51
|
Majumder B, Budhu S, Ganusov VV. Mathematical modeling suggests cytotoxic T lymphocytes control growth of B16 tumor cells in collagin-fibrin gels by cytolytic and non-lytic mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.28.534600. [PMID: 37034693 PMCID: PMC10081166 DOI: 10.1101/2023.03.28.534600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) are important in controlling some viral infections, and therapies involving transfer of large numbers of cancer-specific CTLs have been successfully used to treat several types of cancers in humans. While molecular mechanisms of how CTLs kill their targets are relatively well understood we still lack solid quantitative understanding of the kinetics and efficiency at which CTLs kill their targets in different conditions. Collagen-fibrin gel-based assays provide a tissue-like environment for the migration of CTLs, making them an attractive system to study the cytotoxicity in vitro. Budhu et al. [1] systematically varied the number of peptide (SIINFEKL)- pulsed B16 melanoma cells and SIINFEKL-specific CTLs (OT-1) and measured remaining targets at different times after target and CTL co-inoculation into collagen-fibrin gels. The authors proposed that their data were consistent with a simple model in which tumors grow exponentially and are killed by CTLs at a per capita rate proportional to the CTL density in the gel. By fitting several alternative mathematical models to these data we found that this simple "exponential-growth-mass-action-killing" model does not precisely fit the data. However, determining the best fit model proved difficult because the best performing model was dependent on the specific dataset chosen for the analysis. When considering all data that include biologically realistic CTL concentrations ( E ≤ 10 7 cell/ml) the model in which tumors grow exponentially and CTLs suppress tumor's growth non-lytically and kill tumors according to the mass-action law (SiGMA model) fitted the data with best quality. Results of power analysis suggested that longer experiments (∼ 3 - 4 days) with 4 measurements of B16 tumor cell concentrations for a range of CTL concentrations would best allow to discriminate between alternative models. Taken together, our results suggest that interactions between tumors and CTLs in collagen-fibrin gels are more complex than a simple exponential-growth- mass-action killing model and provide support for the hypothesis that CTLs impact on tumors may go beyond direct cytotoxicity.
Collapse
Affiliation(s)
- Barun Majumder
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | - Sadna Budhu
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Vitaly V. Ganusov
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
- Department of Mathematics, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
52
|
Devi S, Gründemann C, Huber R, Kowarschik S. Characterization of Viscum album L. Effect on Immune Escape Proteins PD-L1, PD-L2, and MHC-I in the Prostate, Colon, Lung, and Breast Cancer Cells. Complement Med Res 2023; 30:386-392. [PMID: 36927644 DOI: 10.1159/000530184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/27/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND Viscum album L. (VA) preparations possess immunomodulatory properties and are used in complementary medicine to support cancer therapy. It is unclear if there is an impact of VA on the expression of immune checkpoint proteins on the surface of cancer cells. This study was designed to investigate the role of commercially available VA preparations on checkpoint programmed death ligand 1, 2 (PD-L1, PD-L2) and on major histocompatibility complex class I (MHC-I). METHODS Four human cancer cell lines (prostate, colon, lung, and breast) were assayed for their PD-L1, PD-L2, and MHC-I level after stimulation with interferon-gamma (IFN-γ). The toxicity of mistletoe preparations for the cells was analysed. Afterwards, the effect of mistletoe preparations on the PD ligands and MHC-I was investigated. RESULTS Surface protein analysis demonstrated that all tested tumour cell lines increased the PD-L1, PD-L2, and MHC-I-expression, but to different extents, after IFN-γ stimulation. Treatment with VA extracts did not influence the viability of the cells. The expression of PD ligands and MHC-I was not affected by incubation with the VA preparations. CONCLUSION Our investigation concludes that VA treatment does not interfere with the expression of PD ligands or MHC-I among selected cancer cells. Hintergrund Viscum album L. (VA)-Präparate besitzen immunmodulatorische Eigenschaften und werden in der Komplementärmedizin zur Unterstützung in der Krebstherapie eingesetzt. Es ist jedoch unklar, ob VA einen Einfluss auf die Expression von Immuncheckpoint-Proteinen auf Krebszellen hat. In der vorliegenden Arbeit wurde ein Einfluss von handelsüblichen VA-Präparaten auf die Checkpoint-Proteine programmed death ligand 1, 2 (PD-L1, PD-L2) und major histocompatibility complex class I (MHC-I) untersucht. Methoden Vier humane Krebszelllinien der Prostata, des Dickdarms, der Lunge und Brust wurden nach Stimulation mit Interferon-gamma (IFN-γ) auf ihre PD-L1, PD-L2 und MHC-I Konzentration untersucht. Zunächst wurde die Toxizität von Mistelpräparaten auf die Tumorzellen analysiert. Anschließend erfolgte eine Charakterisierung der Wirkung von Mistelpräparaten auf die PD-Liganden und MHC-I. Ergebnisse Die Oberflächenproteinanalysen zeigten, dass alle getesteten Tumorzelllinien nach einer IFN-γ-Stimulation die PD-L1, PD-L2 und MHC-I Expression in unterschiedlichem Ausmaß erhöhten. Die Behandlung mit verschiedenen VA-Extrakten hatte keinen Einfluss auf die Viabilität der Zellen, sowie auf die Expression der PD-Liganden und MHC-I. Schlussfolgerung Unsere Untersuchung kommt zu dem Schluss, dass eine VA-Behandlung die Expression von PD-Liganden oder MHC-I in den untersuchten Krebszellen nicht beeinflusst.
Collapse
Affiliation(s)
- Seema Devi
- Centre for Complementary Medicine, Department of Internal Medicine II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carsten Gründemann
- Translational Complementary Medicine, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Roman Huber
- Centre for Complementary Medicine, Department of Internal Medicine II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefanie Kowarschik
- Centre for Complementary Medicine, Department of Internal Medicine II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
53
|
Cheruku S, Rao V, Pandey R, Rao Chamallamudi M, Velayutham R, Kumar N. Tumor-associated macrophages employ immunoediting mechanisms in colorectal tumor progression: Current research in Macrophage repolarization immunotherapy. Int Immunopharmacol 2023; 116:109569. [PMID: 36773572 DOI: 10.1016/j.intimp.2022.109569] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/01/2022] [Accepted: 12/07/2022] [Indexed: 02/11/2023]
Abstract
Tumor-associated macrophages (TAMs) constitute the most prolific resident of the tumor microenvironment (TME) that regulate its TME into tumor suppressive or progressive milieu by utilizing immunoediting machinery. Here, the tumor cells construct an immunosuppressive microenvironment that educates TAMs to polarize from anti-tumor TAM-M1 to pro-tumor TAM-M2 phenotype consequently contributing to tumor progression. In colorectal cancer (CRC), the TME displays a prominent pro-tumorigenic immune profile with elevated expression of immune-checkpoint molecules notably PD-1, CTLA4, etc., in both MSI and ultra-mutated MSS tumors. This authenticated immune-checkpoint inhibition (ICI) immunotherapy as a pre-requisite for clinical benefit in CRC. However, in response to ICI, specifically, the MSIhi tumors evolved to produce novel immune escape variants thus undermining ICI. Lately, TAM-directed therapies extending from macrophage depletion to repolarization have enabled TME alteration. While TAM accrual implicates clinical benefit in CRC, sustained inflammatory insult may program TAMs to shift from M1 to M2 phenotype. Their ability to oscillate on both facets of the spectrum represents macrophage repolarization immunotherapy as an effective approach to treating CRC. In this review, we briefly discuss the differentiation heterogeneity of colonic macrophages that partake in macrophage-directed immunoediting mechanisms in CRC progression and its employment in macrophage re-polarization immunotherapy.
Collapse
Affiliation(s)
- SriPragnya Cheruku
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal- 576104, Karnataka, India
| | - Vanishree Rao
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal- 576104, Karnataka, India
| | - Ruchi Pandey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education, and Research, Hajipur, Export Promotions Industrial Park (EPIP), Industrial area, Hajipur, Vaishali, 844102, Bihar, India
| | - Mallikarjuna Rao Chamallamudi
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal- 576104, Karnataka, India
| | - Ravichandiran Velayutham
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education, and Research, Hajipur, Export Promotions Industrial Park (EPIP), Industrial area, Hajipur, Vaishali, 844102, Bihar, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education, and Research, Hajipur, Export Promotions Industrial Park (EPIP), Industrial area, Hajipur, Vaishali, 844102, Bihar, India.
| |
Collapse
|
54
|
Zhou B, Basu J, Kazmi HR, Chitrala KN, Mo X, Preston-Alp S, Cai KQ, Kappes D, Zaidi MR. Interferon-gamma signaling promotes melanoma progression and metastasis. Oncogene 2023; 42:351-363. [PMID: 36463370 PMCID: PMC9991867 DOI: 10.1038/s41388-022-02561-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022]
Abstract
Interferon-gamma (IFNG) has long been regarded as the flag-bearer for the anti-cancer immunosurveillance mechanisms. However, relatively recent studies have suggested a dual role of IFNG, albeit there is no direct experimental evidence for its potential pro-tumor functions. Here we provide in vivo evidence that treatment of mouse melanoma cell lines with Ifng enhances their tumorigenicity and metastasis in lung colonization allograft assays performed in immunocompetent syngeneic host mice, but not in immunocompromised host mice. We also show that this enhancement is dependent on downstream signaling via Stat1 but not Stat3, suggesting an oncogenic function of Stat1 in melanoma. The experimental results suggest that melanoma cell-specific Ifng signaling modulates the tumor microenvironment and its pro-tumorigenic effects are partially dependent on the γδ T cells, as Ifng-enhanced tumorigenesis was inhibited in the TCR-δ knockout mice. Overall, these results show that Ifng signaling may have tumor-promoting effects in melanoma by modulating the immune cell composition of the tumor microenvironment.
Collapse
Affiliation(s)
- Bo Zhou
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,MEI Pharma, San Diego, CA, USA
| | - Jayati Basu
- Fox Chase Cancer Center, Philadelphia, PA, USA.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Hasan Raza Kazmi
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Kumaraswamy Naidu Chitrala
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Department of Engineering Technology, University of Houston, Houston, TX, USA
| | - Xuan Mo
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Sarah Preston-Alp
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Kathy Q Cai
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | - M Raza Zaidi
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
55
|
Habel A, Xu W, Hadj Ahmed M, Stayoussef M, Bouaziz H, Ayadi M, Mezlini A, Larbi A, Yaacoubi-Loueslati B. Identification of two theranostic biomarker panels for epithelial ovarian cancer. Cytokine 2023; 161:156051. [PMID: 36401984 DOI: 10.1016/j.cyto.2022.156051] [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: 07/01/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Epithelial Ovarian cancer (EOC) is the leading cause of death associated with gynecologic tumors. Because the disease is asymptomatic in early-stage, the majority of patients are not diagnosed until late stages, highlighting the need for the development of novel diagnostic biomarkers. Mediators of tumoral microenvironment may affect EOC progression and resistance to treatment. AIM OF THE STUDY Analysis of serum proteins to identify a panel of theranostic biomarkers for EOC. PATIENTS AND METHODS Serum levels of 65 analytes were determined in EOC patients, and healthy controls with the ProcartaPlex Human Immune Monitoring 65-Plex Panel. RESULTS Twenty-one analytes: 7 cytokines (IFN-γ, IL-12p70, IL-13, IL-18 and TSLP), 7 chemokines (Eotaxin, eotaxin-2, IP-10, BLC, I-TAC, SDF-1α, and fractalkine), 2 growth factors (MMP-1, VEGF-α), and 5 soluble receptors (APRIL, CD40L, TWEAK, CD30 and TNFRII; were significantly differentially expressed between the two groups. ROC curves showed that only seven of them (IL-9, TNF-α, Eotaxin, IP-10, BLC, Fractalkine, and Tweak) had AUC values greater than 0.70 and thus had potential clinical utility. Moreover, five cytokines: IFN-γ, IL-1 β, IL-8, MIP-1β, and TNF-α are positively associated with patients who developed resistance to taxol-platinum-based chemotherapy (CT). CONCLUSION This study has revealed a first panel of 7 analytes (IL-9, TNF-α, Eotaxin, IP-10, BLC, Fractalkine and Tweak) that can be used for early detection of EOC and a second panel of five cytokines (IFN-γ, IL-1β, IL-8, MIP-1β, TNF-α) that can help clinicians to identify EOC patients who are at higher risk to develop resistance to CT of EOC.
Collapse
Affiliation(s)
- Azza Habel
- 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
| | - Mariem Hadj Ahmed
- University of Tunis El Manar (UTM), Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Tunisia
| | - 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
| | - Mouna Ayadi
- 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; Beckman Coulter Life Sciences, Villepinte 93420, France
| | - Basma Yaacoubi-Loueslati
- University of Tunis El Manar (UTM), Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Tunisia.
| |
Collapse
|
56
|
Ujvari D, Malyukova A, Zovko A, Yektaei-Karin E, Madapura HS, Keszei M, Nagy N, Lotfi K, Björn N, Wallvik J, Stenke L, Salamon D. IFNγ directly counteracts imatinib-induced apoptosis of primary human CD34+ CML stem/progenitor cells potentially through the upregulation of multiple key survival factors. Oncoimmunology 2022; 11:2109861. [PMID: 35979386 PMCID: PMC9377247 DOI: 10.1080/2162402x.2022.2109861] [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] [Indexed: 11/12/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have dramatically improved the survival in chronic myeloid leukemia (CML), but residual disease typically persists even after prolonged treatment. Several lines of evidence suggest that TKIs administered to CML patients upregulate interferon γ (IFNγ) production, which may counteract the anti-tumorigenic effects of the therapy. We now show that activated T cell-conditioned medium (TCM) enhanced proliferation and counteracted imatinib-induced apoptosis of CML cells, and addition of a neutralizing anti-IFNγ antibody at least partially inhibited the anti-apoptotic effect. Likewise, recombinant IFNγ also reduced imatinib-induced apoptosis of CML cells. This anti-apoptotic effect of IFNγ was independent of alternative IFNγ signaling pathways, but could be notably diminished by STAT1-knockdown. Furthermore, IFNγ upregulated the expression of several anti-apoptotic proteins, including MCL1, PARP9, and PARP14, both in untreated and imatinib-treated primary human CD34+ CML stem/progenitor cells. Our results suggest that activated T cells in imatinib-treated CML patients can directly rescue CML cells from imatinib-induced apoptosis at least partially through the secretion of IFNγ, which exerts a rapid, STAT1-dependent anti-apoptotic effect potentially through the simultaneous upregulation of several key hematopoietic survival factors. These mechanisms may have a major clinical impact, when targeting residual leukemic stem/progenitor cells in CML.
