1
|
Zhang SW, Wang H, Ding XH, Xiao YL, Shao ZM, You C, Gu YJ, Jiang YZ. Bidirectional crosstalk between therapeutic cancer vaccines and the tumor microenvironment: Beyond tumor antigens. FUNDAMENTAL RESEARCH 2023; 3:1005-1024. [PMID: 38933006 PMCID: PMC11197801 DOI: 10.1016/j.fmre.2022.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 11/20/2022] Open
Abstract
Immunotherapy has rejuvenated cancer therapy, especially after anti-PD-(L)1 came onto the scene. Among the many therapeutic options, therapeutic cancer vaccines are one of the most essential players. Although great progress has been made in research on tumor antigen vaccines, few phase III trials have shown clinical benefits. One of the reasons lies in obstruction from the tumor microenvironment (TME). Meanwhile, the therapeutic cancer vaccine reshapes the TME in an ambivalent way, leading to immune stimulation or immune escape. In this review, we summarize recent progress on the interaction between therapeutic cancer vaccines and the TME. With respect to vaccine resistance, innate immunosuppressive TME components and acquired resistance caused by vaccination are both involved. Understanding the underlying mechanism of this crosstalk provides insight into the treatment of cancer by directly targeting the TME or synergizing with other therapeutics.
Collapse
Affiliation(s)
- Si-Wei Zhang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Han Wang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xiao-Hong Ding
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu-Ling Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Ya-Jia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
2
|
Abascal J, Oh MS, Liclican EL, Dubinett SM, Salehi-Rad R, Liu B. Dendritic Cell Vaccination in Non-Small Cell Lung Cancer: Remodeling the Tumor Immune Microenvironment. Cells 2023; 12:2404. [PMID: 37830618 PMCID: PMC10571973 DOI: 10.3390/cells12192404] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/23/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) remains one of the leading causes of death worldwide. While NSCLCs possess antigens that can potentially elicit T cell responses, defective tumor antigen presentation and T cell activation hinder host anti-tumor immune responses. The NSCLC tumor microenvironment (TME) is composed of cellular and soluble mediators that can promote or combat tumor growth. The composition of the TME plays a critical role in promoting tumorigenesis and dictating anti-tumor immune responses to immunotherapy. Dendritic cells (DCs) are critical immune cells that activate anti-tumor T cell responses and sustain effector responses. DC vaccination is a promising cellular immunotherapy that has the potential to facilitate anti-tumor immune responses and transform the composition of the NSCLC TME via tumor antigen presentation and cell-cell communication. Here, we will review the features of the NSCLC TME with an emphasis on the immune cell phenotypes that directly interact with DCs. Additionally, we will summarize the major preclinical and clinical approaches for DC vaccine generation and examine how effective DC vaccination can transform the NSCLC TME toward a state of sustained anti-tumor immune signaling.
Collapse
Affiliation(s)
- Jensen Abascal
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA; (J.A.); (M.S.O.); (E.L.L.); (S.M.D.)
| | - Michael S. Oh
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA; (J.A.); (M.S.O.); (E.L.L.); (S.M.D.)
| | - Elvira L. Liclican
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA; (J.A.); (M.S.O.); (E.L.L.); (S.M.D.)
| | - Steven M. Dubinett
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA; (J.A.); (M.S.O.); (E.L.L.); (S.M.D.)
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095-1690, USA
| | - Ramin Salehi-Rad
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA; (J.A.); (M.S.O.); (E.L.L.); (S.M.D.)
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Bin Liu
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA; (J.A.); (M.S.O.); (E.L.L.); (S.M.D.)
