1
|
Dong W, Lu J, Li Y, Zeng J, Du X, Yu A, Zhao X, Chi F, Xi Z, Cao S. SIRT1: a novel regulator in colorectal cancer. Biomed Pharmacother 2024; 178:117176. [PMID: 39059350 DOI: 10.1016/j.biopha.2024.117176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/26/2024] [Revised: 07/08/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024] Open
Abstract
The class-III histone deacetylase SIRT1 is the most extensively investigated sirtuin deacetylase. It is resistant to the broad deacetylase inhibitor trichostatin A and depends on oxidized nicotinamide adenine nucleotide (NAD+). SIRT1 plays a crucial role in the tumorigenesis of numerous types of cancers, including colorectal cancer (CRC). Accumulating evidence indicates that SIRT1 is a therapeutic target for CRC; however, the function and underlying mechanism of SIRT1 in CRC still need to be elucidated. Herein, we provide a detailed and updated review to illustrate that SIRT1 regulates many processes that go awry in CRC cells, such as apoptosis, autophagy, proliferation, migration, invasion, metastasis, oxidative stress, resistance to chemo-radio therapy, immune evasion, and metabolic reprogramming. Moreover, we closely link our review to the clinical practice of CRC treatment, summarizing the mechanisms and prospects of SIRT1 inhibitors in CRC therapy. SIRT1 inhibitors as monotherapy in CRC or in combination with chemotherapy, radiotherapy, and immune therapies are comprehensively discussed. From epigenetic regulation to its potential therapeutic effect, we hope to offer novel insights and a comprehensive understanding of SIRT1's role in CRC.
Collapse
Affiliation(s)
- Weiwei Dong
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - Jinjing Lu
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - You Li
- Nursing Department, Liaoning Jinqiu Hospital, Shenyang, Liaoning Province 110016, China
| | - Juan Zeng
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - Xiaoyun Du
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - Ao Yu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - Xuechan Zhao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China
| | - Feng Chi
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China.
| | - Zhuo Xi
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China.
| | - Shuo Cao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province 110004, China.
| |
Collapse
|
2
|
Oshi M, Sarkar J, Wu R, Tokumaru Y, Yan L, Nakagawa K, Ishibe A, Matsuyama R, Endo I, Takabe K. Intratumoral density of regulatory T cells is a predictor of host immune response and chemotherapy response in colorectal cancer. Am J Cancer Res 2022; 12:490-503. [PMID: 35261782 PMCID: PMC8899991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/07/2021] [Accepted: 01/03/2022] [Indexed: 06/14/2023] Open
Abstract
Regulatory T cells (Tregs) are a subset of CD4+ T lymphocytes known to dampen the host immune response against cancer cells. Within the tumor microenvironment, Tregs are potent facilitators of immune tolerance, and a higher proportion of Tregs compared to cytotoxic T cells predicts a worse outcome in most solid tumors. We studied the association between Treg density, and cancer biology and clinical outcome in colorectal cancer (CRC). We used xCell to estimate intratumoral Tregs in total of 898 CRC patients in the Cancer Genome Atlas (TCGA) and GCE39582 cohorts. High-Treg CRCs enriched immune response-related gene sets; inflammatory response, IFN-γ and IFN-α response, IL2/IL6 signaling, and allograft rejection, and had significantly high infiltration of CD8, CD4, M1 and M2 macrophage, and dendritic cells in both cohorts. While high-Treg CRCs enriched multiple pro-cancer signaling pathways compared to low-Treg CRCs, such as Epithelial Mesenchymal Transition, K-ras, Hypoxia, TGF-β, TNF-α, and angiogenesis, Treg infiltration was surprisingly associated with earlier CRC stage in TCGA. Notably, in two separate cohorts a higher proportion of Tregs predicted an improved response to chemotherapy. In the GSE28702 cohort, metastatic CRCs with more Tregs showed a significantly better response to mFOLFOX6 versus low-Treg CRC metastases (88.9% response vs. 16.7%, P<0.001). In the GSE72970 cohort, high-Treg CRCs were found to have a 68.8% response to FOLFOX/FOLFIRI without bevacizumab, compared to 44% response in the low-Treg CRCs. Additionally, high-Treg CRCs were associated with increased expression of immune checkpoint molecules PD-L1/PD-L2, CTLA4, TIGIT and BTLA, implying susceptibility to immunotherapy. We also found that CRCs with higher proportions of Tregs were associated with lower amounts of three microorganisms in the tumor: Lachnoclostridium, flavivirus, and Ornithobacterium. In conclusion, we show that amount of Treg in the tumor is a predictor of host immune response and chemotherapy response in CRC.
