1
|
Lindholm A, Abrahamsen ML, Buch-Larsen K, Marina D, Andersson M, Helge JW, Schwarz P, Dela F, Gillberg L. Pro-inflammatory cytokines increase temporarily after adjuvant treatment for breast cancer in postmenopausal women: a longitudinal study. Breast Cancer Res 2024; 26:142. [PMID: 39415181 PMCID: PMC11481761 DOI: 10.1186/s13058-024-01898-3] [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: 03/05/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Breast cancer patients have an increased risk of cardiometabolic disease and for many patients, adjuvant therapy causes an altered lipid profile, insulin resistance and inflammation. Previous follow-up studies are inconclusive regarding the duration of therapy-induced inflammation. We examined the acute and persistent changes of adjuvant chemotherapy on inflammatory and metabolic health markers in breast cancer patients. METHODS Plasma levels of IL-6, IL-8, IL-10, IFN-γ, TNF-α, high-sensitivity C-reactive protein (hsCRP) and metabolic health parameters were analyzed before, shortly after and every six months up to two years after adjuvant chemotherapy treatment in 51 postmenopausal early breast cancer (EBC) patients, as well as in 41 healthy age- and BMI-matched controls. A target-specific multiplex assay was applied for cytokine measurements. RESULTS Before initiation of adjuvant therapy, plasma IL-8 levels were higher in EBC patients (31%, p = 0.0001). Also, a larger proportion of the patients had a hsCRP level above 2 mg/L (41%) compared to the controls (17%, Χ2 = 5.15, p = 0.023). Plasma levels of all five cytokines, but not hsCRP, were significantly increased after compared to before adjuvant chemotherapy (15-48% increase; all p ≤ 0.05). Already six months after ending chemotherapy treatment, all plasma cytokine levels were significantly reduced and close to pre-chemotherapy levels. Adjuvant chemotherapy caused a worsened lipid profile (increased triglycerides, lower HDL levels), insulin resistance and increased plasma insulin levels that remained high during the first year after chemotherapy. CONCLUSION Postmenopausal women with EBC have temporarily increased plasma levels of pro-inflammatory cytokines after adjuvant chemotherapy. Although transient, the therapy-induced increase in plasma cytokine levels, together with dyslipidemia and insulin resistance, may contribute to cardiometabolic risk in breast cancer patients treated with adjuvant chemotherapy. TRIAL REGISTRATION The clinical trial (registration number NCT03784651) was registered on www. CLINICALTRIALS gov on 24 December 2018.
Collapse
Affiliation(s)
- Agnes Lindholm
- Xlab, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Marie-Louise Abrahamsen
- Xlab, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | | | - Djordje Marina
- Department of Endocrinology, Rigshospitalet, 2100, Copenhagen, Denmark
| | | | - Jørn Wulff Helge
- Xlab, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Peter Schwarz
- Department of Endocrinology, Rigshospitalet, 2100, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Flemming Dela
- Xlab, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
- Department of Biochemistry and Physiology, Riga Stradins University, Riga, Latvia
| | - Linn Gillberg
- Xlab, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark.
| |
Collapse
|
2
|
Garrone O, Paccagnella M, Abbona A, Ruatta F, Vanella P, Denaro N, Tomasello G, Croce N, Barbin F, Rossino MG, La Porta CAM, Sapino A, Torri V, Albini A, Merlano MC. Moderate physical activity during neoadjuvant chemotherapy in breast cancer patients: effect on cancer-related inflammation and pathological complete response-the Neo-Runner study. ESMO Open 2024; 9:103665. [PMID: 39121813 PMCID: PMC11364046 DOI: 10.1016/j.esmoop.2024.103665] [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: 05/04/2024] [Revised: 07/06/2024] [Accepted: 07/15/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Physical activity (PA) reduces the risk of developing breast cancer (BC) and mortality rate in BC patients starting PA after diagnosis. Immunomodulation is considered responsible for these effects. However, limited data exist on the immunomodulation induced by moderate PA (mPA) during neoadjuvant chemotherapy (NACT). We have investigated the longitudinal change of cytokines during NACT alone or combined with mPA. MATERIALS AND METHODS Twenty-three cytokines were analyzed in BC patients at consecutive timepoints: at baseline (T0), before starting mPA (T1), before surgery (T2), and after surgery (T3). mPA consisted of 3-weekly brisk-walking sessions for 9-10 consecutive weeks. RESULTS Ninety-two patients were assessed: 21 patients refused mPA (untrained) and 71 agreed (trained). At T1, NACT induced significant up-regulation of interleukin (IL)-5, IL-6, IL-15, chemokine ligand (CCL)-2, interferon-γ, and C-X-C motif ligand (CXCL)-10 and reduction of expression of IL-13 and CCL-22. At T2, NACT and mPA induced up-regulation of IL-21, CCL-2, and tumor necrosis factor-α and reduction of expression of IL-8, IL-15, vascular endothelial growth factor, and soluble interleukin 6 receptor. Only CXCL-10 increased in untrained patients. A cytokine score (CS) was created to analyze, all together, the changes between T1 and T2. At T2 the CS decreased in trained and increased in untrained patients. We clustered the patients using cytokines and predictive factors and identified two clusters. The cluster A, encompassing 90% of trained patients, showed more pathological complete response (pCR) compared to the cluster B: 78% versus 22%, respectively. CONCLUSIONS mPA interacts with NACT inducing CS reduction in trained patients not observed in untrained patients, suggesting a reduction of inflammation, notwithstanding chemotherapy. This effect may contribute to the higher rate of pCR observed in the cluster A, including most trained patients.
Collapse
Affiliation(s)
- O Garrone
- Department of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan.
| | - M Paccagnella
- Translational Oncology ARCO Foundation, Cuneo. https://twitter.com/matteo_babeuf
| | - A Abbona
- Translational Oncology ARCO Foundation, Cuneo. https://twitter.com/AbbonaAndr36863
| | - F Ruatta
- Department of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan. https://twitter.com/fiorella_ruatta
| | - P Vanella
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, Cuneo
| | - N Denaro
- Department of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan
| | - G Tomasello
- Medical Oncology, ASST Ospedale Maggiore Crema, Crema. https://twitter.com/glucatom
| | - N Croce
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, Cuneo
| | - F Barbin
- Department of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan
| | - M G Rossino
- Department of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan
| | - C A M La Porta
- Center for Complexity and Biosystems, University of Milan, Milan; Department of Environmental Science and Policy, University of Milan, Milan. https://twitter.com/CAMLaPorta
| | - A Sapino
- Scientific Direction, Candiolo Cancer Institute, FPO-IRCCS Candiolo, Turin; Department of Medical Science, University of Turin, Turin. https://twitter.com/sapino58
| | - V Torri
- Mario Negri Institute for Pharmacological Research, Milan. https://twitter.com/ValterTorri
| | - A Albini
- European Institute of Oncology, Milan, Italy. https://twitter.com/adrianaalbini1
| | - M C Merlano
- Scientific Direction, Candiolo Cancer Institute, FPO-IRCCS Candiolo, Turin
| |
Collapse
|
3
|
Torkildsen CF, Austdal M, Jarmund AH, Kleinmanns K, Lamark EK, Nilsen EB, Stefansson I, Sande RK, Iversen AC, Thomsen LCV, Bjørge L. New immune phenotypes for treatment response in high-grade serous ovarian carcinoma patients. Front Immunol 2024; 15:1394497. [PMID: 38947323 PMCID: PMC11211251 DOI: 10.3389/fimmu.2024.1394497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Despite advances in surgical and therapeutic approaches, high-grade serous ovarian carcinoma (HGSOC) prognosis remains poor. Surgery is an indispensable component of therapeutic protocols, as removal of all visible tumor lesions (cytoreduction) profoundly improves the overall survival. Enhanced predictive tools for assessing cytoreduction are essential to optimize therapeutic precision. Patients' immune status broadly reflects the tumor cell biological behavior and the patient responses to disease and treatment. Serum cytokine profiling is a sensitive measure of immune adaption and deviation, yet its integration into treatment paradigms is underexplored. This study is part of the IMPACT trial (NCT03378297) and aimed to characterize immune responses before and during primary treatment for HGSOC to identify biomarkers for treatment selection and prognosis. Longitudinal serum samples from 22 patients were collected from diagnosis until response evaluation. Patients underwent primary cytoreductive surgery or neoadjuvant chemotherapy (NACT) based on laparoscopy scoring. Twenty-seven serum cytokines analyzed by Bio-Plex 200, revealed two immune phenotypes at diagnosis: Immune High with marked higher serum cytokine levels than Immune Low. The immune phenotypes reflected the laparoscopy scoring and allocation to surgical treatment. The five Immune High patients undergoing primary cytoreductive surgery exhibited immune mobilization and extended progression-free survival, compared to the Immune Low patients undergoing the same treatment. Both laparoscopy and cytoreductive surgery induced substantial and transient changes in serum cytokines, with upregulation of the inflammatory cytokine IL-6 and downregulation of the multifunctional cytokines IP-10, Eotaxin, IL-4, and IL-7. Over the study period, cytokine levels uniformly decreased in all patients, leading to the elimination of the initial immune phenotypes regardless of treatment choice. This study reveals distinct pre-treatment immune phenotypes in HGSOC patients that might be informative for treatment stratification and prognosis. This potential novel biomarker holds promise as a foundation for improved assessment of treatment responses in patients with HGSOC. ClinicalTrials.gov Identifier: NCT03378297.
