1
|
Schuch LF, Girardi FM, Kirschnick LB, Chaves ALF, Kowalski LP, Bologna‐Molina R, Junior WNW, Martins MD, Santos‐Silva AR. Targeted Therapy for the Management of Oral Potentially Malignant Disorders: A Systematic Review. Oral Dis 2025. [DOI: 10.1111/odi.15307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/23/2025] [Indexed: 05/14/2025]
Abstract
ABSTRACTObjectiveThis study aimed to integrate the available data published in the literature to identify and elucidate the impact of targeted therapy for oral potentially malignant disorders (OPMDs).Material and MethodsThis systematic review was conducted according to the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta‐analyses (PRISMA) 2020 statement. An electronic search across five databases with no publication date restriction was conducted in January 2023 and subsequently updated in January 2024. Eligibility criteria included studies focusing on targeted therapies. Interventions that either did not align with the specific definition of targeted therapy or failed to meet the criteria for precision‐based treatment approaches were excluded from this review.ResultsSix clinical trials were identified, all conducted in the United States. The sample size ranged from 12 to 150 subjects. Celecoxib, erlotinib, cetuximab, metformin, and nivolumab were employed as targeted systemic therapies at varying doses. Some patients experienced disease progression with no statistically significant differences in histological improvement rates or size increases. Certain studies reported treatment‐related toxicity.ConclusionThe findings of this review do not support the use of targeted therapy in managing OPMDs, highlighting the need for additional clinical trials to further evaluate their efficacy.
Collapse
Affiliation(s)
- Lauren Frenzel Schuch
- Department of Diagnosis in Pathology and Oral Medicine, School of Dentistry Universidad de la República Montevideo Uruguay
| | - Fábio Muradás Girardi
- Department of Oral Pathology, School of Dentistry Universidade Federal do Rio Grande do Sul Porto Alegre RS Brazil
- Head and Neck Department Ana Nery Hospital Santa Cruz do Sul RS Brazil
| | - Laura Borges Kirschnick
- Department of Oral Diagnosis, Piracicaba Dental School Universidade de Campinas Piracicaba SP Brazil
| | | | | | - Ronell Bologna‐Molina
- Department of Diagnosis in Pathology and Oral Medicine, School of Dentistry Universidad de la República Montevideo Uruguay
| | | | - Manoela Domingues Martins
- Department of Oral Pathology, School of Dentistry Universidade Federal do Rio Grande do Sul Porto Alegre RS Brazil
| | - Alan Roger Santos‐Silva
- Department of Oral Diagnosis, Piracicaba Dental School Universidade de Campinas Piracicaba SP Brazil
| |
Collapse
|
2
|
Kubaski Benevides AP, Marin AM, Wosniaki DK, Oliveira RN, Koerich GM, Kusma BN, Munhoz EC, Zanette DL, Aoki MN. Expression of HOTAIR and PTGS2 as potential biomarkers in chronic myeloid leukemia patients in Brazil. Front Oncol 2024; 14:1443346. [PMID: 39450252 PMCID: PMC11499243 DOI: 10.3389/fonc.2024.1443346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024] Open
Abstract
Chronic myeloid leukemia (CML) is a clonal myeloproliferative neoplasm in which all the patients has the translocation (9;22) that generates de BCR::ABL1 tyrosine kinase. Despite this disease possessing a good biomarker (BCR::ABL1 transcripts level) for diagnosis and prognosis, many studies has been performed to investigate other molecules, such as the long noncoding RNAs (lncRNAs) and mRNAs, as potential biomarkers with the aim of predicting a change in BCR::ABL1 levels and as an associated biomarker. A RNAseq was performed comparing 6 CML patients with high BCR::ABL1 expression with 6 healthy control individuals, comprising the investigation cohort to investigate these molecules. To validate the results obtained by RNAseq, samples of 87 CML patients and 42 healthy controls were used in the validation cohort by RT-qPCR assays. The results showed lower expression of HOTAIR and PTGS2 in CML patients. The HOTAIR expression is inversely associated with BCR::ABL1 expression in imatinib-treated CML patients, and to PTGS2 showing that CML patients with high BCR::ABL1 expression showed reduced PTGS2 expression.
Collapse
Affiliation(s)
- Ana Paula Kubaski Benevides
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Anelis Maria Marin
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Denise K. Wosniaki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Rafaela Noga Oliveira
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Gabriela Marino Koerich
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Bianca Nichele Kusma
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | | | - Dalila Luciola Zanette
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Mateus Nóbrega Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| |
Collapse
|
3
|
Papadimitriou E, Chatzellis E, Dimitriadi A, Kaltsas GA, Theocharis S, Alexandraki KI. Prognostic Biomarkers in Pituitary Tumours: A Systematic Review. TOUCHREVIEWS IN ENDOCRINOLOGY 2023; 19:42-53. [PMID: 38187082 PMCID: PMC10769480 DOI: 10.17925/ee.2023.19.2.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/07/2023] [Indexed: 01/09/2024]
Abstract
Pituitary tumours (PTs) are the second most common intracranial tumour. Although the majority show benign behaviour, they may exert aggressive behaviour and can be resistant to treatment. The aim of this review is to report the recently identified biomarkers that might have possible prognostic value. Studies evaluating potentially prognostic biomarkers or a therapeutic target in invasive/recurrent PTs compared with either non-invasive or non-recurrent PTs or normal pituitaries are included in this review. In the 28 included studies, more than 911 PTs were evaluated. A systematic search identified the expression of a number of biomarkers that may be positively correlated with disease recurrence or invasion in PT, grouped according to role: (1) insensitivity to anti-growth signals: minichromosome maintenance protein 7; (2) evasion of the immune system: cyclooxygenase 2, arginase 1, programmed cell death protein 1 (PD-1)/programmed death ligand 2, cluster of differentiation (CD) 80/CD86; (3) sustained angiogenesis: endothelial cell-specific molecule, fibroblast growth factor receptor, matrix metalloproteinase 9, pituitary tumour transforming gene; (4) self-sufficiency in growth signals: epidermal growth factor receptor; and (5) tissue invasion: matrix metalloproteinase 9, fascin protein. Biomarkers with a negative correlation with disease recurrence or invasion include: (1) insensitivity to anti-growth signals: transforming growth factor β1, Smad proteins; (2) sustained angiogenesis: tissue inhibitor of metalloproteinase 1; (3) tissue invasion: Wnt inhibitory factor 1; and (4) miscellaneous: co-expression of glial fibrillary acidic protein and cytokeratin, and oestrogen receptors α36 and α66. PD-1/programmed cell death ligand 1 showed no clear association with invasion or recurrence, while cyclin A, cytotoxic T lymphocyte-associated protein 4, S100 protein, ephrin receptor, galectin-3 , neural cell adhesion molecule, protein tyrosine phosphatase 4A3 and steroidogenic factor 1 had no association with invasion or recurrence of PT. With the aim to develop a more personalized approach to the treatment of PT, and because of the limited number of molecular targets currently studied in the context of recurrent PT and invasion, a better understanding of the most relevant of these biomarkers by well-d esigned interventional studies will lead to a better understanding of the molecular profile of PT. This should also meet the increased need of treatable molecular targets.
Collapse
Affiliation(s)
- Eirini Papadimitriou
- First Department of Propaedeutic Medicine, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleftherios Chatzellis
- Endocrinology Diabetes and Metabolism Department, 251 Hellenic Air Force and VA General Hospital, Athens, Greece
| | | | - Gregory A Kaltsas
- First Department of Propaedeutic Medicine, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | |
Collapse
|
4
|
Zheng Y, Sun L, Guo J, Ma J. The crosstalk between ferroptosis and anti-tumor immunity in the tumor microenvironment: molecular mechanisms and therapeutic controversy. Cancer Commun (Lond) 2023; 43:1071-1096. [PMID: 37718480 PMCID: PMC10565387 DOI: 10.1002/cac2.12487] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/13/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023] Open
Abstract
The advent of immunotherapy has significantly reshaped the landscape of cancer treatment, greatly enhancing therapeutic outcomes for multiple types of cancer. However, only a small subset of individuals respond to it, underscoring the urgent need for new methods to improve its response rate. Ferroptosis, a recently discovered form of programmed cell death, has emerged as a promising approach for anti-tumor therapy, with targeting ferroptosis to kill tumors seen as a potentially effective strategy. Numerous studies suggest that inducing ferroptosis can synergistically enhance the effects of immunotherapy, paving the way for a promising combined treatment method in the future. Nevertheless, recent research has raised concerns about the potential negative impacts on anti-tumor immunity as a consequence of inducing ferroptosis, leading to conflicting views within the scientific community about the interplay between ferroptosis and anti-tumor immunity, thereby underscoring the necessity of a comprehensive review of the existing literature on this relationship. Previous reviews on ferroptosis have touched on related content, many focusing primarily on the promoting role of ferroptosis on anti-tumor immunity while overlooking recent evidence on the inhibitory effects of ferroptosis on immunity. Others have concentrated solely on discussing related content either from the perspective of cancer cells and ferroptosis or from immune cells and ferroptosis. Given that both cancer cells and immune cells exist in the tumor microenvironment, a one-sided discussion cannot comprehensively summarize this topic. Therefore, from the perspectives of both tumor cells and tumor-infiltrating immune cells, we systematically summarize the current conflicting views on the interplay between ferroptosis and anti-tumor immunity, intending to provide potential explanations and identify the work needed to establish a translational basis for combined ferroptosis-targeted therapy and immunotherapy in treating tumors.
Collapse
Affiliation(s)
- Yichen Zheng
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduSichuanP. R. China
| | - Lingqi Sun
- Department of NeurologyAir Force Hospital of the Western Theater of the Chinese People's Liberation ArmyChengduSichuanP. R. China
| | - Jiamin Guo
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduSichuanP. R. China
| | - Ji Ma
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduSichuanP. R. China
| |
Collapse
|
5
|
Kalkavan H, Rühl S, Shaw JJP, Green DR. Non-lethal outcomes of engaging regulated cell death pathways in cancer. NATURE CANCER 2023; 4:795-806. [PMID: 37277528 PMCID: PMC10416134 DOI: 10.1038/s43018-023-00571-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 04/27/2023] [Indexed: 06/07/2023]
Abstract
Regulated cell death (RCD) is essential for successful systemic cancer therapy. Yet, the engagement of RCD pathways does not inevitably result in cell death. Instead, RCD pathways can take part in diverse biological processes if the cells survive. Consequently, these surviving cells, for which we propose the term 'flatliners', harbor important functions. These evolutionarily conserved responses can be exploited by cancer cells to promote their own survival and growth, with challenges and opportunities for cancer therapy.
Collapse
Affiliation(s)
- Halime Kalkavan
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Sebastian Rühl
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
- T3 Pharmaceuticals AG, Allschwil, Switzerland
| | - Jeremy J P Shaw
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Douglas R Green
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
6
|
Maekawa N, Konnai S, Asano Y, Sajiki Y, Deguchi T, Okagawa T, Watari K, Takeuchi H, Takagi S, Hosoya K, Kim S, Ohta H, Kato Y, Suzuki Y, Murata S, Ohashi K. Exploration of serum biomarkers in dogs with malignant melanoma receiving anti-PD-L1 therapy and potential of COX-2 inhibition for combination therapy. Sci Rep 2022; 12:9265. [PMID: 35665759 PMCID: PMC9166720 DOI: 10.1038/s41598-022-13484-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 05/13/2022] [Indexed: 12/15/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) such as anti-PD-L1 antibodies are widely used to treat human cancers, and growing evidence suggests that ICIs are promising treatments for canine malignancies. However, only some canine oral malignant melanoma (OMM) cases respond to ICIs. To explore biomarkers predictive of survival in dogs with pulmonary metastatic OMM receiving the anti-PD-L1 antibody c4G12 (n = 27), serum concentrations of prostaglandin E2 (PGE2), cytokines, chemokines, and growth factors were measured prior to treatment initiation. Among 12 factors tested, PGE2, interleukin (IL)-12p40, IL-8, monocyte chemotactic protein-1 (MCP-1), and stem cell factor (SCF) were higher in OMM dogs compared to healthy dogs (n = 8). Further, lower baseline serum PGE2, MCP-1, and vascular endothelial growth factor (VEGF)-A concentrations as well as higher IL-2, IL-12, and SCF concentrations predicted prolonged overall survival. These observations suggest that PGE2 confers resistance against anti-PD-L1 therapy through immunosuppression and thus is a candidate target for combination therapy. Indeed, PGE2 suppressed IL-2 and interferon (IFN)-γ production by stimulated canine peripheral blood mononuclear cells (PBMCs), while inhibition of PGE2 biosynthesis using the COX-2 inhibitor meloxicam in combination with c4G12 enhanced Th1 cytokine production by PBMCs. Thus, serum PGE2 may be predictive of c4G12 treatment response, and concomitant use of COX-2 inhibitors may enhance ICI antitumor efficacy.
