1
|
Tan G, Spillane KM, Maher J. The Role and Regulation of the NKG2D/NKG2D Ligand System in Cancer. BIOLOGY 2023; 12:1079. [PMID: 37626965 PMCID: PMC10452210 DOI: 10.3390/biology12081079] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/22/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023]
Abstract
The family of human NKG2D ligands (NKG2DL) consists of eight stress-induced molecules. Over 80% of human cancers express these ligands on the surface of tumour cells and/or associated stromal elements. In mice, NKG2D deficiency increases susceptibility to some types of cancer, implicating this system in immune surveillance for malignancy. However, NKG2DL can also be shed, released via exosomes and trapped intracellularly, leading to immunosuppressive effects. Moreover, NKG2D can enhance chronic inflammatory processes which themselves can increase cancer risk and progression. Indeed, tumours commonly deploy a range of countermeasures that can neutralise or even corrupt this surveillance system, tipping the balance away from immune control towards tumour progression. Consequently, the prognostic impact of NKG2DL expression in human cancer is variable. In this review, we consider the underlying biology and regulation of the NKG2D/NKG2DL system and its expression and role in a range of cancer types. We also consider the opportunities for pharmacological modulation of NKG2DL expression while cautioning that such interventions need to be carefully calibrated according to the biology of the specific cancer type.
Collapse
Affiliation(s)
- Ge Tan
- CAR Mechanics Group, Guy’s Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK;
| | | | - John Maher
- CAR Mechanics Group, Guy’s Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK;
- Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne BN21 2UD, UK
- Leucid Bio Ltd., Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
2
|
Kshersagar J, Damle MN, Bedge P, Jagdale R, Tardalkar K, Jadhav D, Jagadale S, Toro Y, Sharma R, Joshi MG. Downregulation of MICA/B tumor surface expressions and augmented soluble MICA serum levels correlate with disease stage in breast cancer. Breast Dis 2023; 41:471-480. [PMID: 36641654 DOI: 10.3233/bd-220023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE In this study, the profiling of the expression of major histocompatibility complex (MHC) class I-related chain A and B (MICA/B) in human breast cancer tumor tissue, saliva, and urine samples of breast cancer patients and control is carried out. MICA/B is ligand of NKG2D receptor expressed on malignant cells. The release of MICA/B from tumor tissue comprises an immune escape mechanism that impairs antitumor immunity. Based on this literature we explored the potential of soluble MICA (sMICA) as a marker in breast cancer (BC). METHODS The expression was profiled by using immunohistochemistry (MICA/B), western blot (MICA/B) and ELISA (MICA). RESULTS The optical density of western blot of MICA/B in different stages of BC illustrated significant difference as per one way analysis of variance and significant difference with stage III and IV by Dunnett's multiple comparisons test respectively. Analysis of sMICA in serum, saliva and urine of BC patients revealed significantly higher levels (median 41.0 ± 4.1 pg/ml in pre-treatment sera, 181.9 ± 1.6 pg/ml in saliva and 90.7 ± 1.7 pg/ml in urine) than in control (median <1.2 pg/ml). The elevated levels of sMICA were related to the cancer stage. CONCLUSIONS The elevated levels of sMICA were observed in patients with well differentiated cancer while the poor expression of sMICA was observed in patients with poorly differentiated tumors. Tumor immunity is impaired by the release of MICA in the biofluids and may be useful for detection and diagnosis of the stage of BC.
Collapse
Affiliation(s)
- Jeevitaa Kshersagar
- Department of Stem Cells & Regenerative Medicine, D. Y. Patil Deemed to be University, D. Y. Patil Vidyanagar, Kasba Bawda, Kolhapur, Maharashtra, India
| | - Mrunal N Damle
- Department of Stem Cells & Regenerative Medicine, D. Y. Patil Deemed to be University, D. Y. Patil Vidyanagar, Kasba Bawda, Kolhapur, Maharashtra, India.,Stem Plus Biotech, SMK Commercial Complex, Near Shivaji Maharaj Putla, Gaon Bhag, Sangli, Maharashtra, India
| | - Poonam Bedge
- Department of Stem Cells & Regenerative Medicine, D. Y. Patil Deemed to be University, D. Y. Patil Vidyanagar, Kasba Bawda, Kolhapur, Maharashtra, India
| | - Rakhi Jagdale
- Department of Pathology, Shri Siddhivinayak Ganpati Cancer Hospital, Miraj, Sangli, Maharashtra, India
| | - Kishor Tardalkar
- Department of Stem Cells & Regenerative Medicine, D. Y. Patil Deemed to be University, D. Y. Patil Vidyanagar, Kasba Bawda, Kolhapur, Maharashtra, India
| | - Dhanaji Jadhav
- Department of Statistics, Yashavantrao Chavan Institute of Science, Satara, Maharashtra, India
| | - Swapnali Jagadale
- Department of Stem Cells & Regenerative Medicine, D. Y. Patil Deemed to be University, D. Y. Patil Vidyanagar, Kasba Bawda, Kolhapur, Maharashtra, India
| | - Yashwant Toro
- Department of Scientific and Industrial Research Organization, Shri Siddhivinayak Ganpati Cancer Hospital, Miraj, Sangli, Maharashtra, India
| | - Rakesh Sharma
- Department of Obstetrics and Gynaecology, Dr. D Y Patil Medical College, Hospital and Research Institute, Kadamwadi, Kolhapur, Maharashtra, India
| | - Meghnad G Joshi
- Department of Stem Cells & Regenerative Medicine, D. Y. Patil Deemed to be University, D. Y. Patil Vidyanagar, Kasba Bawda, Kolhapur, Maharashtra, India.,Stem Plus Biotech, SMK Commercial Complex, Near Shivaji Maharaj Putla, Gaon Bhag, Sangli, Maharashtra, India
| |
Collapse
|
3
|
Association of MICA gene Exon-5 polymorphism in oral submucous fibrosis. Oral Surg Oral Med Oral Pathol Oral Radiol 2023; 135:110-116. [PMID: 36396592 DOI: 10.1016/j.oooo.2022.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 07/10/2022] [Accepted: 08/28/2022] [Indexed: 01/10/2023]
Abstract
OBJECTIVE The present study was conducted to explore the allele frequencies of MICA gene Exon-5 transmembrane and to measure the circulatory MICA levels in various histologic grades of patients with oral submucous fibrosis (OSF) compared to healthy individuals. STUDY DESIGN We enrolled a total of 595 patients for this cross-sectional study and divided them into 2 groups: healthy controls (n = 320) and patients with OSF (n = 275). Further, patients with OSF were subdivided based on their histologic gradings. The genomic DNA was extracted followed by a polymerase chain reaction and genotyping using the ABI Prism DNA Sequencer (ThermoFisher Scientific, Inc., Waltham, MA, USA). RESULTS Our study showed that the A5 allele of the MICA gene in the Exon-5 region conferred significant risk for patients with OSF. With reference to the histologic gradings of OSF, we found that the MICA gene conferred statistically significant risk among patients with grade III OSF. On the other hand, the A8 allele of MICA gene in the Exon-5 region conferred significant protection among the overall OSF cohort and in the grade III of histologic grade. Finally, the circulatory human MICA levels were found to have a stepwise increase from grade I toward grade III in patients with OSF. CONCLUSION Our results suggested that the A5 allele in MICA might confer risk for the progression of OSF among the South Indian ethnic population.