Collapse
Affiliation(s)
- Masanori Oshi
- Department of Surgical Oncology, Roswell Park Cancer InstituteBuffalo, NY, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of MedicineYokohama, Kanagawa, Japan
| | - Joy Sarkar
- Department of Surgical Oncology, Roswell Park Cancer InstituteBuffalo, NY, USA
| | - Rongrong Wu
- Department of Surgical Oncology, Roswell Park Cancer InstituteBuffalo, NY, USA
| | - Yoshihisa Tokumaru
- Department of Surgical Oncology, Roswell Park Cancer InstituteBuffalo, NY, USA
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New YorkBuffalo, NY, USA
| | - Li Yan
- Department of Surgical Oncology, Graduate School of Medicine, Gifu UniversityGifu, Japan
| | - Kazuya Nakagawa
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of MedicineYokohama, Kanagawa, Japan
| | - Atsushi Ishibe
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of MedicineYokohama, Kanagawa, Japan
| | - Ryusei Matsuyama
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of MedicineYokohama, Kanagawa, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of MedicineYokohama, Kanagawa, Japan
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Cancer InstituteBuffalo, NY, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of MedicineYokohama, Kanagawa, Japan
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New YorkBuffalo, NY, USA
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental SciencesNiigata, Japan
- Department of Breast Surgery, Fukushima Medical University School of MedicineFukushima, Japan
- Department of Breast Surgery and Oncology, Tokyo Medical UniversityTokyo, Japan
| |
Collapse
|
3
|
Schnellhardt S, Hirneth J, Büttner-Herold M, Daniel C, Haderlein M, Hartmann A, Fietkau R, Distel L. The Prognostic Value of FoxP3+ Tumour-Infiltrating Lymphocytes in Rectal Cancer Depends on Immune Phenotypes Defined by CD8+ Cytotoxic T Cell Density. Front Immunol 2022; 13:781222. [PMID: 35140715 PMCID: PMC8818710 DOI: 10.3389/fimmu.2022.781222] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/22/2021] [Accepted: 01/04/2022] [Indexed: 12/14/2022] Open
Abstract
Tumour-infiltrating FoxP3+ regulatory T cells have been identified as both positive and negative prognostic factors in colorectal cancer (CRC) and rectal cancer (RC). In this study we investigated whether immune phenotypes, defined by CD8+ cytotoxic T cell density, may influence the prognostic association of FoxP3+ T cell densities in RC. Tissue microarrays from 154 rectal cancer resections were immunohistochemically double stained for CD8 and FoxP3. CD8+ and FoxP3+ cell densities were measured in the stromal and intraepithelial compartment. Stromal FoxP3+ cell densities were not associated with 10-year overall survival (OS). In the “immune-desert” phenotype, defined by very low stromal CD8+ cell density, a high density of stromal FoxP3+ T cells displayed a tendency towards an association with decreased 10-year OS (p = 0.179). In “inflamed” tumours, defined by high intraepithelial CD8+ T cell infiltration, the opposite was the case and high stromal FoxP3+ T cell densities were a positive prognostic factor (p = 0.048). Additionally, patients with an increased FoxP3/CD8 cell density ratio demonstrated a strong trend towards decreased 10-year OS (p = 0.066). These contrasting findings suggest functional heterogeneity within the group of FoxP3+ T cells. They are consistent with experimental studies which reported suppressive and non-suppressive populations of FoxP3+ T cells in CRC. Furthermore, our study demonstrates that CD8 immunohistochemistry may act as an instrument to identify tumours infiltrated by possibly functionally differing FoxP3+ T cell subtypes.
