1
|
Suzuki S, Yashiro M, Izumi N, Tsukioka T, Inoue H, Hara K, Ito R, Tanimura T, Nishiyama N. Impact of CA9 expression in the diagnosis of lymph-node metastases in non-small cell lung cancer based on [18F]FDG PET/CT. PLoS One 2024; 19:e0312846. [PMID: 39471162 PMCID: PMC11521239 DOI: 10.1371/journal.pone.0312846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/15/2024] [Indexed: 11/01/2024] Open
Abstract
BACKGROUND Lung cancer is the leading cause of the global cancer incidence and mortality. It is important to obtain an accurate diagnosis of lymph-node metastasis before surgery to select the therapeutic strategy for non-small cell lung cancer (NSCLC) patients. Carbonic anhydrase 9 (CA9) is considered a marker of hypoxia and it has reported that CA9 is associated with tumor invasion and metastasis. In this study, the correlation between the CA9 expression for lymph-node metastases in NSCLC and [18F]FDG PET/CT results was investigated in order to clarify the efficacy of [18F]FDG PET/CT for detecting lymph-node metastases of NSCLC patients. METHODS Among the 564 patients who underwent surgical treatment for NSCLC between 2010 and 2016 at our hospital, a total of 338 patients who underwent preoperative [18F]FDG PET/CT were included in this study. CA9 expression was evaluated by immunochemistry. A lymph node with maximum standardized uptake value (SUVmax) ≥2.5 on [18F]FDG PET/CT was preoperatively defined as a metastatic lymph node. RESULT CA9 positivity was detected in 122 patients; the other 216 patients were CA9-negative. The CA9-positive NSCLC cases significantly associated with pleural invasion (p = 0.0063), pT-factor (p = 0.0080), pN-factor (p = 0.036) and pStage (p = 0.043). CA9-positive patients presented significantly poorer survival rate for OS than that of the CA9-negative patients (p = 0.0024). In the multivariable analysis, histological SCC and CA9 positivity were independent poor-prognosis factors for OS. For the total patient population, the sensitivity and specificity of [18F]FDG PET/CT for lymph-node metastases were 54% and 89%, respectively. In contrast, the sensitivity and specificity were particularly low in the CA9-positive SCC cases (36% and 69%, respectively). CONCLUSION [18F]FDG PET/CT might not be useful for diagnosing lymph-node metastases of CA9-positive SCC cases of NSCLC.
Collapse
Affiliation(s)
- Satoshi Suzuki
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Nobuhiro Izumi
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Takuma Tsukioka
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hidetoshi Inoue
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Kantaro Hara
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Ryuichi Ito
- Department of Thoracic Surgery, Kansai Rosai Hospital, Hyogo, Japan
| | - Takuya Tanimura
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Noritoshi Nishiyama
- Department of Thoracic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
2
|
Zheng S, Zou Y, Liang J, Xiao W, Yang A, Meng T, Lu S, Luo Z, Xie X. Identification and validation of a combined hypoxia and immune index for triple-negative breast cancer. Mol Oncol 2020; 14:2814-2833. [PMID: 32521117 PMCID: PMC7607163 DOI: 10.1002/1878-0261.12747] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 05/25/2020] [Indexed: 12/21/2022] Open
Abstract
The interaction between hypoxia and immune status has been confirmed in various cancer settings, and corresponding treatments have been investigated. However, reliable biomarkers are needed for individual treatment, so we sought to develop a novel scoring system based on hypoxia and immune status. Prognostic hypoxia-immune status-related signatures of patients with triple-negative breast cancer (TNBC) were identified in The Cancer Genome Atlas (TCGA) (N = 158), Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) (N = 297), and GSE58812 (N = 107). LASSO Cox regression was used for model construction. Hypoxia and immune status expression profiles were analyzed, and infiltrating immune cells were compared. Quantitative real-time PCR (qRT-PCR) was used for validation in the Sun Yat-sen University Cancer Center (SYSUCC) cohort, and immunofluorescence was applied for the detection of hypoxia and immune markers in cancer tissues. Ten cross-cohort prognostic hypoxia-immune signatures were included to construct the comprehensive index of hypoxia and immune (CIHI) in the METABRIC cohort. Two subgroups of patients with distinct hypoxia-immune status conditions were identified using CIHI: hypoxiahigh /immunelow and hypoxialow /immunehigh , with a significantly better overall survival (OS) rate in the latter (P < 0.01). The prognostic value of CIHI was further validated in the TCGA, GSE58812, and SYSUCC cohorts (P < 0.01). Hypoxia-immune signatures were significantly differentially expressed between the two groups, and more active immune responses were observed in the hypoxialow /immunehigh group. Cytotoxic lymphocytes were inversely correlated with CIHI in silico. Differentially expressed CA-IX and stromal PD-L1 were detected between subgroups of the SYSUCC cohort. A hypoxia-immune-based cross-cohort classifier for predicting prognosis was developed and validated, which may guide hypoxia modifier treatment and immunotherapy for TNBC.