Collapse
Affiliation(s)
- Dorina Ujvari
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
- National Pandemic Center, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Alena Malyukova
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Ana Zovko
- Division of Hematology, Karolinska University Hospital Solna, Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Elham Yektaei-Karin
- Division of Hematology, Karolinska University Hospital Solna, Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Harsha S Madapura
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Marton Keszei
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Noemi Nagy
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Kourosh Lotfi
- Department of Hematology, Linköping University Hospital, Linköping, Sweden
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Niclas Björn
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jonas Wallvik
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Leif Stenke
- Division of Hematology, Karolinska University Hospital Solna, Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Daniel Salamon
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
57
|
Liu Q, Tan J, Zhao Z, Li R, Zheng L, Chen X, Li L, Dong X, Wen T, Liu J. Combined Usage of MDK Inhibitor Augments Interferon-γ Anti-Tumor Activity in the SKOV3 Human Ovarian Cancer Cell Line. Biomedicines 2022; 11:biomedicines11010008. [PMID: 36672515 PMCID: PMC9855738 DOI: 10.3390/biomedicines11010008] [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: 11/17/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer (OC) is a particularly lethal disease due to intratumoral heterogeneity, resistance to traditional chemotherapy, and poor response to targeted therapy and immunotherapy. Interferon-γ (IFN-γ) is an attractive therapeutic cytokine, with positive responses achieved in multiple OC clinical trials. However, clinical application of IFN-γ in OC is still hindered, due to the severe toxicity when used at higher levels, as well as the considerable pro-metastatic adverse effect when used at lower levels. Thus, an effective combined intervention is needed to enhance the anti-tumor efficacy of IFN-γ and to suppress the IFN-γ-induced metastasis. Here, we uncovered that OC cells develop an adaptive strategy by upregulating midkine (MDK) to counteract the IFN-γ-induced anti-tumor activity and to fuel IFN-γ-induced metastasis. We showed that MDK is a critical downstream target of IFN-γ in OC, and that this regulation acts in a dose-dependent manner and is mediated by STAT1. Gain-of-function studies showed that MDK overexpression promotes cell proliferation and metastasis in OC, indicating that IFN-γ-activated MDK may antagonize IFN-γ in inhibiting OC proliferation but synergize IFN-γ in promoting OC metastasis. Subsequently, we assessed the influence of MDK inhibition on IFN-γ-induced anti-proliferation and pro-metastasis effects using an MDK inhibitor (iMDK), and we found that MDK inhibition robustly enhanced IFN-γ-induced growth inhibition (all CIs < 0.1) and reversed IFN-γ-driven epithelial-to-mesenchymal transition (EMT) and metastasis in OC in vitro. Collectively, these data identify an IFN-γ responsive protein, MDK, in counteracting anti-proliferation while endowing the pro-metastatic role of IFN-γ in cancer treatment, and we therefore propose the combined utilization of the MDK inhibitor in IFN-γ-based therapies in future OC treatment.
Collapse
Affiliation(s)
- Qun Liu
- Department of Gynaecology and Obstetrics, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Jingyu Tan
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Zhenguo Zhao
- Department of Orthopaedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ruijun Li
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Luyu Zheng
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiangyu Chen
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Lina Li
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xichen Dong
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Tao Wen
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
- Correspondence: (T.W.); (J.L.)
| | - Jian Liu
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
- Correspondence: (T.W.); (J.L.)
| |
Collapse
|
58
|
Rodríguez-Tomàs E, Arenas M, Baiges-Gaya G, Acosta J, Araguas P, Malave B, Castañé H, Jiménez-Franco A, Benavides-Villarreal R, Sabater S, Solà-Alberich R, Camps J, Joven J. Gradient Boosting Machine Identified Predictive Variables for Breast Cancer Patients Pre- and Post-Radiotherapy: Preliminary Results of an 8-Year Follow-Up Study. Antioxidants (Basel) 2022; 11:antiox11122394. [PMID: 36552602 PMCID: PMC9774765 DOI: 10.3390/antiox11122394] [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: 08/25/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/11/2022] Open
Abstract
Radiotherapy (RT) is part of the standard treatment of breast cancer (BC) because of its effects on relapse reduction and survival. However, response to treatment is highly variable, and some patients may develop disease progression (DP), a second primary cancer, or may succumb to the disease. Antioxidant systems and inflammatory processes are associated with the onset and development of BC and play a role in resistance to treatment. Here, we report our investigation into the clinical evolution of BC patients, and the impact of RT on the circulating levels of the antioxidant enzyme paraoxonase-1 (PON1), cytokines, and other standard biochemical and hematological variables. Gradient Boosting Machine (GBM) algorithm was used to identify predictive variables. This was a retrospective study in 237 patients with BC. Blood samples were obtained pre- and post-RT, with samples of healthy women used as control subjects. Results showed that 24 patients had DP eight years post-RT, and eight patients developed a second primary tumor. The algorithm identified interleukin-4 and total lymphocyte counts as the most relevant indices discriminating between BC patients and control subjects, while neutrophils, total leukocytes, eosinophils, very low-density lipoprotein cholesterol, and PON1 activity were potential predictors of fatal outcome.
Collapse
Affiliation(s)
- Elisabet Rodríguez-Tomàs
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Meritxell Arenas
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
- Correspondence: (M.A.); (J.C.); Tel.: +34-977-310-300 (ext. 54132) (M.A.); +34-977-310-300 (ext. 55409) (J.C.)
| | - Gerard Baiges-Gaya
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Johana Acosta
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Pablo Araguas
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Bárbara Malave
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Helena Castañé
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Andrea Jiménez-Franco
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Rocío Benavides-Villarreal
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Sebastià Sabater
- Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43204 Reus, Spain
| | - Rosa Solà-Alberich
- Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-SALUT), Facultat de Medicina i Ciències de La Salut, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
- Correspondence: (M.A.); (J.C.); Tel.: +34-977-310-300 (ext. 54132) (M.A.); +34-977-310-300 (ext. 55409) (J.C.)
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| |
Collapse
|
59
|
CellCallEXT: Analysis of Ligand-Receptor and Transcription Factor Activities in Cell-Cell Communication of Tumor Immune Microenvironment. Cancers (Basel) 2022; 14:cancers14194957. [PMID: 36230879 PMCID: PMC9563271 DOI: 10.3390/cancers14194957] [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: 08/29/2022] [Revised: 10/01/2022] [Accepted: 10/05/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary CellCall is an R package tool that is used to analyze cell–cell communication based on transcription factor (TF) activities calculated by cell-type specificity of target genes and thus cannot directly handle two-condition comparisons. We developed CellCallEXT to complement CellCall. CellCallEXT can directly identify ligand–receptor (L–R) interactions that alter the expression profiles of downstream genes between two conditions, such as tumor and healthy tissue. Scoring in CellCallEXT quantitatively integrates expression of ligands, receptors, TFs, and target genes (TGs). The pathway enrichment analysis and visualization modules allow biologists to investigate how disease alters cell–cell communication. Furthermore, Reactome pathways were added into CellCallEXT to expand the L–R–TF database. Abstract (1) Background: Single-cell RNA sequencing (scRNA-seq) data are useful for decoding cell–cell communication. CellCall is a tool that is used to infer inter- and intracellular communication pathways by integrating paired ligand–receptor (L–R) and transcription factor (TF) activities from steady-state data and thus cannot directly handle two-condition comparisons. For tumor and healthy status, it can only individually analyze cells from tumor or healthy tissue and examine L–R pairs only identified in either tumor or healthy controls, but not both together. Furthermore, CellCall is highly affected by gene expression specificity in tissues. (2) Methods: CellCallEXT is an extension of CellCall that deconvolutes intercellular communication and related internal regulatory signals based on scRNA-seq. Information on Reactome was retrieved and integrated with prior knowledge of L–R–TF signaling and gene regulation datasets of CellCall. (3) Results: CellCallEXT was successfully applied to examine tumors and immune cell microenvironments and to identify the altered L–R pairs and downstream gene regulatory networks among immune cells. Application of CellCallEXT to scRNA-seq data from patients with deficiency of adenosine deaminase 2 demonstrated its ability to impute dysfunctional intercellular communication and related transcriptional factor activities. (4) Conclusions: CellCallEXT provides a practical tool to examine intercellular communication in disease based on scRNA-seq data.
Collapse
|
60
|
Monnier M, Paolini L, Vinatier E, Mantovani A, Delneste Y, Jeannin P. Antitumor strategies targeting macrophages: the importance of considering the differences in differentiation/polarization processes between human and mouse macrophages. J Immunother Cancer 2022; 10:jitc-2022-005560. [PMID: 36270732 PMCID: PMC9594518 DOI: 10.1136/jitc-2022-005560] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2022] [Indexed: 11/05/2022] Open
Abstract
Macrophages are the immune cells that accumulate the most in the majority of established tumors and this accumulation is associated with a poor prognosis. Tumor-associated macrophages (TAMs) produce inflammatory cytokines and growth factors that promote tumor expansion and metastasis. TAMs have recently emerged as targets of choice to restore an efficient antitumor response and to limit tumor growth. Many molecules targeting TAMs are actually evaluated in clinical trials, alone or in combination. While these molecules induce tumor regression and stimulate cytotoxic responses in mouse models of tumor development, results from early clinical trials are less impressive. In this review, we list the biological differences between human and mouse macrophages that help explain the different efficacy of antitumor strategies targeting TAMs between human and animal studies. Differences in the impact of survival and polarization factors and in the cytokines produced and markers expressed as well as the limitations of extrapolations based on in vitro models of TAM-like generation should be considered in order to improve the design and efficacy of antitumor drugs targeting TAMs.
Collapse
Affiliation(s)
- Marine Monnier
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, LabEx IGO, Angers, France.,Univ Angers, SFR ICAT, Angers, France
| | - Léa Paolini
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, LabEx IGO, Angers, France.,Univ Angers, SFR ICAT, Angers, France
| | - Emeline Vinatier
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, LabEx IGO, Angers, France.,Univ Angers, SFR ICAT, Angers, France.,Immunology and Allergology laboratory, University Hospital of Angers, Angers, France
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Milan, Italy.,Humanitas University, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Yves Delneste
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, LabEx IGO, Angers, France.,Univ Angers, SFR ICAT, Angers, France.,Immunology and Allergology laboratory, University Hospital of Angers, Angers, France
| | - Pascale Jeannin
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, LabEx IGO, Angers, France .,Univ Angers, SFR ICAT, Angers, France.,Immunology and Allergology laboratory, University Hospital of Angers, Angers, France
| |
Collapse
|
61
|
Milovanović J, Todorović-Raković N, Vujasinović T, Greenman J, Mandušić V, Radulovic M. Can granulysin provide prognostic value in primary breast cancer? Pathol Res Pract 2022; 237:154039. [PMID: 35905663 DOI: 10.1016/j.prp.2022.154039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 07/20/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Granulysin (GNLY) is a cytolytic and proinflammatory molecule which also acts as an immune alarmin. The multifunctional nature of this molecule has made it challenging to define its full potential as a biomarker in breast cancer. AIM To evaluate the prognostic value of intratumoral GNLY in primary breast cancer patients and its association with established clinicopathological parameters. PATIENTS AND METHODS The study included 69 node-negative breast cancer patients with known clinicopathological parameters, all of whom had not received any prior hormonal or chemotherapeutic systemic therapy that would interfere with the course of disease. The median follow-up period was 144 months. Steroid hormone receptor status was determined by ligand-binding assay and HER2 status by chromogenic in situ hybridisation (CISH). Intratumoral GNLY mRNA levels were determined by RT-qPCR. Prognostic performance was evaluated by the receiver operating characteristic (ROC), Cox proportional hazards regression and Kaplan-Meier analysis. Classification of patients into GNLYlow and GNLYhigh subgroups was performed by the use of the outcome-oriented cut-off point categorisation approach. RESULTS There was a significant difference between GNLY values of patients without any recurrences and those with local or distant recurrences (Mann-Whitney test, p = 0.05 and p = 0.02, respectively). None of the tested parameters showed prognostic significance for local and distant recurrences when combined. When distant metastases and local recurrences were separated as events, the best prognostic performance was observed for GNLY as compared with any clinicopathological parameter (AUC=0.24 and p = 0.04 for local events; AUC=0.71 and p = 0.03 for distant events). Local recurrence incidence was 0% for the GNLYhigh subgroup and 19% for the GNLYlow subgroup; however distant recurrence incidence was 24% for the GNLYhigh subgroup but only 3% for the GNLYlow subgroup (Kaplan-Meier analysis). A significant positive correlation was found between intratumoral ER and GNLY levels, and a significant negative correlation between tumour grade and GNLY levels. CONCLUSION High levels of granulysin prognosticate low risk of local recurrence but a high risk of distant metastasis in primary, untreated, breast cancer patients.