| |
Collapse
|
3
|
Ai H, Yang H, Li L, Ma J, Liu K, Li Z. Cancer/testis antigens: promising immunotherapy targets for digestive tract cancers. Front Immunol 2023; 14:1190883. [PMID: 37398650 PMCID: PMC10311965 DOI: 10.3389/fimmu.2023.1190883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/26/2023] [Indexed: 07/04/2023] Open
Abstract
Digestive tract cancers, including esophageal, gastric, and colorectal cancers, are the major cause of death among cancer patients worldwide due to the heterogeneity of cancer cells, which limits the effectiveness of traditional treatment methods. Immunotherapy represents a promising treatment strategy for improving the prognosis of patients with digestive tract cancers. However, the clinical application of this approach is limited by the absence of optimal targets. Cancer/testis antigens are characterized by low or absent expression in normal tissues, but high expression in tumor tissues, making them an attractive target for antitumor immunotherapy. Recent preclinical trials have shown promising results for cancer/testis antigen-targeted immunotherapy in digestive cancer. However, practical problems and difficulties in clinical application remain. This review presents a comprehensive analysis of cancer/testis antigens in digestive tract cancers, covering their expression, function, and potential as an immunotherapy target. Additionally, the current state of cancer/testis antigens in digestive tract cancer immunotherapy is discussed, and we predict that these antigens hold great promise as an avenue for breakthroughs in the treatment of digestive tract cancers.
Collapse
Affiliation(s)
- Huihan Ai
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Hang Yang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Liang Li
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Jie Ma
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Department of Molecular and Cellular Biology, China-United States (US) Hormel (Henan) Cancer Institute, Zhengzhou, Henan, China
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhi Li
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Ura B, Capaci V, Aloisio M, Di Lorenzo G, Romano F, Ricci G, Monasta L. A Targeted Proteomics Approach for Screening Serum Biomarkers Observed in the Early Stage of Type I Endometrial Cancer. Biomedicines 2022; 10:1857. [PMID: 36009404 PMCID: PMC9405144 DOI: 10.3390/biomedicines10081857] [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: 06/22/2022] [Revised: 07/22/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Endometrial cancer (EC) is the most common gynecologic malignancy, and it arises in the inner part of the uterus. Identification of serum biomarkers is essential for diagnosing the disease at an early stage. In this study, we selected 44 healthy controls and 44 type I EC at tumor stage 1, and we used the Immuno-oncology panel and the Target 96 Oncology III panel to simultaneously detect the levels of 92 cancer-related proteins in serum, using a proximity extension assay. By applying this methodology, we identified 20 proteins, associated with the outcome at binary logistic regression, with a p-value below 0.01 for the first panel and 24 proteins with a p-value below 0.02 for the second one. The final multivariate logistic regression model, combining proteins from the two panels, generated a model with a sensitivity of 97.67% and a specificity of 83.72%. These results support the use of the proposed algorithm after a validation phase.
Collapse
Affiliation(s)
- Blendi Ura
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
| | - Valeria Capaci
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
| | - Michelangelo Aloisio
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
| | - Giovanni Di Lorenzo
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
| | - Federico Romano
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
| | - Giuseppe Ricci
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34129 Trieste, Italy
| | - Lorenzo Monasta
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
| |
Collapse
|
5
|
Gowhari Shabgah A, Amir A, Gardanova ZR, Olegovna Zekiy A, Thangavelu L, Ebrahimi Nik M, Ahmadi M, Gholizadeh Navashenaq J. Interleukin-25: New perspective and state-of-the-art in cancer prognosis and treatment approaches. Cancer Med 2021; 10:5191-5202. [PMID: 34128588 PMCID: PMC8335817 DOI: 10.1002/cam4.4060] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer is a leading cause of death which imposes a substantial financial burden. Among the several mechanisms involved in cancer progression, imbalance of immune cell-derived factors such as cytokines and chemokines plays a central role. IL-25, as a member of the IL-17 cytokine subfamily, exerts a paradoxical role in cancer, including tumor supportive and tumor suppressive. Hence, we have tried to clarify the role of IL-25 and its receptor in tumor progression and cancer prognosis. It has been confirmed that IL-25 exerts a tumor-suppressive role through inducing infiltration of eosinophils and B cells into the tumor microenvironment and activating the apoptotic pathways. In contrast, the tumor-supportive function has been implemented by activating inflammatory cascades, promoting cell cycle, and inducing type-2 immune responses. Since IL-25 has been dysregulated in tumor tissues and this dysregulation is involved in cancer development, its examination can be used as a tumor diagnostic and prognostic biomarker. Moreover, IL-25-based therapeutic approaches have shown promising results in cancer inhibition. In cancers in which IL-25 has a tumor-suppressive function, employing IL-25-enhancing approaches, such as Virulizin® and dihydrobenzofuran administration, has potentially inhibited tumor cell growth. On the other hand, in the case of IL-25-dependent tumor progression, using IL-25 blocking methods, including anti-IL-25 antibodies, might be a complementary approach to the other anticancer agent. Collectively, it is hoped, IL-25 might be a promising target in cancer treatment.