Collapse
Affiliation(s)
- Shaoquan Zheng
- Department of Breast OncologySun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangdongChina
| | - Yutian Zou
- Department of Breast OncologySun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangdongChina
| | - Jie‐ying Liang
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangdongChina
- Department of Medical OncologySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Weikai Xiao
- Department of Breast CancerCancer CenterGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Anli Yang
- Department of Breast OncologySun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangdongChina
| | - Tiebao Meng
- Department of RadiologySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Shilin Lu
- Zhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouChina
| | - Zhongbing Luo
- Department of Breast SurgeryFirst Affiliated Hospital of Gannan Medical CollegeGanzhou CityChina
| | - Xiaoming Xie
- Department of Breast OncologySun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangdongChina
| |
Collapse
|
3
|
Daunys S, Petrikaitė V. The roles of carbonic anhydrases IX and XII in cancer cell adhesion, migration, invasion and metastasis. Biol Cell 2020; 112:383-397. [PMID: 32894882 DOI: 10.1111/boc.201900099] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 09/02/2012] [Indexed: 12/17/2022]
Abstract
The main function of carbonic anhydrases (CAs) in cancer cells is the pH regulation through a conversion of H2 O and CO2 to H+ and HCO3 - . However, the data of in vitro and in vivo studies have demonstrated that transmembrane isoforms of CA IX and CA XII are involved in various steps of cancer cell migration, invasion and metastasis. According to literature, inhibition of these CAs can affect the expression of multiple proteins. Some scientific groups have reported the possible interactions between CA IX and E-cadherin-catenin system, CA IX and integrins, CA IX, CA XII and ion transporters, which all are highly involved in cell-to-cell adhesion, the formation of membrane protrusions and focal adhesions. Nevertheless, CA IX and CA XII have a high impact on tumour growth and metastases formation. The data discussed in this review are quite recent. It highly support the role of CA IX and CA XII in various cancer metastasis processes through their interactions to other invasion proteins. Nevertheless, all findings show the great potential of these CAs in the context of research and application in clinical use.
Collapse
Affiliation(s)
- Simonas Daunys
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, LT-10257, Lithuania
| | - Vilma Petrikaitė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, LT-10257, Lithuania.,Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, LT-50162, Lithuania
| |
Collapse
|
4
|
The Interplay of Dysregulated pH and Electrolyte Imbalance in Cancer. Cancers (Basel) 2020; 12:cancers12040898. [PMID: 32272658 PMCID: PMC7226178 DOI: 10.3390/cancers12040898] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer cells and tissues have an aberrant regulation of hydrogen ion dynamics driven by a combination of poor vascular perfusion, regional hypoxia, and increased the flux of carbons through fermentative glycolysis. This leads to extracellular acidosis and intracellular alkalinization. Dysregulated pH dynamics influence cancer cell biology, from cell transformation and tumorigenesis to proliferation, local growth, invasion, and metastasis. Moreover, this dysregulated intracellular pH (pHi) drives a metabolic shift to increased aerobic glycolysis and reduced mitochondrial oxidative phosphorylation, referred to as the Warburg effect, or Warburg metabolism, which is a selective feature of cancer. This metabolic reprogramming confers a thermodynamic advantage on cancer cells and tissues by protecting them against oxidative stress, enhancing their resistance to hypoxia, and allowing a rapid conversion of nutrients into biomass to enable cell proliferation. Indeed, most cancers have increased glucose uptake and lactic acid production. Furthermore, cancer cells have very dysregulated electrolyte balances, and in the interaction of the pH dynamics with electrolyte, dynamics is less well known. In this review, we highlight the interconnected roles of dysregulated pH dynamics and electrolytes imbalance in cancer initiation, progression, adaptation, and in determining the programming and reprogramming of tumor cell metabolism.