Collapse
Affiliation(s)
- Cecilie Fredvik Torkildsen
- Department of Obstetrics and Gynecology, Stavanger University Hospital, Stavanger, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Marie Austdal
- Department of Research, Stavanger University Hospital, Stavanger, Norway
| | - Anders Hagen Jarmund
- Department of Clinical and Molecular Medicine, and Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Katrin Kleinmanns
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Eva Karin Lamark
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Elisabeth Berge Nilsen
- Department of Obstetrics and Gynecology, Stavanger University Hospital, Stavanger, Norway
| | - Ingunn Stefansson
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Ragnar Kvie Sande
- Department of Obstetrics and Gynecology, Stavanger University Hospital, Stavanger, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ann-Charlotte Iversen
- Department of Clinical and Molecular Medicine, and Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Liv Cecilie Vestrheim Thomsen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Line Bjørge
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
4
|
Chauhan SK, Dunn C, Andresen NK, Røssevold AH, Skorstad G, Sike A, Gilje B, Raj SX, Huse K, Naume B, Kyte JA. Peripheral immune cells in metastatic breast cancer patients display a systemic immunosuppressed signature consistent with chronic inflammation. NPJ Breast Cancer 2024; 10:30. [PMID: 38653982 DOI: 10.1038/s41523-024-00638-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 04/13/2024] [Indexed: 04/25/2024] Open
Abstract
Immunotherapies blocking the PD-1/PD-L1 checkpoint show some efficacy in metastatic breast cancer (mBC) but are often hindered by immunosuppressive mechanisms. Understanding these mechanisms is crucial for personalized treatments, with peripheral blood monitoring representing a practical alternative to repeated biopsies. In the present study, we performed a comprehensive mass cytometry analysis of peripheral blood immune cells in 104 patients with HER2 negative mBC and 20 healthy donors (HD). We found that mBC patients had significantly elevated monocyte levels and reduced levels of CD4+ T cells and plasmacytoid dendritic cells, when compared to HD. Furthermore, mBC patients had more effector T cells and regulatory T cells, increased expression of immune checkpoints and other activation/exhaustion markers, and a shift to a Th2/Th17 phenotype. Furthermore, T-cell phenotypes identified by mass cytometry correlated with functionality as assessed by IFN-γ production. Additional analysis indicated that previous chemotherapy and CDK4/6 inhibition impacted the numbers and phenotype of immune cells. From 63 of the patients, fresh tumor samples were analyzed by flow cytometry. Paired PBMC-tumor analysis showed moderate correlations between peripheral CD4+ T and NK cells with their counterparts in tumors. Further, a CD4+ T cell cluster in PBMCs, that co-expressed multiple checkpoint receptors, was negatively associated with CD4+ T cell tumor infiltration. In conclusion, the identified systemic immune signatures indicate an immune-suppressed environment in mBC patients who had progressed/relapsed on standard treatments, and is consistent with ongoing chronic inflammation. These activated immuno-suppressive mechanisms may be investigated as therapeutic targets, and for use as biomarkers of response or treatment resistance.
Collapse
Affiliation(s)
- Sudhir Kumar Chauhan
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Claire Dunn
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Nikolai Kragøe Andresen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andreas Hagen Røssevold
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gjertrud Skorstad
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Adam Sike
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bjørnar Gilje
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Sunil Xavier Raj
- Department of Oncology, St Olav University Hospital, Trondheim, Norway
| | - Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bjørn Naume
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Jon Amund Kyte
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway.
- Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway.
| |
Collapse
|
5
|
Saldajeno DP, Kawaoka S, Masuda N, Tanaka S, Bando H, Nishimura T, Kadoya T, Yamanaka T, Imoto S, Velaga RM, Tamura N, Aruga T, Ikeda K, Fukui Y, Maeshima Y, Takada M, Suzuki E, Ueno T, Ogawa S, Haga H, Ohno S, Morita S, Kawaguchi K, Toi M. Time-series blood cytokine profiles correlate with treatment responses in triple-negative breast cancer patients. Br J Cancer 2024; 130:1023-1035. [PMID: 38238427 PMCID: PMC10951271 DOI: 10.1038/s41416-023-02527-0] [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: 02/27/2023] [Revised: 10/12/2023] [Accepted: 11/27/2023] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most heterogeneous breast cancer subtype. Partly due to its heterogeneity, it is currently challenging to stratify TNBC patients and predict treatment outcomes. METHODS In this study, we examined blood cytokine profiles of TNBC patients throughout treatments (pre-treatment, during chemotherapy, pre-surgery, and 1 year after the surgery in a total of 294 samples). We analyzed the obtained cytokine datasets using weighted correlation network analyses, protein-protein interaction analyses, and logistic regression analyses. RESULTS We identified five cytokines that correlate with good clinical outcomes: interleukin (IL)-1α, TNF-related apoptosis-inducing ligand (TRAIL), Stem Cell Factor (SCF), Chemokine ligand 5 (CCL5 also known as RANTES), and IL-16. The expression of these cytokines was decreased during chemotherapy and then restored after the treatment. Importantly, patients with good clinical outcomes had constitutively high expression of these cytokines during treatments. Protein-protein interaction analyses implicated that these five cytokines promote an immune response. Logistic regression analyses revealed that IL-1α and TRAIL expression levels at pre-treatment could predict treatment outcomes in our cohort. CONCLUSION We concluded that time-series cytokine profiles in breast cancer patients may be useful for understanding immune cell activity during treatment and for predicting treatment outcomes, supporting precision medicine. TRIAL REGISTRATION The study has been registered with the University Hospital Medical Information Network Clinical Trials Registry ( http://www.umin.ac.jp/ctr/index-j.htm ) with the unique trial number UMIN000023162. The association Japan Breast Cancer Research Group trial number is JBCRG-22. The clinical outcome of the JBCRG-22 study was published in Breast Cancer Research and Treatment on 25 March 2021. https://doi.org/10.1007/s10549-021-06184-w .
Collapse
Affiliation(s)
- Don Pietro Saldajeno
- Inter-Organ Communication Research Team, Institute for Life and Medical Sciences, Kyoto University, 54 Kawahara-cho, Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
- Mathematical Informatics Laboratory, Division of Information Science, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0192, Japan
| | - Shinpei Kawaoka
- Inter-Organ Communication Research Team, Institute for Life and Medical Sciences, Kyoto University, 54 Kawahara-cho, Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Integrative Bioanalytics, Institute of Development, Aging and Cancer, Tohoku University, Sendai, 980-8575, Japan
| | - Norikazu Masuda
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Sunao Tanaka
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, 54 Kawahara-cho, Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroko Bando
- Breast and Endocrine Surgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Tomomi Nishimura
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, 54 Kawahara-cho, Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Next-generation Clinical Genomic Medicine, Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Department of Pathology and Tumor Biology, Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Takayuki Kadoya
- Department of Breast Surgery, Shimane University Hospital, Enyacho 89-1, Izumo, Shimane, 693-0021, Japan
| | - Takashi Yamanaka
- Department of Breast Surgery and Oncology, Kanagawa Cancer Center, 2-3-2 Nakano, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Shigeru Imoto
- Department of Breast Surgery, Kyorin University Hospital, 6-20-2 Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan
| | - Ravindranath M Velaga
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, 54 Kawahara-cho, Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Nobuko Tamura
- Department of Breast and Endocrine Surgery, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Tomoyuki Aruga
- Department of Breast Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Kazushi Ikeda
- Mathematical Informatics Laboratory, Division of Information Science, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0192, Japan
| | - Yukiko Fukui
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, 54 Kawahara-cho, Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yurina Maeshima
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, 54 Kawahara-cho, Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masahiro Takada
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, 54 Kawahara-cho, Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Eiji Suzuki
- Department of Breast Surgery, Kobe City Medical Center General Hospital, 2-1-1 Minatojimaminamimachi Chuo-ku, Kobe-shi, Hyogo, 650-0047, Japan
| | - Takayuki Ueno
- Breast Surgical Oncology, The Cancer Institute Hospital of JFCR, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Department of Pathology and Tumor Biology, Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Kawahara-cho, Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shinji Ohno
- Breast Oncology Center, The Cancer Institute Hospital of JFCR, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kosuke Kawaguchi
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, 54 Kawahara-cho, Shogoin Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Masakazu Toi
- Department of Breast Surgery, Kyoto University Hospital, Graduate School of Medicine, 54 Kawahara-cho, Shogoin Sakyo-ku, Kyoto, 606-8507, Japan.
- Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, 3-18-22, Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan.
| |
Collapse
|
6
|
Bessaad M, Habel A, Hadj Ahmed M, Xu W, Stayoussef M, Bouaziz H, Hachiche M, Mezlini A, Larbi A, Yaacoubi-Loueslati B. Assessing serum cytokine profiles in inflammatory breast cancer patients using Luminex® technology. Cytokine 2023; 172:156409. [PMID: 37918053 DOI: 10.1016/j.cyto.2023.156409] [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: 08/16/2023] [Revised: 10/03/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Inflammatory breast cancer (IBC), accounts for the majority of deaths associated with breast tumors. Because this form is aggressive from its appearance and has a strong metastatic potential. The majority of patients are not diagnosed until late stages, highlighting the need for the development of novel diagnostic biomarkers. Immune mediators may affect IBC progression and metastasis installation. AIM OF THE STUDY Analysis of serum proteins to identify a panel of prognostic biomarkers for IBC. PATIENTS AND METHODS Serum levels of 65 analytes were determined in IBC and Non-IBC patients with the ProcartaPlex Human Immune Monitoring 65-Plex Panel. RESULTS Fifteen analytes: 5 cytokines (IL-8, IL-16, IL-21, IL-22 and MIF), 7 chemokines (Eotaxin, eotaxin-3, Fractalkine, IP-10, MIP-1α, MIP-1β and SDF-1α), One growth factors (FGF-2) and 2 soluble receptors (TNFRII and Tweak); were significantly differentially expressed between the two groups. ROC curves showed that twelve of them (IL-8, IL-16, IL-21, IL-22, MIF, MIP-1α, MIP-1β, SDF-1α, TNFRII, FGF-2, Eotaxin-3, and Fractalkine) had AUC values greater than 0.70 and thus had potential clinical utility. Moreover, seven cytokines: IL-8, IL-16, MIF, Eotaxin-3, MIP-1α, MIP-1β, and CD-30 are positively associated with patients who developed distant metastasis. Ten analytes: Eotaxin-3, Fractalkine, IL-16, IL-1α, IL-22, IL-8, MIF, MIP-1α, MIP-1β, and TNFRII are positively associated with patients who had Lymph-Nodes invasion. CONCLUSION This study has uncovered a set of 8 analytes (Eotaxin-3, Fractalkine, IL-16, IL-8, IL-22, MIF, MIP-1α, MIP-1β) that can be used as biomarkers of IBC, and can be utilized for early detection of IBC, preventing metastasis and lymph-Nodes invasion.