Collapse
Affiliation(s)
- Arezoo Gowhari Shabgah
- School of MedicineBam University of Medical SciencesBamIran
- Student Research CommitteeBam University of Medical SciencesBamIran
| | - Azwar Amir
- Wahidin Sudirohusodo Hospital MakassarMakassarTamalanreaIndonesia
| | - Zhanna R. Gardanova
- Department of PsychotherapyPirogov Russian National Research Medical UniversityMoscowRussia
| | - Angelina Olegovna Zekiy
- Department of Prosthetic DentistrySechenov First Moscow State Medical UniversityMoscowRussia
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of medical and Technical SciencesSaveetha UniversityChennaiIndia
| | - Maryam Ebrahimi Nik
- Nanotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
| | - Majid Ahmadi
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
| | | |
Collapse
|
6
|
Nistor GI, Dillman RO. Cytokine network analysis of immune responses before and after autologous dendritic cell and tumor cell vaccine immunotherapies in a randomized trial. J Transl Med 2020; 18:176. [PMID: 32316978 PMCID: PMC7171762 DOI: 10.1186/s12967-020-02328-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/02/2020] [Indexed: 02/08/2023] Open
Abstract
Background In a randomized phase II trial conducted in patients with metastatic melanoma, patient-specific autologous dendritic cell vaccines (DCV) were associated with longer survival than autologous tumor cell vaccines (TCV). Both vaccines presented antigens from cell-renewing autologous tumor cells. The current analysis was performed to better understand the immune responses induced by these vaccines, and their association with survival. Methods 110 proteomic markers were measured at a week-0 baseline, 1 week before the first of 3 weekly vaccine injections, and at week-4, 1 week after the third injection. Data was presented as a deviation from normal controls. A two-component principal component (PC) statistical analysis and discriminant analysis were performed on this data set for all patients and for each treatment cohort. Results At baseline PC-1 contained 64.4% of the variance and included the majority of cytokines associated with Th1 and Th2 responses, which positively correlated with beta-2-microglobulin (B2M), programmed death protein-1 (PD-1) and transforming growth factor beta (TGFβ1). Results were similar at baseline for both treatment cohorts. After three injections, DCV-treated patients showed correlative grouping among Th1/Th17 cytokines on PC-1, with an inverse correlation with B2M, FAS, and IL-18, and correlations among immunoglobulins in PC-2. TCV-treated patients showed a positive correlation on PC-1 among most of the cytokines and tumor markers B2M and FAS receptor. There were also correlative changes of IL12p40 with both Th1 and Th2 cytokines and TGFβ1. Discriminant analysis provided additional evidence that DCV was associated with innate, Th1/Th17, and Th2 responses while TCV was only associated with innate and Th2 responses. Conclusions These analyses confirm that DCV induced a different immune response than that induced by TCV, and these immune responses were associated with improved survival. Trial registration Clinical trials.gov NCT004936930 retrospectively registered 28 July 2009
Collapse
Affiliation(s)
- Gabriel I Nistor
- AIVITA Biomedical, Inc., 18301 Von Karman, Suite 130, Irvine, CA, 92612, USA
| | - Robert O Dillman
- AIVITA Biomedical, Inc., 18301 Von Karman, Suite 130, Irvine, CA, 92612, USA.
