson S, Toriseva M, Schukov HP, Härmä V, Virtanen J, Sormunen R, Kaakinen M, Kannala J, Eklund L, Heikkilä J, Nees M. Automated tracking of tumor-stroma morphology in microtissues identifies functional targets within the tumor microenvironment for therapeutic intervention. Oncotarget 2016; 6:30035-56. [PMID: 26375443 PMCID: PMC4745780 DOI: 10.18632/oncotarget.5046] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 08/24/2015] [Indexed: 01/01/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) constitute an important part of the tumor microenvironment and promote invasion via paracrine functions and physical impact on the tumor. Although the importance of including CAFs into three-dimensional (3D) cell cultures has been acknowledged, computational support for quantitative live-cell measurements of complex cell cultures has been lacking. Here, we have developed a novel automated pipeline to model tumor-stroma interplay, track motility and quantify morphological changes of 3D co-cultures, in real-time live-cell settings. The platform consists of microtissues from prostate cancer cells, combined with CAFs in extracellular matrix that allows biochemical perturbation. Tracking of fibroblast dynamics revealed that CAFs guided the way for tumor cells to invade and increased the growth and invasiveness of tumor organoids. We utilized the platform to determine the efficacy of inhibitors in prostate cancer and the associated tumor microenvironment as a functional unit. Interestingly, certain inhibitors selectively disrupted tumor-CAF interactions, e.g. focal adhesion kinase (FAK) inhibitors specifically blocked tumor growth and invasion concurrently with fibroblast spreading and motility. This complex phenotype was not detected in other standard in vitro models. These results highlight the advantage of our approach, which recapitulates tumor histology and can significantly improve cancer target validation in vitro.
Collapse
Affiliation(s)
- Malin Åkerfelt
- Turku Centre for Biotechnology, University of Turku, Turku, FI-20520, Finland.,VTT Technical Research Centre of Finland, Turku, FI-20521, Finland
| | - Neslihan Bayramoglu
- Centre for Machine Vision Research, University of Oulu, Oulu, FI-90014, Finland
| | - Sean Robinson
- Department of Mathematics and Statistics, University of Turku, Turku, FI-20014, Finland.,University Grenoble Alpes, iRTSV-BGE, Grenoble, F-38000, France.,CEA, iRTSV-BGE, Grenoble, F-38000, France.,INSERM, BGE, Grenoble, F-38000, France
| | - Mervi Toriseva
- Turku Centre for Biotechnology, University of Turku, Turku, FI-20520, Finland.,VTT Technical Research Centre of Finland, Turku, FI-20521, Finland.,Institute of Biomedicine, University of Turku, Turku, FI-20520, Finland
| | | | - Ville Härmä
- VTT Technical Research Centre of Finland, Turku, FI-20521, Finland
| | | | - Raija Sormunen
- Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Oulu, FI-90220, Finland
| | - Mika Kaakinen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, FI-90014, Finland
| | - Juho Kannala
- Centre for Machine Vision Research, University of Oulu, Oulu, FI-90014, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, FI-90014, Finland
| | - Janne Heikkilä
- Centre for Machine Vision Research, University of Oulu, Oulu, FI-90014, Finland
| | - Matthias Nees
- Turku Centre for Biotechnology, University of Turku, Turku, FI-20520, Finland.,VTT Technical Research Centre of Finland, Turku, FI-20521, Finland
| |
Collapse
|
34
|
Capturing tumor complexity in vitro: Comparative analysis of 2D and 3D tumor models for drug discovery. Sci Rep 2016; 6:28951. [PMID: 27364600 PMCID: PMC4929472 DOI: 10.1038/srep28951] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/27/2016] [Indexed: 01/14/2023] Open
Abstract
Two-dimensional (2D) cell cultures growing on plastic do not recapitulate the three dimensional (3D) architecture and complexity of human tumors. More representative models are required for drug discovery and validation. Here, 2D culture and 3D mono- and stromal co-culture models of increasing complexity have been established and cross-comparisons made using three standard cell carcinoma lines: MCF7, LNCaP, NCI-H1437. Fluorescence-based growth curves, 3D image analysis, immunohistochemistry and treatment responses showed that end points differed according to cell type, stromal co-culture and culture format. The adaptable methodologies described here should guide the choice of appropriate simple and complex in vitro models.