Collapse
Affiliation(s)
- Jelena Milovanović
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia.
| | - Nataša Todorović-Raković
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Tijana Vujasinović
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - John Greenman
- Department of Biomedical Sciences, University of Hull, Hull, UK
| | - Vesna Mandušić
- Department for Radiobiology and Molecular Genetics, Institute of Nuclear Sciences Vinča - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Marko Radulovic
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| |
Collapse
|
62
|
Li L, Li F, Hu X, Wu Z, Ren W, Wang T, Ji Z, Li N, Gu J, Sun C, Feng X, Han W, Huang J, Lei L. LAP3 contributes to IFN-γ-induced arginine depletion and malignant transformation of bovine mammary epithelial cells. BMC Cancer 2022; 22:864. [PMID: 35941558 PMCID: PMC9358085 DOI: 10.1186/s12885-022-09963-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/29/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND IFN-γ has been traditionally recognized as an inflammatory cytokine that involves in inflammation and autoimmune diseases. Previously we have shown that sustained IFN-γ induced malignant transformation of bovine mammary epithelial cells (BMECs) via arginine depletion. However, the molecular mechanism underlying this is still unknown. METHODS In this study, the amino acids contents in BMECs were quantified by a targeted metabolomics method. The acquisition of differentially expressed genes was mined from RNA-seq dataset and analyzed bioinformatically. Quantitative reverse transcription polymerase chain reaction (qRT-PCR), enzyme-linked immunosorbent assay (ELISA), western blotting, and immunohistochemistry (IHC) assay were performed to detect gene mRNA and protein expression levels. CCK-8 and would healing assays were used to detect cell proliferation and migration abilities, respectively. Cell cycle phase alternations were analyzed by flow cytometry. RESULTS The targeted metabolomics analysis specifically discovered IFN-γ induced arginine depletion through accelerating arginine catabolism and inhibiting arginine anabolism in BMECs. Transcriptome analysis identified leucine aminopeptidase 3 (LAP3), which was regulated by p38 and ERK MAPKs, to downregulate arginine level through interfering with argininosuccinate synthetase (ASS1) as IFN-γ stimulated. Moreover, LAP3 also contributed to IFN-γ-induced malignant transformation of BMECs by upregulation of HDAC2 (histone deacetylase 2) expression and promotion of cell cycle proteins cyclin A1 and D1 expressions. Arginine supplementation did not affect LAP3 and HDAC2 expressions, but slowed down cell cycle process of malignant BMECs. In clinical samples of patients with breast cancer, LAP3 was confirmed to be upregulated, while ASS1 was downregulated compared with healthy control. CONCLUSIONS These results demonstrated that LAP3 mediated IFN-γ-induced arginine depletion to malignant transformation of BMECs. Our findings provide a potential therapeutic target for breast cancer both in humans and dairy cows.
Collapse
Affiliation(s)
- Li Li
- Department of First Hospital, Jilin University, Xinmin Street 1, Changchun, China
| | - Fengyang Li
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, China
| | - Xiuhong Hu
- Department of First Hospital, Jilin University, Xinmin Street 1, Changchun, China
- Shannan Hospital, Shannan, China
| | - Zengshuai Wu
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, China
| | - Wenbo Ren
- Department of First Hospital, Jilin University, Xinmin Street 1, Changchun, China
| | - Tingting Wang
- Department of First Hospital, Jilin University, Xinmin Street 1, Changchun, China
| | - Zhengchao Ji
- Department of First Hospital, Jilin University, Xinmin Street 1, Changchun, China
| | - Na Li
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, China
| | - Jingmin Gu
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, China
| | - Changjiang Sun
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, China
| | - Xin Feng
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, China
| | - Wenyu Han
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, China
| | - Jing Huang
- Department of First Hospital, Jilin University, Xinmin Street 1, Changchun, China.
| | - Liancheng Lei
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Xi'an Road 5333, Changchun, China.
| |
Collapse
|
63
|
Gu C, Lin C, Zhu Z, Hu L, Wang F, Wang X, Ruan J, Zhao X, Huang S. The IFN-γ-related long non-coding RNA signature predicts prognosis and indicates immune microenvironment infiltration in uterine corpus endometrial carcinoma. Front Oncol 2022; 12:955979. [PMID: 35957871 PMCID: PMC9360323 DOI: 10.3389/fonc.2022.955979] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 06/30/2022] [Indexed: 12/01/2022] Open
Abstract
Background One of the most common diseases that have a negative impact on women’s health is endometrial carcinoma (EC). Advanced endometrial cancer has a dismal prognosis and lacks solid prognostic indicators. IFN-γ is a key cytokine in the inflammatory response, and it has also been suggested that it has a role in the tumor microenvironment. The significance of IFN-γ-related genes and long non-coding RNAs in endometrial cancer, however, is unknown. Methods The Cancer Genome Atlas (TCGA) database was used to download RNA-seq data from endometrial cancer tissues and normal controls. Genes associated with IFN-γ were retrieved from the gene set enrichment analysis (GSEA) website. Co-expression analysis was performed to find lncRNAs linked to IFN-γ gene. The researchers employed weighted co-expression network analysis (WGCNA) to find lncRNAs that were strongly linked to survival. The prognostic signature was created using univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression. The training cohort, validation cohort, and entire cohort of endometrial cancer patients were then split into high-risk and low-risk categories. To investigate variations across different risk groups, we used survival analysis, enrichment analysis, and immune microenvironment analysis. The platform for analysis is R software (version X64 3.6.1). Results Based on the transcript expression of IFN-γ-related lncRNAs, two distinct subgroups of EC from TCGA cohort were formed, each with different outcomes. Ten IFN-γ-related lncRNAs were used to build a predictive signature using Cox regression analysis and the LASSO regression, including CFAP58, LINC02014, UNQ6494, AC006369.1, NRAV, BMPR1B-DT, AC068134.2, AP002840.2, GS1-594A7.3, and OLMALINC. The high-risk group had a considerably worse outcome (p < 0.05). In the immunological microenvironment, there were also substantial disparities across different risk categories. Conclusion Our findings give a reference for endometrial cancer prognostic type and immunological status assessment, as well as prospective molecular markers for the disease.
Collapse
Affiliation(s)
- Chunyan Gu
- Department of Obstetrics and Gynecology, Nantong Haimen People’s Hospital, Nantong, China
| | - Chen Lin
- Vectors and Parasitosis Control and Prevention Section, Center of Disease Prevention and Control in Pudong New Area of Shanghai, Shanghai, China
| | - Zheng Zhu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Hu
- Department of Medicine, Kangda College of Nanjing Medical University, Lianyungang, China
| | - Fengxu Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Xuehai Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Junpu Ruan
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
- *Correspondence: Xinyuan Zhao, ; Sen Huang,
| | - Sen Huang
- Department of Obstetrics and Gynecology, Nantong Haimen People’s Hospital, Nantong, China
- *Correspondence: Xinyuan Zhao, ; Sen Huang,
| |
Collapse
|
64
|
Baram T, Oren N, Erlichman N, Meshel T, Ben-Baruch A. Inflammation-Driven Regulation of PD-L1 and PD-L2, and Their Cross-Interactions with Protective Soluble TNFα Receptors in Human Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:3513. [PMID: 35884574 PMCID: PMC9323351 DOI: 10.3390/cancers14143513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/04/2022] [Accepted: 07/09/2022] [Indexed: 12/02/2022] Open
Abstract
Pro-inflammatory cytokines play key roles in elevating cancer progression in triple-negative breast cancer (TNBC). We demonstrate that specific combinations between TNFα, IL-1β and IFNγ up-regulated the proportion of human TNBC cells co-expressing the inhibitory immune checkpoints PD-L1 and PD-L2: TNFα + IL-1β in MDA-MB-231 cells and IFNγ + IL-1β in BT-549 cells; in the latter cells, the process depended entirely on STAT1 activation, with no involvement of p65 (CRISPR-Cas9 experiments). Highly significant associations between the pro-inflammatory cytokines and PD-L1/PD-L2 expression were revealed in the TCGA dataset of basal-like breast cancer patients. In parallel, we found that the pro-inflammatory cytokines regulated the expression of the soluble receptors of tumor necrosis factor α (TNFα), namely sTNFR1 and sTNFR2; moreover, we revealed that sTNFR1 and sTNFR2 serve as anti-metastatic and protective factors in TNBC, reducing the TNFα-induced production of inflammatory pro-metastatic chemokines (CXCL8, CXCL1, CCL5) by TNBC cells. Importantly, we found that in the context of inflammatory stimulation and also without exposure to pro-inflammatory cytokines, elevated levels of PD-L1 have down-regulated the production of anti-tumor sTNFR1 and sTNFR2. These findings suggest that in addition to its immune-suppressive activities, PD-L1 may promote disease course in TNBC by inhibiting the protective effects of sTNFR1 and sTNFR2.
Collapse
Affiliation(s)
| | | | | | | | - Adit Ben-Baruch
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (T.B.); (N.O.); (N.E.); (T.M.)
| |
Collapse
|
65
|
Jia H, Xie X, Wang L, Wang L, Che F. IFN- γ induces PD-L1 through p38/JNK/ERK signaling pathways and counteracts the tumor promoting effect mediated by PD-L1 in Glioblastoma. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:5492602. [PMID: 35814563 PMCID: PMC9259257 DOI: 10.1155/2022/5492602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022]
Abstract
Glioblastoma is the most malignant primary glioma. Conventional treatment methods that include surgery, radiotherapy, and chemotherapy have a limited curative effect on the tumor. With the deepening of molecular biology research, molecular targeted therapy has opened a new era of tumor therapy. Programmed death ligand 1 (PD-L1) has been proved to play a pivotal role in the tumor immune evasion process. Previous studies have confirmed the specific expression of PD-L1 in glioblastoma tissues and cells, but there are few studies on inflammation regulating PD-L1 in glioblastoma. In this study, real-time PCR, flow cytometry, and western blot were applied to detect PD-L1 in glioblastoma cells. Short hairpin RNA was used to knock down PD-L1 in glioblastoma cells. Cell counting kit-8 experiment and wound-healing assay were used to detect the proliferation and migration of glioblastoma cells. Here we demonstrated that PD-L1 was overexpressed in glioblastoma cells, and interferon-gamma (IFN-γ) induces PD-L1 in glioblastoma cells via activating p38/JNK/ERK signaling pathways. To summarize, PD-L1 promotes the occurrence and development of glioblastoma. IFN-γ counteracts the tumor-promoting effects mediated by PD-L1 in glioblastoma. IFN-γ regulates PD-L1 through multiple signaling pathways, but the total effect of IFN-γ-mediated inflammatory signals still need to be further explored in glioblastoma. PD-L1 enhances the proliferation and migration of glioblastoma cells by regulating CDK4, CDK6, MMP-2, and vimentin molecules. Most importantly, targeting PD-L1 can be applied in the treatment of glioblastoma. We speculate that IFN-γ may affect glioblastoma through other pathways, and we will continue to further explore the mechanisms in the future.
Collapse
Affiliation(s)
- Huafang Jia
- Department of Neurology, Linyi People's Hospital, Qingdao University, Qingdao 266071, Shandong, China
| | - Xiaoli Xie
- Central Laboratory, Linyi People's Hospital, Linyi 276000, Shandong, China
| | - Long Wang
- Central Laboratory, Linyi People's Hospital, Linyi 276000, Shandong, China
| | - Lijuan Wang
- Central Laboratory, Linyi People's Hospital, Linyi 276000, Shandong, China
| | - Fengyuan Che
- Department of Neurology, Linyi People's Hospital, Qingdao University, Qingdao 266071, Shandong, China
| |
Collapse
|
66
|
IGF1R acts as a cancer-promoting factor in the tumor microenvironment facilitating lung metastasis implantation and progression. Oncogene 2022; 41:3625-3639. [PMID: 35688943 PMCID: PMC9184253 DOI: 10.1038/s41388-022-02376-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 05/27/2022] [Accepted: 06/01/2022] [Indexed: 11/08/2022]
Abstract
Given the long-term ineffectiveness of current therapies and late-stage diagnoses, lung cancer is a leading cause of malignant diseases. Tumor progression is influenced by cancer cell interactions with the tumor microenvironment (TME). Insulin-like growth factor 1 receptor (IGF1R) was reported to affect the TME; however, the role of IGF1R in lung TME has not been investigated. First, we assessed IGF1R genomic alterations and expression in NSCLC patient tissue samples, as well as IGF1R serum levels. Next, we performed tumor heterotopic transplantation and pulmonary metastases in IGF1R-deficient mice using melanoma and Lewis lung carcinoma (LLC) cells. Herein we report increased amplification and mRNA expression, as well as increased protein expression (IGF1R/p-IGF1R) and IGF1R levels in tumor samples and serum from NSCLC patients, respectively. Moreover, IGF1R deficiency in mice reduced tumor growth, proliferation, inflammation and vascularization, and increased apoptosis after tumor heterotopic transplantation. Following induction of lung metastasis, IGF1R-deficient lungs also demonstrated a reduced tumor burden, and decreased expression of tumor progression markers, p-IGF1R and p-ERK1/2. Additionally, IGF1R-deficient lungs showed increased apoptosis and diminished proliferation, vascularization, EMT and fibrosis, along with attenuated inflammation and immunosuppression. Accordingly, IGF1R deficiency decreased expression of p-IGF1R in blood vessels, fibroblasts, tumor-associated macrophages and FOXP3+ tumor-infiltrating lymphocytes. Our results demonstrate that IGF1R promotes metastatic tumor initiation and progression in lung TME. Furthermore, our research indicates that IGF1R could be a potential biomarker for early prediction of drug response and clinical evolution in NSCLC patients.
Collapse
|
67
|
Nayak D, Weadick B, Persaud AK, Raj R, Shakya R, Li J, Campbell MJ, Govindarajan R. EMT alterations in the solute carrier landscape uncover SLC22A10/A15 imposed vulnerabilities in pancreatic cancer. iScience 2022; 25:104193. [PMID: 35479410 PMCID: PMC9036131 DOI: 10.1016/j.isci.2022.104193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 01/31/2022] [Accepted: 03/30/2022] [Indexed: 11/29/2022] Open
Abstract
The involvement of membrane-bound solute carriers (SLCs) in neoplastic transdifferentiation processes is poorly defined. Here, we examined changes in the SLC landscape during epithelial-mesenchymal transition (EMT) of pancreatic cancer cells. We show that two SLCs from the organic anion/cation transporter family, SLC22A10 and SLC22A15, favor EMT via interferon (IFN) α and γ signaling activation of receptor tyrosine kinase-like orphan receptor 1 (ROR1) expression. In addition, SLC22A10 and SLC22A15 allow tumor cell accumulation of glutathione to support EMT via the IFNα/γ-ROR1 axis. Moreover, a pan-SLC22A inhibitor lesinurad reduces EMT-induced metastasis and gemcitabine chemoresistance to prolong survival in mouse models of pancreatic cancer, thus identifying new vulnerabilities for human PDAC.