Collapse
Affiliation(s)
- Naoya Maekawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Satoru Konnai
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.
| | - Yumie Asano
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yamato Sajiki
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Tatsuya Deguchi
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Tomohiro Okagawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kei Watari
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroto Takeuchi
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Satoshi Takagi
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Department of Veterinary Surgery 1, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Kenji Hosoya
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Sangho Kim
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Ohta
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiko Suzuki
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Shiro Murata
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kazuhiko Ohashi
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
7
|
Sun P, Quan JC, Wang S, Zhuang M, Liu Z, Guan X, Wang GY, Wang HY, Wang XS. lncRNA-PACER upregulates COX-2 and PGE2 through the NF-κB pathway to promote the proliferation and invasion of colorectal-cancer cells. Gastroenterol Rep (Oxf) 2021; 9:257-268. [PMID: 34316376 PMCID: PMC8309685 DOI: 10.1093/gastro/goaa060] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/06/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND p50-associated cyclooxygenase-2 extragenic RNA (PACER) is a recently identified antisense long non-coding RNA (lncRNA) located on the upstream of the promoter region of cyclooxygenase-2 (COX-2). Preliminary studies have suggested that PACER is involved in the regulation of COX-2 expression in macrophagocyte and osteosarcoma cells. However, the role of this lncRNA in colorectal cancer (CRC) remains elusive. Here, we investigated the expression of PACER and its effect on cell proliferation and invasion to explore the role of PACER in CRC. METHODS Real-time quantitative PCR (RT-qPCR) analysis was used to evaluate the expression of PACER in CRC tissues and cells. Methyl thiazolyl tetrazolium (MTT) analysis was then used to investigate the inhibition effect of PACER knock-down in cell proliferation. The promoting role of this lncRNA on invasion by CRC cells was analysed by wound-healing assays, colony-formation assay, and transwell assays. We then used fluorescence in situ hybridization (FISH) to establish the subcellular localization of PACER. COX-2 protein levels were quantified by Western blot analysis and grayscale scanning analysis following the knock-down of PACER. Luciferase assay was carried out to monitor the modulation of the COX-2 promoter region by PACER. Tumor xenografts models were used to investigate the impact of PACER on the tumorigenesis of CRC cells in vivo. Enzyme-linked immunosorbent assay (ELISA) was then used to quantify prostaglandin E2 (PGE2) production upon knock-down of PACER. RESULTS RT-qPCR analysis revealed that PACER was highly expressed in CRC tissues and cells, and a high PACER-expression level was associated with poor prognosis. MTT assay, wound-healing assay, colony-formation assay, and transwell assay revealed that PACER enhanced CRC-cell proliferation, invasion, and metastasis in vitro. Analysis of lncRNA localization by FISH showed that it mainly resided in the nucleus. RT-qPCR showed that PACER increased mRNA levels of COX-2. Western blot analysis demonstrated, under normal circumstances, that knock-down of PACER decreased the COX-2 protein level. In the case of p50 absence, COX-2 protein increased rapidly and remained highly expressed after knocking down PACER. Luciferase assay revealed that PACER modulated the COX-2 promoter region. Mouse xenograft models of CRC revealed that PACER promoted colorectal tumorigenesis in vivo. ELISA revealed that PACER knock-down inhibited PGE2 production. CONCLUSIONS PACER modulates COX-2 expression through the nuclear factor kappa B (NF-κB) pathway in CRC. An increased level of PACER enhances proliferation, migration, and invasion of tumor cells by increasing COX-2 and PGE2 synthesis.
Collapse
Affiliation(s)
- Peng Sun
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P. R. China
- Department of Gastrointestinal Surgery, Shenzhen Hospital, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, P. R. China
| | - Ji-Chuan Quan
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Song Wang
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Meng Zhuang
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Zheng Liu
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Xu Guan
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Gui-Yu Wang
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Hong-Ying Wang
- Department of State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Xi-Shan Wang
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P. R. China
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
8
|
Almatroodi SA, Syed MA, Rahmani AH. Potential Therapeutic Targets of Curcumin, Most Abundant Active Compound of Turmeric Spice: Role in the Management of Various Types of Cancer. Recent Pat Anticancer Drug Discov 2021; 16:3-29. [PMID: 33143616 DOI: 10.2174/1574892815999201102214602] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Curcumin, an active compound of turmeric spice, is one of the most-studied natural compounds and has been widely recognized as a chemopreventive agent. Several molecular mechanisms have proven that curcumin and its analogs play a role in cancer prevention through modulating various cell signaling pathways as well as in the inhibition of the carcinogenesis process. OBJECTIVE To study the potential role of curcumin in the management of various types of cancer through modulating cell signalling molecules based on available literature and recent patents. METHODS A wide-ranging literature survey was performed based on Scopus, PubMed, PubMed Central, and Google scholar for the implication of curcumin in cancer management, along with a special emphasis on human clinical trials. Moreover, patents were searched through www.google.com/patents, www.freepatentsonline.com, and www.freshpatents.com. RESULT Recent studies based on cancer cells have proven that curcumin has potential effects against cancer cells as it prevents the growth of cancer and acts as a cancer therapeutic agent. Besides, curcumin exerted anti-cancer effects by inducing apoptosis, activating tumor suppressor genes, cell cycle arrest, inhibiting tumor angiogenesis, initiation, promotion, and progression stages of tumor. It was established that co-treatment of curcumin and anti-cancer drugs could induce apoptosis and also play a significant role in the suppression of the invasion and metastasis of cancer cells. CONCLUSION Accumulating evidences suggest that curcumin has the potential to inhibit cancer growth, induce apoptosis, and modulate various cell signaling pathway molecules. Well-designed clinical trials of curcumin based on human subjects are still needed to establish the bioavailability, mechanism of action, efficacy, and safe dose in the management of various cancers.
Collapse
Affiliation(s)
- Saleh A Almatroodi
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraydah 52571, Saudi Arabia
| | - Mansoor Ali Syed
- Department of Biotechnology, Faculty of Natural Sciences, Translational Research Lab, Jamia Millia Islamia, New Delhi 110025, India
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraydah 52571, Saudi Arabia
| |
Collapse
|
9
|
Parvathareddy SK, Siraj AK, Annaiyappanaidu P, Al-Sobhi SS, Al-Dayel F, Al-Kuraya KS. Prognostic Significance of COX-2 Overexpression in BRAF-Mutated Middle Eastern Papillary Thyroid Carcinoma. Int J Mol Sci 2020; 21:ijms21249498. [PMID: 33327467 PMCID: PMC7764962 DOI: 10.3390/ijms21249498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 11/27/2020] [Accepted: 12/10/2020] [Indexed: 01/05/2023] Open
Abstract
The cyclooxygenase-2 (COX-2)–prostaglandin E2 (PGE2) pathway has been implicated in carcinogenesis, with BRAF mutation shown to promote PGE2 synthesis. This study was conducted to evaluate COX-2 expression in a large cohort of Middle Eastern papillary thyroid carcinoma (PTC), and further evaluate the prognostic significance of COX-2 expression in strata of BRAF mutation status. BRAF mutation analysis was performed using Sanger sequencing, and COX-2 expression was evaluated immunohistochemically using tissue microarray (TMA). COX-2 overexpression, noted in 43.2% (567/1314) of cases, was significantly associated with poor prognostic markers such as extra-thyroidal extension, lymph-node metastasis, and higher tumor stage. COX-2 was also an independent predictor of poor disease-free survival (DFS). Most notably, the association of COX-2 expression with DFS differed by BRAF mutation status. COX-2 overexpression was associated with poor DFS in BRAF-mutant but not BRAF wild-type PTCs, with a multivariate-adjusted hazard ratio of 2.10 (95% CI = 1.52–2.92; p < 0.0001) for COX-2 overexpressed tumors in BRAF-mutant PTC. In conclusion, the current study shows that COX-2 plays a key role in prognosis of PTC patients, especially in BRAF-mutated tumors. Our data suggest the potential therapeutic role of COX-2 inhibition in patients with BRAF-mutated PTC.
Collapse
Affiliation(s)
- Sandeep Kumar Parvathareddy
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia; (S.K.P.); (A.K.S.); (P.A.)
| | - Abdul K. Siraj
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia; (S.K.P.); (A.K.S.); (P.A.)
| | - Padmanaban Annaiyappanaidu
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia; (S.K.P.); (A.K.S.); (P.A.)
| | - Saif S. Al-Sobhi
- Department of Surgery, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia;
| | - Fouad Al-Dayel
- Department of Pathology, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia;
| | - Khawla S. Al-Kuraya
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia; (S.K.P.); (A.K.S.); (P.A.)
- Correspondence: ; Tel.: +966-1-205-5167
| |
Collapse
|
10
|
Lizardo DY, Kuang C, Hao S, Yu J, Huang Y, Zhang L. Immunotherapy efficacy on mismatch repair-deficient colorectal cancer: From bench to bedside. Biochim Biophys Acta Rev Cancer 2020; 1874:188447. [PMID: 33035640 PMCID: PMC7886024 DOI: 10.1016/j.bbcan.2020.188447] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
Abstract
Colorectal cancers (CRCs) with deficient mismatch repair (dMMR) or microsatellite instability-high (MSI-H) often have sustained responses to immune checkpoint inhibitors (ICIs) including selective monoclonal antibodies against Program Death 1 (PD-1), Programmed Death Ligand 1(PD-L1), and cytotoxic T lymphocyte associated antigen 4 (CTLA-4). However, a substantial fraction of dMMR CRCs do not respond or ultimately develop resistance to immunotherapy. The majority (~85%) of CRCs are MMR proficient (pMMR) or microsatellite stable (MSS) and lack response to ICIs. Understanding the biology and mechanisms underlying dMMR-associated immunogenicity is urgently needed for improving the therapeutic efficacy of immunotherapy on CRC. Compared to pMMR/MSS CRCs, dMMR/MSI CRCs typically have increased tumor mutational burden (TMB), lower response rate to 5-fluorouracil-based chemotherapy, distinctive immunological features such as high tumor-infiltrating lymphocytes (TILs), and better prognosis. Here, we review the current understanding of the clinical relevance of dMMR/MSI in CRCs, the molecular basis and rationales for targeting dMMR CRC with immunotherapy, and clinical approaches using ICIs as single agents or in combination with other therapies for MSI-H CRCs. Furthermore, we address the potential strategies to sensitize pMMR/MSS CRC to immunotherapy by converting an immunologically "cold" microenvironment into a "hot" one.