Collapse
|
4
|
Secchiari F, Nuñez SY, Sierra JM, Ziblat A, Regge MV, Raffo Iraolagoitia XL, Rovegno A, Ameri C, Secin FP, Richards N, Ríos Pita H, Vitagliano G, Rico L, Mieggi M, Frascheri F, Bonanno N, Blas L, Trotta A, Friedrich AD, Fuertes MB, Domaica CI, Zwirner NW. The MICA-NKG2D axis in clear cell renal cell carcinoma bolsters MICA as target in immuno-oncology. Oncoimmunology 2022; 11:2104991. [PMID: 35936986 PMCID: PMC9354769 DOI: 10.1080/2162402x.2022.2104991] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
NKG2D is a major natural killer (NK) cell-activating receptor that recognizes eight ligands (NKG2DLs), including MICA, and whose engagement triggers NK cell effector functions. As NKG2DLs are upregulated on tumor cells but tumors can subvert the NKG2D-NKG2DL axis, NKG2DLs constitute attractive targets for antibody (Ab)-based immuno-oncology therapies. However, such approaches require a deep characterization of NKG2DLs and NKG2D cell surface expression on primary tumor and immune cells. Here, using a bioinformatic analysis, we observed that MICA is overexpressed in renal cell carcinoma (RCC), and we also detected an association between the NKG2D-MICA axis and a diminished overall survival of RCC patients. Also, by flow cytometry (FC), we observed that MICA was the only NKG2DL over-expressed on clear cell renal cell carcinoma (ccRCC) tumor cells, including cancer stem cells (CSC) that also coexpressed NKG2D. Moreover, tumor-infiltrating leukocytes (TIL), but not peripheral blood lymphoid cells (PBL) from ccRCC patients, over-expressed MICA, ULBP3 and ULBP4. In addition, NKG2D was downregulated on peripheral blood NK cells (PBNK) from ccRCC patients but upregulated on tumor-infiltrating NK cells (TINK). These TINK exhibited impaired degranulation that negatively correlated with NKG2D expression, diminished IFN-γ production, upregulation of TIM-3, and an impaired glucose intake upon stimulation with cytokines, indicating that they are dysfunctional, display features of exhaustion and an altered metabolic fitness. We conclude that ccRCC patients exhibit a distorted MICA-NKG2D axis, and MICA emerges as the forefront NKG2DL for the development of targeted therapies in ccRCC.
Collapse
Affiliation(s)
- Florencia Secchiari
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Argentina
| | - Sol Yanel Nuñez
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Argentina
| | - Jessica Mariel Sierra
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Argentina
| | - Andrea Ziblat
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Argentina
| | - María Victoria Regge
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Argentina
| | - Ximena Lucía Raffo Iraolagoitia
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Argentina
| | - Agustín Rovegno
- Servicio de Urología, Centro de Educación Médica e Investigaciones Clínicas “Norberto Quirno” (CEMIC)
| | - Carlos Ameri
- Servicio de Urología, Hospital Alemán, Buenos Aires, Argentina
| | - Fernando Pablo Secin
- Servicio de Urología, Centro de Educación Médica e Investigaciones Clínicas “Norberto Quirno” (CEMIC)
| | - Nicolás Richards
- Servicio de Urología, Centro de Educación Médica e Investigaciones Clínicas “Norberto Quirno” (CEMIC)
| | | | | | - Luis Rico
- Servicio de Urología, Hospital Alemán, Buenos Aires, Argentina
| | - Mauro Mieggi
- Servicio de Urología, Hospital Alemán, Buenos Aires, Argentina
| | | | - Nicolás Bonanno
- Servicio de Urología, Hospital Alemán, Buenos Aires, Argentina
| | - Leandro Blas
- Servicio de Urología, Hospital Alemán, Buenos Aires, Argentina
| | - Aldana Trotta
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Argentina
| | - Adrián David Friedrich
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Argentina
| | - Mercedes Beatriz Fuertes
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Argentina
| | - Carolina Inés Domaica
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Argentina
| | - Norberto Walter Zwirner
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Argentina
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Argentina
| |
Collapse
|
5
|
Fuertes MB, Domaica CI, Zwirner NW. Leveraging NKG2D Ligands in Immuno-Oncology. Front Immunol 2021; 12:713158. [PMID: 34394116 PMCID: PMC8358801 DOI: 10.3389/fimmu.2021.713158] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) revolutionized the field of immuno-oncology and opened new avenues towards the development of novel assets to achieve durable immune control of cancer. Yet, the presence of tumor immune evasion mechanisms represents a challenge for the development of efficient treatment options. Therefore, combination therapies are taking the center of the stage in immuno-oncology. Such combination therapies should boost anti-tumor immune responses and/or target tumor immune escape mechanisms, especially those created by major players in the tumor microenvironment (TME) such as tumor-associated macrophages (TAM). Natural killer (NK) cells were recently positioned at the forefront of many immunotherapy strategies, and several new approaches are being designed to fully exploit NK cell antitumor potential. One of the most relevant NK cell-activating receptors is NKG2D, a receptor that recognizes 8 different NKG2D ligands (NKG2DL), including MICA and MICB. MICA and MICB are poorly expressed on normal cells but become upregulated on the surface of damaged, transformed or infected cells as a result of post-transcriptional or post-translational mechanisms and intracellular pathways. Their engagement of NKG2D triggers NK cell effector functions. Also, MICA/B are polymorphic and such polymorphism affects functional responses through regulation of their cell-surface expression, intracellular trafficking, shedding of soluble immunosuppressive isoforms, or the affinity of NKG2D interaction. Although immunotherapeutic approaches that target the NKG2D-NKG2DL axis are under investigation, several tumor immune escape mechanisms account for reduced cell surface expression of NKG2DL and contribute to tumor immune escape. Also, NKG2DL polymorphism determines functional NKG2D-dependent responses, thus representing an additional challenge for leveraging NKG2DL in immuno-oncology. In this review, we discuss strategies to boost MICA/B expression and/or inhibit their shedding and propose that combination strategies that target MICA/B with antibodies and strategies aimed at promoting their upregulation on tumor cells or at reprograming TAM into pro-inflammatory macrophages and remodeling of the TME, emerge as frontrunners in immuno-oncology because they may unleash the antitumor effector functions of NK cells and cytotoxic CD8 T cells (CTL). Pursuing several of these pipelines might lead to innovative modalities of immunotherapy for the treatment of a wide range of cancer patients.