Collapse
Affiliation(s)
- Sören Schnellhardt
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Johannes Hirneth
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Maike Büttner-Herold
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
- Department of Nephropathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Daniel
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
- Department of Nephropathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marlen Haderlein
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Arndt Hartmann
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Luitpold Distel
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
- *Correspondence: Luitpold Distel,
| |
Collapse
|
4
|
Zhu S, Zhang T, Zheng L, Liu H, Song W, Liu D, Li Z, Pan CX. Combination strategies to maximize the benefits of cancer immunotherapy. J Hematol Oncol 2021; 14:156. [PMID: 34579759 PMCID: PMC8475356 DOI: 10.1186/s13045-021-01164-5] [Citation(s) in RCA: 316] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/29/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022] Open
Abstract
Immunotherapies such as immune checkpoint blockade (ICB) and adoptive cell therapy (ACT) have revolutionized cancer treatment, especially in patients whose disease was otherwise considered incurable. However, primary and secondary resistance to single agent immunotherapy often results in treatment failure, and only a minority of patients experience long-term benefits. This review article will discuss the relationship between cancer immune response and mechanisms of resistance to immunotherapy. It will also provide a comprehensive review on the latest clinical status of combination therapies (e.g., immunotherapy with chemotherapy, radiation therapy and targeted therapy), and discuss combination therapies approved by the US Food and Drug Administration. It will provide an overview of therapies targeting cytokines and other soluble immunoregulatory factors, ACT, virotherapy, innate immune modifiers and cancer vaccines, as well as combination therapies that exploit alternative immune targets and other therapeutic modalities. Finally, this review will include the stimulating insights from the 2020 China Immuno-Oncology Workshop co-organized by the Chinese American Hematologist and Oncologist Network (CAHON), the China National Medical Product Administration (NMPA) and Tsinghua University School of Medicine.
Collapse
Affiliation(s)
- Shaoming Zhu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Tian Zhang
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, DUMC 103861, Durham, NC, 27710, USA
| | - Lei Zheng
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,The Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Hongtao Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,University of Chicago, Chicago, IL, USA
| | - Wenru Song
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Kira Pharmaceuticals, Cambridge, MA, USA
| | - Delong Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,New York Medical College, Valhalla, NY, USA
| | - Zihai Li
- Chinese American Hematologist and Oncologist Network, New York, NY, USA. .,Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, USA.
| | - Chong-Xian Pan
- Chinese American Hematologist and Oncologist Network, New York, NY, USA. .,Harvard Medical School, West Roxbury, MA, 02132, USA.
| |
Collapse
|
5
|
Neumeyer S, Hua X, Seibold P, Jansen L, Benner A, Burwinkel B, Halama N, Berndt SI, Phipps AI, Sakoda LC, Schoen RE, Slattery ML, Chan AT, Gala M, Joshi AD, Ogino S, Song M, Herpel E, Bläker H, Kloor M, Scherer D, Ulrich A, Ulrich CM, Win AK, Figueiredo JC, Hopper JL, Macrae F, Milne RL, Giles GG, Buchanan DD, Peters U, Hoffmeister M, Brenner H, Newcomb PA, Chang-Claude J. Genetic Variants in the Regulatory T cell-Related Pathway and Colorectal Cancer Prognosis. Cancer Epidemiol Biomarkers Prev 2020; 29:2719-2728. [PMID: 33008876 PMCID: PMC7976673 DOI: 10.1158/1055-9965.epi-20-0714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2020] [Revised: 07/29/2020] [Accepted: 09/28/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND High numbers of lymphocytes in tumor tissue, including T regulatory cells (Treg), have been associated with better colorectal cancer survival. Tregs, a subset of CD4+ T lymphocytes, are mediators of immunosuppression in cancer, and therefore variants in genes related to Treg differentiation and function could be associated with colorectal cancer prognosis. METHODS In a prospective German cohort of 3,593 colorectal cancer patients, we assessed the association of 771 single-nucleotide polymorphisms (SNP) in 58 Treg-related genes with overall and colorectal cancer-specific survival using Cox regression models. Effect modification by microsatellite instability (MSI) status was also investigated because tumors with MSI show greater lymphocytic infiltration and have been associated with better prognosis. Replication of significant results was attempted in 2,047 colorectal cancer patients of the International Survival Analysis in Colorectal Cancer Consortium (ISACC). RESULTS A significant association of the TGFBR3 SNP rs7524066 with more favorable colorectal cancer-specific survival [hazard ratio (HR) per minor allele: 0.83; 95% confidence interval (CI), 0.74-0.94; P value: 0.0033] was replicated in ISACC (HR: 0.82; 95% CI, 0.68-0.98; P value: 0.03). Suggestive evidence for association was found with two IL7 SNPs, rs16906568 and rs7845577. Thirteen SNPs with differential associations with overall survival according to MSI in the discovery analysis were not confirmed. CONCLUSIONS Common genetic variation in the Treg pathway implicating genes such as TGFBR3 and IL7 was shown to be associated with prognosis of colorectal cancer patients. IMPACT The implicated genes warrant further investigation.