Collapse
Affiliation(s)
- Maryem Bessaad
- University of Tunis El Manar (UTM), Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Tunisia
| | - Azza Habel
- University of Tunis El Manar (UTM), Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Tunisia
| | - Mariem Hadj Ahmed
- University of Tunis El Manar (UTM), Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Tunisia
| | - Weili Xu
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, Singapore 138648, Singapore
| | - Mouna Stayoussef
- University of Tunis El Manar (UTM), Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Tunisia
| | - Hanen Bouaziz
- Salah Azaiez Oncology Institute, Avenue 9 April, 1006, Bab Saadoun, Tunis, Tunisia
| | - Monia Hachiche
- Salah Azaiez Oncology Institute, Avenue 9 April, 1006, Bab Saadoun, Tunis, Tunisia
| | - Amel Mezlini
- Salah Azaiez Oncology Institute, Avenue 9 April, 1006, Bab Saadoun, Tunis, Tunisia
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, Singapore 138648, Singapore
| | - Besma Yaacoubi-Loueslati
- University of Tunis El Manar (UTM), Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Tunisia.
| |
Collapse
|
7
|
Merlano MC, Paccagnella M, Denaro N, Abbona A, Galizia D, Sangiolo D, Gammaitoni L, Fiorino E, Minei S, Bossi P, Licitra L, Garrone O. Baseline Values of Circulating IL-6 and TGF-β Might Identify Patients with HNSCC Who Do Not Benefit from Nivolumab Treatment. Cancers (Basel) 2023; 15:5257. [PMID: 37958430 PMCID: PMC10649732 DOI: 10.3390/cancers15215257] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/20/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND The immunotherapy of head and neck cancer induces a limited rate of long-term survivors at the cost of treating many patients exposed to toxicity without benefit, regardless of PD-L1 expression. The identification of better biomarkers is warranted. We analyzed a panel of cytokines, chemokines and growth factors, hereinafter all referred to as 'cytokines', as potential biomarkers in patients with head and neck cancer treated with nivolumab. MATERIALS AND METHODS A total of 18 circulating cytokines were analyzed. Samples were gathered at baseline (T0) and after 3 courses of nivolumab (T1) in patients with relapsed/metastatic disease. The data extracted at T0 were linked to survival; the comparison of T0-T1 explored the effect of immunotherapy. RESULTS A total of 22 patients were accrued: 64% current heavy smokers, 36% female and 14% had PS = 2. At T0, ROC analysis showed that IL-6, IL-8, IL-10 and TGF-β were higher in patients with poor survival. Cox analysis demonstrated that only patients with the IL-6 and TGF-β discriminate had good or poor survival, respectively. Longitudinal increments of CCL-4, IL-15, IL-2 and CXCL-10 were observed in all patients during nivolumab treatment. CONCLUSION In this small population with poor clinical characteristics, this study highlights the prognostic role of IL-6 and TGF-β. Nivolumab treatment is associated with a positive modulation of some Th1 cytokines, but it does not correlate with the outcome.
Collapse
Affiliation(s)
- Marco Carlo Merlano
- Candiolo Cancer Institute, FPO-IRCCS Candiolo, 10060 Torino, Italy; (M.C.M.); (D.G.); (L.G.)
| | | | - Nerina Denaro
- Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (N.D.); (O.G.)
| | - Andrea Abbona
- Translational Oncology ARCO Foundation, 12100 Cuneo, Italy;
| | - Danilo Galizia
- Candiolo Cancer Institute, FPO-IRCCS Candiolo, 10060 Torino, Italy; (M.C.M.); (D.G.); (L.G.)
| | - Dario Sangiolo
- Department of Oncology, University of Turin, 10060 Torino, Italy; (D.S.); (E.F.)
| | - Loretta Gammaitoni
- Candiolo Cancer Institute, FPO-IRCCS Candiolo, 10060 Torino, Italy; (M.C.M.); (D.G.); (L.G.)
| | - Erika Fiorino
- Department of Oncology, University of Turin, 10060 Torino, Italy; (D.S.); (E.F.)
| | - Silvia Minei
- Post-Graduate School of Specialization Medical Oncology, University of Bari “A. Moro”, 70120 Bari, Italy;
- Medical Oncology, A.U.O. Consorziale Policlinico di Bari, 70120 Bari, Italy
| | - Paolo Bossi
- Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences, Public Health, University of Brescia, 25123 Brescia, Italy;
| | - Lisa Licitra
- Fondazione IRCCS Istituto Nazionale dei Tumori, University of Milan, 20133 Milan, Italy;
| | - Ornella Garrone
- Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (N.D.); (O.G.)
| |
Collapse
|
8
|
Iwata H, Nakamura R, Masuda N, Yamashita T, Yamamoto Y, Kobayashi K, Tsurutani J, Iwasa T, Yonemori K, Tamura K, Aruga T, Tokunaga E, Kaneko K, Lee MJ, Yuno A, Kawabata A, Seike T, Kaneda A, Nishimura Y, Trepel JB, Saji S. Efficacy and exploratory biomarker analysis of entinostat plus exemestane in advanced or recurrent breast cancer: phase II randomized controlled trial. Jpn J Clin Oncol 2023; 53:4-15. [PMID: 36398439 PMCID: PMC9825728 DOI: 10.1093/jjco/hyac166] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 10/07/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND We aimed to confirm the efficacy and safety of the oral histone deacetylase inhibitor entinostat in Japanese patients with hormone receptor-positive advanced/recurrent breast cancer and to explore potential biomarkers. METHODS This phase II, double-blind, randomized, placebo-controlled trial (ClinicalTrials.gov; NCT03291886) was conducted at 28 Japanese sites (September 2017-July 2020; interim analysis cutoff: April 2019). Patients with progression/relapse following non-steroidal aromatase inhibitors were randomized 1:1 to entinostat (5 mg/week) or placebo, plus exemestane (25 mg/day). Primary endpoint was progression-free survival; secondary endpoints included overall survival and safety. Exploratory biomarker outcomes included lysine acetylation, immune cell profiles, estrogen receptor 1 mutations and plasma chemokines. RESULTS Of 133 randomized patients, 131 (65 entinostat, 66 placebo) who received study drug were analyzed. Median (95% confidence interval) progression-free survival was 5.8 (3.2-7.8) months for entinostat and 3.3 (3.1-5.8) months for placebo (hazard ratio [95% confidence interval]: 0.75 [0.50 - 1.14]; P = 0.189). Median overall survival was not reached in either group. Entinostat tended to prolong progression-free survival in patients aged ≥65 years, not endocrine resistant, or with estrogen receptor 1 Y537S mutation. Candidate biomarkers of efficacy (progression-free survival) included lysine acetylation in CD3+ cells, plasma interferon gamma-induced protein 10, dendritic cell CD86 expression, and CD4+ cell expression of human leukocyte antigen-DR and inducible T-cell co-stimulator. Safety was similar to non-Japanese populations; however, seven entinostat-treated patients (10.8%) had reversible lung injury. CONCLUSIONS In Japanese patients, the safety of entinostat plus exemestane was acceptable and progression-free survival was prolonged, although not significantly. Exploratory analyses identified potential biomarkers, including lysine acetylation, of efficacy.
Collapse
Affiliation(s)
- Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Hospital, Aichi, Japan
| | - Rikiya Nakamura
- Division of Breast Surgery, Chiba Cancer Center, Chiba, Japan
| | - Norikazu Masuda
- Department of Surgery, Breast Oncology, National Hospital Organization, Osaka National Hospital, Osaka, Japan
| | - Toshinari Yamashita
- Department of Breast and Endocrine Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Yutaka Yamamoto
- Department of Breast and Endocrine Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kokoro Kobayashi
- Department of Breast Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Junji Tsurutani
- Advanced Cancer Translational Research Institute, Showa University, Tokyo, Japan
| | - Tsutomu Iwasa
- Advanced Cancer Translational Research Institute, Showa University, Tokyo, Japan
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kenji Tamura
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tomoyuki Aruga
- Department of Breast Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Eriko Tokunaga
- Department of Breast Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Koji Kaneko
- Department of Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Akira Yuno
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Ayumi Kaneda
- R&D Division, Kyowa Kirin Co., Ltd, Tokyo, Japan
| | | | - Jane B Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shigehira Saji
- Department of Medical Oncology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
9
|
Ye HM, Lu MJ, Liu Q, Lin Y, Tang LY, Ren ZF. Beneficial Effect of Toxoplasma gondii Infection on the Prognosis of Breast Cancer Was Modified by Cytokines. Clin Epidemiol 2023; 15:469-481. [PMID: 37122480 PMCID: PMC10145453 DOI: 10.2147/clep.s408182] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/11/2023] [Indexed: 05/02/2023] Open
Abstract
Background Animal experiments have shown the anticancer activity of Toxoplasma gondii (T. gondii), but its effect on the prognosis of cancer patients is unclear. Thus, the present study aimed to investigate the prognostic role of anti-T. gondii IgG in breast cancer patients and the modification effect of cytokines. Methods A total of 1121 breast cancer patients were recruited between 2008 and 2018 and followed up until December 31, 2021. Anti-T. gondii IgG and cytokines were measured using an enzyme-linked immunosorbent assay (ELISA) kit and a multiplex assay platform. Endpoints were overall survival (OS) and progression-free survival (PFS). Survival and multiplicative interaction analyses were performed using multivariate Cox regression models. Results According to the cutoff value of optical density (OD=0.111), 900 (80.29%) and 221 (19.71%) patients were divided into two groups: low or high anti-T. gondii IgG. Compared to patients with a low anti-T. gondii IgG level, the adjusted hazard ratios (HRs) of OS and PFS for patients with high anti-T. gondii IgG levels were 0.60 (95% confidence interval (CI): 0.37-0.99) and 0.67 (0.46-0.98), respectively. These associations were profound among patients with a high cytokine score (HR=0.29, 95% CI: 0.10-0.82 for OS; HR=0.30, 95% CI: 0.13-0.69 for PFS), accompanied by a significant interaction between the level of anti-T. gondii IgG and the cytokine score (P interaction=0.019 for PFS); interleukin-17 (IL-17) and interleukin-9 (IL-9) were the main contributors to the interaction. Conclusion Anti-T. gondii IgG was found to be beneficial to breast cancer survival, especially in women with systematic inflammation and high IL-17 or IL-9 levels, suggesting the potential of T. gondii as a prognostic marker and a novel immunotherapy approach for cancer patients.