Collapse
Affiliation(s)
- Debasis Nayak
- Division of Pharmaceutics and Pharmacology, The Ohio State University College of Pharmacy, Columbus, OH 43210, USA
| | - Brenna Weadick
- Division of Pharmaceutics and Pharmacology, The Ohio State University College of Pharmacy, Columbus, OH 43210, USA
| | - Avinash K. Persaud
- Division of Pharmaceutics and Pharmacology, The Ohio State University College of Pharmacy, Columbus, OH 43210, USA
| | - Radhika Raj
- Division of Pharmaceutics and Pharmacology, The Ohio State University College of Pharmacy, Columbus, OH 43210, USA
| | - Reena Shakya
- Target Validation Shared Resource, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Junan Li
- The Ohio State University College of Pharmacy, Columbus, OH 43210, USA
| | - Moray J. Campbell
- Molecular Carcinogenesis and Chemoprevention Program, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
- Biomedical Informatics Shared Resource, The Ohio State University, Columbus, OH 43210, USA
| | - Rajgopal Govindarajan
- Division of Pharmaceutics and Pharmacology, The Ohio State University College of Pharmacy, Columbus, OH 43210, USA
- Translational Therapeutics, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
68
|
Mo X, Kazmi HR, Preston-Alp S, Zhou B, Zaidi MR. Interferon-gamma induces melanogenesis via post-translational regulation of tyrosinase. Pigment Cell Melanoma Res 2022; 35:342-355. [PMID: 35266648 PMCID: PMC9050958 DOI: 10.1111/pcmr.13036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 02/02/2022] [Accepted: 02/24/2022] [Indexed: 11/26/2022]
Abstract
Melanogenesis (melanin pigment production) in melanocytes is canonically stimulated by the alpha melanocyte stimulating hormone (αMSH), which activates the cyclic-AMP-mediated expression of the melanocyte inducing transcription factor (MITF) and its downstream melanogenic genes, including the principal rate-limiting melanogenic enzyme tyrosinase (TYR). Here, we report that interferon-gamma (IFNG; type II interferon), but not interferon-alpha (a type I interferon), induces a noncanonical melanogenic pathway in mouse and human melanocytic cells. Inhibition of IFNG pathway by the JAK1/2 inhibitor ruxolitinib or knocking out Stat1 gene abrogated the IFNG-induced melanogenesis. Interestingly, IFNG-induced melanogenesis was independent of MITF. IFNG markedly increased the TYR protein expression but did not affect the mRNA expression, suggesting a post-translational regulatory mechanism. In contrast, IFNG had no effect on the expression of other melanogenesis-related proteins, for example, tyrosinase-related protein 1 (TYRP1) and dopachrome tautomerase (DCT). Glycosidase digestion assays revealed that IFNG treatment increased the mature glycosylated form of TYR, but not its de novo synthesis. Moreover, cycloheximide chase assay showed that degradation of TYR was decreased in IFNG-treated cells. These results suggest that the IFNG-STAT1 pathway regulates melanogenesis via regulation of the post-translational processing and protein stability of TYR.
Collapse
Affiliation(s)
- Xuan Mo
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Hasan Raza Kazmi
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Sarah Preston-Alp
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Bo Zhou
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - M Raza Zaidi
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
69
|
Immune Checkpoint Inhibitors in Cancer Therapy. Curr Oncol 2022; 29:3044-3060. [PMID: 35621637 PMCID: PMC9139602 DOI: 10.3390/curroncol29050247] [Citation(s) in RCA: 544] [Impact Index Per Article: 181.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
The discovery of immune checkpoint proteins such as PD-1/PDL-1 and CTLA-4 represents a significant breakthrough in the field of cancer immunotherapy. Therefore, humanized monoclonal antibodies, targeting these immune checkpoint proteins have been utilized successfully in patients with metastatic melanoma, renal cell carcinoma, head and neck cancers and non-small lung cancer. The US FDA has successfully approved three different categories of immune checkpoint inhibitors (ICIs) such as PD-1 inhibitors (Nivolumab, Pembrolizumab, and Cemiplimab), PDL-1 inhibitors (Atezolimumab, Durvalumab and Avelumab), and CTLA-4 inhibitor (Ipilimumab). Unfortunately, not all patients respond favourably to these drugs, highlighting the role of biomarkers such as Tumour mutation burden (TMB), PDL-1 expression, microbiome, hypoxia, interferon-γ, and ECM in predicting responses to ICIs-based immunotherapy. The current study aims to review the literature and updates on ICIs in cancer therapy.
Collapse
|
70
|
Todorović-Raković N, Milovanović J, Greenman J, Radulovic M. The prognostic significance of serum interferon-gamma (IFN-γ) in hormonally dependent breast cancer. Cytokine 2022; 152:155836. [PMID: 35219004 DOI: 10.1016/j.cyto.2022.155836] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/15/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Interferon-γ (IFN-γ) is a pleiotropic immunomodulatory cytokine. Because of its contradictory and even dualistic roles in malignancies, its potential as a biomarker remains to be unraveled. AIM To evaluate the prognostic significance of serum IFN-γ in hormonally treated breast cancer patients. MATERIAL AND METHODS The study included 72 premenopausal breast cancer patients with known clinicopathological characteristics. All patients received adjuvant hormonal therapy based on hormone receptor-positivity. The median follow-up period was 93 months. IFN-γ serum protein levels were determined by quantitative ELISA. Prognostic performance was evaluated by the receiver operating characteristic (ROC), Cox proportional hazards regression and Kaplan-Meier analyses. Classification of patients into IFN-γlow and IFN-γhigh subgroups was performed by the use of the outcome-oriented cut-off point categorization approach. RESULTS The best prognostic performance was achieved by IFN-γ (AUC = 0.24 and p = 0.01 for distant events, AUC = 0.29 and p = 0.01 for local and distant events combined). Age and IFN-γ were prognostically significant in instances of all types of outcomes and IFN-γ was the independent prognostic parameter (Cox regression). There was a significant difference between IFN-γ values of patients without any events and those with distant metastases (Mann-Whitney test, p = 0.007). IFN-γ levels correlated significantly with nodal status and tumor stage (Spearman's rank order, r = -0.283 and r = -0.238, respectively). Distant recurrence incidence was 4% for the IFN-γhigh subgroup and 33% for the IFN-γlow subgroup (Kaplan-Meier analysis). CONCLUSIONS Raised serum IFN-γ levels associate independently with favorable disease outcome in hormonally dependent breast cancer.
Collapse
Affiliation(s)
- Nataša Todorović-Raković
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia.
| | - Jelena Milovanović
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia.
| | - John Greenman
- Department of Biomedical Sciences, University of Hull, Hull, UK.
| | - Marko Radulovic
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia.
| |
Collapse
|
71
|
Single-cell transcriptome analysis reveals three sequential phases of gene expression during zebrafish sensory hair cell regeneration. Dev Cell 2022; 57:799-819.e6. [PMID: 35316618 PMCID: PMC9188816 DOI: 10.1016/j.devcel.2022.03.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/19/2021] [Accepted: 02/28/2022] [Indexed: 12/20/2022]
Abstract
Loss of sensory hair cells (HCs) in the mammalian inner ear leads to permanent hearing and vestibular defects, whereas loss of HCs in zebrafish results in their regeneration. We used single-cell RNA sequencing (scRNA-seq) to characterize the transcriptional dynamics of HC regeneration in zebrafish at unprecedented spatiotemporal resolution. We uncovered three sequentially activated modules: first, an injury/inflammatory response and downregulation of progenitor cell maintenance genes within minutes after HC loss; second, the transient activation of regeneration-specific genes; and third, a robust re-activation of developmental gene programs, including HC specification, cell-cycle activation, ribosome biogenesis, and a metabolic switch to oxidative phosphorylation. The results are relevant not only for our understanding of HC regeneration and how we might be able to trigger it in mammals but also for regenerative processes in general. The data are searchable and publicly accessible via a web-based interface.
Collapse
|
72
|
Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B, Sockolosky JT. Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114112. [PMID: 35085624 DOI: 10.1016/j.addr.2022.114112] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Cytokines are a class of potent immunoregulatory proteins that are secreted in response to various stimuli and act locally to regulate many aspects of human physiology and disease. Cytokines play important roles in cancer initiation, progression, and elimination, and thus, there is a long clinical history associated with the use of recombinant cytokines to treat cancer. However, the use of cytokines as therapeutics has been limited by cytokine pleiotropy, complex biology, poor drug-like properties, and severe dose-limiting toxicities. Nevertheless, cytokines are crucial mediators of innate and adaptive antitumor immunity and have the potential to enhance immunotherapeutic approaches to treat cancer. Development of immune checkpoint inhibitors and combination immunotherapies has reinvigorated interest in cytokines as therapeutics, and a variety of engineering approaches are emerging to improve the safety and effectiveness of cytokine immunotherapy. In this review we highlight recent advances in cytokine biology and engineering for cancer immunotherapy.
Collapse
|
73
|
Immunotherapy for Colorectal Cancer: Mechanisms and Predictive Biomarkers. Cancers (Basel) 2022; 14:cancers14041028. [PMID: 35205776 PMCID: PMC8869923 DOI: 10.3390/cancers14041028] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Late-stage colorectal cancer treatment often involves chemotherapy and radiation that can cause dose-limiting toxicity, and therefore there is great interest in developing targeted therapies for this disease. Immunotherapy is a targeted therapy that uses peptides, cells, antibodies, viruses, or small molecules to engage or train the immune system to kill cancer. Here, we discuss the preclinical and clinical development of immunotherapy for treatment of colorectal cancer and provide an overview of predictive biomarkers for such treatments. We also consider open questions including optimal combination treatments and sensitization of colorectal cancer patients with proficient mismatch repair enzymes. Abstract Though early-stage colorectal cancer has a high 5 year survival rate of 65–92% depending on the specific stage, this probability drops to 13% after the cancer metastasizes. Frontline treatments for colorectal cancer such as chemotherapy and radiation often produce dose-limiting toxicities in patients and acquired resistance in cancer cells. Additional targeted treatments are needed to improve patient outcomes and quality of life. Immunotherapy involves treatment with peptides, cells, antibodies, viruses, or small molecules to engage or train the immune system to kill cancer cells. Preclinical and clinical investigations of immunotherapy for treatment of colorectal cancer including immune checkpoint blockade, adoptive cell therapy, monoclonal antibodies, oncolytic viruses, anti-cancer vaccines, and immune system modulators have been promising, but demonstrate limitations for patients with proficient mismatch repair enzymes. In this review, we discuss preclinical and clinical studies investigating immunotherapy for treatment of colorectal cancer and predictive biomarkers for response to these treatments. We also consider open questions including optimal combination treatments to maximize efficacy, minimize toxicity, and prevent acquired resistance and approaches to sensitize mismatch repair-proficient patients to immunotherapy.
Collapse
|
74
|
Mansourian M, Firoozabadi SMP, Hassan ZM. The investigation of Pulse-Modulated GSM-900 MHz electromagnetic field effects on the electrochemotherapy mechanisms in vivo. Electromagn Biol Med 2022; 41:71-79. [PMID: 34839760 DOI: 10.1080/15368378.2021.2006689] [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: 07/26/2021] [Accepted: 11/07/2021] [Indexed: 10/19/2022]
Abstract
Electrochemotherapy (ECT) as a tumor treatment modality is approved for cutaneous and subcutaneous tumors. The purpose of the present study was to examine the effect of 900 MHz radiofrequency (RF) pulse-modulated by 217 Hz EMFs similar to those emitted by mobile phones on the mechanisms of ECT in vivo including: tumor hypoxia and immune system response, and on tumor volume.4 T1 cells were injected subcutaneously into the right flank of Balb/c mice. The mice were exposed to RF fields at specific absorption rate (SAR) 2 W/kg for 10 min/day and then treated with ECT. Two protocols of ECT were used: ((70 V/cm-5 kHz) and 70 V/cm-4 kHz)). Tumor hypoxia was analyzed through HIF-1α immuonohistochemistry assay. Interleukin 4 (IL-4) and IFN-γ levels were estimated by enzyme-linked immunosorbent assay (ELISA) technique to evaluate immune system response. Also, tumors volume changes were measured for 24 days following the treatment. The results showed that pulse-modulated RF fields could increase hypoxia induced by ECT, significantly (about 13% in ECT (70 V/cm-5 kHz) and 11% in ECT (70 V/cm-4 kHz)). However, these fields did not have significant effect on immune system response (the levels of IL-4 and IFN-γ) and tumor volume changes induced by ECT. Our results indicated that pulse-modulated RF fields could not affect tumor volume changes in ECT with the frequency of 5 kHz and voltage of 70 V/cm efficacy in vivo. However, investigating the role of other environmental intervening factors on this protocol of ECT is recommended in further studies.
Collapse
Affiliation(s)
- Mahsa Mansourian
- Department of Medical Physics, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - S M P Firoozabadi
- Biomedical Engineering, Department of Medical Physics, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Zuhair Mohammad Hassan
- Immunonology, Department of Immunology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
75
|
El Bakary NM, Alsharkawy AZ, Shouaib ZA, Barakat EMS. Immune Stimulating Outcome of Chrysin and γ-Irradiation via Apoptotic Activation Against Solid Ehrlich Carcinoma Bearing Mice. Integr Cancer Ther 2022; 21:15347354221096668. [PMID: 35543434 PMCID: PMC9102206 DOI: 10.1177/15347354221096668] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The rising interest in innovative methods of cancer immunotherapy has prompted research into the immunomodulatory mechanisms of natural and synthetic substances. The goal of this study was to assess chrysin immune-stimulating and pro-apoptotic effects on tumor growth and cell susceptibility to ionizing radiation in order to improve cancer therapy. Chrysin (20 mg/kg/day) was intraperitoneally injected to mice bearing 1 cm3 solid tumor of Ehrlich ascites carcinoma (EAC) for 21 consecutive days. Mice were whole body exposed to 1 Gy of gamma radiation (2 fractionated dose 0.5 Gy each). Treatment with chrysin dramatically reduces tumor proliferation in EAC mice; furthermore, IFN-γ activity is significantly reduced when compared to EAC mice. When compared to EAC mice, the expression of TNF-α, free radicals, and nitric oxide (NO) levels were considerably reduced, along with improvements in apoptotic regulators (caspase-3 activity). Moreover, the histopathological investigation confirms the improvement exerted by chrysin even in the EAC mice group or the EAC + R group. What is more, exposure to gamma radiation sustained the modulatory effect of chrysin on tumor when compared with EAC + Ch mice. Hence, chrysin might represent a potential therapeutic strategy for increasing the radiation response of solid tumor.