Collapse
Affiliation(s)
- Darleny Y Lizardo
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chaoyuan Kuang
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Suisui Hao
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jian Yu
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Yi Huang
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Lin Zhang
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
11
|
Lipid metabolic Reprogramming: Role in Melanoma Progression and Therapeutic Perspectives. Cancers (Basel) 2020; 12:cancers12113147. [PMID: 33121001 PMCID: PMC7692067 DOI: 10.3390/cancers12113147] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Melanoma is a devastating skin cancer characterized by an impressive metabolic plasticity. Melanoma cells are able to adapt to the tumor microenvironment by using a variety of fuels that contribute to tumor growth and progression. In this review, the authors summarize the contribution of the lipid metabolic network in melanoma plasticity and aggressiveness, with a particular attention to specific lipid classes such as glycerophospholipids, sphingolipids, sterols and eicosanoids. They also highlight the role of adipose tissue in tumor progression as well as the potential antitumor role of drugs targeting critical steps of lipid metabolic pathways in the context of melanoma. Abstract Metabolic reprogramming contributes to the pathogenesis and heterogeneity of melanoma. It is driven both by oncogenic events and the constraints imposed by a nutrient- and oxygen-scarce microenvironment. Among the most prominent metabolic reprogramming features is an increased rate of lipid synthesis. Lipids serve as a source of energy and form the structural foundation of all membranes, but have also emerged as mediators that not only impact classical oncogenic signaling pathways, but also contribute to melanoma progression. Various alterations in fatty acid metabolism have been reported and can contribute to melanoma cell aggressiveness. Elevated expression of the key lipogenic fatty acid synthase is associated with tumor cell invasion and poor prognosis. Fatty acid uptake from the surrounding microenvironment, fatty acid β-oxidation and storage also appear to play an essential role in tumor cell migration. The aim of this review is (i) to focus on the major alterations affecting lipid storage organelles and lipid metabolism. A particular attention has been paid to glycerophospholipids, sphingolipids, sterols and eicosanoids, (ii) to discuss how these metabolic dysregulations contribute to the phenotype plasticity of melanoma cells and/or melanoma aggressiveness, and (iii) to highlight therapeutic approaches targeting lipid metabolism that could be applicable for melanoma treatment.
Collapse
|
12
|
Zhao G, Chen W, He J, Cui C, Zhao L, Zhao Y, Sun C, Nie D, Jin F, Kong L. Analysis of Cyclooxygenase 2, Programmed Cell Death Ligand 1, and Arginase 1 Expression in Human Pituitary Adenoma. World Neurosurg 2020; 144:e660-e673. [PMID: 32920160 DOI: 10.1016/j.wneu.2020.09.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Cyclooxygenase 2 (COX-2) is a key enzyme in the synthesis of prostaglandins. Recent studies have shown that overexpression of COX-2 can reduce the antitumor effect of the immune system by inhibiting the proliferation of B and T lymphocytes. Programmed cell death ligand 1 (PD-L1) was the first functionally characterized ligand of programmed cell death protein 1. It plays an important role in maintaining peripheral and central immune tolerance by combining with programmed cell death protein 1. Arginase 1 (ARG1) can process L-arginine in the local microenvironment and affect the function of T cells, resulting in immune escape. In this study, COX-2, PD-L1, and ARG1 expression in human pituitary adenoma (PA) and their relationship were investigated, which provided an initial theoretic basis for further study of the immune escape mechanism in PA in cellular and animal experiments. METHODS The protein expression of COX-2, PD-L1, and ARG1 in 55 PA samples was detected by immunohistochemistry, with 10 normal brain tissues as the control group. The location of COX-2, PD-L1, and ARG1 in PA cells was studied by double immunofluorescence colocalization. The results of immunohistochemistry were further verified by Western blot. RESULTS The expression of COX-2, PD-L1, and ARG1 in PA was significantly higher than that in normal brain tissue. In functional PA (FPA) and nonfunctional PA (NFPA), there was no significant difference in the expression of COX-2 and PD-L1, whereas ARG1 was higher in NFPA. Moreover, the protein expression level of COX-2 was positively correlated with that of PD-L1 and ARG1, and the expression of PD-L1 was positively correlated with that of ARG1. Immunofluorescence confocal imaging showed that COX-2, PD-L1, and ARG1 were all expressed in the cytoplasm of PA cells, and the physical positions of COX-2, PD-L1, and ARG1 were partially coincident. CONCLUSIONS These findings indicate that overexpression of COX-2, PD-L1, and ARG1 may be involved in the pathogenesis of PA. ARG1 plays a more important role in the development of NFPA. By upregulating the expression of PD-L1, COX-2 may promote the expression of ARG1, forming the COX-2/PD-L1/ARG1 signal pathway in promoting the occurrence and development of PA. Perhaps further study of the pathogenesis of PA can start with the mechanism of immune escape.
Collapse
Affiliation(s)
- Guodong Zhao
- Clinical Medical College, Jining Medical University, Jining, Shandong Province, China
| | - Weike Chen
- Clinical Medical College, Jining Medical University, Jining, Shandong Province, China
| | - Juanjuan He
- Clinical Medical College, Jining Medical University, Jining, Shandong Province, China
| | - Changmeng Cui
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China
| | - Lihua Zhao
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China
| | - Yueshu Zhao
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China
| | - Cuilian Sun
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China
| | - Dongli Nie
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China
| | - Feng Jin
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China
| | - Lingsheng Kong
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China.
| |
Collapse
|
13
|
Longhi G, van Sinderen D, Ventura M, Turroni F. Microbiota and Cancer: The Emerging Beneficial Role of Bifidobacteria in Cancer Immunotherapy. Front Microbiol 2020; 11:575072. [PMID: 33013813 PMCID: PMC7507897 DOI: 10.3389/fmicb.2020.575072] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/17/2020] [Indexed: 12/15/2022] Open
Abstract
Many intestinal bacteria are believed to be involved in various inflammatory and immune processes that influence tumor etiology because of their metabolic properties and their ability to alter the microbiota homeostasis. Although many functions of the microbiota are still unclear, there is compelling experimental evidence showing that the intestinal microbiota is able to modulate carcinogenesis and the response to anticancer therapies, both in the intestinal tract and other body sites. Among the wide variety of gut-colonizing microorganisms, various species belonging to the Bifidobacterium genus are believed to elicit beneficial effects on human physiology and on the host-immune system. Recent findings, based on preclinical mouse models and on human clinical trials, have demonstrated the impact of gut commensals including bifidobacteria on the efficacy of tumor-targeting immunotherapy. Although the underlying molecular mechanisms remain obscure, bifidobacteria and other microorganisms have become a promising aid to immunotherapeutic procedures that are currently applied to treat cancer. The present review focuses on strategies to recruit the microbiome in order to enhance anticancer responses and develop therapies aimed at fighting the onset and progression of malignancies.
Collapse
Affiliation(s)
- Giulia Longhi
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- Alimentary Pharmabotic Centre (APC) Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.,Microbiome Research Hub, University of Parma, Parma, Italy
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.,Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
14
|
Smith PG, Roque D, Ching MM, Fulton A, Rao G, Reader JC. The Role of Eicosanoids in Gynecological Malignancies. Front Pharmacol 2020; 11:1233. [PMID: 32982722 PMCID: PMC7479818 DOI: 10.3389/fphar.2020.01233] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/28/2020] [Indexed: 12/20/2022] Open
Abstract
Eicosanoids, bio-active lipid molecules, evoke a multitude of biological effects that directly affect cancer cells and indirectly affect tumor microenvironment. An emerging role has been shown for eicosanoids in the pathogenesis of gynecological malignancies which include cancers of the vulva, vagina, cervix, uterine, and ovary. Eicosanoid biosynthesis pathways start at the metabolism of phospholipids by phospholipase A2 then proceeding to one of three pathways: the cyclooxygenase (COX), lipoxygenase (LOX), or P450 epoxygenase pathways. The most studied eicosanoid pathways include COX and LOX; however, more evidence is appearing to support further study of the P450 epoxygenase pathway in gynecologic cancers. In this review, we present the current knowledge of the role of COX, LOX and P450 pathways in the pathogenesis of gynecologic malignancies. Vulvar and vaginal cancer, the rarest subtypes, there is association of COX-2 expression with poor disease specific survival in vulvar cancer and, in vaginal cancer, COX-2 expression has been found to play a role in mucosal inflammation leading to disease susceptibility and transmission. Cervical cancer is associated with COX-2 levels 7.4 times higher than in healthy tissues. Additionally, HPV elevates COX-2 levels through the EGFR pathway and HIV promotes elevated COX-2 levels in cervical tissue as well as increases PGE2 levels eliciting inflammation and progression of cancer. Evidence supports significant roles for both the LOX and COX pathways in uterine cancer. In endometrial cancer, there is increased expression of 5-LOX which is associated with adverse outcomes. Prostanoids in the COX pathway PGE2 and PGF2α have been shown to play a significant role in uterine cancer including alteration of proliferation, adhesion, migration, invasion, angiogenesis, and the inflammatory microenvironment. The most studied gynecological malignancy in regard to the potential role of eicosanoids in tumorigenesis is ovarian cancer in which all three pathways have shown to be associated or play a role in ovarian tumorigenesis directly on the tumor cell or through modulation of the tumor microenvironment. By identifying the gaps in knowledge, additional pathways and targets could be identified in order to obtain a better understanding of eicosanoid signaling in gynecological malignancies and identify potential new therapeutic approaches.
Collapse
Affiliation(s)
- Paige G. Smith
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Dana Roque
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Mc Millan Ching
- Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amy Fulton
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
- Baltimore Veterans Administration Medical Center, Baltimore, MD, United States
| | - Gautam Rao
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Jocelyn C. Reader
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
15
|
Tudor DV, Bâldea I, Lupu M, Kacso T, Kutasi E, Hopârtean A, Stretea R, Gabriela Filip A. COX-2 as a potential biomarker and therapeutic target in melanoma. Cancer Biol Med 2020; 17:20-31. [PMID: 32296574 PMCID: PMC7142851 DOI: 10.20892/j.issn.2095-3941.2019.0339] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/15/2020] [Indexed: 12/15/2022] Open
Abstract
With a constantly increasing incidence, cutaneous melanoma has raised the need for a better understanding of its complex microenvironment that may further guide therapeutic options. Melanoma is a model tumor in immuno-oncology. Inflammation represents an important hallmark of cancer capable of inducing and sustaining tumor development. The inflammatory process also orchestrates the adaptative immunosuppression of tumor cells that helps them to evade immune destruction. Besides its role in proliferation, angiogenesis, and apoptosis, cyclooxygenase-2 (COX-2) is a well-known promoter of immune suppression in melanoma. COX-2 inhibitors are closely involved in this condition. This review attempts to answer two controversial questions: is COX-2 a valuable prognostic factor? Among all COX-2 inhibitors, is celecoxib a suitable adjuvant in melanoma therapy?
Collapse
Affiliation(s)
- Diana Valentina Tudor
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Ioana Bâldea
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Mihai Lupu
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Teodor Kacso
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Eniko Kutasi
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Andreea Hopârtean
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Roland Stretea
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Adriana Gabriela Filip
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| |
Collapse
|
16
|
Tang B, Guo ZS, Bartlett DL, Yan DZ, Schane CP, Thomas DL, Liu J, McFadden G, Shisler JL, Roy EJ. Synergistic Combination of Oncolytic Virotherapy and Immunotherapy for Glioma. Clin Cancer Res 2020; 26:2216-2230. [PMID: 32019860 DOI: 10.1158/1078-0432.ccr-18-3626] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/05/2019] [Accepted: 01/30/2020] [Indexed: 01/10/2023]
Abstract
PURPOSE We hypothesized that the combination of a local stimulus for activating tumor-specific T cells and an anti-immunosuppressant would improve treatment of gliomas. Virally encoded IL15Rα-IL15 as the T-cell activating stimulus and a prostaglandin synthesis inhibitor as the anti-immunosuppressant were combined with adoptive transfer of tumor-specific T cells. EXPERIMENTAL DESIGN Two oncolytic poxviruses, vvDD vaccinia virus and myxoma virus, were each engineered to express the fusion protein IL15Rα-IL15 and a fluorescent protein. Viral gene expression (YFP or tdTomato Red) was confirmed in the murine glioma GL261 in vitro and in vivo. GL261 tumors in immunocompetent C57BL/6J mice were treated with vvDD-IL15Rα-YFP vaccinia virus or vMyx-IL15Rα-tdTr combined with other treatments, including vaccination with GARC-1 peptide (a neoantigen for GL261), rapamycin, celecoxib, and adoptive T-cell therapy. RESULTS vvDD-IL15Rα-YFP and vMyx-IL15Rα-tdTr each infected and killed GL261 cells in vitro. In vivo, NK cells and CD8+ T cells were increased in the tumor due to the expression of IL15Rα-IL15. Each component of a combination treatment contributed to prolonging survival: an oncolytic virus, the IL15Rα-IL15 expressed by the virus, a source of T cells (whether by prevaccination or adoptive transfer), and prostaglandin inhibition all synergized to produce elimination of gliomas in a majority of mice. vvDD-IL15Rα-YFP occasionally caused ventriculitis-meningitis, but vMyx-IL15Rα-tdTr was safe and effective, causing a strong infiltration of tumor-specific T cells and eliminating gliomas in 83% of treated mice. CONCLUSIONS IL15Rα-IL15-armed oncolytic poxviruses provide potent antitumor effects against brain tumors when combined with adoptive T-cell therapy, rapamycin, and celecoxib.