| |
Collapse
|
7
|
Cytokine and Cancer Biomarkers Detection: The Dawn of Electrochemical Paper-Based Biosensor. SENSORS 2020; 20:s20071854. [PMID: 32230808 PMCID: PMC7180619 DOI: 10.3390/s20071854] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/24/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022]
Abstract
Although the established ELISA-based sensing platforms have many benefits, the importance of cytokine and cancer biomarkers detection for point-of-care diagnostics has propelled the search for more specific, sensitive, simple, accessible, yet economical sensor. Paper-based biosensor holds promise for future in-situ applications and can provide rapid analysis and data without the need to conduct in a laboratory. Electrochemical detection plays a vital role in interpreting results obtained from qualitative assessment to quantitative determination. In this review, various factors affecting the design of an electrochemical paper-based biosensor are highlighted and discussed in depth. Different detection methods, along with the latest development in utilizing them in cytokine and cancer biomarkers detection, are reviewed. Lastly, the fabrication of portable electrochemical paper-based biosensor is ideal in deliberating positive societal implications in developing countries with limited resources and accessibility to healthcare services.
Collapse
|
8
|
Seim-Wikse T, Skancke E, Nødtvedt A, Jörundsson E, Grotmol T, Kristensen AT, Bjørnvad CR. Comparison of body condition score and other minimally invasive biomarkers between dogs with gastric carcinoma and dogs with chronic gastritis. J Am Vet Med Assoc 2020; 254:226-235. [PMID: 30605380 DOI: 10.2460/javma.254.2.226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To identify minimally invasive biomarkers to help differentiate dogs with gastric carcinoma from those with chronic gastritis. DESIGN Prospective study. ANIMALS 15 dogs with gastric carcinoma, 29 dogs with chronic gastritis, and 7 healthy dogs. PROCEDURES Dogs with clinical signs of upper gastrointestinal tract disease for > 14 days that underwent gastroscopy or necropsy for collection of gastric biopsy specimens for histologic evaluation were prospectively enrolled. Gastric carcinoma and chronic gastritis were diagnosed on the basis of histologic findings. Additionally, gastric biopsy specimens were collected endoscopically from 7 healthy (control) dogs while they were anesthetized for a routine neutering procedure. Prior to being anesthetized for gastroscopy or euthanized, all dogs underwent a physical examination, and a blood sample was collected for quantification of select serum biomarker concentrations. Histologic findings, body condition score (BCS), and serum biomarker concentrations were compared among the 3 groups. RESULTS Dogs with gastric carcinoma were significantly older and had a significantly lower BCS, lower serum folate concentration, and greater serum C-reactive protein (CRP) concentration, compared with dogs with chronic gastritis and control dogs. CONCLUSIONS AND CLINICAL RELEVANCE Results suggested that age > 8 years, BCS < 4, serum CRP concentration > 25 mg/L, and an abnormally low serum folate concentration might be useful noninvasive biomarkers for identification of dogs with gastric carcinoma. For underweight older dogs with signs of upper gastrointestinal tract disease and high serum CRP and low serum folate concentrations, gastric biopsy specimens should be obtained and evaluated so that a prompt definitive diagnosis can be made and appropriate treatment initiated.
Collapse
|
9
|
Sun S, Gong C, Yuan K. LncRNA UCA1 promotes cell proliferation, invasion and migration of laryngeal squamous cell carcinoma cells by activating Wnt/β-catenin signaling pathway. Exp Ther Med 2018; 17:1182-1189. [PMID: 30679991 PMCID: PMC6327537 DOI: 10.3892/etm.2018.7097] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 07/31/2018] [Indexed: 12/11/2022] Open
Abstract