Collapse
|
5
|
Vergara D, Ravaioli S, Fonzi E, Adamo L, Damato M, Bravaccini S, Pirini F, Gaballo A, Barbano R, Pasculli B, Franck J, Fournier I, Salzet M, Maffia M. Carbonic Anhydrase XII Expression Is Modulated during Epithelial Mesenchymal Transition and Regulated through Protein Kinase C Signaling. Int J Mol Sci 2020; 21:ijms21030715. [PMID: 31979064 PMCID: PMC7037142 DOI: 10.3390/ijms21030715] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 01/08/2023] Open
Abstract
Members of the carbonic anhydrase family are functionally involved in the regulation of intracellular and extracellular pH in physiological and pathological conditions. Their expression is finely regulated to maintain a strict control on cellular homeostasis, and it is dependent on the activation of extracellular and intracellular signaling pathways. Combining RNA sequencing (RNA-seq), NanoString, and bioinformatics data, we demonstrated that the expression of carbonic anhydrase 12 (CAXII) is significantly different in luminal and triple negative breast cancer (BC) models and patients, and is associated with the activation of an epithelial mesenchymal transition (EMT) program. In BC models, the phorbol ester 12-myristate 13-acetate (PMA)-mediated activation of protein kinase C (PKC) induced a down-regulation of CAXII with a concomitant modulation of other members of the transport metabolon, including CAIX and the sodium bicarbonate cotransporter 3 (NBCn1). This is associated with a remodeling of tumor glycolytic metabolism induced after PKC activation. Overall, this analysis highlights the dynamic nature of transport metabolom and identifies signaling pathways finely regulating this plasticity.
Collapse
Affiliation(s)
- Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.A.); (M.D.)
- Laboratory of Clinical Proteomics, “Giovanni Paolo II” Hospital, 73100 ASL-Lecce, Italy
- Correspondence: (D.V.); (M.M.); Tel.: +39-0832-661915 (D.V.); +39-0832-298670 (M.M.)
| | - Sara Ravaioli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (S.R.); (E.F.); (S.B.); (F.P.)
| | - Eugenio Fonzi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (S.R.); (E.F.); (S.B.); (F.P.)
| | - Loredaria Adamo
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.A.); (M.D.)
- Laboratory of Clinical Proteomics, “Giovanni Paolo II” Hospital, 73100 ASL-Lecce, Italy
| | - Marina Damato
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.A.); (M.D.)
- Laboratory of Clinical Proteomics, “Giovanni Paolo II” Hospital, 73100 ASL-Lecce, Italy
| | - Sara Bravaccini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (S.R.); (E.F.); (S.B.); (F.P.)
| | - Francesca Pirini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (S.R.); (E.F.); (S.B.); (F.P.)
| | - Antonio Gaballo
- CNR-NANOTEC, Institute of Nanotechnology c/o Campus Ecotekne, 73100 Lecce, Italy;
| | - Raffaela Barbano
- Fondazione IRCCS Casa Sollievo della Sofferenza Laboratorio di Oncologia, 71013 San Giovanni Rotondo, Italy; (R.B.); (B.P.)
| | - Barbara Pasculli
- Fondazione IRCCS Casa Sollievo della Sofferenza Laboratorio di Oncologia, 71013 San Giovanni Rotondo, Italy; (R.B.); (B.P.)
| | - Julien Franck
- Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Université de Lille, INSERM, U1192 F-59000 Lille, France; (J.F.); (I.F.); (M.S.)
| | - Isabelle Fournier
- Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Université de Lille, INSERM, U1192 F-59000 Lille, France; (J.F.); (I.F.); (M.S.)
| | - Michel Salzet
- Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), Université de Lille, INSERM, U1192 F-59000 Lille, France; (J.F.); (I.F.); (M.S.)
| | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.A.); (M.D.)
- Laboratory of Clinical Proteomics, “Giovanni Paolo II” Hospital, 73100 ASL-Lecce, Italy
- Correspondence: (D.V.); (M.M.); Tel.: +39-0832-661915 (D.V.); +39-0832-298670 (M.M.)