Collapse
Affiliation(s)
- Bingtao Tang
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Zong Sheng Guo
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David L Bartlett
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David Z Yan
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Claire P Schane
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Diana L Thomas
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Jia Liu
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Grant McFadden
- Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Joanna L Shisler
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Edward J Roy
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois.
| |
Collapse
|
17
|
COX-2 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1277:87-104. [PMID: 33119867 DOI: 10.1007/978-3-030-50224-9_6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumorigenesis is a multistep, complicated process, and many studies have been completed over the last few decades to elucidate this process. Increasingly, many studies have shifted focus toward the critical role of the tumor microenvironment (TME), which consists of cellular players, cell-cell communications, and extracellular matrix (ECM). In the TME, cyclooxygenase-2 (COX-2) has been found to be a key molecule mediating the microenvironment changes. COX-2 is an inducible form of the enzyme that converts arachidonic acid into the signal transduction molecules (thromboxanes and prostaglandins). COX-2 is frequently expressed in many types of cancers and has been closely linked to its occurrence, progression, and prognosis. For example, COX-2 has been shown to (1) regulate tumor cell growth, (2) promote tissue invasion and metastasis, (3) inhibit apoptosis, (4) suppress antitumor immunity, and (5) promote sustainable angiogenesis. In this chapter, we summarize recent advances of studies that have evaluated COX-2 signaling in TME.
Collapse
|
18
|
Abstract
New therapies that promote antitumour immunity have been recently developed. Most of these immunomodulatory approaches have focused on enhancing T-cell responses, either by targeting inhibitory pathways with immune checkpoint inhibitors, or by targeting activating pathways, as with chimeric antigen receptor T cells or bispecific antibodies. Although these therapies have led to unprecedented successes, only a minority of patients with cancer benefit from these treatments, highlighting the need to identify new cells and molecules that could be exploited in the next generation of immunotherapy. Given the crucial role of innate immune responses in immunity, harnessing these responses opens up new possibilities for long-lasting, multilayered tumour control.
Collapse
|
19
|
Tumour-infiltrating lymphocytes, programmed death ligand 1 and cyclooxygenase-2 expression in skin melanoma of elderly patients: clinicopathological correlations. Melanoma Res 2019; 28:547-554. [PMID: 29794562 DOI: 10.1097/cmr.0000000000000462] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Age is an important prognostic factor in melanoma; notably, elderly patients tend to present with advanced stage skin melanoma (SM) and worse outcome. Moreover, SM is an immunogenic cancer, and its interaction with the aging immune system could have an effect on biologic behaviour of this disease. Tumour-infiltrating lymphocytes (TILs) could represent the host response in SM; it has been shown that higher grade of TILs is associated with better survival. Moreover, programmed death ligand 1 (PD-L1) and cyclooxygenase-2 (COX-2) are potential markers of host immune response and inflammation. We retrospectively reviewed 113 consecutive cases of early-stage SM that occurred in patients aged greater than or equal to 65 years at the time of diagnosis, followed between January 2010 and March 2014 at the University and General Hospital of Udine, Italy. The aim of this study was to evaluate TILs grade, PD-L1 expression on TILs and tumour expression of PD-L1 and COX-2 and their prognostic value in elderly patients with early SM. A better disease-free survival as well as melanoma-specific survival (MSS) was significantly associated with TILs [hazard ratios (HR): 0.41, 95% confidence interval (CI): 0.20-0.84, P=0.02 and HR: 0.37, 95% CI: 0.17-0.82, P=0.01, respectively]. PD-L1 positivity on TILs was associated with a better MSS (HR: 0.41, 95% CI: 0.17-0.97, P=0.04). Moreover, among patients with TILs, those showing COX-2 positivity on tumour cells and no PD-L1 expression on TILs had a worse disease-free survival and MSS (HR: 5.18, 95% CI: 1.33-20.23, P=0.018; HR: 6.21, 95% CI: 1.20-32.24, P=0.03; respectively). Immune and inflammatory markers deserve further investigation in aging patients with melanoma.
Collapse
|
20
|
Tai Y, Zhang LH, Gao JH, Zhao C, Tong H, Ye C, Huang ZY, Liu R, Tang CW. Suppressing growth and invasion of human hepatocellular carcinoma cells by celecoxib through inhibition of cyclooxygenase-2. Cancer Manag Res 2019; 11:2831-2848. [PMID: 31114336 PMCID: PMC6497485 DOI: 10.2147/cmar.s183376] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 01/23/2019] [Indexed: 02/05/2023] Open
Abstract
Purpose: Biomarkers are lacking in hepatocellular carcinoma (HCC). Cyclooxygenase-2 (COX-2) and its metabolites play crucial roles in the process of inflammation-tumor transformation. This study was aimed to detect COX-2 expression in HCC tissues and evaluate the effects of a COX-2 inhibitor, celecoxib, on biological behaviors of HCC cell lines in vitro. Methods: COX-2 expression was detected by immunohistochemistry on a human HCC tissue microarray. The correlations of COX-2 expression with tumor clinicopathological variables and overall survival were analyzed. The proliferation, apoptosis, cell cycle distribution, invasion capacity, and related signaling molecules of HCC cells after incubated with COX-2 inhibitor celecoxib were evaluated in vitro. Results: Expression levels of COX-2 in HCC tissues were significantly higher than those in paracancerous tissues. The TNM stage III-IV, tumor size >5 cm, lymphovascular invasion and distant metastasis was higher in high COX-2 expression group compared with that in low COX-2 expression group. Patients with low COX-2 expression achieved better 5-year overall survival than those with high COX-2 expression. Treatment with celecoxib was sufficient to inhibit cell proliferation, promote apoptosis, and induce G0/G1 cell cycle arrest in HCC cells with concentration- and time-dependent manners. Celecoxib up-regulated E-cadherin protein through inhibiting COX-2-prostaglandin E2 (PGE2)-PGE2 receptor 2 (EP2)-p-Akt/p-ERK signaling pathway to suppress HCC cells migration and invasion. Conclusion: High COX-2 expression was associated with advanced TNM stage, larger tumor size, increased lymphovascular invasion and short survival. Targeting inhibition of COX-2 by celecoxib exhibited anti-tumor activities by suppressing proliferation, promoting apoptosis, and inhibiting the aggressive properties of HCC cells.
Collapse
Affiliation(s)
- Yang Tai
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Lin-Hao Zhang
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Jin-Hang Gao
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Chong Zhao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Huan Tong
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Cheng Ye
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Zhi-Yin Huang
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Rui Liu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Cheng-Wei Tang
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| |
Collapse
|
21
|
Hays E, Bonavida B. YY1 regulates cancer cell immune resistance by modulating PD-L1 expression. Drug Resist Updat 2019; 43:10-28. [PMID: 31005030 DOI: 10.1016/j.drup.2019.04.001] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/03/2019] [Accepted: 04/05/2019] [Indexed: 02/08/2023]
Abstract
Recent advances in the treatment of various cancers have resulted in the adaptation of several novel immunotherapeutic strategies. Notably, the recent intervention through immune checkpoint inhibitors has resulted in significant clinical responses and prolongation of survival in patients with several therapy-resistant cancers (melanoma, lung, bladder, etc.). This intervention was mediated by various antibodies directed against inhibitory receptors expressed on cytotoxic T-cells or against corresponding ligands expressed on tumor cells and other cells in the tumor microenvironment (TME). However, the clinical responses were only observed in a subset of the treated patients; it was not clear why the remaining patients did not respond to checkpoint inhibitor therapies. One hypothesis stated that the levels of PD-L1 expression correlated with poor clinical responses to cell-mediated anti-tumor immunotherapy. Hence, exploring the underlying mechanisms that regulate PD-L1 expression on tumor cells is one approach to target such mechanisms to reduce PD-L1 expression and, therefore, sensitize the resistant tumor cells to respond to PD-1/PD-L1 antibody treatments. Various investigations revealed that the overexpression of the transcription factor Yin Yang 1 (YY1) in most cancers is involved in the regulation of tumor cells' resistance to cell-mediated immunotherapies. We, therefore, hypothesized that the role of YY1 in cancer immune resistance may be correlated with PD-L1 overexpression on cancer cells. This hypothesis was investigated and analysis of the reported literature revealed that several signaling crosstalk pathways exist between the regulations of both YY1 and PD-L1 expressions. Such pathways include p53, miR34a, STAT3, NF-kB, PI3K/AKT/mTOR, c-Myc, and COX-2. Noteworthy, many clinical and pre-clinical drugs have been utilized to target these above pathways in various cancers independent of their roles in the regulation of PD-L1 expression. Therefore, the direct inhibition of YY1 and/or the use of the above targeted drugs in combination with checkpoint inhibitors should result in enhancing the cell-mediated anti-tumor cell response and also reverse the resistance observed with the use of checkpoint inhibitors alone.
Collapse
Affiliation(s)
- Emily Hays
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, United States
| | - Benjamin Bonavida
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, United States.
| |
Collapse
|
22
|
Kosumi K, Hamada T, Zhang S, Liu L, da Silva A, Koh H, Twombly TS, Mima K, Morikawa T, Song M, Nowak JA, Nishihara R, Saltz LB, Niedzwiecki D, Ou FS, Zemla T, Mayer RJ, Baba H, Ng K, Giannakis M, Zhang X, Wu K, Giovannucci EL, Chan AT, Fuchs CS, Meyerhardt JA, Ogino S. Prognostic association of PTGS2 (COX-2) over-expression according to BRAF mutation status in colorectal cancer: Results from two prospective cohorts and CALGB 89803 (Alliance) trial. Eur J Cancer 2019; 111:82-93. [PMID: 30826660 PMCID: PMC6436990 DOI: 10.1016/j.ejca.2019.01.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/09/2019] [Accepted: 01/20/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Prostaglandin-endoperoxide synthase 2 (PTGS2, cyclooxygenase-2, COX-2)-prostaglandin E2 (PGE2) pathway promotes tumour progression. Considering evidence suggesting increased PGE2 synthesis by BRAF mutation in tumour cells, we hypothesised that the association of tumour PTGS2 (COX-2) expression with colorectal cancer mortality might be stronger in BRAF-mutated tumours than in BRAF-wild-type tumours. METHODS Using 1708 patients, including 1200 stage I-IV colorectal carcinoma cases in the Nurses' Health Study (NHS) and the Health Professionals Follow-up Study (HPFS) and 508 stage III colon cancer cases in a National Cancer Institute-sponsored randomised controlled trial of adjuvant therapy (CALGB/Alliance 89803), we evaluated tumour PTGS2 (COX-2) expression status using immunohistochemistry. We examined the prognostic association of PTGS2 (COX-2) expression in strata of BRAF mutation status by multivariable Cox proportional hazards regression models to adjust for potential confounders, including disease stage, tumour differentiation, microsatellite instability status and KRAS and PIK3CA mutations. RESULTS In NHS and HPFS, the association of PTGS2 (COX-2) expression with colorectal cancer-specific survival differed by BRAF mutation status (Pinteraction = 0.0005); compared with PTGS2 (COX-2)-negative/low carcinomas, the multivariable-adjusted hazard ratios for PTGS2 (COX-2)-high carcinomas were 2.44 (95% confidence interval, 1.39-4.28) in BRAF-mutated cases and 0.82 (95% confidence interval, 0.65-1.04) in BRAF-wild-type cases. Differential prognostic associations of PTGS2 (COX-2) expression in strata of BRAF mutation status were similarly observed in CALGB/Alliance 89803 trial (Pinteraction = 0.03). CONCLUSIONS The association of tumour PTGS2 (COX-2) expression with colorectal cancer mortality is stronger in BRAF-mutated tumours than in BRAF-wild-type tumours, supporting interactive roles of PTGS2 (COX-2) expression and BRAF mutation statuses in prognostication of patients with colorectal cancer; ClinicalTrials.gov Identifier, NCT00003835.