Collapse
|
35
|
Santo VE, Estrada MF, Rebelo SP, Abreu S, Silva I, Pinto C, Veloso SC, Serra AT, Boghaert E, Alves PM, Brito C. Adaptable stirred-tank culture strategies for large scale production of multicellular spheroid-based tumor cell models. J Biotechnol 2016; 221:118-29. [PMID: 26815388 DOI: 10.1016/j.jbiotec.2016.01.031] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/15/2016] [Accepted: 01/21/2016] [Indexed: 12/29/2022]
Abstract
Currently there is an effort toward the development of in vitro cancer models more predictive of clinical efficacy. The onset of advanced analytical tools and imaging technologies has increased the utilization of spheroids in the implementation of high throughput approaches in drug discovery. Agitation-based culture systems are commonly proposed as an alternative method for the production of tumor spheroids, despite the scarce experimental evidence found in the literature. In this study, we demonstrate the robustness and reliability of stirred-tank cultures for the scalable generation of 3D cancer models. We developed standardized protocols to a panel of tumor cell lines from different pathologies and attained efficient tumor cell aggregation by tuning hydrodynamic parameters. Large numbers of spheroids were obtained (typically 1000-1500 spheroids/mL) presenting features of native tumors, namely morphology, proliferation and hypoxia gradients, in a cell line-dependent mode. Heterotypic 3D cancer models, based on co-cultures of tumor cells and fibroblasts, were also established in the absence or presence of additional physical support from an alginate matrix, with maintenance of high cell viability. Altogether, we demonstrate that 3D tumor cell model production in stirred-tank culture systems is a robust and versatile approach, providing reproducible tools for drug screening and target verification in pre-clinical oncology research.
Collapse
Affiliation(s)
- Vítor E Santo
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Marta F Estrada
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Sofia P Rebelo
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Sofia Abreu
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Inês Silva
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Catarina Pinto
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Susana C Veloso
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Ana Teresa Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | | | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal.
| |
Collapse
|
36
|
Osswald A, Sun Z, Grimm V, Ampem G, Riegel K, Westendorf AM, Sommergruber W, Otte K, Dürre P, Riedel CU. Three-dimensional tumor spheroids for in vitro analysis of bacteria as gene delivery vectors in tumor therapy. Microb Cell Fact 2015; 14:199. [PMID: 26655167 PMCID: PMC4676896 DOI: 10.1186/s12934-015-0383-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/11/2015] [Indexed: 12/31/2022] Open
Abstract
Background Several studies in animal models demonstrated that obligate and facultative anaerobic bacteria of the genera Bifidobacterium, Salmonella, or Clostridium specifically colonize solid tumors. Consequently, these and other bacteria are discussed as live vectors to deliver therapeutic genes to inhibit tumor growth. Therapeutic approaches for cancer treatment using anaerobic bacteria have been investigated in different mouse models. In the present study, solid three-dimensional (3D) multicellular tumor spheroids (MCTS) of the colorectal adenocarcinoma cell line HT-29 were generated and tested for their potential to study prodrug-converting enzyme therapies using bacterial vectors in vitro. Results HT-29 MCTS resembled solid tumors displaying all relevant features with an outer zone of proliferating cells and hypoxic and apoptotic regions in the core. Upon incubation with HT-29 MCTS, Bifidobacterium bifidum S17 and Salmonella typhimurium YB1 selectively localized, survived and replicated in hypoxic areas inside MCTS. Furthermore, spores of the obligate anaerobe Clostridium sporogenes germinated in these hypoxic areas. To further evaluate the potential of MCTS to investigate therapeutic approaches using bacteria as gene delivery vectors, recombinant bifidobacteria expressing prodrug-converting enzymes were used. Expression of a secreted cytosine deaminase in combination with 5-fluorocytosine had no effect on growth of MCTS due to an intrinsic resistance of HT-29 cells to 5-fluorouracil, i.e. the converted drug. However, a combination of the prodrug CB1954 and a strain expressing a secreted chromate reductase effectively inhibited MCTS growth. Conclusions Collectively, the presented results indicate that MCTS are a suitable and reliable model to investigate live bacteria as gene delivery vectors for cancer therapy in vitro. Electronic supplementary material The online version of this article (doi:10.1186/s12934-015-0383-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Annika Osswald
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany. .,Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, 1121, Vienna, Austria.