In view of the poor prognosis of laryngeal squamous cell carcinoma (LSCC) and the functionality of long non-coding (lnc)RNA UCA1 in different types of cancer, the present study aimed to investigate the role of UCA1 in the development and progression of LSCC. A total of 90 patients with LSCC and 90 healthy subjects were enrolled in the present study. Expression levels of UCA1 in tumor tissues and adjacent healthy tissues, as well as serum of patients with LSCC and normal controls were detected by reverse transcription-quantitative polymerase chain reaction. Receiver operating characteristic curve analysis was performed to evaluate the diagnostic value of serum UCA1 for LSCC. Survival curves were plotted using the Kaplan-Meier method and employed to evaluate the prognosic values of serum UCA1 for LSCC. Cell proliferation, migration and invasion were detected using the cell proliferation assay, and Transwell migration and invasion assays, respectively. Expression levels of Wnt/β-catenin-associated proteins were detected by western blot analysis. Results indicated that the expression levels of UCA1 were significantly higher in tumor tissues compared with adjacent healthy tissues in the majority of patients with LSCC. In addition, serum levels of UCA1 were significantly higher in patients with LSCC coapred with healthy controls. UCA1 overexpression promoted, whereas UCA1 knockdown inhibited the proliferation, migration and invasion of LSCC cells. UCA1 overexpression activated the Wnt/β-catenin signaling pathway in LSCC cells, whereas treatment with Wnt inhibitor reduced the enhancing effects of UCA1 overexpression on the proliferation, migration and invasion of LSCC cells. The present findings suggest that UCA1 can promote cell proliferation, invasion and migration of LSCC cells by activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Suguang Sun
- Department of Otolaryngology-Head and Neck Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Cheng Gong
- Department of Otolaryngology-Head and Neck Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Kun Yuan
- Department of Otolaryngology-Head and Neck Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
10
|
Butterfield LH, Zhao F, Lee S, Tarhini AA, Margolin KA, White RL, Atkins MB, Cohen GI, Whiteside TL, Kirkwood JM, Lawson DH. Immune Correlates of GM-CSF and Melanoma Peptide Vaccination in a Randomized Trial for the Adjuvant Therapy of Resected High-Risk Melanoma (E4697). Clin Cancer Res 2017; 23:5034-5043. [PMID: 28536308 DOI: 10.1158/1078-0432.ccr-16-3016] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/31/2017] [Accepted: 05/19/2017] [Indexed: 12/23/2022]
Abstract
Purpose: E4697 was a multicenter intergroup randomized placebo-controlled phase III trial of adjuvant GM-CSF and/or a multiepitope melanoma peptide vaccine for patients with completely resected, high-risk stage III/IV melanoma.Experimental Design: A total of 815 patients were enrolled from December 1999 to October 2006 into this six-arm study. GM-CSF was chosen to promote the numbers and functions of dendritic cells (DC). The melanoma antigen peptide vaccine (Tyrosinase368-376 (370D), gp100209-217 (210M), MART-127-35) in montanide was designed to promote melanoma-specific CD8+ T-cell responses.Results: Although the overall RFS and OS were not significantly improved with the vaccine or GM-CSF when compared with placebo, immunomodulatory effects were observed in peripheral blood and served as important correlates to this therapeutic study. Peripheral blood was examined to evaluate the impact of GM-CSF and/or the peptide vaccine on peripheral blood immunity and to investigate potential predictive or prognostic biomarkers. A total of 11.3% of unvaccinated patients and 27.1% of vaccinated patients developed peptide-specific CD8+ T-cell responses. HLA-A2+ patients who had any peptide-specific CD8+ T-cell response at day +43 tended to have poorer OS in univariate analysis. Patients receiving GM-CSF had significant reduction in percentages of circulating myeloid dendritic cells (mDC) and plasmacytoid DC (pDC) at day +43. In a subset of patients who received GM-CSF, circulating myeloid-derived suppressor cells (MDSC), and anti-GM-CSF-neutralizing antibodies (Nabs) were also modulated. The majority of patients developed anti-GM-CSF Nabs, which correlated with improved RFS and OS.Conclusions: The assessment of cellular and humoral responses identified counterintuitive immune system changes correlating with clinical outcome. Clin Cancer Res; 23(17); 5034-43. ©2017 AACR.