Collapse
Affiliation(s)
- Keisuke Kosumi
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Sui Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Li Liu
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology and Biostatistics, and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, PR China
| | - Annacarolina da Silva
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Hideo Koh
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Department of Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Tyler S Twombly
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Kosuke Mima
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Teppei Morikawa
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mingyang Song
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Reiko Nishihara
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Leonard B Saltz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Donna Niedzwiecki
- Alliance Statistics and Data Center and Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Fang-Shu Ou
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Tyler Zemla
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Robert J Mayer
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Charles S Fuchs
- Yale Cancer Center, New Haven, CT, USA; Department of Medicine, Yale School of Medicine, New Haven, CT, USA; Smilow Cancer Hospital, New Haven, CT, USA
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Shuji Ogino
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
23
|
Abstract
The documented efficacy of COX-2 inhibitors in cancer chemoprevention and in suppression of metastasis is predominantly attributed to inflammatory responses, whereas their effects on tumor-stromal interaction are poorly understood. Through single-cell transcriptome analyses in an immune-compromised mouse xenograft model and in vitro reconstitution experiments, we uncover a tumor-stromal paracrine pathway in which secretion by tumor cells of the COX-2 product prostaglandin E2 induces prolactin production by stromal cells, which activates signaling in disseminated tumor cells with upregulated prolactin receptor expression. Analysis of multiple human cancers confirms differential tumor and stromal cell expression of COX-2, prolactin, and prolactin receptor. Together, these findings may provide novel biomarkers to inform the selective application of COX-2 inhibitors and point to additional targets for suppressing metastasis recurrence. Tumor-stromal communication within the microenvironment contributes to initiation of metastasis and may present a therapeutic opportunity. Using serial single-cell RNA sequencing in an orthotopic mouse prostate cancer model, we find up-regulation of prolactin receptor as cancer cells that have disseminated to the lungs expand into micrometastases. Secretion of the ligand prolactin by adjacent lung stromal cells is induced by tumor cell production of the COX-2 synthetic product prostaglandin E2 (PGE2). PGE2 treatment of fibroblasts activates the orphan nuclear receptor NR4A (Nur77), with prolactin as a major transcriptional target for the NR4A-retinoid X receptor (RXR) heterodimer. Ectopic expression of prolactin receptor in mouse cancer cells enhances micrometastasis, while treatment with the COX-2 inhibitor celecoxib abrogates prolactin secretion by fibroblasts and reduces tumor initiation. Across multiple human cancers, COX-2, prolactin, and prolactin receptor show consistent differential expression in tumor and stromal compartments. Such paracrine cross-talk may thus contribute to the documented efficacy of COX-2 inhibitors in cancer suppression.
Collapse
|
24
|
Verschuur A, Heng-Maillard MA, Dory-Lautrec P, Truillet R, Jouve E, Chastagner P, Leblond P, Aerts I, Honoré S, Entz-Werle N, Sirvent N, Gentet JC, Corradini N, André N. Metronomic Four-Drug Regimen Has Anti-tumor Activity in Pediatric Low-Grade Glioma; The Results of a Phase II Clinical Trial. Front Pharmacol 2018; 9:00950. [PMID: 30319400 PMCID: PMC6171442 DOI: 10.3389/fphar.2018.00950] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/02/2018] [Indexed: 12/14/2022] Open
Abstract
Background: Metronomic chemotherapy (MC) is defined as the frequent administration of chemotherapy at doses below the maximal tolerated dose and with no prolonged drug-free break. MC has shown its efficacy in adult tumor types such as breast and ovarian cancer and has to some extent been studied in pediatrics. Objective: To assess the anti-tumor activity and toxicity of a four-drug metronomic regimen in relapsing/refractory pediatric brain tumors (BT) with progression-free survival (PFS) after two cycles as primary endpoint. Methods: Patients ≥4 to 25 years of age were included with progressing BT. Treatment consisted of an 8-week cycle of celecoxib, vinblastine, and cyclophosphamide alternating with methotrexate. Kepner and Chang two-steps model was used with 10 patients in the first stage. If stabilization was observed in ≥2 patients, 8 additional patients were recruited. Assessment was according WHO criteria with central radiology review. Results: Twenty-nine patients (27 evaluable) were included in two groups: ependymoma (group 1, N = 8), and miscellaneous BT (group 2): 3 medulloblastoma (MB), 5 high grade glioma (HGG), 11 low grade glioma (LGG), 2 other BT. After first stage, recruitment for ependymoma was closed [one patient had stable disease (SD) for 4 months]. Cohort 2 was opened for second stage since 1 HGG and 3 LGG patients had SD after two cycles. Recruitment was limited to LGG for the second stage and 2 partial responses (PR), 6 SD and 2 progressive disease (PD) were observed after two cycles. Of these patients with LGG, median age was 10 years, nine patients received vinblastine previously. Median number of cycles was 6.8 (range: 1–12). Treatment was interrupted in five patients for grade 3/4 toxicity. Conclusion: This regimen is active in patients with LGG, even if patients had previously received vinblastine. Toxicity is acceptable. Trial Registration: This study was registered under clinicaltrials.gov – NCT01285817; EUDRACT nr: 2010-021792-81.
Collapse
Affiliation(s)
- Arnauld Verschuur
- Department of Pediatric Oncology, La Timone Children's Hospital, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Marie-Amélie Heng-Maillard
- Department of Pediatric Oncology, La Timone Children's Hospital, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Philippe Dory-Lautrec
- Department of Neuroradiology, La Timone Hospital, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Romain Truillet
- CIC-CPCET, La Timone Hospital, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Elisabeth Jouve
- CIC-CPCET, La Timone Hospital, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Pascal Chastagner
- Department of Pediatric Oncology, Children's Hospital, Nancy, France
| | - Pierre Leblond
- Pediatric Oncology Unit, Oscar Lambret Centre, Lille, France
| | | | - Stéphane Honoré
- Department of Clinical Pharmacy, La Timone Hospital, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | | | - Nicolas Sirvent
- Department of Pediatric and Adolescent Hematology-Oncology, Children's Hospital Arnaud de Villeneuve, Montpellier, France
| | - Jean-Claude Gentet
- Department of Pediatric Oncology, La Timone Children's Hospital, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Nadège Corradini
- Department of Pediatric and Adolescent Hematology-Oncology, Hôpital Mère-Enfant, Nantes, France
| | - Nicolas André
- Department of Pediatric Oncology, La Timone Children's Hospital, Assistance Publique Hôpitaux de Marseille, Marseille, France.,Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Institut Paoli Calmettes, Centre de Recherche en Cancérologie de Marseille, Aix-Marseille Université, Marseille, France.,Metronomics Global Health Initiative, Marseille, France
| |
Collapse
|
25
|
Sun Y, Dai H, Chen S, Zhang Y, Wu T, Cao X, Zhao G, Xu A, Wang J, Wu L. Disruption of Chromosomal Architecture of cox2 Locus Sensitizes Lung Cancer Cells to Radiotherapy. Mol Ther 2018; 26:2456-2465. [PMID: 30131302 PMCID: PMC6171098 DOI: 10.1016/j.ymthe.2018.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/27/2018] [Accepted: 08/01/2018] [Indexed: 11/15/2022] Open
Abstract
Despite treatment of lung cancer with radiotherapy and chemotherapy, the survival rate of lung cancer patients remains poor. Previous studies demonstrated the importance of upregulation of inflammatory factors, such as cyclooxygenase 2 (cox2), in tumor tolerance. In the present study, we investigated the role of cox2 in radiosensitivity of lung cancer. Our results showed that the combination treatment of radiation with aspirin, an anti-inflammatory drug, induced a synergistic reduction of cell survival in A549 and H1299 lung cancer cells. In comparison with normal human lung fibroblasts (NHLFs), the cell viability was significantly decreased and the level of apoptosis was remarkably enhanced in A549 cells. Mechanistic studies revealed that the reduction of cox2 by aspirin in A549 and H1299 was caused by disruption of the chromosomal architecture of the cox2 locus. Moreover, the disruption of chromatin looping was mediated by the inhibition of nuclear translocation of p65 and decreased enrichment of p65 at cox2-regulatory elements. Importantly, disorganization of the chromosomal architecture of cox2 triggered A549 cells sensitive to γ-radiation by the induction of apoptosis. In conclusion, we present evidence of an effective therapeutic treatment targeting the epigenetic regulation of lung cancer and a potential strategy to overcome radiation resistance in cancer cells.
Collapse
Affiliation(s)
- Yuxiang Sun
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Hui Dai
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Shaopeng Chen
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China.
| | - Yajun Zhang
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Tao Wu
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Xianbin Cao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Guoping Zhao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - An Xu
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Jun Wang
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China
| | - Lijun Wu
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China; Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, China.
| |
Collapse
|
26
|
Hazama S, Tamada K, Yamaguchi Y, Kawakami Y, Nagano H. Current status of immunotherapy against gastrointestinal cancers and its biomarkers: Perspective for precision immunotherapy. Ann Gastroenterol Surg 2018; 2:289-303. [PMID: 30003192 PMCID: PMC6036392 DOI: 10.1002/ags3.12180] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 05/21/2018] [Indexed: 12/14/2022] Open
Abstract
Immunotherapy has shown encouraging results for some types of tumor. Although enormous efforts have been made toward the development of specific immunotherapeutic strategies against gastrointestinal cancers, such as adoptive T-cell transfer, peptide vaccines, or dendritic cell vaccines, the efficacy of immunotherapies prior to the introduction of immune checkpoint inhibitors was not substantial. This article reviews immunotherapy for gastrointestinal malignancies, including cell therapy, peptide vaccine, and immune checkpoint inhibitors, and attempts to resolve the immunosuppressive conditions surrounding the tumor microenvironment, and to construct novel combination immunotherapies beyond immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Shoichi Hazama
- Department of Translational Research and Developmental Therapeutics against CancerYamaguchi University School of MedicineUbeJapan
| | - Koji Tamada
- Department of ImmunologyYamaguchi University Graduate School of MedicineUbeJapan
| | | | - Yutaka Kawakami
- Division of Cellular SignalingInstitute for Advanced Medical ResearchKeio University School of MedicineTokyoJapan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| |
Collapse
|
27
|
Shang FM, Liu HL. Fusobacterium nucleatum and colorectal cancer: A review. World J Gastrointest Oncol 2018; 10:71-81. [PMID: 29564037 PMCID: PMC5852398 DOI: 10.4251/wjgo.v10.i3.71] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/09/2018] [Accepted: 03/06/2018] [Indexed: 02/05/2023] Open
Abstract
Fusobacterium nucleatum (F. nucleatum) is a Gram-negative obligate anaerobe bacterium in the oral cavity and plays a role in several oral diseases, including periodontitis and gingivitis. Recently, several studies have reported that the level of F. nucleatum is significantly elevated in human colorectal adenomas and carcinomas compared to that in adjacent normal tissue. Several researchers have also demonstrated that F. nucleatum is obviously associated with colorectal cancer and promotes the development of colorectal neoplasms. In this review, we have summarized the recent reports on F. nucleatum and its role in colorectal cancer and have highlighted the methods of detecting F. nucleatum in colorectal cancer, the underlying mechanisms of pathogenesis, immunity status, and colorectal cancer prevention strategies that target F. nucleatum.
Collapse
Affiliation(s)
- Fu-Mei Shang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Hong-Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
28
|
|
29
|
Dai JJ, Jiang MJ, Wang XP, Tian L. Inflammation-Related Pancreatic Carcinogenesis: Mechanisms and Clinical Potentials in Advances. Pancreas 2017; 46:973-985. [PMID: 28796135 DOI: 10.1097/mpa.0000000000000886] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Chronic inflammation has long been considered critical in pancreatic carcinogenesis, and recently studies showed that some anti-inflammatory agents such as aspirin could potentially be used to attenuate pancreatic carcinogenesis. Several inflammation-related critical transcription factors and pathways such as NF-κB (nuclear factor κ-light-chain enhancer of activated B cells) and reactive oxygen species have been confirmed to be involved in carcinogenesis. However, its underlying mechanisms are far from clear, which largely limits further development of potential anticarcinogenesis drugs. As a result, it is of great importance for us to better understand and gain a better perspective in inflammation-related pancreatic carcinogenesis. In this review, we systematically analyzed recent advances concerning inflammation-related pancreatic carcinogenesis and brought out the possible underlying mechanisms. Potential preventive and therapeutic strategies based on anti-inflammatory agents have also been further discussed.