Collapse
Affiliation(s)
- Heng-Ming Ye
- The School of Public Health, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
| | - Min-Jie Lu
- The School of Public Health, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
| | - Qiang Liu
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
| | - Ying Lin
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
| | - Lu-Ying Tang
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, People’s Republic of China
| | - Ze-Fang Ren
- The School of Public Health, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
- Correspondence: Ze-Fang Ren, The School of Public Health, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, People’s Republic of China, Tel/Fax +86-20-87332577, Email
| |
Collapse
|
10
|
Lee MH, Laajala E, Kreutzman A, Järvinen P, Nísen H, Mirtti T, Hollmén M, Mustjoki S. The tumor and plasma cytokine profiles of renal cell carcinoma patients. Sci Rep 2022; 12:13416. [PMID: 35927313 PMCID: PMC9352752 DOI: 10.1038/s41598-022-17592-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/27/2022] [Indexed: 11/16/2022] Open
Abstract
Renal cell carcinoma (RCC) accounts for 90% of all renal cancers and is considered highly immunogenic. Although many studies have reported the circulating peripheral cytokine profiles, the signatures between the tumor tissue and matching healthy adjacent renal tissue counterparts have not been explored. We aimed to comprehensively investigate the cytokine landscape of RCC tumors and its correlation between the amount and phenotype of the tumor infiltrating lymphocytes (TILs). We analyzed the secretion of 42 cytokines from the tumor (n = 46), adjacent healthy kidney tissues (n = 23) and matching plasma samples (n = 33) with a Luminex-based assay. We further explored the differences between the tissue types, as well as correlated the findings with clinical data and detailed immunophenotyping of the TILs. Using an unsupervised clustering approach, we observed distinct differences in the cytokine profiles between the tumor and adjacent renal tissue samples. The tumor samples clustered into three distinct profiles based on the cytokine expressions: high (52.2% of the tumors), intermediate (26.1%), and low (21.7%). Most of the tumor cytokines positively correlated with each other, except for IL-8 that showed no correlation with any of the measured cytokine expressions. Furthermore, the quantity of lymphocytes in the tumor samples analyzed with flow cytometry positively correlated with the chemokine-family of cytokines, CXCL10 (IP-10) and CXCL9 (MIG). No significant correlations were found between the tumor and matching plasma cytokines, suggesting that circulating cytokines poorly mirror the tumor cytokine environment. Our study highlights distinct cytokine profiles in the RCC tumor microenvironment and provides insights to potential biomarkers for the treatment of RCC.
Collapse
Affiliation(s)
- Moon Hee Lee
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Haartmaninkatu 8, N00290, Helsinki, Finland.,Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Essi Laajala
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Haartmaninkatu 8, N00290, Helsinki, Finland.,Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Anna Kreutzman
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Haartmaninkatu 8, N00290, Helsinki, Finland.,Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Petrus Järvinen
- Abdominal Center, Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Harry Nísen
- Abdominal Center, Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tuomas Mirtti
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Maija Hollmén
- Medicity Research Laboratory, University of Turku, Turku, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Haartmaninkatu 8, N00290, Helsinki, Finland. .,Translational Immunology Research Program, University of Helsinki, Helsinki, Finland. .,iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| |
Collapse
|
11
|
Plasma profile of immune determinants predicts pathological complete response in locally advanced breast cancer patients: a pilot study. Clin Breast Cancer 2022; 22:705-714. [DOI: 10.1016/j.clbc.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 05/02/2022] [Accepted: 05/19/2022] [Indexed: 11/24/2022]
|
12
|
Chen Q, Wei Z, Wang L, Xu X, Wei Z, Zheng P, Cao K, Zhang Z, Chen K, Liang Q. Dry Eye Disease in Patients With Schizophrenia: A Case-Control Study. Front Med (Lausanne) 2022; 9:831337. [PMID: 35223927 PMCID: PMC8864171 DOI: 10.3389/fmed.2022.831337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022] Open
Abstract
Objective To evaluate the clinical features and inflammatory cytokines of dry eye disease (DED) in patients with schizophrenia. Methods This is a case-control study. The modified self-rating depression scale (M-SDS) and the ocular surface disease index (OSDI) were used to evaluate the symptoms of depression and DED, respectively. Lipid layer thickness (LLT), partial blink rate (PBR), meibomian gland loss (MGL), tear break-up time (TBUT), corneal fluorescein staining, Schirmer I-test, and eyelid margin abnormalities were also measured. A multiplex ELISA Quantibody array was used to detect the inflammatory cytokines in the tears of all participants. Results Forty schizophrenic patients and 20 control subjects were included. The mean age was 45.0 ± 9.5 years (range, 22–63 years) in schizophrenic patients and 45.4 ± 16.2 years (range, 23–76 years) in controls (P = 0.914). The ratio of male to female was 1.1 in schizophrenic patients and 1.0 in controls (P = 0.914). Ten women (52.6%) with schizophrenia and 2 (20%) in the control group (P = 0.096) were menopausal or post-menopausal. The OSDI [0.0 (0.0–4.2) vs. 7.3 (2.1–14.6)] and TBUT [4.5 (3.0–6.0) vs. 10.0 (3.5–11.0)] were significantly lower in patients with schizophrenia than in controls (P = 0.003 and P = 0.009, respectively). The rate of MGL [36.5 (17.5–47.5) vs. 8.5 (0.0–17.5)] increased in schizophrenic patients (P < 0.001). Among pro-inflammatory cytokines, the levels of interleukin (IL)-1α, IL-6, IL-11, IL-12A, IL-15, IL-17A, and granulocyte colony-stimulating factor (G-CSF) in tears were elevated in the schizophrenia group (all P < 0.01). Most of the chemokines examined were at increased levels in the tears of schizophrenics (all P < 0.05). The levels of matrix metalloproteinases-9 (MMP-9) and intercellular adhesion molecule-1 (ICAM-1) were also higher in the schizophrenic patients (all P < 0.001). The concentrations of IL-1Ra, tissue-inhibitor of metalloproteinase-1 (TIMP-1), and TIMP-2 in the schizophrenia group were decreased (all P < 0.001). In schizophrenic patients, the level of CCL2 in tears was positively correlated with OSDI (R = 0.34, P = 0.03). The increasing TIMP-1 and decreasing IL-5 were correlated with increasing LLT (R = 0.33, P = 0.035; R = −0.35, P = 0.027, respectively). The level of ICAM-1 was then positively correlated with partial blink rate (PBR) (R = 0.33, P = 0.035). There was a negative correlation between IL-8 and the Schirmer I-test (R = −0.41, P = 0.009). Conclusions Patients with schizophrenia were more likely to experience asymptomatic DED, with mild symptoms and obvious signs. The inflammatory cytokines in the tears of schizophrenic patients differed greatly from that of non-schizophrenic patients.
Collapse
Affiliation(s)
- Qiankun Chen
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Zhengjiang Wei
- Beijing Miyun Mental Health Prevention and Treatment Hospital, Beijing, China
| | - Leying Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Xizhan Xu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Zhenyu Wei
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Panpan Zheng
- National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Kai Cao
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Zijun Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Kexin Chen
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Qingfeng Liang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| |
Collapse
|
13
|
Paccagnella M, Abbona A, Michelotti A, Geuna E, Ruatta F, Landucci E, Denaro N, Vanella P, Lo Nigro C, Galizia D, Merlano M, Garrone O. Circulating Cytokines in Metastatic Breast Cancer Patients Select Different Prognostic Groups and Patients Who Might Benefit from Treatment beyond Progression. Vaccines (Basel) 2022; 10:78. [PMID: 35062739 PMCID: PMC8781714 DOI: 10.3390/vaccines10010078] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/28/2021] [Accepted: 01/03/2022] [Indexed: 12/28/2022] Open
Abstract
Cancer induces immune suppression to overcome its recognition and eradication by the immune system. Cytokines are messengers able to modulate immune response or suppression. There is great interest in the evaluation of their changes during treatment in order to identify their relationship with clinical outcome. We evaluated 18 cytokines in breast cancer patients treated with eribulin before starting treatment (T0) and after four courses of therapy (T1). Longitudinal modifications were considered and cytokine clusters through PCA and HCPC correlated to patients' outcomes were identified. Forty-one metastatic breast cancer patients and fifteen healthy volunteers were included. After clustering, we identified at T0 six patient clusters with different risk of relapse and death. At T1, only four clusters were identified, and three of them accounted for thirty-eight of forty-one patients, suggesting a possible role of treatment in reducing heterogeneity. The cluster with the best survival at T1 was characterized by low levels of IL-4, IL-6, IL-8, IL-10, CCL-2, CCL-4, and TGF-β. The cluster showing the worst survival encompassed high levels of IL-4, IL-6, IL-8, IL-10, CCL-2, and IFN-γ. A subgroup of patients with short progression-free survival (PFS) and long overall survival (OS) was comprised in the cluster characterized by low levels of CCL-2, IL-6, IL-8, IL-10, and IL-12 at T0. Our data support the prognostic significance of longitudinal serum cytokine analysis. This approach may help identify patients for whom early treatment stop avoids needless toxicity or might justify treatment beyond early progression. Further investigations are required to validate this hypothesis.