| | - Zhongke Sun
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany. .,College of Life Sciences and Agriculture, Zhoukou Normal University, Chuanhui District, Zhoukou, 466001, People's Republic of China.
| | - Verena Grimm
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| | - Grace Ampem
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| | - Karin Riegel
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | - Wolfgang Sommergruber
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, 1121, Vienna, Austria.
| | - Kerstin Otte
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, 88400, Biberach, Germany.
| | - Peter Dürre
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| | - Christian U Riedel
- Institute of Microbiology and Biotechnology, University of Ulm, 89069, Ulm, Germany.
| |
Collapse
|
37
|
Stadler M, Walter S, Walzl A, Kramer N, Unger C, Scherzer M, Unterleuthner D, Hengstschläger M, Krupitza G, Dolznig H. Increased complexity in carcinomas: Analyzing and modeling the interaction of human cancer cells with their microenvironment. Semin Cancer Biol 2015; 35:107-24. [DOI: 10.1016/j.semcancer.2015.08.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/19/2015] [Accepted: 08/21/2015] [Indexed: 02/08/2023]
|
38
|
Shan C, Wei J, Hou R, Wu B, Yang Z, Wang L, Lei D, Yang X. Schwann cells promote EMT and the Schwann-like differentiation of salivary adenoid cystic carcinoma cells via the BDNF/TrkB axis. Oncol Rep 2015; 35:427-35. [PMID: 26530352 DOI: 10.3892/or.2015.4366] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 09/23/2015] [Indexed: 11/06/2022] Open
Abstract
Perineural invasion (PNI) is a striking biological behavior observed in salivary adenoid cystic carcinoma (SACC). The present study was designed to establish a co-culture model of SACC cells with Schwann cells (SCs), and then study epithelial-mesenchymal transition (EMT) and the Schwann-like differentiation of SACC cells to investigate the likely molecular mechanism of PNI. The co-culture models of SCs with tumor cells (SACC-83, SACC-LM and MEC-1) were established using a Transwell system. An elevated concentration of brain-derived neurotrophic factor (BDNF) was detected by ELISA assay in the co-cultured medium of the SACC-83 group and SACC-LM group rather than the MEC-1 group. The EMT process and Schwann-like differentiation in SACC-83 cells were analyzed by RT-PCR, western blotting, immunofluorescence, photography, and migration and perineural invasion assays. The SACC-83 cells under the co-culture condition with SCs changed to a mesenchymal morphology and had higher migration and invasion capabilities compared with the solely cultured SACC-83 cells, accompanied by the downregulation of E-cadherin and upregulation of N-cadherin and vimentin. The co-cultured SACC-83 cells also developed Schwann-like differentiation with increased expression of SC markers, S100A4 and GFAP. However, inhibition of tropomyosin-related kinase B (TrkB) by K252a markedly blocked these effects. Additionally, the expression and correlation of TrkB, E-cadherin and S100A4 were analyzed by immunohistochemistry in 187 primary SACC cases. The levels of TrkB and S100A4 expression were both positively associated with PNI in the SACC cases, while E-cadherin expression was negatively associated with PNI. Elevated expression of TrkB was significantly correlated with the downregulated expression of E-cadherin and the upregulated expression of S100A4 in the SACC cases. Our results suggest that SCs play a pivotal role in the PNI process by inducing the EMT process and the Schwann-like differentiation of SACC cells via the BDNF/TrkB axis. Interruption of the interreaction between SACC cells and SCs by targeting the BDNF/TrkB axis may be a potential strategy for anti-PNI therapy in SACC.
Collapse
Affiliation(s)
- Chun Shan
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jianhua Wei
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Rui Hou
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Baolei Wu
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zihui Yang
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lei Wang
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Delin Lei
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xinjie Yang
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|