Collapse
Affiliation(s)
| | - Fengmin Zhao
- Dana Farber Cancer Institute - ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | - Sandra Lee
- Dana Farber Cancer Institute - ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | | | | | - Richard L White
- Levine Cancer Institute, Carolinas Healthcare System, Charlotte, North Carolina
| | | | - Gary I Cohen
- Greater Baltimore Medical Center, Baltimore, Maryland
| | | | | | - David H Lawson
- Winship Cancer institute of Emory University, Atlanta, Georgia
| |
Collapse
|
11
|
Li Y, Yang F, Sang L, Zhu J, Han X, Shan F, Li S, Zhai J, Wang D, Lu C, Sun X. Enhanced therapeutic effects against murine colon carcinoma induced by a Colon 26/Ag85A-CD226 tumor cell vaccine. Oncol Rep 2015; 34:1795-804. [PMID: 26238268 DOI: 10.3892/or.2015.4137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/22/2015] [Indexed: 11/06/2022] Open
Abstract
Genetically modified tumor cells represent one of the most effective cancer vaccine strategies. In the present study, we describe our approach for inducing an immune response against a colon carcinoma in BALB/c mice, using a Colon 26 tumor cell line expressing Ag85A and CD226. We investigated whether CD226 plays a promotive role for Ag85A against Colon 26 colon carcinoma. The therapeutic efficacy was investigated. The cytotoxic T lymphocyte (CTL) and natural killer (NK) cell cytotoxicity were assessed. Dynamic changes in interferon (IFN)-γ levels in the spleen and the number of IFN-γ-producing CD4+ or CD8+ T cells in the spleen or mesenteric lymph nodes were detected by enzyme-linked immunoabsorbent assay or flow cytometry. Extended survival times, delayed appearances of tumors, and reduced tumor volumes were achieved by preventive vaccination with the Colon 26/Ag85A-CD226 tumor cell vaccine. NK cell or CTL cytotoxicity in the spleens of mice immunized with the Colon 26/Ag85A-CD226 tumor cell vaccine was significantly higher than that in the other treatment groups. The numbers of CD4+ IFN-γ+ and CD8+ IFN-γ+ T cells were both significantly increased in mice immunized with the Colon 26/Ag85A-CD226 tumor cell vaccine in both the spleen and mesenteric lymph nodes. Our results indicated that the tumor vaccine expressing Ag85A and CD226 induced more intensive antitumor immunity than tumor vaccine expressing Ag85A or CD226 only. Moreover, the results suggest that Ag85A and CD226 play a synergistic antitumor effect and CD226 could be used as a genetic adjuvant to enhance the effects of Ag85A vaccine against murine colon carcinoma.
Collapse
Affiliation(s)
- Yan Li
- Department of Immunology, School of Basic Medical Science, China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Fangli Yang
- Department of Immunology, School of Basic Medical Science, China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Lixuan Sang
- Department of Immunology, School of Basic Medical Science, China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Junfeng Zhu
- Department of Immunology, School of Basic Medical Science, China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Xue Han
- Department of Immunology, School of Basic Medical Science, China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Shengjun Li
- Department of Immunology, School of Basic Medical Science, China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Jingbo Zhai
- Department of Immunology, School of Basic Medical Science, China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Danan Wang
- Department of Immunology, School of Basic Medical Science, China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Changlong Lu
- Department of Immunology, School of Basic Medical Science, China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| | - Xun Sun
- Department of Immunology, School of Basic Medical Science, China Medical University, Heping, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
12
|
Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2014; 1:1111-1134. [PMID: 23170259 PMCID: PMC3494625 DOI: 10.4161/onci.21494] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) occupy a central position in the immune system, orchestrating a wide repertoire of responses that span from the development of self-tolerance to the elicitation of potent cellular and humoral immunity. Accordingly, DCs are involved in the etiology of conditions as diverse as infectious diseases, allergic and autoimmune disorders, graft rejection and cancer. During the last decade, several methods have been developed to load DCs with tumor-associated antigens, ex vivo or in vivo, in the attempt to use them as therapeutic anticancer vaccines that would elicit clinically relevant immune responses. While this has not always been the case, several clinical studies have demonstrated that DC-based anticancer vaccines are capable of activating tumor-specific immune responses that increase overall survival, at least in a subset of patients. In 2010, this branch of clinical research has culminated with the approval by FDA of a DC-based therapeutic vaccine (sipuleucel-T, Provenge®) for use in patients with asymptomatic or minimally symptomatic metastatic hormone-refractory prostate cancer. Intense research efforts are currently dedicated to the identification of the immunological features of patients that best respond to DC-based anticancer vaccines. This knowledge may indeed lead to personalized combination strategies that would extend the benefit of DC-based immunotherapy to a larger patient population. In addition, widespread enthusiasm has been generated by the results of the first clinical trials based on in vivo DC targeting, an approach that holds great promises for the future of DC-based immunotherapy. In this Trial Watch, we will summarize the results of recently completed clinical trials and discuss the progress of ongoing studies that have evaluated/are evaluating DC-based interventions for cancer therapy.