Collapse
Affiliation(s)
- Juan-Juan Dai
- From the *Shanghai Key Laboratory of Pancreatic Diseases, †Institute of Translational Medicine, and ‡Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | |
Collapse
|
30
|
Hennequart M, Pilotte L, Cane S, Hoffmann D, Stroobant V, Plaen ED, Van den Eynde B. Constitutive IDO1 Expression in Human Tumors Is Driven by Cyclooxygenase-2 and Mediates Intrinsic Immune Resistance. Cancer Immunol Res 2017; 5:695-709. [PMID: 28765120 DOI: 10.1158/2326-6066.cir-16-0400] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 05/02/2017] [Accepted: 06/30/2017] [Indexed: 12/16/2022]
Abstract
Tumors use various mechanisms to avoid immune destruction. Cyclooxygenase-2 (COX-2) expression may be a driver of immune suppression in melanoma, but the mechanisms involved remain elusive. Here, we show that COX-2 expression drives constitutive expression of indoleamine 2,3-dioxygenase 1 (IDO1) in human tumor cells. IDO1 is an immunosuppressive enzyme that degrades tryptophan. In a series of seven human tumor lines, constitutive IDO1 expression depends on COX-2 and prostaglandin E2 (PGE2), which, upon autocrine signaling through the EP receptor, activates IDO1 via the PKC and PI3K pathways. COX-2 expression itself depends on the MAPK pathway, which therefore indirectly controls IDO1 expression. Most of these tumors carry PI3K or MAPK oncogenic mutations, which may favor constitutive IDO1 expression. Celecoxib treatment promoted immune rejection of IDO1-expressing human tumor xenografts in immunodeficient mice reconstituted with human allogeneic lymphocytes. This effect was associated with a reduced expression of IDO1 in those ovarian SKOV3 tumors and an increased infiltration of CD3+ and CD8+ cells. Our results highlight the role of COX-2 in constitutive IDO1 expression by human tumors and substantiate the use of COX-2 inhibitors to improve the efficacy of cancer immunotherapy, by reducing constitutive IDO1 expression, which contributes to the lack of T-cell infiltration in "cold" tumors, which fail to respond to immunotherapy. Cancer Immunol Res; 5(8); 695-709. ©2017 AACR.
Collapse
Affiliation(s)
- Marc Hennequart
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Luc Pilotte
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Stefania Cane
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Delia Hoffmann
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Vincent Stroobant
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Etienne De Plaen
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Benoît Van den Eynde
- Ludwig Institute for Cancer Research, Brussels, Belgium. .,de Duve Institute, Université catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium
| |
Collapse
|
31
|
Langhammer S, Scheerer J. Breaking the crosstalk of the cellular tumorigenic network: Hypothesis for addressing resistances to targeted therapies in advanced NSCLC. Oncotarget 2017; 8:43555-43570. [PMID: 28402937 PMCID: PMC5522169 DOI: 10.18632/oncotarget.16674] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/13/2017] [Indexed: 12/26/2022] Open
Abstract
In the light of current treatment developments for non-small cell lung cancer (NSCLC), the idea of a plastic cellular tumorigenic network bound by key paracrine signaling pathways mediating resistances to targeted therapies is brought forward. Based on a review of available preclinical and clinical data in NSCLC combinational approaches to address drivers of this network with marketed drugs are discussed. Five criteria for selecting drug combination regimens aiming at its disruption and thereby overcoming resistances are postulated.
Collapse
|
32
|
Korczak-Kowalska G, Stelmaszczyk-Emmel A, Bocian K, Kiernozek E, Drela N, Domagała-Kulawik J. Expanding Diversity and Common Goal of Regulatory T and B Cells. II: In Allergy, Malignancy, and Transplantation. Arch Immunol Ther Exp (Warsz) 2017; 65:523-535. [PMID: 28470464 PMCID: PMC5688211 DOI: 10.1007/s00005-017-0471-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/19/2017] [Indexed: 01/03/2023]
Abstract
Regulation of immune response was found to play an important role in the course of many diseases such as autoimmune diseases, allergy, malignancy, organ transplantation. The studies on immune regulation focus on the role of regulatory cells (Tregs, Bregs, regulatory myeloid cells) in these disorders. The number and function of Tregs may serve as a marker of disease activity. As in allergy, the depletion of Tregs is observed and the results of allergen-specific immunotherapy could be measured by an increase in the population of IL-10+ regulatory cells. On the basis of the knowledge of anti-cancer immune response regulation, new directions in therapy of tumors are introduced. As the proportion of regulatory cells is increased in the course of neoplasm, the therapeutic action is directed at their inhibition. The depletion of Tregs may be also achieved by an anti-check-point blockade, anti-CD25 agents, and inhibition of regulatory cell recruitment to the tumor site by affecting chemokine pathways. However, the possible favorable role of Tregs in cancer development is considered and the plasticity of immune regulation should be taken into account. The new promising direction of the treatment based on regulatory cells is the prevention of transplant rejection. A different way of production and implementation of classic Tregs as well as other cell types such as double-negative cells, Bregs, CD4+ Tr1 cells are tested in ongoing trials. On the basis of the results of current studies, we could show in this review the significance of therapies based on regulatory cells in different disorders.
Collapse
Affiliation(s)
- Grażyna Korczak-Kowalska
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Warsaw, Poland
| | - Anna Stelmaszczyk-Emmel
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Bocian
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Ewelina Kiernozek
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Nadzieja Drela
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
33
|
Hamada T, Cao Y, Qian ZR, Masugi Y, Nowak JA, Yang J, Song M, Mima K, Kosumi K, Liu L, Shi Y, da Silva A, Gu M, Li W, Keum N, Zhang X, Wu K, Meyerhardt JA, Giovannucci EL, Giannakis M, Rodig SJ, Freeman GJ, Nevo D, Wang M, Chan AT, Fuchs CS, Nishihara R, Ogino S. Aspirin Use and Colorectal Cancer Survival According to Tumor CD274 (Programmed Cell Death 1 Ligand 1) Expression Status. J Clin Oncol 2017; 35:1836-1844. [PMID: 28406723 DOI: 10.1200/jco.2016.70.7547] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Blockade of the programmed cell death 1 (PDCD1, PD-1) immune checkpoint pathway can improve clinical outcomes in various malignancies. Evidence suggests that aspirin (a widely used nonsteroidal anti-inflammatory drug) not only prolongs colorectal cancer survival, but can also activate T cell-mediated antitumor immunity and synergize with immunotherapy through inhibition of prostaglandin E2 production. We hypothesized that the survival benefit associated with aspirin might be stronger in colorectal carcinoma with a lower CD274 (PDCD1 ligand 1, PD-L1) expression level that resulted in lower signaling of the immune checkpoint pathway. Patients and Methods Using data from 617 patients with rectal and colon cancer in the Nurses' Health Study and the Health Professionals Follow-Up Study, we examined the association of postdiagnosis aspirin use with patient survival in strata of tumor CD274 expression status measured by immunohistochemistry. We used multivariable Cox proportional hazards regression models to control for potential confounders, including disease stage, microsatellite instability status, CpG island methylator phenotype, long interspersed nucleotide element-1 methylation, cyclooxygenase-2 (PTGS2), and CDX2 expression, and KRAS, BRAF, and PIK3CA mutations. Results The association of postdiagnosis aspirin use with colorectal cancer-specific survival differed by CD274 expression status ( Pinteraction < .001); compared with aspirin nonusers; multivariable-adjusted hazard ratios for regular aspirin users were 0.16 (95% CI, 0.06 to 0.41) in patients with low CD274 and 1.01 (95% CI, 0.61 to 1.67) in patients with high CD274. This differential association seemed consistent in patients with microsatellite-stable or PIK3CA wild-type disease and in strata of PTGS2 expression, CDX2 expression, tumor-infiltrating lymphocytes, or prediagnosis aspirin use status. Conclusion The association of aspirin use with colorectal cancer survival is stronger in patients with CD274-low tumors than CD274-high tumors. Our findings suggest a differential antitumor effect of aspirin according to immune checkpoint status.
Collapse
Affiliation(s)
- Tsuyoshi Hamada
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Yin Cao
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Zhi Rong Qian
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Yohei Masugi
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Jonathan A Nowak
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Juhong Yang
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Mingyang Song
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Kosuke Mima
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Keisuke Kosumi
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Li Liu
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Yan Shi
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Annacarolina da Silva
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Mancang Gu
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Wanwan Li
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - NaNa Keum
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Xuehong Zhang
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Kana Wu
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Jeffrey A Meyerhardt
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Edward L Giovannucci
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Marios Giannakis
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Scott J Rodig
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Gordon J Freeman
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Daniel Nevo
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Molin Wang
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Andrew T Chan
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Charles S Fuchs
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Reiko Nishihara
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Shuji Ogino
- Tsuyoshi Hamada, Zhi Rong Qian, Yohei Masugi, Juhong Yang, Kosuke Mima, Keisuke Kosumi, Li Liu, Yan Shi, Annacarolina da Silva, Mancang Gu, Wanwan Li, Jeffrey A. Meyerhardt, Marios Giannakis, Scott J. Rodig, Gordon J. Freeman, Charles S. Fuchs, Reiko Nishihara, and Shuji Ogino, Dana-Farber Cancer Institute and Harvard Medical School; Yin Cao, Mingyang Song, and Andrew T. Chan, Massachusetts General Hospital and Harvard Medical School; Yin Cao, Mingyang Song, Li Liu, NaNa Keum, Kana Wu, Edward L. Giovannucci, Daniel Nevo, Molin Wang, Reiko Nishihara, and Shuji Ogino, Harvard T.H. Chan School of Public Health; Jonathan A. Nowak, Xuehong Zhang, Edward L. Giovannucci, Marios Giannakis, Gordon J. Freeman, Molin Wang, Andrew T. Chan, Charles S. Fuchs, and Shuji Ogino, Brigham and Women's Hospital and Harvard Medical School, Boston; Marios Giannakis and Andrew T. Chan, Broad Institute of MIT and Harvard University, Cambridge, MA; Li Liu, Huazhong University of Science and Technology, Wuhan; Yan Shi, Chinese People's Liberation Army General Hospital, Beijing; and Mancang Gu, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| |
Collapse
|
34
|
Botti G, Fratangelo F, Cerrone M, Liguori G, Cantile M, Anniciello AM, Scala S, D'Alterio C, Trimarco C, Ianaro A, Cirino G, Caracò C, Colombino M, Palmieri G, Pepe S, Ascierto PA, Sabbatino F, Scognamiglio G. COX-2 expression positively correlates with PD-L1 expression in human melanoma cells. J Transl Med 2017; 15:46. [PMID: 28231855 PMCID: PMC5324267 DOI: 10.1186/s12967-017-1150-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 02/17/2017] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The resistance to PD-1/PD-L1 inhibitors for the treatment of melanoma have prompted investigators to implement novel clinical trials which combine immunotherapy with different treatment modalities. Moreover is also important to investigate the mechanisms which regulate the dynamic expression of PD-L1 on tumor cells and PD-1 on T cells in order to identify predictive biomarkers of response. COX-2 is currently investigated as a major player of tumor progression in several type of malignancies including melanoma. In the present study we investigated the potential relationship between COX-2 and PD-L1 expression in melanoma. METHODS Tumor samples obtained from primary melanoma lesions and not matched lymph node metastases were analyzed for both PD-L1 and COX-2 expression by IHC analysis. Status of BRAF and NRAS mutations was analyzed by sequencing and PCR. Co-localization of PD-L1 and COX-2 expression was analyzed by double fluorescence staining. Lastly the BRAFV600E A375 and NRASQ61R SK-MEL-2 melanoma cell lines were used to evaluate the effect of COX-2 inhibition by celecoxib on expression of PD-L1 in vitro. RESULTS BRAFV600E/V600K and NRASQ61R/Q61L were detected in 57.8 and 8.9% of the metastatic lesions, and in 65.9 and 6.8% of the primary tumors, respectively. PD-L1 and COX-2 expression were heterogeneously expressed in both primary melanoma lesions and not matched lymph node metastases. A significantly lower number of PD-L1 negative lesions was found in primary tumors as compared to not matched metastatic lesions (P = 0.002). COX-2 expression significantly correlated with PD-L1 expression in both primary (P = 0.001) and not matched metastatic (P = 0.048) lesions. Furthermore, in melanoma tumors, cancer cells expressing a higher levels of COX-2 also co-expressed a higher level of PD-L1. Lastly, inhibition of COX-2 activity by celecoxib down-regulated the expression of PD-L1 in both BRAFV600E A375 and NRASQ61R SK-MEL-2 melanoma cell lines. CONCLUSIONS COX-2 expression correlates with and modulates PD-L1 expression in melanoma cells. These findings have clinical relevance since they provide a rationale to implement novel clinical trials to test COX-2 inhibition as a potential treatment to prevent melanoma progression and immune evasion as well as to enhance the anti-tumor activity of PD-1/PD-L1 based immunotherapy for the treatment of melanoma patients with or without BRAF/NRAS mutations.