Collapse
Affiliation(s)
| | - Andrea Abbona
- Translational Oncology ARCO Foundation, 12100 Cuneo, Italy;
| | - Andrea Michelotti
- Department of Medical Oncology, Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy; (A.M.); (E.L.)
| | - Elena Geuna
- Multidisciplinary Oncology Outpatient Clinic, Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo, Italy; (E.G.); (D.G.)
| | - Fiorella Ruatta
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, 12100 Cuneo, Italy; (F.R.); (N.D.); (P.V.)
| | - Elisabetta Landucci
- Department of Medical Oncology, Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy; (A.M.); (E.L.)
| | - Nerina Denaro
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, 12100 Cuneo, Italy; (F.R.); (N.D.); (P.V.)
| | - Paola Vanella
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, 12100 Cuneo, Italy; (F.R.); (N.D.); (P.V.)
| | | | - Danilo Galizia
- Multidisciplinary Oncology Outpatient Clinic, Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo, Italy; (E.G.); (D.G.)
| | - Marco Merlano
- Experimental Cell Therapy Lab, Candiolo Cancer Institute, FPO-IRCCS Candiolo, 10060 Torino, Italy;
| | - Ornella Garrone
- Department of Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Policlinico Maggiore, 20122 Milano, Italy;
| |
Collapse
|
14
|
Sirven P, Faucheux L, Grandclaudon M, Michea P, Vincent-Salomon A, Mechta-Grigoriou F, Scholer-Dahirel A, Guillot-Delost M, Soumelis V. Definition of a novel breast tumor-specific classifier based on secretome analysis. Breast Cancer Res 2022; 24:94. [PMID: 36539890 PMCID: PMC9764559 DOI: 10.1186/s13058-022-01590-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND During cancer development, the normal tissue microenvironment is shaped by tumorigenic events. Inflammatory mediators and immune cells play a key role during this process. However, which molecular features most specifically characterize the malignant tissue remains poorly explored. METHODS Within our institutional tumor microenvironment global analysis (T-MEGA) program, we set a prospective cohort of 422 untreated breast cancer patients. We established a dedicated pipeline to generate supernatants from tumor and juxta-tumor tissue explants and quantify 55 soluble molecules using Luminex or MSD. Those analytes belonged to five molecular families: chemokines, cytokines, growth factors, metalloproteinases, and adipokines. RESULTS When looking at tissue specificity, our dataset revealed some breast tumor-specific characteristics, as IL-16, as well as some juxta-tumor-specific secreted molecules, as IL-33. Unsupervised clustering analysis identified groups of molecules that were specific to the breast tumor tissue and displayed a similar secretion behavior. We identified a tumor-specific cluster composed of nine molecules that were secreted fourteen times more in the tumor supernatants than the corresponding juxta-tumor supernatants. This cluster contained, among others, CCL17, CCL22, and CXCL9 and TGF-β1, 2, and 3. The systematic comparison of tumor and juxta-tumor secretome data allowed us to mathematically formalize a novel breast cancer signature composed of 14 molecules that segregated tumors from juxta-tumors, with a sensitivity of 96.8% and a specificity of 96%. CONCLUSIONS Our study provides the first breast tumor-specific classifier computed on breast tissue-derived secretome data. Moreover, our T-MEGA cohort dataset is a freely accessible resource to the biomedical community to help advancing scientific knowledge on breast cancer.
Collapse
Affiliation(s)
- Philémon Sirven
- grid.418596.70000 0004 0639 6384INSERM Unit U932, Immunity and Cancer, Institut Curie, Paris, France ,grid.440907.e0000 0004 1784 3645Paris Sciences Lettres (PSL) University, Paris, France ,Center of Clinical Investigation, CIC IGR-Curie 1428, Paris, France
| | - Lilith Faucheux
- INSERM U976, Université de Paris, IRSLHôpital Saint Louis, 75006 Paris, France ,INSERM UMR1153, Université de Paris, ECSTRRA Team, 75006 Paris, France
| | - Maximilien Grandclaudon
- grid.418596.70000 0004 0639 6384INSERM Unit U932, Immunity and Cancer, Institut Curie, Paris, France ,grid.440907.e0000 0004 1784 3645Paris Sciences Lettres (PSL) University, Paris, France ,Center of Clinical Investigation, CIC IGR-Curie 1428, Paris, France
| | - Paula Michea
- grid.418596.70000 0004 0639 6384INSERM Unit U932, Immunity and Cancer, Institut Curie, Paris, France ,grid.440907.e0000 0004 1784 3645Paris Sciences Lettres (PSL) University, Paris, France ,Center of Clinical Investigation, CIC IGR-Curie 1428, Paris, France
| | - Anne Vincent-Salomon
- grid.440907.e0000 0004 1784 3645Paris Sciences Lettres (PSL) University, Paris, France ,grid.418596.70000 0004 0639 6384Diagnostic and Theranostic Medicine Division, Institut Curie, Paris, France
| | - Fatima Mechta-Grigoriou
- grid.440907.e0000 0004 1784 3645Paris Sciences Lettres (PSL) University, Paris, France ,grid.418596.70000 0004 0639 6384Centre de Recherche, Stress and Cancer Laboratory, U830 Genetics and Biology of Cancers, INSERM, Institut Curie, Paris, France
| | - Alix Scholer-Dahirel
- grid.418596.70000 0004 0639 6384INSERM Unit U932, Immunity and Cancer, Institut Curie, Paris, France ,grid.440907.e0000 0004 1784 3645Paris Sciences Lettres (PSL) University, Paris, France ,Center of Clinical Investigation, CIC IGR-Curie 1428, Paris, France
| | - Maude Guillot-Delost
- grid.418596.70000 0004 0639 6384INSERM Unit U932, Immunity and Cancer, Institut Curie, Paris, France ,grid.440907.e0000 0004 1784 3645Paris Sciences Lettres (PSL) University, Paris, France ,Center of Clinical Investigation, CIC IGR-Curie 1428, Paris, France
| | - Vassili Soumelis
- grid.418596.70000 0004 0639 6384INSERM Unit U932, Immunity and Cancer, Institut Curie, Paris, France ,grid.440907.e0000 0004 1784 3645Paris Sciences Lettres (PSL) University, Paris, France ,Center of Clinical Investigation, CIC IGR-Curie 1428, Paris, France ,INSERM U976, Université de Paris, IRSLHôpital Saint Louis, 75006 Paris, France ,grid.413328.f0000 0001 2300 6614Department of Immunology-Histocompatibility, AP-HP, Hôpital Saint-Louis, 75010 Paris, France
| |
Collapse
|
15
|
Zemlin C, Stuhlert C, Schleicher JT, Wörmann C, Altmayer L, Lang M, Scherer LS, Thul IC, Müller C, Kaiser E, Stutz R, Goedicke-Fritz S, Ketter L, Zemlin M, Wagenpfeil G, Steffgen G, Solomayer EF. Longitudinal Assessment of Physical Activity, Fitness, Body Composition, Immunological Biomarkers, and Psychological Parameters During the First Year After Diagnosis in Women With Non-Metastatic Breast Cancer: The BEGYN Study Protocol. Front Oncol 2021; 11:762709. [PMID: 34737966 PMCID: PMC8560964 DOI: 10.3389/fonc.2021.762709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/01/2021] [Indexed: 12/24/2022] Open
Abstract
Background Moderate physical activity is associated with an improved prognosis and psychosocial outcome in breast cancer patients. Although exercise and physical activity are associated with multiple physiological and psychological effects, many of the underlying mechanisms remain obscure. The BEGYN study (Influence of physical activity in breast cancer patients on physiological and psychological parameters and on biomarkers) aims at identifying potential associations between the extent of physical activity, fitness, body composition, immunological biomarkers, psycho-emotional parameters, and the course of treatment during the first year after diagnosis of breast cancer. Methods The prospective observational BEGYN study will include 110 non-metastatic breast cancer patients. The patients will be assessed during a base line visit prior to the initiation of the antineoplastic therapy and after 3, 6, 9 and 12 months. The physical activity will be measured using a fitness tracker and a self-assessment diary during the entire study. Each visit will include the assessment of (i) cardiorespiratory fitness measured by spiroergometry, (ii) body composition, (iii) psycho-emotional parameters (quality of life, mental health, fatigue, depression, distress, anxiety, well-being), and (iv) extensive blood tests including routine laboratory, vitamin D, selenium and immunologically relevant biomarkers (e.g., leukocyte subpopulations and cytokine profiles). Discussion Whereas most studies investigating the influence of physical activity in breast cancer patients focus on specific activities for three months or less, the BEGYN study will quantify the daily physical activity and cardiorespiratory fitness of breast cancer patients based on objective measurements in the context of the oncological therapy for 12 months after diagnosis. The study will reveal potential associations between exercise, immune status and physical as well as psycho-emotional outcome and the clinical course of the disease. Moreover, complementary therapies such as Vit D and Selenium supplementation and parameters investigating the motivation of the patients are part of the study. Due to this holistic approach, the BEGYN study will guide towards confirmatory studies on the role of physical activity in breast cancer patients to develop individualized counselling regarding the recommended type and extent of exercise. Trial Registration This study has been registered at the German Clinical Trials Register DRKS00024829.