Collapse
Affiliation(s)
- Sonja Neumeyer
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Xinwei Hua
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- School of Public Health, University of Washington, Seattle, Washington
| | - Petra Seibold
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lina Jansen
- Division of Clinical Epidemiology and Aging Research, DKFZ, Heidelberg, Germany
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara Burwinkel
- Division of Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Gynecology and Obstetrics, Molecular Biology of Breast Cancer, University of Heidelberg, Heidelberg, Germany
| | - Niels Halama
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Tissue Imaging and Analysis Center, National Center for Tumor Diseases, BIOQUANT, University of Heidelberg, Heidelberg, Germany
- Institute for Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| | - Amanda I Phipps
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Epidemiology Department, University of Washington, Seattle, Washington
| | - Lori C Sakoda
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Robert E Schoen
- Department of Medicine and Epidemiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Martha L Slattery
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Manish Gala
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Amit D Joshi
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Shuji Ogino
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Mingyang Song
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Esther Herpel
- NCT Tissue Bank, National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hendrik Bläker
- Institute of Pathology, Charité University Medicine, Berlin, Germany
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Dominique Scherer
- Institute of Medical Biometry and Informatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Germany
| | - Cornelia M Ulrich
- Huntsman Cancer Institute, Population Sciences, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Aung K Win
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, Melbourne, Australia
| | - Jane C Figueiredo
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles California
| | - John L Hopper
- Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Victoria, Australia
| | - Finlay Macrae
- Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Victoria, Australia
| | - Roger L Milne
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, Melbourne, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
| | - Graham G Giles
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, Melbourne, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, Victoria, Australia
- Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Epidemiology, University of Washington, Seattle, Washington
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, DKFZ, Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, DKFZ, Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Polly A Newcomb
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Cancer Epidemiology Group, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
6
|
Rizeq B, Malki MI. The Role of CCL21/CCR7 Chemokine Axis in Breast Cancer Progression. Cancers (Basel) 2020; 12:E1036. [PMID: 32340161 PMCID: PMC7226115 DOI: 10.3390/cancers12041036] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/28/2020] [Revised: 04/01/2020] [Accepted: 04/11/2020] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is a leading cause of cancer-related deaths worldwide, predominantly caused by metastasis. It is generally accepted that the pattern of breast cancer metastasis is largely determined by the interaction between the chemokine receptors on cancer cells and the chemokines expressed at the sites of metastatic disease. Chemokine receptors belong to the G-protein-coupled receptors (GPCRs) family that appear to be implicated in inflammatory diseases, tumor growth and metastasis. One of its members, C-C Chemokine receptor 7 (CCR7), binds chemokines CCL19 and CCL21, which are important for tissue homeostasis, immune surveillance and tumorigenesis. These receptors have been shown to induce the pathobiology of breast cancer due to their ability to induce cellular proliferation and migration upon the binding of the cognate chemokine receptors. The underlying signaling pathways and exact cellular interactions within this biological system are not fully understood and need further insights. Thus, in this review, we summarize the essential roles of CCR7 and its receptors in breast cancer progression. Furthermore, we discuss the mechanisms of regulation that may lead to novel opportunities for therapeutic intervention. Despite the enormous advances in our knowledge of the nature of the chemokines in breast cancer metastasis, research about the involvement of CCR7 in cancer progression is still limited. Therefore, further studies are essential to illustrate the distinct roles of CCR7 in cancer progression and validate its potential as a preventive bio-factor for human breast cancer metastasis by targeting chemokine receptor genes.