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Institut Gustave Roussy; Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Nikolic Nielsen L, Kjelgaard-Hansen M, Kristensen AT. Monocyte chemotactic protein-1 and other inflammatory parameters in Bernese Mountain dogs with disseminated histiocytic sarcoma. Vet J 2013; 198:424-8. [DOI: 10.1016/j.tvjl.2013.07.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 07/19/2013] [Accepted: 07/21/2013] [Indexed: 12/31/2022]
|
14
|
Status of Active Specific Immunotherapy for Stage II, Stage III, and Resected Stage IV Colon Cancer. CURRENT COLORECTAL CANCER REPORTS 2013. [DOI: 10.1007/s11888-013-0182-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
15
|
Zeestraten ECM, Speetjens FM, Welters MJP, Saadatmand S, Stynenbosch LFM, Jongen R, Kapiteijn E, Gelderblom H, Nijman HW, Valentijn ARPM, Oostendorp J, Fathers LM, Drijfhout JW, van de Velde CJH, Kuppen PJK, van der Burg SH, Melief CJM. Addition of interferon-α to the p53-SLP® vaccine results in increased production of interferon-γ in vaccinated colorectal cancer patients: a phase I/II clinical trial. Int J Cancer 2012; 132:1581-91. [PMID: 22948952 DOI: 10.1002/ijc.27819] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 08/09/2012] [Indexed: 12/25/2022]
Abstract
We previously established safety and immunogenicity of a p53 synthetic long peptides (p53-SLP®) vaccine. In the current trial, we investigated whether combination of interferon-alpha (IFN-α) with p53-SLP® is both safe and able to improve the induced p53-specific IFN-γ response. Eleven colorectal cancer patients successfully treated for metastatic disease were enrolled in this study. Of these, nine patients completed follow-up after two injections with p53-SLP® together with IFN-α. Safety and p53-specific immune responses were determined before and after vaccination. Furthermore, cryopreserved PBMCs were compared head-to-head to cryopreserved PBMCs obtained in our previous trial with p53-SLP® only. Toxicity of p53-SLP® vaccination in combination with IFN-α was limited to Grade 1 or 2, with predominantly small ongoing swellings at the vaccination site. All patients harbored p53-specific T cells after vaccination and most patients showed p53-specific antibodies. Compared to the previous trial, addition of IFN-α significantly improved the frequency of p53-specific T cells in IFN-γ ELISPOT. Moreover, in this trial, p53-specific T cells were detectable in blood samples of all patients in a direct ex vivo multiparameter flowcytometric assay, opposed to only 2 of 10 patients vaccinated with p53-SLP® only. Finally, patients in this trial displayed a broader p53-specific immunoglobulin-G response, indicating an overall better p53-specific T-helper response. Our study shows that p53-SLP® vaccination combined with IFN-α injection is safe and capable of inducing p53-specific immunity. When compared to a similar trial with p53-SLP® vaccination alone the combination was found to induce significantly more IFN-γ producing p53-specific T cells.
Collapse
|
16
|
Yang Z, Chen B, Pei X, Shangguan F. Multiplex analysis of tumor multidrug-resistance genes expression with photonic suspension array. Analyst 2012; 137:3343-8. [PMID: 22683740 DOI: 10.1039/c2an35243e] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An easy-operated suspension array based on silica colloidal crystal beads is developed for multiplex analysis of tumor multidrug-resistance genes expression, such as multidrug resistance 1 (MDR1) and multidrug resistance-associated protein 1 (MRP1), and potentially single nucleotide polymorphism. In order to obtain high fluorescence intensity, controlled PCR was used to amplify targets at the samples pretreatment stage. By optimizing the conditions a hybridization procedure, which is similar to nucleic acids analysis with binary probes, was established. Small amounts of analytes 10(-19) M could be detected by the method. The K562 cell, human myeloma cell, and its multidrug-resistance string, adriamycin-selected P-glycoprotein-overexpressed K562/A02, were analyzed by using an established procedure to validate feasibility. Clinical blood samples were detected by our method and real-time PCR simultaneously to validate accuracy. Moreover, when combined with multiplex controlled PCR, the method successfully meets the requirements of multiplex analysis. Hence, the method presented is a good method for multiplex analysis of tumor multidrug-resistance genes expression.