Collapse
Affiliation(s)
- Cosima Zemlin
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Homburg, Germany
| | - Caroline Stuhlert
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Homburg, Germany
| | - Julia Theresa Schleicher
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Homburg, Germany
| | - Carolin Wörmann
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Homburg, Germany
| | - Laura Altmayer
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Homburg, Germany
| | - Marina Lang
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Homburg, Germany
| | - Laura-Sophie Scherer
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Homburg, Germany
| | - Ida Clara Thul
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Homburg, Germany
| | - Carolin Müller
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Homburg, Germany
| | - Elisabeth Kaiser
- Department for General Pediatrics, Saarland University Medical Center, Homburg, Germany
| | - Regine Stutz
- Department for General Pediatrics, Saarland University Medical Center, Homburg, Germany
| | | | - Laura Ketter
- Department of Behavioural and Cognitive Sciences, Institute for Health and Behaviour, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Michael Zemlin
- Department for General Pediatrics, Saarland University Medical Center, Homburg, Germany
| | - Gudrun Wagenpfeil
- Institute for Medical Biometry, Epidemiology and Medical Informatics (IMBEI), Saarland University, Homburg, Germany
| | - Georges Steffgen
- Department of Behavioural and Cognitive Sciences, Institute for Health and Behaviour, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Erich-Franz Solomayer
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
16
|
Al-Rashidi HE, Refaat S, Ahmed E, Hussein DT, Eltantawy FM, Hamed S. Involvement of INF-γ functional single nucleotide polymorphism +874 T/A (rs2430561) in breast cancer risk. Saudi J Biol Sci 2021; 28:6289-6296. [PMID: 34759748 PMCID: PMC8568710 DOI: 10.1016/j.sjbs.2021.06.083] [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: 04/13/2021] [Revised: 06/27/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023] Open
Abstract
According Global Cancer Statistics 2020 GLOBOCAN estimates female breast cancer was found as the most commonly diagnosed cancer, with an estimated 2.3 million new cases (11.7%), and the fourth leading cause (6.9%) of cancer death among women worldwide. Identification of new diagnostic marker sharply characterize the tumor feature is intensive need. The present work was performed to investigate the involvement of the INF-γ + 874 T/A gene polymorphism in different breast cancer prognostic factors. Polymorphism detection analysis was performed on 163 subjects from breast cancer patients, 79 with inflamed cells of breast patients and 144 controls. The gene polymorphism was detected using the amplification refractory mutation system- polymerase chain reaction method (ARMS-PCR). The distribution of INF-γ T + 874A gene polymorphism shows strong significant association between INF-γ + 874 T/A genotypes TT in BC patients (ORTT: 6.41 [95% CI = 2.72-15.1] P < 0.0001) as well as strong significant association regarding T allele (ORT: 1.99 [95% CI = 1.43-2.76] P < 0.0001) when compared to the healthy control. In ICB group the strong association was noted with INF-γ + 874 T/A genotypes AT genotype (ORAT: 2.28 [95% CI = 1.22-4.29] P = 0.007). From the different histological BC hormonal markers the human epidermal growth factor receptor 2 (HER2) was showing significant association in INF-γ + 874 T/A genotypes TT (P = 0.03) and recessive model (TT versus AA + AT P = 0.03). Concerning different BC prognostic models, the poor prognostic one of luminal B, (ER+ve PR+ve Her2+ve) show significant association in the host INF-γ + 874 T/A genotype (TT, P = 0.03) and recessive model (TT versus AA + AT P = 0.02) when compared to the good prognostic hormonal status luminal A model, (ER+ve PR+ve Her2-ve). It seems that this is the first study that interested in correlate the INF-γ + 874 T/A gene polymorphisms in Egyptian BC patients. T allele, TT genotype and recessive model of the INF-γ + 874 T/A gene variants were documented as risk factors for BC pathogenesis. It may be used as practical biomarker to guide the BC carcinogenesis and risk process.
Collapse
Key Words
- ARMS-PCR, amplification refractory mutation system, polymerase chain reaction method
- BC, Breast cancer
- Breast cancer
- C, controls
- CD, cluster of differentiation
- CI, 95% confidence intervals
- ER, estrogen receptor
- GPI, good prognostic index
- Genotypes
- HER2, human epidermal growth factor receptor 2
- ICB, inflamed cells of breast
- IL, interleukin
- INF-γ
- INF-γ, Interferon-γ
- IRB, Institutional Review Board
- ISGs, INF-stimulated genes
- MPI, moderate prognostic index
- NK, natural killer cells
- NPI, the mandatory prognostic index
- OR, odds ratio
- PAM50, Prediction Analysis of Microarray 50
- PPI, poor prognostic index
- PR, progesterone receptor
- Polymorphism
- Risk factor
- SNPs, single nucleotide polymorphism
- TGF-β, transforming growth factor-β
- TNBC, Triple Negative BC
- TNF-α, tumor necrosis factor-α
- Th1, T helper1
Collapse
Affiliation(s)
- Hanan E Al-Rashidi
- Medical Laboratory Technology Department, College of Applied Medical Science, Taibah University, Madinah, Saudi Arabia
| | | | - Enas Ahmed
- Emergency Hospital, Mansoura University, Egypt
| | | | | | - Sahar Hamed
- Urology and Nephrology Center, Mansoura University, Egypt
| |
Collapse
|
17
|
Kochumon S, Al-Sayyar A, Jacob T, Hasan A, Al-Mulla F, Sindhu S, Ahmad R. TNF-α Increases IP-10 Expression in MCF-7 Breast Cancer Cells via Activation of the JNK/c-Jun Pathways. Biomolecules 2021; 11:biom11091355. [PMID: 34572567 PMCID: PMC8464892 DOI: 10.3390/biom11091355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 12/28/2022] Open
Abstract
IP-10 (also called CXCL10) plays a significant role in leukocyte homing to inflamed tissues, and increased IP-10 levels are associated with the pathologies of various inflammatory disorders, including type 2 diabetes, atherosclerosis, and cancer. TNF-α is a potent activator of immune cells and induces inflammatory cytokine expression in these cells. However, it is unclear whether TNF-α is able to induce IP-10 expression in MCF-7 breast cancer cells. We therefore determined IP-10 expression in TNF-α-treated MCF-7 cells and investigated the mechanism involved. Our data show that TNF-α induced/upregulated the IP-10 expression at both mRNA and protein levels in MCF-7 cells. Inhibition of JNK (SP600125) significantly suppressed the TNF-α-induced IP-10 in MCF-7 cells, while the inhibition of p38 MAPK (SB203580), MEK1/2 (U0126), and ERK1/2 (PD98059) had no significant effect. Furthermore, TNF-α-induced IP-10 expression was abolished in MCF-7 cells deficient in JNK. Similar results were obtained using MCF-7 cells deficient in c-Jun. Moreover, the JNK kinase inhibitor markedly reduced the TNF-α-induced JNK and c-Jun phosphorylation. The kinase activity of JNK induced by TNF-α stimulation of MCF-7 cells was significantly inhibited by SP600125. Altogether, our novel findings provide the evidence that TNF-α induces IP-10 expression in MCF-7 breast cancer cells via activation of the JNK/c-Jun signaling pathway.
Collapse
Affiliation(s)
- Shihab Kochumon
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
| | - Amnah Al-Sayyar
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
| | - Texy Jacob
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
| | - Amal Hasan
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
| | - Fahd Al-Mulla
- Genetics & Bioinformatics Department, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Sardar Sindhu
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
- Animal and Imaging Core Facility, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
- Correspondence:
| |
Collapse
|
18
|
Dymicka-Piekarska V, Koper-Lenkiewicz OM, Zińczuk J, Kratz E, Kamińska J. Inflammatory cell-associated tumors. Not only macrophages (TAMs), fibroblasts (TAFs) and neutrophils (TANs) can infiltrate the tumor microenvironment. The unique role of tumor associated platelets (TAPs). Cancer Immunol Immunother 2021; 70:1497-1510. [PMID: 33146401 PMCID: PMC8139882 DOI: 10.1007/s00262-020-02758-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022]
Abstract
It is well known that various inflammatory cells infiltrate cancer cells. Next to TAMs (tumor-associated macrophages), TAFs (tumor-associated fibroblasts) and TANs (tumor-associated neutrophils) also platelets form the tumor microenvironment. Taking into account the role of platelets in the development of cancer, we have decided to introduce a new term: tumor associated platelets-TAPs. To the best of our knowledge, thus far this terminology has not been employed by anyone. Platelets are the first to appear at the site of the inflammatory process that accompanies cancer development. Within the first few hours from the start of the colonization of cancer cells platelet-tumor aggregates are responsible for neutrophils recruitment, and further release a number of factors associated with tumor growth, metastasis and neoangiogenesis. On the other hand, it also has been indicated that factors delivered from platelets can induce a cytotoxic effect on the proliferating neoplastic cells, and even enhance apoptosis. Undoubtedly, TAPs' role seems to be more complex when compared to tumor associated neutrophils and macrophages, which do not allow for their division into TAP P1 and TAP P2, as in the case of TANs and TAMs. In this review we discuss the role of TAPs as an important element of tumor invasiveness and as a potentially new therapeutic target to prevent cancer development. Nevertheless, better exploring the interactions between platelets and tumor cells could help in the formulation of new therapeutic goals that support or improve the effectiveness of cancer treatment.
Collapse
Affiliation(s)
- Violetta Dymicka-Piekarska
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| | - Olga M. Koper-Lenkiewicz
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| | - Justyna Zińczuk
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| | - Ewa Kratz
- Department of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wrocław, Poland
| | - Joanna Kamińska
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| |
Collapse
|
19
|
Pharmacological inhibition of MDA-9/Syntenin blocks breast cancer metastasis through suppression of IL-1β. Proc Natl Acad Sci U S A 2021; 118:2103180118. [PMID: 34016751 PMCID: PMC8166168 DOI: 10.1073/pnas.2103180118] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Melanoma differentiation associated gene-9 (MDA-9), Syntenin-1, or syndecan binding protein is a differentially regulated prometastatic gene with elevated expression in advanced stages of melanoma. MDA-9/Syntenin expression positively associates with advanced disease stage in multiple histologically distinct cancers and negatively correlates with patient survival and response to chemotherapy. MDA-9/Syntenin is a highly conserved PDZ-domain scaffold protein, robustly expressed in a spectrum of diverse cancer cell lines and clinical samples. PDZ domains interact with a number of proteins, many of which are critical regulators of signaling cascades in cancer. Knockdown of MDA-9/Syntenin decreases cancer cell metastasis, sensitizing these cells to radiation. Genetic silencing of MDA-9/Syntenin or treatment with a pharmacological inhibitor of the PDZ1 domain, PDZ1i, also activates the immune system to kill cancer cells. Additionally, suppression of MDA-9/Syntenin deregulates myeloid-derived suppressor cell differentiation via the STAT3/interleukin (IL)-1β pathway, which concomitantly promotes activation of cytotoxic T lymphocytes. Biologically, PDZ1i treatment decreases metastatic nodule formation in the lungs, resulting in significantly fewer invasive cancer cells. In summary, our observations indicate that MDA-9/Syntenin provides a direct therapeutic target for mitigating aggressive breast cancer and a small-molecule inhibitor, PDZ1i, provides a promising reagent for inhibiting advanced breast cancer pathogenesis.