Collapse
Affiliation(s)
- Mercedes Beatriz Fuertes
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Carolina Inés Domaica
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Norberto Walter Zwirner
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina.,Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
6
|
Rittmann MC, Hussung S, Braun LM, Klar RFU, Biesel EA, Fichtner-Feigl S, Fritsch R, Wittel UA, Ruess DA. Plasma biomarkers for prediction of early tumor recurrence after resection of pancreatic ductal adenocarcinoma. Sci Rep 2021; 11:7499. [PMID: 33820913 PMCID: PMC8021576 DOI: 10.1038/s41598-021-86779-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/19/2021] [Indexed: 01/01/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a disease with a very unfavorable prognosis. Surgical resection represents the only potentially curative treatment option, but recurrence after complete resection is almost certain. In an exploratory attempt we here aimed at identifying preoperative plasma protein biomarkers with the potential to predict early recurrence after resection of PDAC. Peripheral blood samples from 14 PDAC patients divided into three groups according to their time to tumor recurrence after curatively intended resection (early: < 6 months, medium: 6–12 months, late: > 12 months) underwent targeted proteome analysis. Proteins most strongly discriminating early and late recurrence were then examined in a number of established PDAC cell lines and their culture supernatants. Finally, PDAC organoid lines from primary tumors of patients with early and late recurrence were analyzed for confirmation and validation of results. In total, 23 proteins showed differential abundance in perioperative plasma from PDAC patients with early recurrence when compared to patients with late recurrence. Following confirmation of expression on a transcriptional and translational level in PDAC cell lines we further focused on three upregulated (MAEA, NT5E, AZU1) and two downregulated proteins (ATP6AP2, MICA). Increased expression of NT5E was confirmed in a subset of PDAC organoid cultures from tumors with early recurrence. MICA expression was heterogeneous and ATP6AP2 levels were very similar in both organoids from early and late recurrent tumors. Most strikingly, we observed high MAEA expression in all tested PDAC (n = 7) compared to a non-cancer ductal organoid line. MAEA also demonstrated potential to discriminate early recurrence from late recurrence PDAC organoids. Our study suggests that identification of plasma protein biomarkers released by tumor cells may be feasible and of value to predict the clinical course of patients. Prediction of recurrence dynamics would help to stratify up-front resectable PDAC patients for neoadjuvant chemotherapy approaches in an individualized fashion. Here, MAEA and NT5E were the most promising candidates for further evaluation.
Collapse
Affiliation(s)
- Marie-Claire Rittmann
- Department of General and Visceral Surgery, Medical Center and Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Saskia Hussung
- Department of Medicine I, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Medical Oncology and Hematology, Zurich University Hospital, Zurich, Switzerland
| | - Lukas M Braun
- Department of General and Visceral Surgery, Medical Center and Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.,Department of Medicine I, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rhena F U Klar
- Department of Medicine I, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Esther A Biesel
- Department of General and Visceral Surgery, Medical Center and Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Stefan Fichtner-Feigl
- Department of General and Visceral Surgery, Medical Center and Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Ralph Fritsch
- Department of Medicine I, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Medical Oncology and Hematology, Zurich University Hospital, Zurich, Switzerland
| | - Uwe A Wittel
- Department of General and Visceral Surgery, Medical Center and Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
| | - Dietrich A Ruess
- Department of General and Visceral Surgery, Medical Center and Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| |
Collapse
|
7
|
Pastò A, Consonni FM, Sica A. Influence of Innate Immunity on Cancer Cell Stemness. Int J Mol Sci 2020; 21:ijms21093352. [PMID: 32397392 PMCID: PMC7247585 DOI: 10.3390/ijms21093352] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/02/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Even if cancer stem cells (CSCs) represent only a small proportion of the tumor mass, they significantly account for tumor maintenance, resistance to therapies, relapse and metastatic spread, due to their increased capacity of self-renewal, multipotency, tumorigenicity and quiescence. Emerging evidence suggests that the immune contexture within the tumor microenvironment (TME) determines both the response to therapy and the clinical outcome. In this context, CSCs acquire immune evasion skills by editing immune cell functions and sculpting the immunosuppressive landscape of TME. Reciprocally, infiltrating immune cells influence CSCs self-renewal, tumorigenicity and metastasis. In this review, we summarize the immunomodulatory properties of CSCs, as well as the impact of innate immune cells on cancer cells stemness in the different phases of cancer immunoediting process and neoplastic progression.