| |
Collapse
|
6
|
Xu G, Fang Z, Clark LH, Sun W, Yin Y, Zhang R, Sullivan SA, Tran AQ, Kong W, Wang J, Zhou C, Bae-Jump VL. Topiramate exhibits anti-tumorigenic and metastatic effects in ovarian cancer cells. Am J Transl Res 2018; 10:1663-1676. [PMID: 30018708 PMCID: PMC6038080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/19/2018] [Indexed: 06/08/2023]
Abstract
Ovarian cancer is one of the leading causes of cancer related deaths among women worldwide, with an overall 5-year survival of only 30-40%. Carbonic anhydrases are up-regulated in many types of cancer and play an important role in tumor progression and metastasis. Carbonic anhydrase 9 has been implicated as a potential anti-tumorigenic target. Topiramate (TPM) is a potent inhibitor of carbonic anhydrase isozymes, including carbonic anhydrase 9, and has been shown to have anti-tumorigenic activity in several cancer types. Our goal was to evaluate the effect of TPM on cell proliferation and to identify possible mechanisms by which TPM inhibits cell growth in ovarian cancer. TPM significantly inhibited ovarian cancer cell proliferation and induced cell cycle G1 arrest, cellular stress and apoptosis through the AKT/mTOR and MAPK pathways. TPM also exerted anti-metastatic effects by decreasing the adhesion and invasion of ovarian cancer cells and affecting the expression of critical regulators of the epithelial-mesenchymal transition (EMT). Our findings demonstrate that TPM has anti-tumorigenic effects in ovarian cancer and is worthy of further exploration in clinical trials.
Collapse
Affiliation(s)
- Guangxu Xu
- Department of Gynecology, Shanghai Jiaotong University Affiliated Sixth People Hospital South CampusShanghai, P. R. China
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Ziwei Fang
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, P. R. China
| | - Leslie H Clark
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Yajie Yin
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Rong Zhang
- Department of Gynecology, Shanghai Jiaotong University Affiliated Sixth People Hospital South CampusShanghai, P. R. China
| | - Stephanie A Sullivan
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Arthur-Quan Tran
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Weimin Kong
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, P. R. China
| | - Jiandong Wang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, P. R. China
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Victoria L Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC, USA
| |
Collapse
|
7
|
Lei B, Zhang XY, Zhou JP, Mu GN, Li YW, Zhang YX, Pang D. Transcriptome sequencing of HER2-positive breast cancer stem cells identifies potential prognostic marker. Tumour Biol 2016; 37:14757-14764. [PMID: 27629143 DOI: 10.1007/s13277-016-5351-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 09/07/2016] [Indexed: 12/11/2022] Open
Abstract
In cancer stem cell theory, breast cancer stem cells (BCSCs) are postulated to be the root cause of recurrence and metastasis in breast cancer. Discovery of new biomarkers and development of BCSC-targeted therapy are practical issues that urgently need to be addressed in the clinic. However, few breast cancer stem cell targets are known. Given that there are few BCSCs, performing transcriptome sequencing on them thus far has not been possible. With the emergence of single-cell sequencing technology, we have now undertaken such a study. We prepared single-cell suspensions, which were sorted using flow cytometry from breast tumor tissue and adjacent normal breast tissue from two HER2-positive patients. We obtained BCSCs, breast cancer cells, mammary cells, and CD44+ mammary cells. Transcriptome sequencing was then performed on these four cell types. Using bioinformatics, we identified 404 differentially expressed BCSC genes from the HER2-positive tumors and preliminary explored transcriptome characteristics of BCSCs. Finally, by querying a public database, we found that CA12 was a novel prognostic biomarker in HER2-positive breast cancer, which also had prognostic value in all breast cancer types. In conclusion, our results suggest that CA12 may be associated with BCSCs, especially HER2-positive BCSCs, and is a potential novel therapeutic target and biomarker.