Collapse
|
20
|
Decreased levels of circulating cytokines VEGF, TNF-β and IL-15 indicate PD-L1 overexpression in tumours of primary breast cancer patients. Sci Rep 2021; 11:1294. [PMID: 33446741 PMCID: PMC7809365 DOI: 10.1038/s41598-020-80351-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/18/2020] [Indexed: 12/13/2022] Open
Abstract
Programmed death ligand 1 (PD-L1) overexpression has been associated with poor clinical outcomes in several human cancers whose increased malignant behaviour might be related to PD-L1 mediated systemic immunological tolerance. This study aims to verify if circulating cytokines may serve as a proxy for non-invasive identification of sensitive prognostic biomarkers reflecting tumour and its microenvironment. Immunohistochemistry was used to measure PD-L1 expression in tumour tissue sections of 148 chemonaïve breast cancer (BC) patients. The panel of 51 cytokines was analysed using multiplex bead arrays. High PD-L1 expression in tumours was associated with shorter progression-free survival (HR 3.25; 95% CI 1.39–7.61; P = 0.006) and low circulating levels of three multifunctional molecules; VEGF, TNF-β and IL-15 (P = 0.001). In multivariate analysis, patients with low VEGF had 4.6-fold increased risk of PD-L1 overexpression (P = 0.008), present in 76.5% of patients with all these three cytokines below the median (vs. 35.6% among the others; P = 0.002). The area under the curve value of 0.722 (95% CI 0.59–0.85; P = 0.004) shows that this combination of cytokines has a moderate ability to discriminate between PD-L1 high vs. PD-L1 low patients. Plasma cytokines, therefore, could serve as potential non-invasive biomarkers for the identification of high-risk BC cases.
Collapse
|
21
|
Terkelsen T, Pernemalm M, Gromov P, Børresen-Dale AL, Krogh A, Haakensen VD, Lethiö J, Papaleo E, Gromova I. High-throughput proteomics of breast cancer interstitial fluid: identification of tumor subtype-specific serologically relevant biomarkers. Mol Oncol 2021; 15:429-461. [PMID: 33176066 PMCID: PMC7858121 DOI: 10.1002/1878-0261.12850] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 08/13/2020] [Accepted: 11/09/2020] [Indexed: 12/24/2022] Open
Abstract
Despite significant advancements in breast cancer (BC) research, clinicians lack robust serological protein markers for accurate diagnostics and tumor stratification. Tumor interstitial fluid (TIF) accumulates aberrantly externalized proteins within the local tumor space, which can potentially gain access to the circulatory system. As such, TIF may represent a valuable starting point for identifying relevant tumor-specific serological biomarkers. The aim of the study was to perform comprehensive proteomic profiling of TIF to identify proteins associated with BC tumor status and subtype. A liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis of 35 TIFs of three main subtypes: luminal (19), Her2 (4), and triple-negative (TNBC) (12) resulted in the identification of > 8800 proteins. Unsupervised hierarchical clustering segregated the TIF proteome into two major clusters, luminal and TNBC/Her2 subgroups. High-grade tumors enriched with tumor infiltrating lymphocytes (TILs) were also stratified from low-grade tumors. A consensus analysis approach, including differential abundance analysis, selection operator regression, and random forest returned a minimal set of 24 proteins associated with BC subtypes, receptor status, and TIL scoring. Among them, a panel of 10 proteins, AGR3, BCAM, CELSR1, MIEN1, NAT1, PIP4K2B, SEC23B, THTPA, TMEM51, and ULBP2, was found to stratify the tumor subtype-specific TIFs. In particular, upregulation of BCAM and CELSR1 differentiates luminal subtypes, while upregulation of MIEN1 differentiates Her2 subtypes. Immunohistochemistry analysis showed a direct correlation between protein abundance in TIFs and intratumor expression levels for all 10 proteins. Sensitivity and specificity were estimated for this protein panel by using an independent, comprehensive breast tumor proteome dataset. The results of this analysis strongly support our data, with eight of the proteins potentially representing biomarkers for stratification of BC subtypes. Five of the most representative proteomics databases currently available were also used to estimate the potential for these selected proteins to serve as putative serological markers.
Collapse
Affiliation(s)
- Thilde Terkelsen
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Maria Pernemalm
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Pavel Gromov
- Breast Cancer Biology Group, Genome Integrity Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Anna-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway
| | - Anders Krogh
- Department of Computer Science, University of Copenhagen, Denmark.,Department of Biology, University of Copenhagen, Denmark
| | - Vilde D Haakensen
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway
| | - Janne Lethiö
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Elena Papaleo
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark.,Translational Disease System Biology, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Denmark
| | - Irina Gromova
- Breast Cancer Biology Group, Genome Integrity Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| |
Collapse
|
22
|
Soheilifar MH, Vaseghi H, Seif F, Ariana M, Ghorbanifar S, Habibi N, Papari Barjasteh F, Pornour M. Concomitant overexpression of mir-182-5p and mir-182-3p raises the possibility of IL-17-producing Treg formation in breast cancer by targeting CD3d, ITK, FOXO1, and NFATs: A meta-analysis and experimental study. Cancer Sci 2020; 112:589-603. [PMID: 33283362 PMCID: PMC7893989 DOI: 10.1111/cas.14764] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022] Open
Abstract
T cells are polarized toward regulatory T cells (Tregs) in tumor microenvironment by the shuttling of microRNAs that target T cell–activating signaling pathways. We evaluated the expression of the miR‐182 cluster (miR‐96, 182, and 183) in peripheral blood mononuclear cells (PBMCs) of patients with breast cancer (BC), and T cell polarization by the expression of FOXO1, NFATs, ITK, TCR/CD3 complex, and IL‐2/IL‐2RA. Twenty‐six microRNAs overexpressed in tumor tissues and sera of these patients were extracted by a meta‐analysis. Then, the expression of the miR‐182 cluster was investigated in PBMCs and sera of these patients and correlated with their targets in PBMCs. Finally, miR‐182 was cloned into Jurkat cells to evaluate its effects on T cell polarization. FOXO1, CD3d, ITK, NFATc3, NFATc4, and IL‐2RA were targeted by miR‐182, due to which their expression decreased in PBMCs of patients. Although IL‐6, IL‐17, and TGF‐β increased after miR‐182 transduction, IL‐2 dramatically decreased. We revealed CD4+FOXP3+ T cell differentiation in the miR‐182–transduced group. Although miR‐182 has inhibitory effects on T cells by the inhibition of FOXO1, TCR/CD3 complex, NFATs, and IL‐2/IL‐2RA signaling pathways, it increases FOXP3, TGF‐β, and IL‐17 expression to possibly drive T cell deviation toward the transitional state of IL‐17–producing Tregs and Treg formation in the end.
Collapse
Affiliation(s)
- Mohammad Hasan Soheilifar
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, Academic Center for Education, Culture, and Research (ACECR), Tehran, Iran
| | - Hajar Vaseghi
- Medical Genetics Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Seif
- Department of Immunology and Allergy, Academic Center for Education, Culture, and Research (ACECR), Tehran, Iran
| | - Mehdi Ariana
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shima Ghorbanifar
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, Academic Center for Education, Culture, and Research (ACECR), Tehran, Iran
| | - Nazanin Habibi
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, Academic Center for Education, Culture, and Research (ACECR), Tehran, Iran
| | - Fatemeh Papari Barjasteh
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, Academic Center for Education, Culture, and Research (ACECR), Tehran, Iran
| | - Majid Pornour
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, Academic Center for Education, Culture, and Research (ACECR), Tehran, Iran
| |
Collapse
|
23
|
Smolkova B, Cierna Z, Kalavska K, Miklikova S, Plava J, Minarik G, Sedlackova T, Cholujova D, Gronesova P, Cihova M, Majerova K, Karaba M, Benca J, Pindak D, Mardiak J, Mego M. Increased Stromal Infiltrating Lymphocytes Are Associated with the Risk of Disease Progression in Mesenchymal Circulating Tumor Cell-Positive Primary Breast Cancer Patients. Int J Mol Sci 2020; 21:ijms21249460. [PMID: 33322711 PMCID: PMC7763628 DOI: 10.3390/ijms21249460] [Citation(s) in RCA: 7] [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: 10/01/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/27/2022] Open
Abstract
Circulating tumor cells (CTCs) and the immune infiltration of tumors are closely related to clinical outcomes. This study aimed to verify the influence of stromal lymphocyte infiltration and the immune context of tumor microenvironment on the hematogenous spread and prognosis of 282 chemotherapy naïve primary BC patients. To detect the presence of mesenchymal CTCs, RNA extracted from CD45-depleted peripheral blood was interrogated for the expression of mesenchymal gene transcripts. Tumor-infiltrating lymphocytes (TILs) were detected in the stromal areas by immunohistochemistry, using CD3, CD8, and CD45RO antibodies. The concentrations of 51 plasma cytokines were measured by multiplex bead arrays. TILs infiltration in mesenchymal CTC-positive patients significantly decreased their progression-free survival (HR = 4.88, 95% CI 2.30–10.37, p < 0.001 for CD3high; HR = 6.17, 95% CI 2.75–13.80, p < 0.001 for CD8high; HR = 6.93, 95% CI 2.86–16.81, p < 0.001 for CD45ROhigh). Moreover, the combination of elevated plasma concentrations of transforming growth factor beta-3 (cut-off 662 pg/mL), decreased monocyte chemotactic protein-3 (cut-off 52.5 pg/mL) and interleukin-15 (cut-off 17.1 pg/mL) significantly increased the risk of disease recurrence (HR = 4.838, 95% CI 2.048–11.427, p < 0.001). Our results suggest a strong impact of the immune tumor microenvironment on BC progression, especially through influencing the dissemination and survival of more aggressive, mesenchymal CTC subtypes.