Collapse
Affiliation(s)
- Anna Pastò
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center–IRCCS–, via Manzoni 56, 20089 Rozzano (MI), Italy;
| | - Francesca Maria Consonni
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, A. Avogadro, via Bovio 6, 28100 Novara, Italy;
| | - Antonio Sica
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center–IRCCS–, via Manzoni 56, 20089 Rozzano (MI), Italy;
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, A. Avogadro, via Bovio 6, 28100 Novara, Italy;
- Correspondence: ; Tel.: +39-0321-375-881; Fax: +39-0321-375-621
| |
Collapse
|
8
|
Onyeaghala G, Lane J, Pankratz N, Nelson HH, Thyagarajan B, Walcheck B, Anderson KE, Prizment AE. Association between MICA polymorphisms, s-MICA levels, and pancreatic cancer risk in a population-based case-control study. PLoS One 2019; 14:e0217868. [PMID: 31166958 PMCID: PMC6550421 DOI: 10.1371/journal.pone.0217868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/20/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Pancreatic tumor cells may avoid immune surveillance by releasing the transmembrane major histocompatibility complex class I chain-related A (MICA) protein in soluble form (s-MICA). We hypothesized that the presence of the A5.1 polymorphism in the MICA gene, which encodes a truncated MICA protein, is associated with higher s-MICA levels and increased pancreatic cancer risk. METHODS MICA alleles and s-MICA levels were measured in 121 pancreatic cancer cases and 419 controls. General linear regression with a log transformation assessed geometric means of s-MICA levels across MICA alleles. Unconditional logistic regression was used to calculate the odds ratio (OR) and 95% confidence intervals (CI) for pancreatic cancer associated with MICA alleles. RESULTS After multivariate adjustment, participants with at least one copy of the A5.1 allele versus no A5.1 allele had 1.35 (95% CI: 1.05-1.74) times greater s-MICA levels (1.65 times higher for cases and 1.28, for controls) and increased risk of pancreatic cancer (OR = 1.91, 95% CI: 1.05-3.48). CONCLUSIONS Our study suggests higher risk of pancreatic cancer among those with the MICA A5.1 polymorphism, which may be explained by an increase in s-MICA secretion and impaired immune response. IMPACT These findings provide further evidence at the genetic and molecular level of the important role of MICA in pancreatic cancer development, and may have important implications with regards to pancreatic cancer screening.
Collapse
Affiliation(s)
- Guillaume Onyeaghala
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, United States of America
| | - John Lane
- University of Minnesota Masonic Cancer Center, Minneapolis, MN, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States of America
| | - Nathan Pankratz
- University of Minnesota Masonic Cancer Center, Minneapolis, MN, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States of America
| | - Heather H. Nelson
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, United States of America
- University of Minnesota Masonic Cancer Center, Minneapolis, MN, United States of America
| | - Bharat Thyagarajan
- University of Minnesota Masonic Cancer Center, Minneapolis, MN, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States of America
| | - Bruce Walcheck
- University of Minnesota Masonic Cancer Center, Minneapolis, MN, United States of America
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN, United States of America
| | - Kristin E. Anderson
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, United States of America
- University of Minnesota Masonic Cancer Center, Minneapolis, MN, United States of America
| | - Anna E. Prizment
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, United States of America
- University of Minnesota Masonic Cancer Center, Minneapolis, MN, United States of America
| |
Collapse
|
9
|
Sakiyama MJ, Espinoza I, Reddy A, de Carlo F, Kumar A, Levenson AS, Bae S, Zhou X, Claudio PP, Lewin J, Manucha V, Pound CR, Vijayakumar S, Gomez CR. Race-associated expression of MHC class I polypeptide-related sequence A (MICA) in prostate cancer. Exp Mol Pathol 2019; 108:173-182. [PMID: 31004600 DOI: 10.1016/j.yexmp.2019.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/04/2019] [Accepted: 04/16/2019] [Indexed: 01/07/2023]
Abstract
Despite the lack of a complete understanding of the disparities involved, prostate cancer (PCa) has both higher incidence and death rates in African American Men (AAM) relative to those of Caucasian American Men (CAM). MHC class I polypeptide related sequence A (MICA) is an innate immunity protein involved in tumor immunoevasion. Due to a lack of reports of race-specific expression of MICA in PCa, we evaluated MICA expression in patients' tumors and in cell lines from a racially diverse origin. Immunohistochemistry was done on a tissue microarray (TMA) with antibodies against MICA. Tumor MICA mRNA was assessed by data mining using Oncomine and PROGeneV2. Surface MICA and release rate of soluble (s) MICA was evaluated in PCa cell lines originally derived from African American (MDA-PCa-2b) or Caucasian (LNCaP and DU-145) PCa patients. Prostate tumor tissue had a 1.7-fold higher MICA expression relative to normal tissue (p < .0001). MICA immunoreactivity in PCa tissue from AAM was 24% lower (p = .002) compared to CAM. Survival analysis revealed a marginal association of low MICA with poor overall survival (OS) (p = .058). By data mining analysis, a 2.9-fold higher level of MICA mRNA was evidenced in tumor compared to normal tissue (p < .0001). Tumors from AAM had 24% lower levels of MICA mRNA compared to tumors from CAM (p = .038), and poor prognosis was found for patients with lower MICA mRNA (p = .028). By flow cytometry analysis, cell fraction positive for surface MICA was of 3% in MDA-PCa-2b cells, 54% in DU-145 cells, and 67% in LNCaP cells (p < .0001). sMICA was detected in DU-145 and LNCaP cells, but was not detected in MDA-PCa-2b cells. Both LNCaP and DU-145 cells were sensitive to cytolysis mediated by Natural killer (NK) cells. MDA-PCa-2b cells, however were between 1.3-fold at 10:1 Effector:Target (E:T) ratio (p < .0001) and 2-fold at 50:1 E:T ratio (p < .0001) more resistant to NK-mediated cytolysis relative to cells from Caucasian origin. These results suggest that MICA expression may be related to the aggressive nature of PCa. Our findings also demonstrate for the first time that there are variations in MICA expression in the context of racial differences. This study establishes a rationale for further investigation of MICA as a potential race-specific prognostic marker in PCa.