Collapse
Affiliation(s)
- Nermeen M El Bakary
- National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | | | | | | |
Collapse
|
76
|
Padmanabhan S, Gaire B, Zou Y, Uddin MM, DeLeon D, Vancurova I. IFNγ induces JAK1/STAT1/p65 NFκB-dependent interleukin-8 expression in ovarian cancer cells, resulting in their increased migration. Int J Biochem Cell Biol 2021; 141:106093. [PMID: 34626802 PMCID: PMC8639749 DOI: 10.1016/j.biocel.2021.106093] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/20/2021] [Accepted: 10/04/2021] [Indexed: 01/05/2023]
Abstract
Interferon-γ (IFNγ) is a pleiotropic cytokine that has a crucial role in immune response and tumor immunity. Because of its anti-tumor effects, IFNγ has been used in cancer treatment. However, IFNγ also has tumor-promoting functions that are less well understood. Here, we show that IFNγ induces expression of the pro-inflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8) in ovarian cancer (OC) cells. The IFNγ-induced IL-8 expression is dependent on JAK1, STAT1, and p65 NFκB, and is associated with an increased occupancy of K314/315 acetylated p65 NFκB and Ser-727 phosphorylated STAT1 at the IL-8 promoter. Neutralization of IL-8 using anti-IL-8 antibody reduces IFNγ-induced migration of OC cells, and their invasion ability in 3D spheroids. Together, these findings identify IL-8 as a novel target induced by IFNγ/JAK1/STAT1/p65 NFκB signaling, and indicate that the IFNγ-induced IL-8 contributes to IFNγ pro-tumorigenic effects in ovarian cancer cells.
Collapse
Affiliation(s)
- Sveta Padmanabhan
- Department of Biological Sciences, St. John's University, NY 11439, USA
| | - Bijaya Gaire
- Department of Biological Sciences, St. John's University, NY 11439, USA
| | - Yue Zou
- Department of Biological Sciences, St. John's University, NY 11439, USA
| | - Mohammad M Uddin
- Department of Biological Sciences, St. John's University, NY 11439, USA
| | - Daniel DeLeon
- Department of Biological Sciences, St. John's University, NY 11439, USA
| | - Ivana Vancurova
- Department of Biological Sciences, St. John's University, NY 11439, USA.
| |
Collapse
|
77
|
Ikewaki N, Dedeepiya VD, Raghavan K, Rao KS, Vaddi S, Osawa H, Kisaka T, Kurosawa G, Srinivasan S, Kumar SRB, Senthilkumar R, Iwasaki M, Preethy S, Abraham SJK. β‑glucan vaccine adjuvant approach for cancer treatment through immune enhancement (B‑VACCIEN) in specific immunocompromised populations (Review). Oncol Rep 2021; 47:14. [PMID: 34779494 DOI: 10.3892/or.2021.8225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/07/2021] [Indexed: 11/06/2022] Open
Abstract
The incidence of cancer, which is the second leading cause of mortality globally, continues to increase, although continued efforts are being made to identify effective treatments with fewer side‑effects. Previous studies have reported that chronic microinflammation, which occurs in diseases, including diabetes, along with weakened immune systems, may ultimately lead to cancer development. Chemotherapy, radiotherapy and surgery are the mainstream approaches to treatment; however, they all lead to immune system weakness, which in turn increases the metastatic spread. The aim of the present review was to provide evidence of a biological response modifier β‑glucan [β‑glucan vaccine adjuvant approach to treating cancer via immune enhancement (B‑VACCIEN)] and its beneficial effects, including vaccine‑adjuvant potential, balancing metabolic parameters (including blood glucose and lipid levels), increasing peripheral blood cell cytotoxicity against cancer and alleviating chemotherapy side effects in animal models. This suggests its value as a potential strategy to provide long‑term prophylaxis in immunocompromised individuals or genetically prone to cancer.
Collapse
Affiliation(s)
- Nobunao Ikewaki
- Department of Medical Life Science, Kyushu University of Health and Welfare, Nobeoka, Miyazaki 882‑8508, Japan
| | | | - Kadalraja Raghavan
- Department of Paediatric Neurology, Kenmax Medical Service Private Limited, Tallakulam, Madurai 625002, India
| | - Kosagi-Sharaf Rao
- Institute of Scientific Research and High Technology Services of Panama (INDICASAT‑AIP), Clayton 88888, Republic of Panama
| | - Suryaprakash Vaddi
- Department of Urology, Yashoda Hospitals, Hyderabad, Telangana 50008, India
| | - Hiroshi Osawa
- Clinical Services Department, Omote Medical Clinic, Chiba 296‑8602, Japan
| | - Tomohiko Kisaka
- Division of Biodesign, Office of Research and Academic‑Government‑Community Collaboration, Hiroshima University, Higashihiroshima, Hiroshima 739‑8511, Japan
| | - Gene Kurosawa
- Department of Academic Research Support Promotion Facility, Center for Research Promotion and Support, Fujita Health University, Toyoake, Aichi 470‑1192, Japan
| | - Subramaniam Srinivasan
- The Mary‑Yoshio Translational Hexagon (MYTH), Nichi‑In Centre for Regenerative Medicine (NCRM), Chennai 600034, India
| | | | - Rajappa Senthilkumar
- The Fujio‑Eiji Academic Terrain (FEAT), Nichi‑In Centre for Regenerative Medicine (NCRM), Chennai 600034, India
| | - Masaru Iwasaki
- Centre for Advancing Clinical Research (CACR), University of Yamanashi‑ School of Medicine, Chuo, Yamanashi 409‑3898, Japan
| | - Senthilkumar Preethy
- The Fujio‑Eiji Academic Terrain (FEAT), Nichi‑In Centre for Regenerative Medicine (NCRM), Chennai 600034, India
| | - Samuel J K Abraham
- The Mary‑Yoshio Translational Hexagon (MYTH), Nichi‑In Centre for Regenerative Medicine (NCRM), Chennai 600034, India
| |
Collapse
|
78
|
Cho JS, Seon K, Kim MY, Kim SW, Yoo YC. Effects of Perioperative Dexmedetomidine on Immunomodulation in Uterine Cancer Surgery: A Randomized, Controlled Trial. Front Oncol 2021; 11:749003. [PMID: 34868950 PMCID: PMC8635094 DOI: 10.3389/fonc.2021.749003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/28/2021] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE Dexmedetomidine has sympatholytic, anti-inflammatory, and analgesic effects and may exert anti-tumor effect by acting on α2A adrenoreceptor. We investigated whether perioperative dexmedetomidine preserves immune function in patients undergoing uterine cancer surgery. METHODS One hundred patients were randomly assigned to the control or dexmedetomidine groups (50 patients each). Dexmedetomidine was infused at rates of 0.4 μg/kg/h intraoperatively and 0.15 μg/kg/h during the first 24 h postoperatively. The primary outcome was natural killer (NK) cell activity, which was measured preoperatively and 1, 3, and 5 days postoperatively. The inflammatory response was measured by interleukin-6, interferon-γ, and neutrophil/lymphocyte ratio, and pain scores and opioid consumption were assessed. Cancer recurrence or metastasis and death were evaluated 2 years postoperatively. RESULTS NK cell activity decreased postoperatively in both groups and changes over time were not different between groups (P=0.496). Interferon-γ increased postoperatively in the dexmedetomidine group, whereas it maintained at the baseline value in the control group. Change in interferon-γ differed significantly between groups (P=0.003). Changes in interleukin-6 and neutrophil-lymphocyte ratio were comparable between groups. Both pain score with activity during the first 1 h and opioid consumption during the first 1-24 h postoperatively were lower in the dexmedetomidine group. Rates of cancer recurrence/metastasis (16.3% vs. 8.7%, P=0.227) and death within 2 years postoperatively (6.7% vs. 2.2%, P=0.318) were not different between groups. CONCLUSIONS Perioperative dexmedetomidine had no favorable impacts on NK cell activity, inflammatory responses, or prognosis, whereas it increased interferon-γ and reduced early postoperative pain severity and opioid consumption in uterine cancer surgery patients.
Collapse
Affiliation(s)
- Jin Sun Cho
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Kieun Seon
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Institute of Women’s Life Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Min-Yu Kim
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Wun Kim
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Institute of Women’s Life Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Young Chul Yoo
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
79
|
Wang Y, Xu L, Zhao W, Chen X, Wen L, Duan W, Yu X, De Zhou F, Liu Y, Hao J, Huang X, Lu J, Ge Q. T cell landscape and dynamics in immunoglobulin light chain amyloidosis before and after daratumumab-based therapy. Clin Transl Med 2021; 11:e582. [PMID: 34845849 PMCID: PMC8630449 DOI: 10.1002/ctm2.582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/05/2021] [Accepted: 09/10/2021] [Indexed: 11/07/2022] Open
Abstract
Amyloid light-chain (AL) is characterized by the presence of small, poorly proliferating plasma cell clones with the production and deposition of light chains into tissues. T cell changes within the tumour microenvironment in AL are poorly understood. By sequencing at a single-cell level of CD3+ T cells purified from bone marrow (BM) and blood of newly diagnosed AL patients before and after a combination of daratumumab with cyclophosphamide, bortezomib, and dexamethasone (Dara-BCD), we analysed the transcriptomic features of T cells and found an expansion, activation and type I cytokine upregulation in BM and circulating T cells after the treatment. More prominent changes were shown in CD8+ T cells. In particular, we found the presence of CD8+ BM resident memory T cells (TRM ) with high expression of inhibitory molecules in AL patients at diagnosis. After Dara-BCD, these TRM cells were quickly activated with downregulation of suppressive molecules and upregulation of IFNG expression. These data collectively demonstrate that Dara-based therapy in patients with AL amyloidosis promotes anti-tumour T cell responses. The similar transcriptomic features of BM and circulating T cells before and after therapy further provide a less invasive approach for molecular monitoring of T cell response in AL amyloidosis.
Collapse
Affiliation(s)
- Yujia Wang
- Department of ImmunologySchool of Basic Medical SciencesPeking University. NHC Key Laboratory of Medical Immunology (Peking University)BeijingChina
| | - Lushuang Xu
- Department of ImmunologySchool of Basic Medical SciencesPeking University. NHC Key Laboratory of Medical Immunology (Peking University)BeijingChina
| | - Weijia Zhao
- Department of ImmunologySchool of Basic Medical SciencesPeking University. NHC Key Laboratory of Medical Immunology (Peking University)BeijingChina
| | | | - Lei Wen
- Beijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's Hospital & Institute of HematologyBeijingChina
| | - Wenbing Duan
- Beijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's Hospital & Institute of HematologyBeijingChina
| | - Xiao‐Juan Yu
- Renal DivisionDepartment of MedicineInstitute of NephrologyPeking University First Hospital, & Renal Pathology CenterPeking UniversityBeijingChina
- Renal Pathology CenterInstitute of NephrologyPeking UniversityBeijingChina
- Key Laboratory of Renal DiseaseMinistry of Health of ChinaBeijingChina
- Key Laboratory of CKD Prevention and TreatmentMinistry of Education of ChinaBeijingChina
| | - Fu‐ De Zhou
- Renal DivisionDepartment of MedicineInstitute of NephrologyPeking University First Hospital, & Renal Pathology CenterPeking UniversityBeijingChina
- Renal Pathology CenterInstitute of NephrologyPeking UniversityBeijingChina
- Key Laboratory of Renal DiseaseMinistry of Health of ChinaBeijingChina
- Key Laboratory of CKD Prevention and TreatmentMinistry of Education of ChinaBeijingChina
| | - Yang Liu
- Beijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's Hospital & Institute of HematologyBeijingChina
| | - Jie Hao
- Department of ImmunologySchool of Basic Medical SciencesPeking University. NHC Key Laboratory of Medical Immunology (Peking University)BeijingChina
| | - Xiaojun Huang
- Beijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's Hospital & Institute of HematologyBeijingChina
| | - Jin Lu
- Beijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's Hospital & Institute of HematologyBeijingChina
- Collaborative Innovation Center of HaematologySoochow UniversitySuzhouJiangsuChina
| | - Qing Ge
- Department of ImmunologySchool of Basic Medical SciencesPeking University. NHC Key Laboratory of Medical Immunology (Peking University)BeijingChina
- Department of Integration of Chinese and Western MedicineSchool of Basic Medical SciencesPeking UniversityBeijingChina
- National Key Laboratory of Human Factors EngineeringChina Astronauts Research and Training CenterBeijingChina
| |
Collapse
|
80
|
Alphavirus-Driven Interferon Gamma (IFNg) Expression Inhibits Tumor Growth in Orthotopic 4T1 Breast Cancer Model. Vaccines (Basel) 2021; 9:vaccines9111247. [PMID: 34835178 PMCID: PMC8620866 DOI: 10.3390/vaccines9111247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/10/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022] Open
Abstract
Interferon gamma (IFNg) is a pleiotropic cytokine that can potentially reprogram the tumor microenvironment; however, the antitumor immunomodulatory properties of IFNg still need to be validated due to variable therapeutic outcomes in preclinical and clinical studies. We developed a replication-deficient Semliki Forest virus vector expressing IFNg (SFV/IFNg) and evaluated its immunomodulatory antitumor potential in vitro in a model of 3D spheroids and in vivo in an immunocompetent 4T1 mouse breast cancer model. We demonstrated that SFV-derived, IFN-g-stimulated bone marrow macrophages can be used to acquire the tumoricidal M1 phenotype in 3D nonattached conditions. Coculturing SFV/IFNg-infected 4T1 spheroids with BMDMs inhibited spheroid growth. In the orthotopic 4T1 mouse model, intratumoral administration of SFV/IFNg virus particles alone or in combination with the Pam3CSK4 TLR2/1 ligand led to significant inhibition of tumor growth compared to the administration of the control SFV/Luc virus particles. Analysis of the composition of intratumoral lymphoid cells isolated from tumors after SFV/IFNg treatment revealed increased CD4+ and CD8+ and decreased T-reg (CD4+/CD25+/FoxP3+) cell populations. Furthermore, a significant decrease in the populations of cells bearing myeloid cell markers CD11b, CD38, and CD206 was observed. In conclusion, the SFV/IFNg vector induces a therapeutic antitumor T-cell response and inhibits myeloid cell infiltration in treated tumors.