Collapse
Affiliation(s)
| | - Mohammed Imad Malki
- College of Medicine, QU Health, Qatar University, P. O. Box. 2713, Doha, Qatar;
| |
Collapse
|
7
|
Caraglia M, Correale P, Giannicola R, Staropoli N, Botta C, Pastina P, Nesci A, Caporlingua N, Francini E, Ridolfi L, Mini E, Roviello G, Ciliberto D, Agostino RM, Strangio A, Azzarello D, Nardone V, Falzea A, Cappabianca S, Bocchetti M, D'Arrigo G, Tripepi G, Tassone P, Addeo R, Giordano A, Pirtoli L, Francini G, Tagliaferri P. GOLFIG Chemo-Immunotherapy in Metastatic Colorectal Cancer Patients. A Critical Review on a Long-Lasting Follow-Up. Front Oncol 2019; 9:1102. [PMID: 31781481 PMCID: PMC6857002 DOI: 10.3389/fonc.2019.01102] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2019] [Accepted: 10/07/2019] [Indexed: 01/19/2023] Open
Abstract
Background: GOLFIG is a chemo-immunotherapy regimen established in preclinical models that combines gemcitabine + FOLFOX (fluoropyrimidine backbone coupled to oxaliplatin) poly-chemotherapy with low-dose s. c. recombinant interleukin-2 (rIL-2) and granulocyte-macrophage colony stimulating factor (GM-CSF). Promising antitumor effects in metastatic colorectal cancer (mCRC) patients were obtained in previous phase II and III trials. Here we report the results of 15 years of follow-up. Methods: This is a multi-institutional retrospective analysis including 179 mCRC patients receiving GOLFIG regimen between June 2002 and June 2018. Sixty-two of them received the treatment as frontline (enrolled in the GOLFIG-2 phase III trial) and 117 as second/third line (49 enrolled in the GOLFIG-1 phase II trial and 68 as compassionate use). One hundred twelve patients showed a primary left side and 67 a primary right side; K/N-ras mutational status was available in 74 cases, and an activating mutation was detected in 33. Kaplan-Meier and Cox regression analyses were carried out to relate PFS and OS with different parameters. Results: Overall, we recorded a mean PFS and OS of 15.28 (95% CI: 10.36-20.20) and 24.6 (95% CI: 19.07-30.14) months, respectively, with 14 patients surviving free of progression for 10 years. This regimen, in our updated survey of the GOLFIG-2 trial, confirmed superiority over FOLFOX in terms of PFS (hazard ratio (HR) = 0.58, p = 0.006) with a trend to a longer OS (HR = 0.69, P = 0.06) in the first line. Our analysis also confirmed significant antitumor activity in pre-treated patients, reporting a mean PFS and OS of 12.55 (95% CI: 7.19-17.9) and 20.28 (95% CI: 14.4-26.13) months, respectively. Immune-related adverse events (irAEs) were recorded in 24% of the cases and were related to a longer survival (HR = 0.36; P = 0.0001). Finally, patients' outcome was not correlated to sex, sidedness, and MT-K/N-ras. Conclusions: The GOLFIG regimen is a reliable underestimated therapeutic option in pre-treated mCRC patients and offers a strong rationale to design further trials.
Collapse
Affiliation(s)
- Michele Caraglia
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Naples, Italy.,Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, Ariano Irpino, Italy
| | - Pierpaolo Correale
- Medical Oncology Unit, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Rocco Giannicola
- Medical Oncology Unit, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Nicoletta Staropoli
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Cirino Botta
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | | | - Antonello Nesci
- Unit of Pharmacy, Section of Anti-blastic Drugs, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Nadia Caporlingua
- Unit of Pharmacy, Section of Anti-blastic Drugs, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | | | - Laura Ridolfi
- Immunotherapy, Cell Therapy and Biobank, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy
| | - Enrico Mini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, School of Medicine/Translational Oncology Unit, Careggi University Hospital, University of Florence, Florence, Italy
| | - Giandomenico Roviello
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, School of Medicine/Translational Oncology Unit, Careggi University Hospital, University of Florence, Florence, Italy
| | - Domenico Ciliberto
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Rita Maria Agostino
- Medical Oncology Unit, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Alessandra Strangio
- Medical Oncology Unit, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Domenico Azzarello
- Medical Oncology Unit, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Valerio Nardone
- Radiation Oncology Unit, Siena University Hospital, Siena, Italy
| | - Antonella Falzea
- Medical Oncology Unit, "Bianchi-Melacrino-Morelli" Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Naples, Italy.,Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, Ariano Irpino, Italy