Collapse
Affiliation(s)
- Marcelo J Sakiyama
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA; CAPES Foundation, Ministry of Education of Brazil, Brasília, DF 70040-020, Brazil
| | - Ingrid Espinoza
- Department of Preventive Medicine, University of Mississippi Medical Center, Jackson, MS, USA; Center for Clinical and Translational Science (CCTS), University of Mississippi School of Pharmacy & University of Mississippi Medical Center, Jackson, MS, USA
| | - Amit Reddy
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Flavia de Carlo
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA; Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, USA; National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, MS, USA; Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Avinash Kumar
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA
| | - Anait S Levenson
- College of Veterinary Medicine, Long Island University, Brookville, NY, USA
| | - Sejong Bae
- Division of Preventive Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xinchun Zhou
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Pier Paolo Claudio
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA; Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, USA; National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, MS, USA; Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jack Lewin
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Varsha Manucha
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Charles R Pound
- Department of Urology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Srinivasan Vijayakumar
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Christian R Gomez
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA; Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, USA; Center for Clinical and Translational Science (CCTS), University of Mississippi School of Pharmacy & University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
10
|
Interactions between cancer stem cells, immune system and some environmental components: Friends or foes? Immunol Lett 2019; 208:19-29. [DOI: 10.1016/j.imlet.2019.03.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/02/2019] [Accepted: 03/08/2019] [Indexed: 12/17/2022]
|
11
|
Vilhav C, Engström C, Naredi P, Novotny A, Bourghardt-Fagman J, Iresjö BM, Asting AG, Lundholm K. Fractional uptake of circulating tumor cells into liver-lung compartments during curative resection of periampullary cancer. Oncol Lett 2018; 16:6331-6338. [PMID: 30405768 PMCID: PMC6202519 DOI: 10.3892/ol.2018.9435] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/02/2018] [Indexed: 12/25/2022] Open
Abstract
Circulating tumor cells (CTCs) are able to predict outcome in patients with breast, colon and prostate cancer and appear to be promising biomarkers of pancreatic carcinoma. The aim of the present study was to demonstrate a statistically significant portal-arterial difference of CTCs during curative resection of periampullary cancer. A commercially available instrument (IsofluxR) was used to quantify blood content of CTC in 10 patients with periampullary cancer according to preoperative diagnostics. Portal and arterial blood samples (~8 ml each) were simultaneously collected intra-operatively following surgical dissection prior to division of the pancreas for tumor removal. Quantitative CTC analyses were performed according to standardized protocols for immune-magnetic enrichment of CTC. Flow cytometry was applied for qualitative evaluations of various CTC markers in 7 patients. There was a statistically significant difference in the number of CTCs collected in the portal blood [58±14 cells per 100 ml; mean ± standard error (SE)] vs. arterial blood [24±7 cells per 100 ml (SE), P<0.025]. A fractional uptake of ≥40% across liver and lung compartments of assumed malignant CTC was estimated to correspond to the appearance of ~410 tumor cells per minute during pancreatic resections based on estimated hepatic blood flow, measured tumor cell mass and tumor cell proliferation activity. Complications in the collection of portal blood were not observed. A significant uptake across liver or lung compartments of potentially malignant tumor CTCs from periampullary carcinoma may represent a model to capture, define and characterize cell clones with metastatic potential in liver and lung tissues following surgical resection.
Collapse
Affiliation(s)
- Caroline Vilhav
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Cecilia Engström
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Peter Naredi
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Ann Novotny
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Johan Bourghardt-Fagman
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Britt-Marie Iresjö
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Annika G Asting
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Kent Lundholm
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| |
Collapse
|
12
|
Abstract
Pancreatic cancer is among the three deadliest cancers worldwide with the lowest 5-year survival of all cancers. Despite all efforts, therapeutic improvements have barely been made over the last decade. Even recent highly promising targeted and immunotherapeutic approaches did not live up to their expectations. Therefore, other horizons have to be explored. Natural Killer (NK) cells are gaining more and more interest as a highly attractive target for cancer immunotherapies, both as pharmaceutical target and for cell therapies. In this systematic review we summarise the pathophysiological adaptions of NK cells in pancreatic cancer and highlight possible (future) therapeutic NK cell-related targets. Furthermore, an extensive overview of recent therapeutic approaches with an effect on NK cells is given, including cytokine-based, viro- and bacteriotherapy and cell therapy. We also discuss ongoing clinical trials that might influence NK cells. In conclusion, although several issues regarding NK cells in pancreatic cancer remain unsolved and need further investigation, extensive evidence is already provided that support NK cell oriented approaches in pancreatic cancer.