Collapse
|
81
|
Nakamura-Shinya Y, Iguchi-Manaka A, Murata R, Sato K, Van Vo A, Kanemaru K, Shibuya A, Shibuya K. DNAM-1 promotes inflammation-driven tumor development via enhancing IFN-γ production. Int Immunol 2021; 34:149-157. [PMID: 34672321 DOI: 10.1093/intimm/dxab099] [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: 07/15/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
DNAM-1 is an activating immunoreceptor on T cells and natural killer (NK) cells. Expression levels of its ligands, CD155 and CD112, are upregulated on tumor cells. The interaction of DNAM-1 on CD8 + T cells and NK cells with the ligands on tumor cells plays an important role in tumor immunity. We previously reported that mice deficient in DNAM-1 showed accelerated growth of tumors induced by the chemical carcinogen 7,12-dimethylbenz[a]anthracene (DMBA). Contrary to those results, we show here that tumor development induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) together with DMBA was suppressed in DNAM-1-deficient mice. In this model, DNAM-1 enhanced IFN-γ secretion from conventional CD4 + T cells to promote inflammation-related tumor development. These findings suggest that, under inflammatory conditions, DNAM-1 contributes to tumor development via conventional CD4 + T cells.
Collapse
Affiliation(s)
- Yuho Nakamura-Shinya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.,Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Akiko Iguchi-Manaka
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.,Breast and Endocrine Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Rikito Murata
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.,Ph.D. Program in Human Biology, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kazuki Sato
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.,R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Anh Van Vo
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kazumasa Kanemaru
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.,R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.,R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kazuko Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.,R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| |
Collapse
|
82
|
Johns DN, Lucas CG, Pfeiffer CA, Chen PR, Meyer AE, Perry SD, Spate LD, Cecil RF, Fudge MA, Samuel MS, Spinka CM, Liu H, Lucy MC, Wells KD, Prather RS, Spencer TE, Geisert RD. Conceptus interferon gamma is essential for establishment of pregnancy in the pig. Biol Reprod 2021; 105:1577-1590. [PMID: 34608481 DOI: 10.1093/biolre/ioab186] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/10/2021] [Accepted: 09/28/2021] [Indexed: 01/04/2023] Open
Abstract
Establishment and maintenance of pregnancy in the pig is a complex process that relies on conceptus regulation of the maternal proinflammatory response to endometrial attachment. Following elongation, pig conceptuses secrete interferon gamma (IFNG) during attachment to the endometrial luminal epithelium. The objective here was to determine if conceptus production of IFNG is important for early development and establishment of pregnancy. CRISPR/Cas9 gene editing and somatic cell nuclear transfer technologies were used to create an IFNG loss-of-function study in pigs. Wild-type (IFNG+/+) and null (IFNG-/-) fibroblast cells were used to create embryos through somatic cell nuclear transfer. IFNG expression was not detected in IFNG-/- conceptuses on either day 15 or day 17 of pregnancy. Ablation of conceptus IFNG production resulted in the reduction of stromal CD3+ and mast cells which localized to the site of conceptus attachment on day 15. The uteri of recipients with IFNG-/- conceptuses were inflamed, hyperemic and there was an abundance of erythrocytes in the uterine lumen associated with the degenerating conceptuses. The endometrial stromal extracellular matrix was altered in the IFNG-/- embryo pregnancies and there was an increased endometrial mRNA levels for collagen XVII (COL17A1), matrilin 1 (MATN1), secreted phosphoprotein 1 (SPP1) and cysteine-rich secretory protein 3 (CRISP3), which are involved with repair and remodeling of the extracellular matrix. These results indicate conceptus IFNG production is essential in modulating the endometrial proinflammatory response for conceptus attachment and survival in pigs.
Collapse
Affiliation(s)
- Destiny N Johns
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Caroline G Lucas
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | | | - Paula R Chen
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Ashley E Meyer
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Shelbi D Perry
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Lee D Spate
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Raissa F Cecil
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Melissa A Fudge
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Melissa S Samuel
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | | | - Hongyu Liu
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Matthew C Lucy
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Kevin D Wells
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Randall S Prather
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Thomas E Spencer
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Rodney D Geisert
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| |
Collapse
|
83
|
Baxter MA, Middleton F, Cagney HP, Petty RD. Resistance to immune checkpoint inhibitors in advanced gastro-oesophageal cancers. Br J Cancer 2021; 125:1068-1079. [PMID: 34230609 PMCID: PMC8505606 DOI: 10.1038/s41416-021-01425-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/17/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have altered the treatment paradigm across a range of tumour types, including gastro-oesophageal cancers. For patients with any cancer type who respond, ICIs can confer long-term disease control and significantly improve survival and quality of life, but for patients with gastro-oesophageal cancer, ICIs can be transformative, as durable responses in advanced disease have hitherto been rare, especially in those patients who are resistant to first-line cytotoxic therapies. Results from trials in patients with advanced-stage gastro-oesophageal cancer have raised hopes that ICIs will be successful as adjuvant and neoadjuvant treatments in early-stage disease, when the majority of patients relapse after potential curative treatments, and several trials are ongoing. Unfortunately, however, ICI-responding patients appear to constitute a minority subgroup within gastro-oesophageal cancer, and resistance to ICI therapy (whether primary or acquired) is common. Understanding the biological mechanisms of ICI resistance is a current major research challenge and involves investigation of both tumour and patient-specific factors. In this review, we discuss the mechanisms underlying ICI resistance and their potential specific applications of this knowledge towards precision medicine strategies in the management of gastro-oesophageal cancers in clinical practice.
Collapse
Affiliation(s)
- Mark A Baxter
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK.
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee, UK.
| | - Fearghas Middleton
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee, UK
| | - Hannah P Cagney
- School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Russell D Petty
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK.
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee, UK.
| |
Collapse
|
84
|
Kang M, Lee SH, Kwon M, Byun J, Kim D, Kim C, Koo S, Kwon SP, Moon S, Jung M, Hong J, Go S, Song SY, Choi JH, Hyeon T, Oh YK, Park HH, Kim BS. Nanocomplex-Mediated In Vivo Programming to Chimeric Antigen Receptor-M1 Macrophages for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2103258. [PMID: 34510559 DOI: 10.1002/adma.202103258] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/06/2021] [Indexed: 06/13/2023]
Abstract
Chimeric antigen receptor-T (CAR-T) cell immunotherapy has shown impressive clinical outcomes for hematologic malignancies. However, its broader applications are challenged due to its complex ex vivo cell-manufacturing procedures and low therapeutic efficacy against solid tumors. The limited therapeutic effects are partially due to limited CAR-T cell infiltration to solid tumors and inactivation of CAR-T cells by the immunosuppressive tumor microenvironment. Here, a facile approach is presented to in vivo program macrophages, which can intrinsically penetrate solid tumors, into CAR-M1 macrophages displaying enhanced cancer-directed phagocytosis and anti-tumor activity. In vivo injected nanocomplexes of macrophage-targeting nanocarriers and CAR-interferon-γ-encoding plasmid DNA induce CAR-M1 macrophages that are capable of CAR-mediated cancer phagocytosis, anti-tumor immunomodulation, and inhibition of solid tumor growth. Together, this study describes an off-the-shelf CAR-macrophage therapy that is effective for solid tumors and avoids the complex and costly processes of ex vivo CAR-cell manufacturing.
Collapse
Affiliation(s)
- Mikyung Kang
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seong Ho Lee
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Miji Kwon
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Junho Byun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dongyoon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sagang Koo
- Center for Nanoparticle Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seuk Young Song
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jae Hyun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
- Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, 04763, Republic of Korea
| | - Byung-Soo Kim
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Chemical Processes, Institute of Engineering Research, BioMAX, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
85
|
Todorović-Raković N, Whitfield JR. Between immunomodulation and immunotolerance: The role of IFNγ in SARS-CoV-2 disease. Cytokine 2021; 146:155637. [PMID: 34242899 PMCID: PMC8253693 DOI: 10.1016/j.cyto.2021.155637] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/21/2022]
Abstract
Interferons have prominent roles in various pathophysiological conditions, mostly related to inflammation. Interferon-gamma (IFNγ) was, initially discovered as a potent antiviral agent, over 50 years ago, and has recently garnered renewed interest as a promising factor involved in both innate and adaptive immunity. When new disease epidemics appear such as SARS-CoV (severe acute respiratory syndrome coronavirus), MERS-CoV (Middle East respiratory syndrome coronavirus), IAV (Influenza A virus), and in particular the current SARS-CoV-2 pandemic, it is especially timely to review the complexity of immune system responses to viral infections. Here we consider the controversial roles of effectors like IFNγ, discussing its actions in immunomodulation and immunotolerance. We explore the possibility that modulation of IFNγ could be used to influence the course of such infections. Importantly, not only could endogenous expression of IFNγ influence the outcome, there are existing IFNγ therapeutics that can readily be applied in the clinic. However, our understanding of the molecular mechanisms controlled by IFNγ suggests that the exact timing for application of IFNγ-based therapeutics could be crucial: it should be earlier to significantly reduce the viral load and thus decrease the overall severity of the disease.
Collapse
Affiliation(s)
- Nataša Todorović-Raković
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia.
| | - Jonathan R Whitfield
- Vall d'Hebron Institute of Oncology (VHIO), Carrer Natzaret 115, Vall d'Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| |
Collapse
|
86
|
Lee KS, Chung WY, Park JE, Jung YJ, Park JH, Sheen SS, Park KJ. Interferon-γ-Inducible Chemokines as Prognostic Markers for Lung Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18179345. [PMID: 34501934 PMCID: PMC8431216 DOI: 10.3390/ijerph18179345] [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] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 11/16/2022]
Abstract
Interferon (IFN)-γ-inducible chemokines in the CXCR3/ligand axis are involved in cell-mediated immunity and play a significant role in the progression of cancer. We enrolled patients with lung cancer (n = 144) and healthy volunteers as the controls (n = 140). Initial blood samples were collected and concentrations of IFN-γ and IFN-γ-inducible chemokines CXCL9, CXCL10, and CXCL11 were measured using enzyme-linked immunosorbent assay. Of patients with lung cancer, 125 had non-small cell lung cancer (NSCLC) and 19 had small cell lung cancer. The area under the curve (AUC) (95% CI) of CXCL9 was 0.83 (0.80-0.89) for differentiating lung cancer patients from controls. The levels of all the markers were significantly higher in NSCLC patients with stage IV than in those with stages I-III. A Kaplan-Meier survival analysis showed that NSCLC cancer patients with higher levels of all markers showed poorer survival than those with lower levels. In Cox multivariate analysis of patients with NSCLC, independent prognostic factors for overall survival were CXCL9 and CXCL11. CXCL9 was the only independent prognostic factor for cancer-specific survival. Serum IFN-γ-inducible chemokines may be useful as clinical markers of metastasis and prognosis in NSCLC, and CXCL9 levels showed the most significant results.
Collapse
|
87
|
Dobson HE, Ruan S, Chang AE, Wicha MS, Li Q. Targeting cancer stem cells via integrin β4. Oncotarget 2021; 12:1850-1858. [PMID: 34504657 PMCID: PMC8416562 DOI: 10.18632/oncotarget.27977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Integrins mediate cell-cell interactions and communication with the extracellular matrix (ECM). These transmembrane protein receptors allow binding between a cell and its surroundings, initiating a breadth of intracellular signaling resulting in proliferation, differentiation, survival, or migration. Such responses have made integrins an attractive target for cancer therapy. Self-renewing and highly tumorigenic cancer stem cells (CSCs) are most resistant to traditional radiation treatment and chemotherapy, and therefore may contribute directly to the metastasis and relapse of the disease. In both the 4T1 mouse metastatic mammary tumor model and SCC7 head and neck squamous cell carcinoma model, integrin β4 (ITGB4) was expressed on ALDHhigh 4T1 and SCC7 CSCs. Using two immunological approaches, we targeted ITGB4 through 1) ITGB4 protein-pulsed dendritic cell (ITGB4-DC) vaccination or 2) via anti-CD3/anit-ITGB4 bispecific antibody (ITGB4 BiAb)-armed T cell adoptive transfer. These two therapies reduced ITGB4-expressing CSCs and inhibited local tumor growth and lung metastasis through ITGB4 specific cellular and humoral immune responses. Additionally, the combination of anti-PD-L1 immunotherapy with our two ITGB4-targeted approaches significantly improved treatment efficacy. We also found increased concentrations of serum IFN-γ and IL-6 in the 4T1 and SCC7 models which may help define future directions of this ITGB4-targeted study. Together, these results emphasize ITGB4 as a practical CSC immunological target with possible therapeutic benefits across tumor types with high ITGB4 expression.