Collapse
Affiliation(s)
- Gerardo Botti
- Dipartimento di Patologia Diagnostica e di Laboratorio: SC di Anatomia Patologica e Citopatologia, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Federica Fratangelo
- Struttura Complessa di Oncologia Medica e Terapie Innovative, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Margherita Cerrone
- Dipartimento di Patologia Diagnostica e di Laboratorio: SC di Anatomia Patologica e Citopatologia, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Giuseppina Liguori
- Dipartimento di Patologia Diagnostica e di Laboratorio: SC di Anatomia Patologica e Citopatologia, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Monica Cantile
- Dipartimento di Patologia Diagnostica e di Laboratorio: SC di Anatomia Patologica e Citopatologia, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Anna Maria Anniciello
- Dipartimento di Patologia Diagnostica e di Laboratorio: SC di Anatomia Patologica e Citopatologia, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Stefania Scala
- Genomica Funzionale, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Crescenzo D'Alterio
- Genomica Funzionale, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Chiara Trimarco
- Dipartimento di Patologia Diagnostica e di Laboratorio: SC di Anatomia Patologica e Citopatologia, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Angela Ianaro
- Department of Pharmacy, University of Naples "Federico II", 80131, Naples, Italy
| | - Giuseppe Cirino
- Department of Pharmacy, University of Naples "Federico II", 80131, Naples, Italy
| | - Corrado Caracò
- Melanoma and Sarcoma Surgery Unit, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Maria Colombino
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry, National Research Council, 07100, Sassari, Italy
| | - Giuseppe Palmieri
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry, National Research Council, 07100, Sassari, Italy
| | - Stefano Pepe
- Department of Medicine and Surgery, University of Salerno, Baronissi, 84081, Salerno, Italy
| | - Paolo Antonio Ascierto
- Struttura Complessa di Oncologia Medica e Terapie Innovative, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Francesco Sabbatino
- Department of Medicine and Surgery, University of Salerno, Baronissi, 84081, Salerno, Italy.
| | - Giosuè Scognamiglio
- Dipartimento di Patologia Diagnostica e di Laboratorio: SC di Anatomia Patologica e Citopatologia, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy.
| |
Collapse
|
35
|
Suzuki N, Hazama S, Iguchi H, Uesugi K, Tanaka H, Hirakawa K, Aruga A, Hatori T, Ishizaki H, Umeda Y, Fujiwara T, Ikemoto T, Shimada M, Yoshimatsu K, Shimizu R, Hayashi H, Sakata K, Takenouchi H, Matsui H, Shindo Y, Iida M, Koki Y, Arima H, Furukawa H, Ueno T, Yoshino S, Nakamura Y, Oka M, Nagano H. Phase II clinical trial of peptide cocktail therapy for patients with advanced pancreatic cancer: VENUS-PC study. Cancer Sci 2017; 108:73-80. [PMID: 27783849 PMCID: PMC5276830 DOI: 10.1111/cas.13113] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 10/12/2016] [Accepted: 10/24/2016] [Indexed: 12/15/2022] Open
Abstract
We previously conducted a phase I clinical trial combining the HLA-A*2402-restricted KIF20A-derived peptide vaccine with gemcitabine for advanced pancreatic cancer (PC) and confirmed its safety and immunogenicity in cancer patients. In this study, we conducted a multicenter, single-armed, phase II trial using two antiangiogenic cancer vaccines targeting VEGFR1 and VEGFR2 in addition to the KIF20A peptide. We attempted to evaluate the clinical benefit of the cancer vaccination in combination with gemcitabine. Chemotherapy naïve PC patients were enrolled to evaluate primarily the 1-year survival rate, and secondarily overall survival (OS), progression free survival (PFS), response rate (RR), disease control rate (DCR) and the peptide-specific immune responses. All enrolled patients received therapy without the HLA-A information, and the HLA genotypes were used for classification of the patients. Between June 2012 and May 2013, a total of 68 patients were enrolled. No severe systemic adverse effects of Grade 3 or higher related to these three peptides were observed. The 1-year survival rates between the HLA-A*2402-matched and -unmatched groups were not significantly different. In the HLA-A*2402 matched group, patients showing peptide-specific CTL induction for KIF20A or VEGFR1 showed a better prognosis compared to those without such induction (P = 0.023, P = 0.009, respectively). In the HLA-A*2402-matched group, the patients who showed a strong injection site reaction had a better survival rate (P = 0.017) compared to those with a weak or no injection site reaction. This phase II study demonstrated that this therapeutic peptide cocktail might be effective in patients who demonstrate peptide-specific immune reactions although predictive biomarkers are needed for patient selection in its further clinical application.
Collapse
Affiliation(s)
- Nobuaki Suzuki
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Shoichi Hazama
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Haruo Iguchi
- Clinical Research CenterShikoku Cancer Center, NHOMatsuyamaJapan
| | - Kazuhiro Uesugi
- Clinical Research CenterShikoku Cancer Center, NHOMatsuyamaJapan
| | - Hiroaki Tanaka
- Department of Surgical OncologyOsaka City University Graduate School of MedicineOsakaJapan
| | - Kosei Hirakawa
- Department of Surgical OncologyOsaka City University Graduate School of MedicineOsakaJapan
| | - Atsushi Aruga
- Institute of GastroenterologyTokyo Women's Medical UniversityTokyoJapan
| | - Takashi Hatori
- Institute of GastroenterologyTokyo Women's Medical UniversityTokyoJapan
| | - Hidenobu Ishizaki
- Department of Surgical Oncology and Regulation of Organ FunctionMiyazaki University School of MedicineMiyazakiJapan
| | - Yuzo Umeda
- Department of Gastroenterological SurgeryOkayama University Graduate School of MedicineOkayamaJapan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological SurgeryOkayama University Graduate School of MedicineOkayamaJapan
| | - Tetsuya Ikemoto
- Department of Digestive and Transplant SurgeryTokushima University Graduate School of MedicineTokushimaJapan
| | - Mitsuo Shimada
- Department of Digestive and Transplant SurgeryTokushima University Graduate School of MedicineTokushimaJapan
| | - Kazuhiko Yoshimatsu
- Department of SurgeryTokyo Women's Medical University Medical Center EastTokyoJapan
| | - Ryoichi Shimizu
- Department of SurgeryOgori Dai‐ichi General HospitalYamaguchiJapan
| | - Hiroto Hayashi
- Department of SurgeryKanmon Medical Center, NHOShimonosekiJapan
| | - Koichiro Sakata
- Department of SurgeryShimonoseki Medical Center, JCHOShimonosekiJapan
| | - Hiroko Takenouchi
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Hiroto Matsui
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Yoshitaro Shindo
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Michihisa Iida
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Yasunobu Koki
- Department of PharmacyYamaguchi University HospitalUbeJapan
| | - Hideki Arima
- Department of PharmacyYamaguchi University HospitalUbeJapan
| | | | - Tomio Ueno
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Shigefumi Yoshino
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Yusuke Nakamura
- Section of Hematology/OncologyDepartment of MedicineThe University of ChicagoChicagoIllinoisUSA
| | | | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| |
Collapse
|
36
|
Kitahara M, Hazama S, Tsunedomi R, Takenouchi H, Kanekiyo S, Inoue Y, Nakajima M, Tomochika S, Tokuhisa Y, Iida M, Sakamoto K, Suzuki N, Takeda S, Ueno T, Yamamoto S, Yoshino S, Nagano H. Prediction of the efficacy of immunotherapy by measuring the integrity of cell-free DNA in plasma in colorectal cancer. Cancer Sci 2016; 107:1825-1829. [PMID: 27663862 PMCID: PMC5199104 DOI: 10.1111/cas.13085] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/18/2016] [Accepted: 09/21/2016] [Indexed: 12/14/2022] Open
Abstract
We previously reported a phase II study of a cancer vaccine using five novel peptides recognized by HLA‐A*2402‐restricted CTL in combination with oxaliplatin‐containing chemotherapy (FXV study) as first‐line therapy for patients with metastatic colorectal cancer and demonstrated the safety and promising potential of our five‐peptide cocktail. The objective of this analysis was to identify predictive biomarkers for identifying patients who are likely to receive a clinical benefit from immunochemotherapy. Circulating cell‐free DNA (cfDNA) in plasma has been reported to be a candidate molecular biomarker for the efficacy of anticancer therapy. Unlike uniformly truncated small‐sized DNA released from apoptotic normal cells, DNA released from necrotic cancer cells varies in size. The integrity of plasma cfDNA (i.e. the ratio of longer fragments [400 bp] to shorter fragments [100 bp] of cfDNA), may be clinically useful for detecting colorectal cancer progression. We assessed plasma samples collected from 93 patients prior to receiving immunochemotherapy. The cfDNA levels and integrity were analyzed by semi‐quantitative real‐time PCR. Progression‐free survival was significantly better in patients with a low plasma cfDNA integrity value than in those with a high value (P = 0.0027). Surprisingly, in the HLA‐A*2402‐matched group, patients with a low plasma cfDNA integrity value had significantly better progression‐free survival than those with a high value (P = 0.0015). This difference was not observed in the HLA‐A*2402‐unmatched group. In conclusion, the integrity of plasma cfDNA may provide important clinical information and may be a useful predictive biomarker of the outcome of immunotherapy in metastatic colorectal cancer.