Collapse
|
13
|
Yan J, Chen G, Zhao X, Chen F, Wang T, Miao F. High expression of diffuse panbronchiolitis critical region 1 gene promotes cell proliferation, migration and invasion in pancreatic ductal adenocarcinoma. Biochem Biophys Res Commun 2017; 495:1908-1914. [PMID: 29242154 DOI: 10.1016/j.bbrc.2017.12.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 12/06/2017] [Indexed: 12/19/2022]
Abstract
Diffuse panbronchiolitis critical region 1 (DPCR1) is located in the major histocompatibility complex (MHC) class I. It was reported to be downregulated in invasive pituitary adenoma compared with that in non-invasive tumors, but upregulated in the precursor of gastric carcinogenesis. However, the direct effect of DPCR1 on cancer cells has rarely been reported, and the role DPCR1 in pancreatic ductal adenocarcinoma (PDAC) remains unclear. The clinical sample validation and public data analysis of the present study demonstrated that DPCR1 was upregulated markedly in PDAC and this high expression was negatively correlated with the patient prognosis. Functionally, knocking down DPCR1 in PDAC cell lines inhibited cell proliferation, migration and invasion in vitro. Tumor xenograft experiments further showed that suppression of DPCR1 inhibited tumor growth in vivo. In addition, the results of RNA deep sequencing and qRT-PCR assay showed that DPCR1 participated in PADC progression by regulating nuclear factor-kappa B signaling pathway, suggesting that it might be a novel oncogene in tumor progression and a potential therapeutic target in PDAC as well.
Collapse
Affiliation(s)
- Jiayi Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai 200025, China
| | - Guanghui Chen
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Xuesong Zhao
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai 200025, China
| | - Fangying Chen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai 200025, China
| | - Ting Wang
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China.
| | - Fei Miao
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai 200025, China.
| |
Collapse
|
14
|
Onyeaghala G, Nelson HH, Thyagarajan B, Linabery AM, Panoskaltsis-Mortari A, Gross M, Anderson KE, Prizment AE. Soluble MICA is elevated in pancreatic cancer: Results from a population based case-control study. Mol Carcinog 2017; 56:2158-2164. [PMID: 28470829 PMCID: PMC5590635 DOI: 10.1002/mc.22667] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 04/20/2017] [Accepted: 05/01/2017] [Indexed: 01/23/2023]
Abstract
Pancreatic cancer is diagnosed at a late stage and has one of the highest cancer mortality rates in the United States, creating an urgent need for novel early detection tools. A candidate biomarker for use in early detection is the soluble MHC class I-related chain A (s-MICA) ligand, which pancreatic tumors shed to escape immune detection. The objective of this study was to define the association between s-MICA levels and pancreatic cancer, in a population-based case-control study. S-MICA was measured in 143 pancreatic cancer cases and 459 controls. Unconditional logistic regression was used to calculate odds ratio (OR) for pancreatic cancer and 95% confidence intervals (CI). There was a positive association between increasing s-MICA levels and pancreatic cancer: compared to the lowest tertile, the ORs for pancreatic cancer were 1.25 (95%CI: 0.75-2.07) and 2.10 (95%CI: 1.29-3.42) in the second and highest tertiles, respectively (P-trend = 0.02). Our study supports previous work demonstrating a positive association between plasma s-MICA levels and pancreatic cancer.
Collapse
Affiliation(s)
- Guillaume Onyeaghala
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Heather H Nelson
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
- University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
| | - Bharat Thyagarajan
- University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Amy M Linabery
- University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
| | - Angela Panoskaltsis-Mortari
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Myron Gross
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Kristin E Anderson
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
- University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
| | - Anna E Prizment
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
- University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
| |
Collapse
|
15
|
Luna JI, Grossenbacher SK, Murphy WJ, Canter RJ. Targeting Cancer Stem Cells with Natural Killer Cell Immunotherapy. Expert Opin Biol Ther 2016; 17:313-324. [PMID: 27960589 DOI: 10.1080/14712598.2017.1271874] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Standard cytoreductive cancer therapy, such as chemotherapy and radiotherapy, are frequently resisted by a small portion of cancer cells with 'stem-cell' like properties including quiescence and repopulation. Immunotherapy represents a breakthrough modality for improving oncologic outcomes in cancer patients. Since the success of immunotherapy is not contingent on target cell proliferation, it may also be uniquely suited to address the problem of resistance and repopulation exerted by cancer stem cells (CSCs). Areas covered: Natural killer (NK) cells have long been known for their ability to reject allogeneic hematopoietic stem cells, and there are increasing data demonstrating that NK cells can selectively identify and lyse CSCs. The authors review the current knowledge of CSCs and NK cells and highlight recent studies that support the concept that NK cells are capable of targeting CSC in solid tumors, especially in the context of combination therapy simultaneously targeting non-CSCs and CSCs. Expert opinion: Unlike cytotoxic cancer treatments, NK cells can target and eliminate quiescent/non-proliferating cells such as CSCs, and these enigmatic cells are an important source of relapse and metastasis. NK targeting of CSCs represents a novel and potentially high impact method to capitalize on the intrinsic therapeutic potential of NK cells.
Collapse
Affiliation(s)
- Jesus I Luna
- a Department of Dermatology , University of California Davis School of Medicine , Sacramento , CA USA
| | - Steven K Grossenbacher
- a Department of Dermatology , University of California Davis School of Medicine , Sacramento , CA USA
| | - William J Murphy
- a Department of Dermatology , University of California Davis School of Medicine , Sacramento , CA USA.,b Department of Internal Medicine , University of California Davis Medical Center , Sacramento , CA USA
| | - Robert J Canter
- c Division of Surgical Oncology, Department of Surgery , University of California Davis School of Medicine , Sacramento , CA USA
| |
Collapse
|
16
|
Xu H, Zhu XX, Chen J. DNT cell inhibits the growth of pancreatic carcinoma via abnormal expressions of NKG2D and MICA in vivo. Biochem Biophys Res Commun 2016; 469:145-50. [PMID: 26616050 DOI: 10.1016/j.bbrc.2015.11.085] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 11/19/2015] [Indexed: 01/19/2023]
Abstract
This research aimed to investigate the effects of natural killer group 2 member D (NKG2D) and its ligands major histocompatibility complex class I chain-related molecules A(MICA) in DNT cell killing pancreatic carcinoma. Antibodies adsorption was used to separate DNT cell from human peripheral blood. Human pancreatic tumor models were established via implanting BXPC-3 cells into nude mice. Then randomly divided mice into blank group, gemcitabine group and DNT group. Mice weights and mice tumor volumes were measured every 5 days. 50 days later mice were euthanized at cervical dislocation method. Tumor weights were measured. Relative tumor volume and tumor inhibition rate were calculated. Western blot and qPCR were used to detect the expressions of NKG2D and MICA in the transplanted tumors of the three groups. DNT cell significantly increased over time. The blank group tumor volume and weight were significantly larger than the other groups (p < 0.001, p < 0.001), but there were no significantly difference between DNT group and gemcitabine group (p > 0.05). Gemcitabine and DNT cell tumor inhibition rate were 40.4% and 35.5%. Western blot and qPCR showed that MICA mRNA and protein levels in blank group were significantly higher than DNT group (p = 0.001, p = 0.003). NKG2D mRNA and protein levels in blank group were significantly lower than DNT cells group (p < 0.001, p = 0.001). In conclusion DNT cell can significantly inhibit the growth of pancreatic carcinoma in vivo, and the mechanism may be involved in abnormal expressions of MICA and NKG2D.