Collapse
Affiliation(s)
- Bo Lei
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Xian-Yu Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Jia-Peng Zhou
- Department of Internal Medicine-Neurology, General Hospital of Chinese People's Armed Police Forces, 69 Yongding Road, Beijing, 100039, China
| | - Guan-Nan Mu
- Department of Biotherapy Center, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Yi-Wen Li
- Department of Medical Oncology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, China
| | - You-Xue Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| |
Collapse
|
8
|
Singh M, Mukundan S, Jaramillo M, Oesterreich S, Sant S. Three-Dimensional Breast Cancer Models Mimic Hallmarks of Size-Induced Tumor Progression. Cancer Res 2016; 76:3732-43. [PMID: 27216179 DOI: 10.1158/0008-5472.can-15-2304] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 03/07/2016] [Indexed: 11/16/2022]
Abstract
Tumor size is strongly correlated with breast cancer metastasis and patient survival. Increased tumor size contributes to hypoxic and metabolic gradients in the solid tumor and to an aggressive tumor phenotype. Thus, it is important to develop three-dimensional (3D) breast tumor models that recapitulate size-induced microenvironmental changes and, consequently, natural tumor progression in real time without the use of artificial culture conditions or gene manipulations. Here, we developed size-controlled multicellular aggregates ("microtumors") of subtype-specific breast cancer cells by using non-adhesive polyethylene glycol dimethacrylate hydrogel microwells of defined sizes (150-600 μm). These 3D microtumor models faithfully represent size-induced microenvironmental changes, such as hypoxic gradients, cellular heterogeneity, and spatial distribution of necrotic/proliferating cells. These microtumors acquire hallmarks of tumor progression in the same cell lines within 6 days. Of note, large microtumors of hormone receptor-positive cells exhibited an aggressive phenotype characterized by collective cell migration and upregulation of mesenchymal markers at mRNA and protein level, which was not observed in small microtumors. Interestingly, triple-negative breast cancer (TNBC) cell lines did not show size-dependent upregulation of mesenchymal markers. In conclusion, size-controlled microtumor models successfully recapitulated clinically observed positive association between tumor size and aggressive phenotype in hormone receptor-positive breast cancer while maintaining clinically proven poor correlation of tumor size with aggressive phenotype in TNBC. Such clinically relevant 3D models generated under controlled experimental conditions can serve as precise preclinical models to study mechanisms involved in breast tumor progression as well as antitumor drug effects as a function of tumor progression. Cancer Res; 76(13); 3732-43. ©2016 AACR.
Collapse
Affiliation(s)
- Manjulata Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shilpaa Mukundan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Maria Jaramillo
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Steffi Oesterreich
- Women's Cancer Research Center, Magee-Womens Research Institute, University of Pittsburgh Cancer Institute, School of Medicine, Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania. Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania. McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
9
|
Jin MS, Lee H, Park IA, Chung YR, Im SA, Lee KH, Moon HG, Han W, Kim K, Kim TY, Noh DY, Ryu HS. Overexpression of HIF1α and CAXI predicts poor outcome in early-stage triple negative breast cancer. Virchows Arch 2016; 469:183-90. [PMID: 27184798 DOI: 10.1007/s00428-016-1953-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 05/03/2016] [Indexed: 11/29/2022]
Abstract
Dysregulated energy metabolism is one of the main mechanisms for uncontrolled growth in solid tumors. Hypoxia-inducible factor 1-alpha (HIF1α) is a transcription factor implicated in regulating several genes that are responsible for cell metabolism, including carbonic anhydrase IX (CAIX). The aim of this study is to determine the clinical significance of immunohistochemical metabolic alteration in early-stage triple negative breast cancer (TNBC) patients who received cyclophosphamide-based chemotherapy or radiotherapy and those with basal phenotype. Immunohistochemical staining for HIF1α and CAIX was performed to determine the correlation with clinicopathologic variables and survival outcome on tissue microarrays from 270 early-stage TNBC patients. In vitro experiments with multiple human TNBC cell lines, suppression of HIF1α by small interfering RNA (siRNA) significantly reduced CAIX protein expression in all cell lines. In multivariate analyses for different therapeutic modalities and basal phenotype, combined HIF1α and CAIX protein overexpression was significantly associated with disease-free survival in the total cohort (OR = 2.583, P = 0.002), stratified cohorts expressing basal phenotype (OR = 2.234, P = 0.021), and in those patients who received adjuvant chemotherapy (OR = 3.078, P = 0.023) and adjuvant radiotherapy (OR = 2.111, P = 0.050), respectively. In early TNBC, combined HIF1α and CAIX protein expression may serve as an unfavorable prognostic indicator particularly in patients treated with cyclophosphamide-based chemotherapy or radiotherapy as well as those with basal phenotype of breast cancer.
Collapse
Affiliation(s)
- Min-Sun Jin
- Department of Pathology, Bucheon St. Mary's Hospital, Catholic University, Gyeonggi-do, South Korea
| | - Hyebin Lee
- Department of Radiation Oncology, Kangbuk Samsung Medical Center, Seoul, South Korea
| | - In Ae Park
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea
| | - Yul Ri Chung
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Hyeong-Gon Moon
- Department of Surgery, Seoul National University Hospital, Seoul, South Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University Hospital, Seoul, South Korea
| | - Kyubo Kim
- Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Dong-Young Noh
- Department of Surgery, Seoul National University Hospital, Seoul, South Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea.
| |
Collapse
|