Collapse
Affiliation(s)
- Bozena Smolkova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; (B.S.); (S.M.); (J.P.); (D.C.); (P.G.); (M.C.); (K.M.)
| | - Zuzana Cierna
- Department of Pathology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia;
- Department of Pathology, Faculty Hospital, A. Zarnova 11, 917 75 Trnava, Slovakia
| | - Katarina Kalavska
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia; (K.K.); (J.M.)
- Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia
| | - Svetlana Miklikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; (B.S.); (S.M.); (J.P.); (D.C.); (P.G.); (M.C.); (K.M.)
| | - Jana Plava
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; (B.S.); (S.M.); (J.P.); (D.C.); (P.G.); (M.C.); (K.M.)
| | - Gabriel Minarik
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 842 15 Bratislava, Slovakia;
| | - Tatiana Sedlackova
- Comenius University Science Park, Ilkovicova 8, 841 04 Bratislava, Slovakia;
- Geneton Ltd., Ilkovicova 8, 841 04 Bratislava, Slovakia
| | - Dana Cholujova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; (B.S.); (S.M.); (J.P.); (D.C.); (P.G.); (M.C.); (K.M.)
| | - Paulina Gronesova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; (B.S.); (S.M.); (J.P.); (D.C.); (P.G.); (M.C.); (K.M.)
| | - Marina Cihova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; (B.S.); (S.M.); (J.P.); (D.C.); (P.G.); (M.C.); (K.M.)
| | - Karolina Majerova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; (B.S.); (S.M.); (J.P.); (D.C.); (P.G.); (M.C.); (K.M.)
| | - Marian Karaba
- Department of Oncosurgery, National Cancer Institute, Klenova 1, 83310 Bratislava, Slovakia; (M.K.); (J.B.); (D.P.)
| | - Juraj Benca
- Department of Oncosurgery, National Cancer Institute, Klenova 1, 83310 Bratislava, Slovakia; (M.K.); (J.B.); (D.P.)
- Department of Medicine, St. Elizabeth University, Namestie 1. maja 1, 811 02 Bratislava, Slovakia
| | - Daniel Pindak
- Department of Oncosurgery, National Cancer Institute, Klenova 1, 83310 Bratislava, Slovakia; (M.K.); (J.B.); (D.P.)
- Department of Oncosurgery, Slovak Medical University, Limbova 12, 83103 Bratislava, Slovakia
| | - Jozef Mardiak
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia; (K.K.); (J.M.)
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia; (K.K.); (J.M.)
- Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia
- Correspondence:
| |
Collapse
|
24
|
Prognosis of Macrophage Density in the Absence of Neutrophils in Differentiated Thyroid Cancer. J Surg Res 2020; 256:458-467. [PMID: 32798993 DOI: 10.1016/j.jss.2020.07.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 06/29/2020] [Accepted: 07/18/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND Despite the advances in treatment of differentiated thyroid cancer (DTC), predicting prognosis remains a challenge. Immune cells in the tumor microenvironment may provide an insight to predicting recurrence. Therefore, the objective of this study was to investigate the association of tumor-associated macrophages (TAMs) and tumor-associated neutrophils (TANs) with recurrence in DTC and to identify serum cytokines that correlate with the presence of these immune cells in the tumor. MATERIALS AND METHODS Forty-two DTC tissues from our institutional neoplasia repository were stained for immunohistochemistry markers for TAMs and TANs. In addition, cytokine levels were analyzed from these patients from preoperative blood samples. TAM and TAN staining were compared with clinical data and serum cytokine levels. RESULTS Neither TAM nor TAN scores alone correlated with tumor size, the presence of lymph node metastases, multifocal tumors, lymphovascular or capsular invasion, or the presence of BRAFV600E mutation (all P > 0.05). There was no association with recurrence-free survival (RFS) in TAN density (mean RFS, 169.1 versus 148.1 mo, P = 0.23) or TAM density alone (mean RFS, 121.3 versus 205.2 mo, P = 0.54). However, when scoring from both markers were combined, patients with high TAM density and TAN negative scores had significantly lower RFS (mean RFS, 50.7 versus 187.3 mo, P = 0.04) compared with the remaining cohort. Patients with high TAM/negative TAN tumors had significantly lower serum levels of interleukin 12p70, interleukin 8, tumor necrosis factor alpha, and tumor necrosis factor beta. CONCLUSIONS In DTCs, high density of TAMs in the absence of TANs is associated with worse outcome. Assessment of multiple immune cell types and serum cytokines may predict outcomes in DTC.
Collapse
|
25
|
Terkelsen T, Russo F, Gromov P, Haakensen VD, Brunak S, Gromova I, Krogh A, Papaleo E. Secreted breast tumor interstitial fluid microRNAs and their target genes are associated with triple-negative breast cancer, tumor grade, and immune infiltration. Breast Cancer Res 2020; 22:73. [PMID: 32605588 PMCID: PMC7329449 DOI: 10.1186/s13058-020-01295-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 05/14/2020] [Indexed: 12/21/2022] Open
Abstract
Background Studies on tumor-secreted microRNAs point to a functional role of these in cellular communication and reprogramming of the tumor microenvironment. Uptake of tumor-secreted microRNAs by neighboring cells may result in the silencing of mRNA targets and, in turn, modulation of the transcriptome. Studying miRNAs externalized from tumors could improve cancer patient diagnosis and disease monitoring and help to pinpoint which miRNA-gene interactions are central for tumor properties such as invasiveness and metastasis. Methods Using a bioinformatics approach, we analyzed the profiles of secreted tumor and normal interstitial fluid (IF) microRNAs, from women with breast cancer (BC). We carried out differential abundance analysis (DAA), to obtain miRNAs, which were enriched or depleted in IFs, from patients with different clinical traits. Subsequently, miRNA family enrichment analysis was performed to assess whether any families were over-represented in the specific sets. We identified dysregulated genes in tumor tissues from the same cohort of patients and constructed weighted gene co-expression networks, to extract sets of co-expressed genes and co-abundant miRNAs. Lastly, we integrated miRNAs and mRNAs to obtain interaction networks and supported our findings using prediction tools and cancer gene databases. Results Network analysis showed co-expressed genes and miRNA regulators, associated with tumor lymphocyte infiltration. All of the genes were involved in immune system processes, and many had previously been associated with cancer immunity. A subset of these, BTLA, CXCL13, IL7R, LAMP3, and LTB, was linked to the presence of tertiary lymphoid structures and high endothelial venules within tumors. Co-abundant tumor interstitial fluid miRNAs within this network, including miR-146a and miR-494, were annotated as negative regulators of immune-stimulatory responses. One co-expression network encompassed differences between BC subtypes. Genes differentially co-expressed between luminal B and triple-negative breast cancer (TNBC) were connected with sphingolipid metabolism and predicted to be co-regulated by miR-23a. Co-expressed genes and TIF miRNAs associated with tumor grade were BTRC, CHST1, miR-10a/b, miR-107, miR-301a, and miR-454. Conclusion Integration of IF miRNAs and mRNAs unveiled networks associated with patient clinicopathological traits, and underlined molecular mechanisms, specific to BC sub-groups. Our results highlight the benefits of an integrative approach to biomarker discovery, placing secreted miRNAs within a biological context.
Collapse
Affiliation(s)
- Thilde Terkelsen
- Computational Biology Laboratory, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Francesco Russo
- Computational Biology Laboratory, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pavel Gromov
- Breast Cancer Biology Group, Genome Integrity Unit, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Vilde Drageset Haakensen
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Irina Gromova
- Breast Cancer Biology Group, Genome Integrity Unit, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Anders Krogh
- Unit of Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Elena Papaleo
- Computational Biology Laboratory, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark. .,Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
26
|
Terkelsen T, Krogh A, Papaleo E. CAncer bioMarker Prediction Pipeline (CAMPP)-A standardized framework for the analysis of quantitative biological data. PLoS Comput Biol 2020; 16:e1007665. [PMID: 32176694 PMCID: PMC7108742 DOI: 10.1371/journal.pcbi.1007665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 03/31/2020] [Accepted: 01/18/2020] [Indexed: 01/21/2023] Open
Abstract
With the improvement of -omics and next-generation sequencing (NGS) methodologies, along with the lowered cost of generating these types of data, the analysis of high-throughput biological data has become standard both for forming and testing biomedical hypotheses. Our knowledge of how to normalize datasets to remove latent undesirable variances has grown extensively, making for standardized data that are easily compared between studies. Here we present the CAncer bioMarker Prediction Pipeline (CAMPP), an open-source R-based wrapper (https://github.com/ELELAB/CAncer-bioMarker-Prediction-Pipeline -CAMPP) intended to aid bioinformatic software-users with data analyses. CAMPP is called from a terminal command line and is supported by a user-friendly manual. The pipeline may be run on a local computer and requires little or no knowledge of programming. To avoid issues relating to R-package updates, a renv .lock file is provided to ensure R-package stability. Data-management includes missing value imputation, data normalization, and distributional checks. CAMPP performs (I) k-means clustering, (II) differential expression/abundance analysis, (III) elastic-net regression, (IV) correlation and co-expression network analyses, (V) survival analysis, and (VI) protein-protein/miRNA-gene interaction networks. The pipeline returns tabular files and graphical representations of the results. We hope that CAMPP will assist in streamlining bioinformatic analysis of quantitative biological data, whilst ensuring an appropriate bio-statistical framework.
Collapse
Affiliation(s)
- Thilde Terkelsen
- Computational Biology Laboratory, Danish Cancer Society Research Center and Center for Autophagy, Recycling and Disease, Copenhagen, Denmark
| | - Anders Krogh
- Unit of Computational and RNA biology, Department of Biology, University of Copenhagen, Copenhagen Denmark
| | - Elena Papaleo
- Computational Biology Laboratory, Danish Cancer Society Research Center and Center for Autophagy, Recycling and Disease, Copenhagen, Denmark
- Translational Disease System Biology, Faculty of Health and Medical Science, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|