Collapse
Affiliation(s)
- Hannah E Dobson
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shasha Ruan
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Alfred E Chang
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Max S Wicha
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Qiao Li
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
88
|
Qi J, Crinier A, Escalière B, Ye Y, Wang Z, Zhang T, Batista L, Liu H, Hong L, Wu N, Zhang M, Chen L, Liu Y, Shen L, Narni-Mancinelli E, Vivier E, Su B. Single-cell transcriptomic landscape reveals tumor specific innate lymphoid cells associated with colorectal cancer progression. CELL REPORTS MEDICINE 2021; 2:100353. [PMID: 34467243 PMCID: PMC8385246 DOI: 10.1016/j.xcrm.2021.100353] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/29/2021] [Accepted: 06/24/2021] [Indexed: 02/08/2023]
Abstract
Innate lymphoid cells (ILCs) are tissue-resident lymphocytes differing from conventional T lymphocytes in having no antigen-specific receptors. ILCs include natural killer (NK) cells, helper-like ILC1s, ILC2s, and ILC3s, and lymphoid tissue-inducer (LTi) cells. Tumor ILCs are frequently found in various cancers, but their roles in cancer immunity and immunotherapy remain largely unclear. We report here the single-cell characterization of blood and gut helper-like ILC subsets in healthy conditions and in colorectal cancer (CRC). The healthy gut contains ILC1s, ILC3s, and ILC3/NKs, but no ILC2s. Additional tumor-specific ILC1-like and ILC2 subsets were identified in CRC patients. Signaling lymphocytic activation molecule family member 1 (SLAMF1) was found to be selectively expressed on tumor-specific ILCs, and higher levels of SLAMF1+ ILCs were observed in the blood of CRC patients. The SLAMF1-high group of CRC patients had a significantly higher survival rate than the SLAMF1-low group, suggesting that SLAMF1 is an anti-tumor biomarker in CRC. Healthy gut contains ILC1s, ILC3s, and ILC3/NKs, but no ILC2s Blood and tumor ILCs from CRC patients have unique transcriptomic features Tumor tissue from CRC patients contains a tumor specific ILC1-like subset and ILC2s SLAMF1 is identified as an anti-tumor biomarker in CRC
Collapse
Affiliation(s)
- Jingjing Qi
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Adeline Crinier
- Aix-Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, 13009 Marseille, France
| | - Bertrand Escalière
- Aix-Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, 13009 Marseille, France
| | - Youqiong Ye
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhengting Wang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tianyu Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Luciana Batista
- Innate Pharma Research Laboratories, Innate Pharma, 13009 Marseille, France
| | - Hongzhi Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Liwen Hong
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ningbo Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Mingnan Zhang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lei Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yingbin Liu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Lei Shen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Emilie Narni-Mancinelli
- Aix-Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, 13009 Marseille, France
| | - Eric Vivier
- Aix-Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, 13009 Marseille, France.,Innate Pharma Research Laboratories, Innate Pharma, 13009 Marseille, France.,Immunology, Marseille Immunopole, Hôpital de la Timone, Assistance Publique des Hôpitaux de 13005 Marseille, France
| | - Bing Su
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
89
|
Fusion Protein Vaccine Based on Ag85B and STEAP1 Induces a Protective Immune Response against Prostate Cancer. Vaccines (Basel) 2021; 9:vaccines9070786. [PMID: 34358202 PMCID: PMC8310044 DOI: 10.3390/vaccines9070786] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 01/05/2023] Open
Abstract
(1) Background: There are currently limited treatments for castration-resistant prostate cancer. Immunotherapy involving Sipuleucel-T has increasingly drawn attention for prostate cancer management. BCG plays a vital role in treating bladder cancer, mainly by inducing immune activation, but is rarely used for prostate cancer. (2) Methods: The TCGA database, PCR, and Western blotting were used to analyze the expression of STEAP1 in mouse and human tissues. Then, we constructed a fusion protein vaccine with Mycobacterium tuberculosis Ag85B and three repeated octapeptide epitopes of a six-transmembrane epithelial antigen of the prostate 1 (STEAP1186-193), Ag85B-3×STEAP1186-193. The uptake of the fusion protein vaccine by DCs was evaluated by confocal microscopy, and DC markers were detected using flow cytometry after incubation with the fusion protein. The immune response against prostate cancer was evaluated by the LDH assay and xenografts in vitro and in vivo. Then, the tumor microenvironment was determined using IHC and ELISA. In addition, the epitope was mutated using CRISPR-Cas9 to illustrate that the fusion protein elicited immunization against STEAP1. (3) Results: The TCGA database analysis, PCR, and Western blotting showed that STEAP1 was highly expressed in human and murine prostate cancer. After the uptake of the purified fusion protein vaccine by DCs, CD11c, CD80, CD86, and MHC II were upregulated and triggered a cytotoxic T lymphocyte (CTL) response against TRAMP-C1 and RM1 cells in vitro. Furthermore, the fusion protein vaccine inhibited tumor growth and improved the tumor microenvironment in vivo, with more CD3+ cells and fewer FOXP3+ cells in the tumor. Serum IFN-γ and IL-2 were significantly higher than in the control group, while IL-4 expression was lower, indicating that the fusion protein vaccine activated Th1 immunity. The immune response against prostate cancer was greatly suppressed when the antigen targets were knocked out using CRISPR-Cas9. (4) Conclusion: In summary, our results provide the first evidence that a vaccine based on a fusion protein consisting of Ag85B and a prostate cancer octapeptide epitope with complete Freund’s adjuvant (CFA), triggers a robust immune response and inhibits tumor growth in murine prostate cancer.
Collapse
|
90
|
Puah BP, Jalil J, Attiq A, Kamisah Y. New Insights into Molecular Mechanism behind Anti-Cancer Activities of Lycopene. Molecules 2021; 26:molecules26133888. [PMID: 34202203 PMCID: PMC8270321 DOI: 10.3390/molecules26133888] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/05/2021] [Accepted: 06/17/2021] [Indexed: 12/15/2022] Open
Abstract
Lycopene is a well-known compound found commonly in tomatoes which brings wide range of health benefits against cardiovascular diseases and cancers. From an anti-cancer perspective, lycopene is often associated with reduced risk of prostate cancer and people often look for it as a dietary supplement which may help to prevent cancer. Previous scientific evidence exhibited that the anti-cancer activity of lycopene relies on its ability to suppress oncogene expressions and induce proapoptotic pathways. To further explore the real potential of lycopene in cancer prevention, this review discusses the new insights and perspectives on the anti-cancer activities of lycopene which could help to drive new direction for research. The relationship between inflammation and cancer is being highlighted, whereby lycopene suppresses cancer via resolution of inflammation are also discussed herein. The immune system was found to be a part of the anti-cancer system of lycopene as it modulates immune cells to suppress tumor growth and progression. Lycopene, which is under the family of carotenoids, was found to play special role in suppressing lung cancer.
Collapse
Affiliation(s)
- Boon-Peng Puah
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Juriyati Jalil
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
- Correspondence: ; Tel.: +603-9289-7533
| | - Ali Attiq
- Faculty of Pharmacy, MAHSA University, Bandar Saujana Putra, Jenjarom 42610, Malaysia;
| | - Yusof Kamisah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
91
|
Tecalco-Cruz AC, Macías-Silva M, Ramírez-Jarquín JO, Méndez-Ambrosio B. Identification of genes modulated by interferon gamma in breast cancer cells. Biochem Biophys Rep 2021; 27:101053. [PMID: 34189281 PMCID: PMC8220005 DOI: 10.1016/j.bbrep.2021.101053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 11/04/2022] Open
Abstract
Interferon gamma (IFNγ) plays a context-dependent dual tumor-suppressor and pro-tumorigenic roles in cancer. IFNγ induces morphological changes in breast cancer (BC) cells with or without estrogen receptor alpha (ERα) expression. However, IFNγ-regulated genes in BC cells remain unexplored. Here, we performed a cDNA microarray analysis of MCF-7 (ERα+) and MDA-MB-231 (HER2-/PR-/ERα-) cells with and without IFNγ treatment. We identified specific IFNγ−modulated genes in each cell type, and a small group of genes regulated by IFNγ common in both cell types. IFNγ treatment for an extended time mainly repressed gene expression shared by both cell types. Nonetheless, some of these IFNγ-repressed genes were seemingly deregulated in human mammary tumor samples, along with decreased IFNGR1 (an IFNγ receptor) expression. Thus, IFNγ signaling-elicited anti-tumor activities may be mediated by the downregulation of main IFNγ target genes in BC; however, it may be deregulated by the tumor microenvironment in a tumor stage-dependent manner. Identification of new potential genes regulated by IFNγ in breast cancer cells. A small group of common genes is regulated by IFNγ in ERα- and ERα+ breast cancer cells. IFNγ treatment for a long time mainly represses gene expression in breast cancer cells. The tumor environment may lead to a decrease in IFNGR1 expression in mammary tumors.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), Ciudad de México, Mexico
| | | | | | | |
Collapse
|
92
|
Morein D, Rubinstein-Achiasaf L, Brayer H, Dorot O, Pichinuk E, Ben-Yaakov H, Meshel T, Pasmanik-Chor M, Ben-Baruch A. Continuous Inflammatory Stimulation Leads via Metabolic Plasticity to a Prometastatic Phenotype in Triple-Negative Breast Cancer Cells. Cells 2021; 10:cells10061356. [PMID: 34072893 PMCID: PMC8229065 DOI: 10.3390/cells10061356] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/23/2021] [Accepted: 05/28/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic inflammation promotes cancer progression by affecting the tumor cells and their microenvironment. Here, we demonstrate that a continuous stimulation (~6 weeks) of triple-negative breast tumor cells (TNBC) by the proinflammatory cytokines tumor necrosis factor α (TNFα) + interleukin 1β (IL-1β) changed the expression of hundreds of genes, skewing the cells towards a proinflammatory phenotype. While not affecting stemness, the continuous TNFα + IL-1β stimulation has increased tumor cell dispersion and has induced a hybrid metabolic phenotype in TNBC cells; this phenotype was indicated by a transcription-independent elevation in glycolytic activity and by increased mitochondrial respiratory potential (OXPHOS) of TNBC cells, accompanied by elevated transcription of mitochondria-encoded OXPHOS genes and of active mitochondria area. The continuous TNFα + IL-1β stimulation has promoted in a glycolysis-dependent manner the activation of p65 (NF-κB), and the transcription and protein expression of the prometastatic and proinflammatory mediators sICAM-1, CCL2, CXCL8 and CXCL1. Moreover, when TNBC cells were stimulated continuously by TNFα + IL-1β in the presence of a glycolysis inhibitor, their conditioned media had reduced ability to recruit monocytes and neutrophils in vivo. Such inflammation-induced metabolic plasticity, which promotes prometastatic cascades in TNBC, may have important clinical implications in treatment of TNBC patients.
Collapse
Affiliation(s)
- Dina Morein
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (D.M.); (L.R.-A.); (H.B.); (H.B.-Y.); (T.M.)
| | - Linor Rubinstein-Achiasaf
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (D.M.); (L.R.-A.); (H.B.); (H.B.-Y.); (T.M.)
| | - Hadar Brayer
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (D.M.); (L.R.-A.); (H.B.); (H.B.-Y.); (T.M.)
| | - Orly Dorot
- Blavatnik Center for Drug Discovery, Tel Aviv University, Tel Aviv 6997801, Israel; (O.D.); (E.P.)
| | - Edward Pichinuk
- Blavatnik Center for Drug Discovery, Tel Aviv University, Tel Aviv 6997801, Israel; (O.D.); (E.P.)
| | - Hagar Ben-Yaakov
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (D.M.); (L.R.-A.); (H.B.); (H.B.-Y.); (T.M.)
| | - Tsipi Meshel
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (D.M.); (L.R.-A.); (H.B.); (H.B.-Y.); (T.M.)
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| | - Adit Ben-Baruch
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (D.M.); (L.R.-A.); (H.B.); (H.B.-Y.); (T.M.)
- Correspondence: ; Tel.: +972-3-6405491; Fax: +972-3-6422046
| |
Collapse
|
93
|
Kamimaki C, Kobayashi N, Hirata M, Somekawa K, Fukuda N, Kubo S, Katakura S, Teranishi S, Watanabe K, Horita N, Hara Y, Yamamoto M, Kudo M, Piao H, Kaneko T. T-cell response to phytohemagglutinin in the interferon-γ release assay as a potential biomarker for the response to immune checkpoint inhibitors in patients with non-small cell lung cancer. Thorac Cancer 2021; 12:1726-1734. [PMID: 33943031 PMCID: PMC8169292 DOI: 10.1111/1759-7714.13978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 12/23/2022] Open
Abstract
Background Immune checkpoint inhibitors are a standard treatment for advanced lung cancer, although it remains important to identify biomarkers that can accurately predict treatment response. Immune checkpoint inhibitors enhance the antitumor T‐cell response, and interferon‐γ plays an important role in this process. Therefore, this study evaluated whether the number of interferon‐γ‐releasing peripheral T cells after phytohemagglutinin stimulation in the interferon‐γ release assay might act as a biomarker for the response of non‐small cell lung cancer to immune checkpoint inhibitor treatment. Methods Data were retrospectively collected regarding 74 patients with non‐small cell lung cancer who had received immune checkpoint inhibitors. Pretreatment screening tests had been performed using the T‐SPOT.TB assay, which quantifies the number of interferon‐γ‐releasing T cells (as immunospots) in response to phytohemagglutinin and tuberculosis‐specific antigen stimulation. Clinical factors and the number of spots in the T‐SPOT fields were evaluated for associations with patient outcomes. The median number of spots was used to categorize patients as having high or low values, and the two groups were compared. Results Relative to patients with a low ratio, patients with a high ratio of phytohemagglutinin/tuberculosis‐specific antigen spots (i.e. more responsive T cells) had significantly better progression‐free survival after immune checkpoint inhibitor treatment. When we only considered patients with negative T‐SPOT results, a high number of phytohemagglutinin‐stimulated spots corresponded to significantly longer progression‐free survival. Conclusion The T‐SPOT.TB assay can be used to quantify the number of immunospots in response to antigen stimulation, which may predict the response to immune checkpoint inhibitors in patients with non‐small cell lung cancer.
Collapse
Affiliation(s)
- Chisato Kamimaki
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Nobuaki Kobayashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Momo Hirata
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kohei Somekawa
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Nobuhiko Fukuda
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sousuke Kubo
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Seigo Katakura
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shuhei Teranishi
- Department of Respiratory Medicine, Yokohama City Medical Center, Yokohama, Japan
| | - Keisuke Watanabe
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Nobuyuki Horita
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yu Hara
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masaki Yamamoto
- Department of Respiratory Medicine, Yokohama City Medical Center, Yokohama, Japan
| | - Makoto Kudo
- Department of Respiratory Medicine, Yokohama City Medical Center, Yokohama, Japan
| | - Hongmei Piao
- Department of Respiratory Medicine, Yanbian University Hospital, Yanji, China
| | - Takeshi Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
94
|
Lee S, Kim KM, Lee SY, Jung J. Estrogen Aggravates Tumor Growth in a Diffuse Gastric Cancer Xenograft Model. Pathol Oncol Res 2021; 27:622733. [PMID: 34257587 PMCID: PMC8262185 DOI: 10.3389/pore.2021.622733] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/01/2021] [Indexed: 12/18/2022]
Abstract
Gastric cancer has the fifth-highest incidence rate and is the third leading cause of cancer-related deaths worldwide. The incidence of gastric cancer is higher in men than in women, but for the diffuse types of gastric cancer, the trend is opposite. Estrogen is considered the prime culprit behind these differences. Nevertheless, the action of estrogen in gastric cancers remains unclear. In this study, we investigated the effect of estrogen on diffuse-type gastric cancer. Human female diffuse gastric cancer SNU-16 cells were transplanted into male and female mice to analyze the effect of endogenous estrogen on tumor growth. Furthermore, the effect of exogenous estrogen was evaluated in ovariectomized mice. Expressed genes were compared between female and male xenograft models using RNA sequencing analysis. Furthermore, human gene expression omnibus databases were utilized to examine the effect of our target genes on overall survival. SNU-16-derived tumor growth was faster in female mice than in male mice. In total RNA sequencing, interferon gamma receptor 2 (IFNGR2), IQ motif containing E (IQCE), transient receptor potential cation channel subfamily M member 4 (TRPM4), and structure-specific endonuclease subunit SLX4 (SLX4) were found. These genes could be associated with the tumor growth in female diffuse-type gastric cancer which was affected by endogenous estrogen. In an ovariectomized gastric cancer xenograft model, exogenous estrogen promoted tumor growth. Especially, our results indicated that estrogen induced G protein-coupled estrogen receptor expression in these mice. These results suggest that estrogen aggravates tumor progression in female diffuse gastric cancer.