Collapse
Affiliation(s)
- Masahiro Kitahara
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Shoichi Hazama
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan.,Department of Translational Research and Developmental Therapeutics against Cancer, Yamaguchi University School of Medicine, Ube, Japan
| | - Ryouichi Tsunedomi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Hiroko Takenouchi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Shinsuke Kanekiyo
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Yuka Inoue
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Masao Nakajima
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Shinobu Tomochika
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Yoshihiro Tokuhisa
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Michihisa Iida
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Kazuhiko Sakamoto
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Nobuaki Suzuki
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Shigeru Takeda
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Tomio Ueno
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Shigeru Yamamoto
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Shigefumi Yoshino
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan.,Oncology Center, Yamaguchi University Hospital, Ube, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| |
Collapse
|
37
|
Cao Y, Nishihara R, Qian ZR, Song M, Mima K, Inamura K, Nowak JA, Drew DA, Lochhead P, Nosho K, Morikawa T, Zhang X, Wu K, Wang M, Garrett WS, Giovannucci EL, Fuchs CS, Chan AT, Ogino S. Regular Aspirin Use Associates With Lower Risk of Colorectal Cancers With Low Numbers of Tumor-Infiltrating Lymphocytes. Gastroenterology 2016; 151:879-892.e4. [PMID: 27475305 PMCID: PMC5159194 DOI: 10.1053/j.gastro.2016.07.030] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 06/26/2016] [Accepted: 07/19/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Aspirin use reduces colorectal cancer risk. Aspirin, a nonsteroidal anti-inflammatory drug, inhibits prostaglandin-endoperoxide synthase 2 (PTGS2 or cyclooxygenase-2); PTGS2 promotes inflammation and suppresses T-cell-mediated adaptive immunity. We investigated whether the inverse association of aspirin use with colorectal carcinoma risk was stronger for tumors with lower degrees of lymphocytic infiltrates than for tumors with higher degrees of lymphocytic infiltrates. METHODS We collected aspirin use data biennially from participants in the Nurses' Health Study and Health Professionals Follow-up Study. Participants were asked whether they took aspirin in most weeks, the number of tablets taken per week, and years of aspirin use. We collected available tumor specimens (n = 1458) from pathology laboratories in the United States. A pathologist confirmed the diagnosis of colorectal adenocarcinoma (excluding anal squamous cell carcinoma), and evaluated histopathology features, including patterns and degrees of lymphocytic infiltrates within and around tumor areas. Person-years of follow-up evaluation were accrued from the date of return of questionnaires until dates of colorectal cancer diagnosis, death, or the end of follow-up evaluation (June 2010). Duplication-method Cox proportional hazards regression was used to assess the association of aspirin with the incidence of colorectal carcinoma subgroups according to the degree of tumor-infiltrating lymphocytes (TILs), intratumoral periglandular reaction, peritumoral reaction, or Crohn's-like reaction. RESULTS We documented 1458 rectal and colon cancers. The inverse association between regular aspirin use and colorectal cancer risk significantly differed by concentrations of TILs (Pheterogeneity = .007). Compared with nonregular use, regular aspirin use was associated with a lower risk of tumors that had low levels of TILs (relative risk, 0.72; 95% confidence interval, 0.63-0.81), and strength of the association depended on aspirin dose and duration (both Ptrend < .001). In contrast, aspirin use was not associated with a risk of tumors having intermediate or high levels of TILs. This differential association was consistent regardless of the status of tumor microsatellite instability, mutations in BRAF, or expression of PTGS2. Regular aspirin use was associated with a lower risk of tumors that contained low levels of CD3+ T cells, CD8+ T cells, or CD45RO (PTPRC)+ T cells (measured by immunohistochemistry and computer-assisted image analysis). CONCLUSIONS Based on data from the prospective cohort studies, regular use of aspirin is associated with a lower risk of colorectal carcinomas with low concentrations of TILs. These findings indicate that the immune response in the tumor microenvironment could be involved in the chemopreventive effects of aspirin.
Collapse
Affiliation(s)
- Yin Cao
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Division of Gastroenterology, Massachusetts General Hospital, Boston, MA,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Reiko Nishihara
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Zhi Rong Qian
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Mingyang Song
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Division of Gastroenterology, Massachusetts General Hospital, Boston, MA,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Kosuke Mima
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | | | - Jonathan A. Nowak
- Division of MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - David A. Drew
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Paul Lochhead
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Katsuhiko Nosho
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Teppei Morikawa
- Department of Pathology, University of Tokyo Hospital, Tokyo, Japan
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Molin Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Wendy S. Garrett
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Edward L. Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Charles S. Fuchs
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Andrew T. Chan
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Shuji Ogino
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Division of MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
38
|
Santhanam S, Alvarado DM, Ciorba MA. Therapeutic targeting of inflammation and tryptophan metabolism in colon and gastrointestinal cancer. Transl Res 2016; 167:67-79. [PMID: 26297050 PMCID: PMC4684437 DOI: 10.1016/j.trsl.2015.07.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/14/2015] [Accepted: 07/23/2015] [Indexed: 12/19/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide and the second leading cause of cancer death in the United States. Cytotoxic therapies cause significant adverse effects for most patients and do not offer cure in many advanced cases of CRC. Immunotherapy is a promising new approach to harness the body's own immune system and inflammatory response to attack and clear the cancer. Tryptophan metabolism along the kynurenine pathway (KP) is a particularly promising target for immunotherapy. Indoleamine 2,3-dioxygenase 1 (IDO1) is the most well studied of the enzymes that initiate this pathway and it is commonly overexpressed in CRC. Herein, we provide an in-depth review of how tryptophan metabolism and KP metabolites shape factors important to CRC pathogenesis including the host mucosal immune system, pivotal transcriptional pathways of neoplastic growth, and luminal microbiota. This pathway's role in other gastrointestinal (GI) malignancies such as gastric, pancreatic, esophageal, and GI stromal tumors is also discussed. Finally, we highlight how currently available small molecule inhibitors and emerging methods for therapeutic targeting of IDO1 might be applied to colon, rectal, and colitis-associated cancer.
Collapse
Affiliation(s)
- Srikanth Santhanam
- Division of Gastroenterology, Washington University School of Medicine, Saint Louis, Mo
| | - David M Alvarado
- Division of Gastroenterology, Washington University School of Medicine, Saint Louis, Mo
| | - Matthew A Ciorba
- Division of Gastroenterology, Washington University School of Medicine, Saint Louis, Mo.
| |
Collapse
|
39
|
Tuncer S, Banerjee S. Eicosanoid pathway in colorectal cancer: Recent updates. World J Gastroenterol 2015; 21:11748-11766. [PMID: 26557000 PMCID: PMC4631974 DOI: 10.3748/wjg.v21.i41.11748] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/25/2015] [Accepted: 08/31/2015] [Indexed: 02/06/2023] Open
Abstract
Enzymatic metabolism of the 20C polyunsaturated fatty acid (PUFA) arachidonic acid (AA) occurs via the cyclooxygenase (COX) and lipoxygenase (LOX) pathways, and leads to the production of various bioactive lipids termed eicosanoids. These eicosanoids have a variety of functions, including stimulation of homeostatic responses in the cardiovascular system, induction and resolution of inflammation, and modulation of immune responses against diseases associated with chronic inflammation, such as cancer. Because chronic inflammation is essential for the development of colorectal cancer (CRC), it is not surprising that many eicosanoids are implicated in CRC. Oftentimes, these autacoids work in an antagonistic and highly temporal manner in inflammation; therefore, inhibition of the pro-inflammatory COX-2 or 5-LOX enzymes may subsequently inhibit the formation of their essential products, or shunt substrates from one pathway to another, leading to undesirable side-effects. A better understanding of these different enzymes and their products is essential not only for understanding the importance of eicosanoids, but also for designing more effective drugs that solely target the inflammatory molecules found in both chronic inflammation and cancer. In this review, we have evaluated the cancer promoting and anti-cancer roles of different eicosanoids in CRC, and highlighted the most recent literature which describes how those molecules affect not only tumor tissue, but also the tumor microenvironment. Additionally, we have attempted to delineate the roles that eicosanoids with opposing functions play in neoplastic transformation in CRC through their effects on proliferation, apoptosis, motility, metastasis, and angiogenesis.
Collapse
|
40
|
Liu B, Qu L, Yan S. Cyclooxygenase-2 promotes tumor growth and suppresses tumor immunity. Cancer Cell Int 2015; 15:106. [PMID: 26549987 PMCID: PMC4635545 DOI: 10.1186/s12935-015-0260-7] [Citation(s) in RCA: 298] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 10/28/2015] [Indexed: 01/05/2023] Open
Abstract
Cyclooxygenase-2 (COX-2), an inducible form of the enzyme that catalyzes the first step in the synthesis of prostanoids, is associated with inflammatory diseases and carcinogenesis, which is suspected to promote angiogenesis and tissue invasion of tumors and resistance to apoptosis. Meanwhile, COX-2 contributes to immune evasion and resistance to cancer immunotherapy, which plays a crucial role in the innate and adaptive immune response. The activity of COX-2-PGE2-EP signal pathway can suppress Dendritic cells (DCs), natural killer (NK), T cells, type-1 immunity excluding type-2 immunity which promote tumor immune evasion. COX-2 and the prostaglandin cascade play important roles in the "inflammogenesis of cancer". In addition, COX-inhibitors can inhibit tumor immune evasion. Therefore, we can exert the COX-inhibitors to facilitate the patients to benefit from addition of COX-inhibitors to standard cytotoxic therapy.
Collapse
Affiliation(s)
- Bing Liu
- />Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, 310009 Hangzhou, Zhejiang People’s Republic of China
| | - Liyan Qu
- />Clinical Laboratory Centre, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, 310009 Hangzhou, Zhejiang People’s Republic of China
- />Clinical Laboratory Centre, Binjiang Hospital of Hangzhou, Hangzhou, Zhejiang People’s Republic of China
| | - Shigui Yan
- />Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, 310009 Hangzhou, Zhejiang People’s Republic of China
| |
Collapse
|
41
|
Wang Y, Jiang H, Liu T, Tang W, Ma Z. Cyclooxygenase-2 -1195G>A (rs689466) polymorphism and cancer susceptibility: an updated meta-analysis involving 50,672 subjects. Int J Clin Exp Med 2015; 8:12448-12462. [PMID: 26550156 PMCID: PMC4612841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 07/11/2015] [Indexed: 06/05/2023]
Abstract
The association between cyclooxygenase-2 (COX-2) -1195G>A (rs689466) polymorphism and cancer risk has been extensively explored. However, the results of previous studies remain controversial. To address this gap, we performed an updated meta-analysis of fifty-eight studies involving a total of 50,672 subjects. Searching of PubMed and Embase databases was performed for publications on the association between COX-2 -1195G>A polymorphism and the risk of cancer. Statistical correlation was identified between COX-2 -1195G>A variants and overall cancer risk in five genetic models. In a sub-group analysis based on cancer type, significant association between COX-2 -1195G>A polymorphism and increased risk of gastric cancer, pancreatic cancer, hepatocellular carcinoma and other cancers was found. In a sub-group analysis by ethnicity, increased cancer risk was observed among Asians instead of Caucasians, Africans and mixed populations. Furthermore, in a sub-group analysis based on cancer system, increased cancer risk was found in digestive system cancer and other system cancer. Non-parametric "trim-and-fill" method was harnessed as a sensitivity analysis method and the results suggested our findings reliable. In summary, the results of our meta-analysis highlight that COX-2 -1195G>A polymorphism may be a risk factor for cancer.
Collapse
Affiliation(s)
- Yafeng Wang
- Department of Cardiology, The People’s Hospital of Xishuangbanna Dai Autonomous PrefectureJinghong, Yunnan Province, China
| | - Heping Jiang
- Emergency Department, Affiliated Jintan People’s Hospital of Jiangsu UniversityJintan, China
| | - Tianyun Liu
- Department of Cardiology, The Second Clinical Medical College of Fujian Medical UniversityQuanzhou, Fujian Province, China
| | - Weifeng Tang
- Department of Cardiothoracic Surgery, Affiliated People’s Hospital of Jiangsu UniversityZhenjiang, Jiangsu Province, China
| | - Zhiqiang Ma
- Department of Cardiothoracic Surgery, The People’s Hospital of Xishuangbanna Dai Autonomous PrefectureJinghong, Yunnan Province, China
| |
Collapse
|
42
|
Beavis PA, Slaney CY, Kershaw MH, Neeson PJ, Darcy PK. Enhancing the efficacy of adoptive cellular therapy by targeting tumor-induced immunosuppression. Immunotherapy 2015; 7:499-512. [DOI: 10.2217/imt.15.16] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Strategies aimed at stimulating the immune system against cancer have signaled a new era for designing new effective therapies for patients. Recent breakthroughs in adoptive cellular therapy and in using checkpoint inhibitors for some patients have renewed much enthusiasm in this field. However, it has become apparent that tumors can use a multitude of inhibitory networks to effectively reduce antitumor immunity. This review discusses our current knowledge of these immune suppressive mechanisms used by tumors and describes potential new strategies that may counteract this problem resulting in significantly increasing therapeutic outcomes of adoptive immunotherapy in a higher proportion of patients.
Collapse
Affiliation(s)
- Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Australia
| | - Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Australia
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Australia
- Department of Pathology, University of Melbourne, Parkville, Australia
- Department of Immunology, Monash University, Clayton, Australia
| | - Paul J Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Australia
- Department of Pathology, University of Melbourne, Parkville, Australia
- Department of Immunology, Monash University, Clayton, Australia
| |
Collapse
|