Collapse
Affiliation(s)
- Hong Xu
- Department of General Surgery, Affiliated Provincial Hospital, Anhui Medical University, Hefei 230001, Anhui Province, PR China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei 230001, PR China
| | - Xing-Xing Zhu
- Department of General Surgery, Affiliated Provincial Hospital, Anhui Medical University, Hefei 230001, Anhui Province, PR China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei 230001, PR China
| | - Jiong Chen
- Department of General Surgery, Affiliated Provincial Hospital, Anhui Medical University, Hefei 230001, Anhui Province, PR China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei 230001, PR China.
| |
Collapse
|
17
|
Chen J, Xu H, Zhu XX. Abnormal expression levels of sMICA and NKG2D are correlated with poor prognosis in pancreatic cancer. Ther Clin Risk Manag 2015; 12:11-8. [PMID: 26730197 PMCID: PMC4694694 DOI: 10.2147/tcrm.s96869] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Soluble major histocompatibility complex class I-related chain A molecules (sMICA) and natural-killer group 2 member D (NKG2D) not only correlate with tumorigenesis and progression, but also with tumor invasion and metastasis. In this study, we used immunohistochemistry to investigate the correlation and prognostic significance of the differential expression of sMICA and NKG2D in pancreatic carcinoma and paracarcinoma tissues from 70 patients with pancreatic carcinomas. The results showed that sMICA expression was significantly (P<0.05) higher in tumor tissues (67.1%) than that in adjacent nontumor tissues (31.4%), whereas NKG2D expression was significantly (P<0.001) lower in tumor tissues (32.9%) than that in adjacent nontumor tissues (60.0%). Spearman’s rank correlation test showed a negative correlation between the expression of sMICA and that of NKG2D (r=−0.676, P<0.001). Kaplan–Meier survival analysis showed that a high sMICA expression was significantly correlated with decreased disease-free survival (DFS) (P<0.001) and overall survival (OS) (P<0.001), while a high NKG2D expression was significantly associated with increased DFS (P=0.001) and OS (P=0.001) of the patients. Multivariate analysis showed that a high sMICA expression was an independent predictive factor for poor DFS (P<0.001) and OS (P=0.012); but low NKG2D expression was not an independent prognostic factor for poor DFS (P=0.238) and OS (P=0.574). In conclusion, our findings suggest that the expression levels of sMICA and NKG2D are abnormal and negatively correlated with one another in pancreatic carcinoma tissues; they may be considered as valuable biomarkers for the prognosis of pancreatic carcinoma.
Collapse
Affiliation(s)
- Jiong Chen
- Department of General Surgery, Affiliated Provincial Hospital, Anhui Medical University, Hefei, People's Republic of China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, People's Republic of China
| | - Hong Xu
- Department of General Surgery, Affiliated Provincial Hospital, Anhui Medical University, Hefei, People's Republic of China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, People's Republic of China
| | - Xing-Xing Zhu
- Department of General Surgery, Affiliated Provincial Hospital, Anhui Medical University, Hefei, People's Republic of China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, People's Republic of China
| |
Collapse
|
18
|
Process of hepatic metastasis from pancreatic cancer: biology with clinical significance. J Cancer Res Clin Oncol 2015; 142:1137-61. [PMID: 26250876 DOI: 10.1007/s00432-015-2024-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 07/23/2015] [Indexed: 12/14/2022]
Abstract
PURPOSE Pancreatic cancer shows a remarkable preference for the liver to establish secondary tumors. Selective metastasis to the liver is attributed to the development of potential microenvironment for the survival of pancreatic cancer cells. This review aims to provide a full understanding of the hepatic metastatic process from circulating pancreatic cancer cells to their settlement in the liver, serving as a basic theory for efficient prediction and treatment of metastatic diseases. METHODS A systematic search of relevant original articles and reviews was performed on PubMed, EMBASE and Cochrane Library for the purpose of this review. RESULTS Three interrelated phases are delineated as the contributions of the interaction between pancreatic cancer cells and the liver to hepatic metastasis process. Chemotaxis of disseminated pancreatic cancer cells and simultaneous defensive formation of platelets or neutrophils facilitate specific metastasis toward the liver. Remodeling of extracellular matrix and stromal cells in hepatic lobules and angiogenesis induced by proangiogenic factors support the survival and growth of clinical micrometastasis colonizing the liver. The bimodal role of the immune system or prevalence of cancer cells over the immune system makes metastatic progression successfully proceed from micrometastasis to macrometastasis. CONCLUSIONS Pancreatic cancer is an appropriate research object of cancer metastasis representing more than a straight cascade. If any of the successive or simultaneous phases, especially tumor-induced immunosuppression, is totally disrupted, hepatic metastasis will be temporarily under control or even cancelled forever. To shrink cancers on multiple fronts and prolong survival for patients, novel oral or intravenous anti-cancer agents covering one or different phases of metastatic pancreatic cancer are expected to be integrated into innovative strategies on the premise of safety and efficacious biostability.