Collapse
Affiliation(s)
- Sunyi Lee
- Duksung Innovative Drug Center, Duksung Women’s University, Seoul, Korea
| | - Kyoung Mee Kim
- Duksung Innovative Drug Center, Duksung Women’s University, Seoul, Korea
| | - Seung Yeon Lee
- College of Pharmacy, Duksung Women’s University, Seoul, Korea
| | - Joohee Jung
- Duksung Innovative Drug Center, Duksung Women’s University, Seoul, Korea
- College of Pharmacy, Duksung Women’s University, Seoul, Korea
| |
Collapse
|
95
|
Zhou ZH, Zhao TC, Liang SY, Zhang ZY, Zhu DW, Ju WT, Zhong LP. A therapeutic approach with combination of interferon-gamma and autophagy inhibitor for oral squamous cell carcinoma. Am J Cancer Res 2021; 11:1503-1521. [PMID: 33948370 PMCID: PMC8085849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/28/2020] [Indexed: 06/12/2023] Open
Abstract
Former clinical trials and experimental research have indicated that Interferon-gamma therapy does not achieve an ideal effect in solid tumors. Autophagy has been associated with tumor chemoresistance. The aim of this study was to explore the efficacy of Interferon-gamma and autophagy inhibitor in the combination treatment of oral squamous cell carcinoma. Interferon-gamma-induced apoptosis was evaluated by the expression of relative proteins (cleaved-PARP and caspase-3) and flow cytometry. Interferon-gamma induced autophagy was assessed by the expression of Beclin1, LC3B, and P62. The synergistic effect of interferon-gamma and autophagy inhibitor (chloroquine) was evaluated in vitro and in vivo. Interferon-gamma induced anti-proliferation, apoptosis, and autophagy in oral squamous cell carcinoma cells. Autophagy-related protein 5 was a key feature in Interferon-gamma-induced autophagy flux. Interferon-gamma and chloroquine had obvious synergistic effects on cellular growth inhibition and apoptosis promotion in oral squamous cell carcinoma cells and xenograft models. Our findings suggest that Interferon-gamma-induced autophagy plays a cellular protective role, and blocking autophagy flux can promote Interferon-gamma mediated oral squamous cell carcinoma cell apoptosis. The combination of Interferon-gamma and autophagy inhibitors represents a novel strategy for oral squamous cell carcinoma therapy.
Collapse
Affiliation(s)
- Zhi-Hang Zhou
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of StomatologyShanghai, China
| | - Tong-Chao Zhao
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of StomatologyShanghai, China
| | - Si-Yuan Liang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of StomatologyShanghai, China
| | - Zhi-Yuan Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of StomatologyShanghai, China
| | - Dong-Wang Zhu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of StomatologyShanghai, China
| | - Wu-Tong Ju
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of StomatologyShanghai, China
| | - Lai-Ping Zhong
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of StomatologyShanghai, China
| |
Collapse
|
96
|
Ruiz-Ordoñez I, Piedrahita JM, Arévalo JA, Agualimpia A, Tobón GJ. Lymphomagenesis predictors and related pathogenesis. J Transl Autoimmun 2021; 4:100098. [PMID: 33889831 PMCID: PMC8050773 DOI: 10.1016/j.jtauto.2021.100098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 11/23/2022] Open
Abstract
Sjögren's syndrome (SS) is a systemic autoimmune disease characterised by a wide range of clinical manifestations and complications, including B-cell lymphoma. This study aims to describe the predictors associated with lymphomagenesis in patients with Sjögren's syndrome, emphasising the pathophysiological bases that support this association. We performed a review of the literature published through a comprehensive search strategy in PubMed/MEDLINE, Scopus, and Web of science. Forty publications describing a total of 45,208 patients with SS were retrieved. The predictors were grouped according to their pathophysiological role in the lymphoproliferation process. Also, some new biomarkers such as MicroRNAs, P2X7 receptor-NLRP3 inflammasome, Thymic stromal lymphopoietin, and Three-prime repair exonuclease 1 (TREX1) were identified. The knowledge of the pathophysiology allows the discrimination of markers that participate in the initial stages. Considering that the lymphoproliferation process includes the progression of lymphoma towards more aggressive subtypes, it is essential to recognise biomarkers associated with a worse prognosis.
Collapse
Affiliation(s)
- Ingrid Ruiz-Ordoñez
- Fundación Valle del Lili, Centro de Investigaciones Clínicas, Cra 98 No. 18-49, Cali, 760032, Colombia
- Universidad Icesi, Centro de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Cali, Colombia
| | - Juan-Manuel Piedrahita
- Universidad Icesi, Centro de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Cali, Colombia
- Universidad Icesi, Calle 18 No. 122-135, Cali, Colombia
| | - Javier-Andrés Arévalo
- Universidad Icesi, Centro de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Cali, Colombia
- Universidad Icesi, Calle 18 No. 122-135, Cali, Colombia
| | - Andrés Agualimpia
- Universidad Icesi, Centro de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Cali, Colombia
- Fundación Valle del Lili, Unidad de Reumatología, Cra 98 No. 18-49, Cali. 760032, Colombia
| | - Gabriel J Tobón
- Universidad Icesi, Centro de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Cali, Colombia
- Fundación Valle del Lili, Unidad de Reumatología, Cra 98 No. 18-49, Cali. 760032, Colombia
| |
Collapse
|
97
|
Lan M, Lu W, Zou T, Li L, Liu F, Cai T, Cai Y. Role of inflammatory microenvironment: potential implications for improved breast cancer nano-targeted therapy. Cell Mol Life Sci 2021; 78:2105-2129. [PMID: 33386887 PMCID: PMC11073202 DOI: 10.1007/s00018-020-03696-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 09/20/2020] [Accepted: 10/31/2020] [Indexed: 02/06/2023]
Abstract
Tumor cells, inflammatory cells and chemical factors work together to mediate complex signaling networks, which forms inflammatory tumor microenvironment (TME). The development of breast cancer is closely related to the functional activities of TME. This review introduces the origins of cancer-related chronic inflammation and the main constituents of inflammatory microenvironment. Inflammatory microenvironment plays an important role in breast cancer growth, metastasis, drug resistance and angiogenesis through multifactorial mechanisms. It is suggested that inflammatory microenvironment contributes to providing possible mechanisms of drug action and modes of drug transport for anti-cancer treatment. Nano-drug delivery system (NDDS) becomes a popular topic for optimizing the design of tumor targeting drugs. It is seen that with the development of therapeutic approaches, NDDS can be used to achieve drug-targeted delivery well across the biological barriers and into cells, resulting in superior bioavailability, drug dose reduction as well as off-target side effect elimination. This paper focuses on the review of modulation mechanisms of inflammatory microenvironment and combination with nano-targeted therapeutic strategies, providing a comprehensive basis for further research on breast cancer prevention and control.
Collapse
Affiliation(s)
- Meng Lan
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Wenping Lu
- Guang an'men Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Tengteng Zou
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Lihong Li
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Fengjie Liu
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Tiange Cai
- College of Life Sciences, Liaoning University, Shenyang, 110036, China.
| | - Yu Cai
- College of Pharmacy, Jinan University, Guangzhou, 510632, China.
- Cancer Research Institute of Jinan University, Guangzhou, China.
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, Guangzhou, China.
| |
Collapse
|
98
|
Maire CL, Mohme M, Bockmayr M, Fita KD, Riecken K, Börnigen D, Alawi M, Failla A, Kolbe K, Zapf S, Holz M, Neumann K, Dührsen L, Lange T, Fehse B, Westphal M, Lamszus K. Glioma escape signature and clonal development under immune pressure. J Clin Invest 2021; 130:5257-5271. [PMID: 32603315 DOI: 10.1172/jci138760] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/24/2020] [Indexed: 12/21/2022] Open
Abstract
Immunotherapeutic strategies are increasingly important in neuro-oncology, and the elucidation of escape mechanisms that lead to treatment resistance is crucial. We investigated the impact of immune pressure on the clonal dynamics and immune escape signature by comparing glioma growth in immunocompetent versus immunodeficient mice. Glioma-bearing WT and Pd-1-/- mice survived significantly longer than immunodeficient Pfp-/- Rag2-/- mice. While tumors in Pfp-/- Rag2-/- mice were highly polyclonal, immunoedited tumors in WT and Pd-1-/- mice displayed reduced clonality with emergence of immune escape clones. Tumor cells in WT mice were distinguished by an IFN-γ-mediated response signature with upregulation of genes involved in immunosuppression. Tumor-infiltrating stromal cells, which include macrophages/microglia, contributed even more strongly to the immunosuppressive signature than the actual tumor cells. The identified murine immune escape signature was reflected in human patients and correlated with poor survival. In conclusion, immune pressure profoundly shapes the clonal composition and gene regulation in malignant gliomas.
Collapse
Affiliation(s)
| | | | - Michael Bockmayr
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt University Berlin and Berlin Institute of Health, Berlin, Germany
| | | | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation
| | | | | | | | | | | | | | | | | | - Tobias Lange
- Institutes of Anatomy, Experimental Morphology and Pathology, University Cancer Center Hamburg, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation
| | | | | |
Collapse
|
99
|
Shukla A, Cloutier M, Appiya Santharam M, Ramanathan S, Ilangumaran S. The MHC Class-I Transactivator NLRC5: Implications to Cancer Immunology and Potential Applications to Cancer Immunotherapy. Int J Mol Sci 2021; 22:ijms22041964. [PMID: 33671123 PMCID: PMC7922096 DOI: 10.3390/ijms22041964] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/03/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
The immune system constantly monitors the emergence of cancerous cells and eliminates them. CD8+ cytotoxic T lymphocytes (CTLs), which kill tumor cells and provide antitumor immunity, select their targets by recognizing tumor antigenic peptides presented by MHC class-I (MHC-I) molecules. Cancer cells circumvent immune surveillance using diverse strategies. A key mechanism of cancer immune evasion is downregulation of MHC-I and key proteins of the antigen processing and presentation machinery (APM). Even though impaired MHC-I expression in cancers is well-known, reversing the MHC-I defects remains the least advanced area of tumor immunology. The discoveries that NLRC5 is the key transcriptional activator of MHC-I and APM genes, and genetic lesions and epigenetic modifications of NLRC5 are the most common cause of MHC-I defects in cancers, have raised the hopes for restoring MHC-I expression. Here, we provide an overview of cancer immunity mediated by CD8+ T cells and the functions of NLRC5 in MHC-I antigen presentation pathways. We describe the impressive advances made in understanding the regulation of NLRC5 expression, the data supporting the antitumor functions of NLRC5 and a few reports that argue for a pro-tumorigenic role. Finally, we explore the possible avenues of exploiting NLRC5 for cancer immunotherapy.
Collapse
Affiliation(s)
- Akhil Shukla
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (A.S.); (M.C.); (M.A.S.); (S.R.)
| | - Maryse Cloutier
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (A.S.); (M.C.); (M.A.S.); (S.R.)
| | - Madanraj Appiya Santharam
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (A.S.); (M.C.); (M.A.S.); (S.R.)
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (A.S.); (M.C.); (M.A.S.); (S.R.)
- CRCHUS, Centre Hospitalier de l’Université de Sherbrooke, Sherbrooke, QC J1H5N4, Canada
| | - Subburaj Ilangumaran
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (A.S.); (M.C.); (M.A.S.); (S.R.)
- CRCHUS, Centre Hospitalier de l’Université de Sherbrooke, Sherbrooke, QC J1H5N4, Canada
- Correspondence: ; Tel.: +1-819-346-1110 (ext. 14834)
| |
Collapse
|
100
|
Ramesh P, Shivde R, Jaishankar D, Saleiro D, Le Poole IC. A Palette of Cytokines to Measure Anti-Tumor Efficacy of T Cell-Based Therapeutics. Cancers (Basel) 2021; 13:821. [PMID: 33669271 PMCID: PMC7920025 DOI: 10.3390/cancers13040821] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
Cytokines are key molecules within the tumor microenvironment (TME) that can be used as biomarkers to predict the magnitude of anti-tumor immune responses. During immune monitoring, it has been customary to predict outcomes based on the abundance of a single cytokine, in particular IFN-γ or TGF-β, as a readout of ongoing anti-cancer immunity. However, individual cytokines within the TME can exhibit dual opposing roles. For example, both IFN-γ and TGF-β have been associated with pro- and anti-tumor functions. Moreover, cytokines originating from different cellular sources influence the crosstalk between CD4+ and CD8+ T cells, while the array of cytokines expressed by T cells is also instrumental in defining the mechanisms of action and efficacy of treatments. Thus, it becomes increasingly clear that a reliable readout of ongoing immunity within the TME will have to include more than the measurement of a single cytokine. This review focuses on defining a panel of cytokines that could help to reliably predict and analyze the outcomes of T cell-based anti-tumor therapies.
Collapse
Affiliation(s)
- Prathyaya Ramesh
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA; (P.R.); (R.S.); (D.J.); (D.S.)
- Department of Dermatology, Northwestern University, Chicago, IL 60611, USA
| | - Rohan Shivde
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA; (P.R.); (R.S.); (D.J.); (D.S.)
- Department of Dermatology, Northwestern University, Chicago, IL 60611, USA
| | - Dinesh Jaishankar
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA; (P.R.); (R.S.); (D.J.); (D.S.)
- Department of Dermatology, Northwestern University, Chicago, IL 60611, USA
| | - Diana Saleiro
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA; (P.R.); (R.S.); (D.J.); (D.S.)
- Division of Hematology-Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - I. Caroline Le Poole
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA; (P.R.); (R.S.); (D.J.); (D.S.)
- Department of Dermatology, Northwestern University, Chicago, IL 60611, USA
- Department of Microbiology and Immunology, Northwestern University at Chicago, Chicago, IL 60611, USA
| |
Collapse
|