Collapse
Affiliation(s)
- Zixue Yang
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Nanjing, China 210009
| | | | | | | |
Collapse
|
17
|
Speetjens FM, Zeestraten ECM, Kuppen PJK, Melief CJM, van der Burg SH. Colorectal cancer vaccines in clinical trials. Expert Rev Vaccines 2011; 10:899-921. [PMID: 21692708 DOI: 10.1586/erv.11.63] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This article elucidates current strategies of active immunotherapy for colorectal cancer patients with a focus on T-cell mediated immunotherapy. Poor prognosis of especially stage III and IV colorectal cancer patients emphasizes the need for advanced therapeutic intervention. Here, we refer to clinical trials using either tumor cell-derived vaccines or tumor antigen vaccines with a special interest on safety, induced immune responses, clinical benefit and efforts to improve the clinical impact of these vaccines in the context of colorectal cancer treatment.
Collapse
Affiliation(s)
- Frank M Speetjens
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
18
|
The immuno-regulatory effects of Schisandra chinensis and its constituents on human monocytic leukemia cells. Molecules 2011; 16:4836-49. [PMID: 21666550 PMCID: PMC6264271 DOI: 10.3390/molecules16064836] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/07/2011] [Accepted: 06/09/2011] [Indexed: 11/17/2022] Open
Abstract
Many diseases occur when the immune system is weakened. Intracellular signals activate immuno-responsive cells to produce cytokines that modulate the immune response. Schisandra chinensis has been used traditionally to treat general fatigue, neurasthenia, and spontaneous sweating. In the present study, the effect of constituents of S. chinensis on cytokine release by human monocytic leukemia cells (THP-1) was tested using microparticle-based flow cytometric analysis. Two major lignans, schizandrin (Sch) and gomisin A (Gom A), were identified and shown to induce interleukin (IL)-8, macrophage inflammatory protein-1β (MIP-1β), and granulocyte-macrophage-colony stimulating factor (GM-CSF) release by THP-1 cells. By reverse transcription polymerase chain reaction (RT-PCR) or quantitative real-time PCR, there was a dose-dependent increase of IL-8, MIP-1β and GM-CSF mRNA levels. Thus, Sch and Gom A from S. chinensis enhance cytokine release by THP-1 cells and this effect occurs through mRNA upregulation. Upregulation of MIP-1β and GM-CSF in particular may have clinical applications. Therefore, S. chinensis may be therapeutically beneficial by promoting humoral and cell-mediated immune responses.
Collapse
|
19
|
Abstract
Measuring cytokine production is an integral part of measuring immune response during immunotherapy. Current technologies allow the simultaneous quantification of multiple cytokines in a variety of tissues. Patterns of cytokine response can be referred to as cytokine profiles. This article discusses the experimental design and data analysis of a number of studies that examined cytokine profiles in humans. We highlight potential sources of variability, both due to assay nuances and the diversity of human populations. We present strategies for analyzing data, emphasizing both multidimensional analysis and the value of treating each donor as his or her own control.
Collapse
Affiliation(s)
- Janet C Siebert
- Robert W Franz Cancer Research Center, Earle A Chiles Research Institute, Providence Cancer Center, Portland, OR 97213, USA.
| | | |
Collapse
|
20
|
Abstract
Multiplex technologies at both the mRNA and protein level have given researchers the ability to determine the co-ordinated cellular response to any given stimuli, in both biological and laboratory-derived fluids. This article examines some of the different mRNA and protein multiplex platforms available and how they may be used in assessing vaccine immunity.
Collapse
Affiliation(s)
- Gendie E Lash
- Reproductive and Vascular Biology Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| | | |
Collapse
|