Collapse
|
19
|
Zamora AE, Grossenbacher SK, Aguilar EG, Murphy WJ. Models to Study NK Cell Biology and Possible Clinical Application. ACTA ACUST UNITED AC 2015; 110:14.37.1-14.37.14. [PMID: 26237009 DOI: 10.1002/0471142735.im1437s110] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Natural killer (NK) cells are large granular lymphocytes of the innate immune system, responsible for direct targeting and killing of both virally infected and transformed cells. NK cells rapidly recognize and respond to abnormal cells in the absence of prior sensitization due to their wide array of germline-encoded inhibitory and activating receptors, which differs from the receptor diversity found in B and T lymphocytes that is due to the use of recombination-activation gene (RAG) enzymes. Although NK cells have traditionally been described as natural killers that provide a first line of defense prior to the induction of adaptive immunity, a more complex view of NK cells is beginning to emerge, indicating they may also function in various immunoregulatory roles and have the capacity to shape adaptive immune responses. With the growing appreciation for the diverse functions of NK cells, and recent technological advancements that allow for a more in-depth understanding of NK cell biology, we can now begin to explore new ways to manipulate NK cells to increase their clinical utility. In this overview unit, we introduce the reader to various aspects of NK cell biology by reviewing topics ranging from NK cell diversity and function, mouse models, and the roles of NK cells in health and disease, to potential clinical applications. © 2015 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Anthony E Zamora
- Department of Dermatology, University of California, Davis, Sacramento, California
| | | | - Ethan G Aguilar
- Department of Dermatology, University of California, Davis, Sacramento, California
| | - William J Murphy
- Department of Dermatology, University of California, Davis, Sacramento, California.,Department of Internal Medicine, University of California, Davis, Sacramento, California
| |
Collapse
|
20
|
Effect of surgery on pancreatic tumor-dependent lymphocyte asset: modulation of natural killer cell frequency and cytotoxic function. Pancreas 2015; 44:386-93. [PMID: 25621568 PMCID: PMC4358707 DOI: 10.1097/mpa.0000000000000288] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Tumor burden and invasiveness establish a microenvironment that surgery could alter. This study shows a comprehensive analysis of size, dynamics, and function of peripheral lymphocyte subsets in pancreatic cancer patients before and at different times after duodenopancreatectomy. METHODS Lymphocyte frequency and natural cytotoxicity were evaluated by flow cytometry and in vitro assay on peripheral blood from initial and advanced-stage pancreatic cancer patients before (BS), at day 7 (PS7), and at day 30 (PS30) after surgery. RESULTS An increase in natural killer (NK) cells and the diminution of B-cells occurred at PS30, whereas cytotoxicity decreased at PS7. The positive correlation between NK frequency and cytotoxicity at BS and PS7 revealed an altered NK behavior. The elevation of NK cell frequency at PS30, an initial defect in CD56bright NK, and the aberrant correlation between NK frequency and cytotoxicity remained significant in advanced-stage patients, whereas the diminution of NK cytotoxicity only affected initial stage patients. CONCLUSIONS The NK cell functional ability is altered in presurgery patients; duodenopancreatectomy is associated with short-term impairment of NK function and with a long-term NK cell augmentation and reversion of the aberrant NK behavior, which may impact on immunosurveillance against residual cancer.
Collapse
|
21
|
Fang L, Gong J, Wang Y, Liu R, Li Z, Wang Z, Zhang Y, Zhang C, Song C, Yang A, Ting JPY, Jin B, Chen L. MICA/B expression is inhibited by unfolded protein response and associated with poor prognosis in human hepatocellular carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:76. [PMID: 25228093 PMCID: PMC4174668 DOI: 10.1186/s13046-014-0076-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/06/2014] [Indexed: 12/19/2022]
Abstract
Background MICA/B are major ligands for NK cell activating receptor NKG2D and previous studies showed that the serum level of soluble MICA (sMICA) is an independent prognostic factor for advanced human hepatocellular carcinoma. However, the correlation between cellular MICA/B expression pattern and human hepatocellular carcinoma progression has not been well explored. The unfolded protein response is one of the main causes of resistance to chemotherapy and radiotherapy in tumor cells. However, whether the UPR in HCC could regulate the expression levels of MICA/B and affect the sensitivity of HCC cells to NK cell cytolysis has not been established yet. Methods MICA/B expression pattern was evaluated by immunohistochemistry and Kaplan-Meier survival analysis was done to explore the relationship between MICA/B expression level and patient survival. The protein and mRNA expression levels of MICA/B in SMMC7721 and HepG2 cells treated by tunicamycin were evaluated by flow cytometry, Western Blot and RT-PCR. The cytotoxicity analysis was performed with the CytoTox 96 Non-Radioactive LDH Cytotoxicity Assay. Results MICA/B was highly expressed in human hepatocellular carcinoma and the expression level was significantly and negatively associated with tumor-node metastasis (TNM) stages. Patients with low level of MICA/B expression showed a trend of shorter survival time. The unfolded protein response (UPR) downregulated the expression of MICA/B. This decreased protein expression occurred via post-transcriptional regulation and was associated with proteasomal degradation. Moreover, decreased expression level of MICA/B led to the attenuated sensitivity of human HCC to NK cell cytotoxicity. Conclusion These new findings of the connection of MICA/B, UPR and NK cells may represent a new concrete theory of NK cell regulation in HCC, and suggest that targeting this novel NK cell-associated immune evasion pathway may be meaningful in treating patients with HCC. Electronic supplementary material The online version of this article (doi:10.1186/s13046-014-0076-7) contains supplementary material, which is available to authorized users.
Collapse
|
22
|
Cui J, Wang H, Liu S, Qiu X, Jiang Z, Wang X. Transcriptome analysis of the gill of Takifugu rubripes using Illumina sequencing for discovery of SNPs. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2014; 10:44-51. [DOI: 10.1016/j.cbd.2014.03.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/18/2014] [Accepted: 03/20/2014] [Indexed: 12/01/2022]
|