| | - Marco Bocchetti
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Naples, Italy.,Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, Ariano Irpino, Italy
| | - Graziella D'Arrigo
- Statistical Unit, IFC-CNR (CNR), Grand Metropolitan Hospital-IFC, Reggio Calabria, Italy
| | - Giovanni Tripepi
- Statistical Unit, IFC-CNR (CNR), Grand Metropolitan Hospital-IFC, Reggio Calabria, Italy
| | - Pierfrancesco Tassone
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Raffaele Addeo
- Oncology Unit, Day Hospital, San Giovanni di Dio Hospital, ASL Napoles 2 Nord, Frattamaggiore, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States.,Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Luigi Pirtoli
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Guido Francini
- Medical Oncology Unit, Siena University Hospital, Siena, Italy
| | - Pierosandro Tagliaferri
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| |
Collapse
|
8
|
Yang Y, Nam GH, Kim GB, Kim YK, Kim IS. Intrinsic cancer vaccination. Adv Drug Deliv Rev 2019; 151-152:2-22. [PMID: 31132376 DOI: 10.1016/j.addr.2019.05.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/13/2019] [Revised: 05/22/2019] [Accepted: 05/22/2019] [Indexed: 12/15/2022]
Abstract
Immunotherapy is revolutionizing the treatment of cancer, and the current immunotherapeutics have remarkably improved the outcomes for some cancer patients. However, we still need answers for patients with immunologically cold tumors that do not benefit from the current immunotherapy treatments. Here, we suggest a novel strategy that is based on using a very old and sophisticated system for cancer immunotherapy, namely "intrinsic cancer vaccination", which seeks to awaken our own immune system to activate tumor-specific T cells. To do this, we must take advantage of the genetic instability of cancer cells and the expression of cancer cell neoantigens to trigger immunity against cancer cells. It will be necessary to not only enhance the phagocytosis of cancer cells by antigen presenting cells but also induce immunogenic cancer cell death and the subsequent immunogenic clearance, cross-priming and generation of tumor-specific T cells. This strategy will allow us to avoid using known tumor-specific antigens, ex vivo manipulation or adoptive cell therapy; rather, we will efficiently present cancer cell neoantigens to our immune system and propagate the cancer-immunity cycle. This strategy simply follows the natural cycle of cancer-immunity from its very first step, and therefore could be combined with any other treatment modality to yield enhanced efficacy.
Collapse
Affiliation(s)
- Yoosoo Yang
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Gi-Hoon Nam
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Gi Beom Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Yoon Kyoung Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea.
| |
Collapse
|
9
|
Karkeni E, Morin SO, Bou Tayeh B, Goubard A, Josselin E, Castellano R, Fauriat C, Guittard G, Olive D, Nunès JA. Vitamin D Controls Tumor Growth and CD8+ T Cell Infiltration in Breast Cancer. Front Immunol 2019; 10:1307. [PMID: 31244851 PMCID: PMC6563618 DOI: 10.3389/fimmu.2019.01307] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/15/2019] [Accepted: 05/22/2019] [Indexed: 02/06/2023] Open
Abstract
Women with low levels of vitamin D have a higher risk of developing breast cancer. Numerous studies associated the presence of a CD8+ T cell infiltration with a good prognosis. As vitamin D may play a key role in the modulation of the immune system, the objective of this work was to evaluate the impact of vitamin D on the breast cancer progression and mammary tumor microenvironment. We show that vitamin D decreases breast cancer tumor growth. Immunomonitoring of the different immune subsets in dissociated tumors revealed an increase in tumor infiltrating CD8+ T cells in the vitamin D-treated group. Interestingly, these CD8+ T cells exhibited a more active T cell (TEM/CM) phenotype. However, in high-fat diet conditions, we observed an opposite effect of vitamin D on breast cancer tumor growth, associated with a reduction of CD8+ T cell infiltration. Our data show that vitamin D is able to modulate breast cancer tumor growth and inflammation in the tumor microenvironment in vivo. Unexpectedly, this effect is reversed in high-fat diet conditions, revealing the importance of diet on tumor growth. We believe that supplementation with vitamin D can in certain conditions represent a new adjuvant in the treatment of breast cancers.
Collapse
Affiliation(s)
- Esma Karkeni
- Immunity and Cancer Team, Centre de Recherche en Cancérologie de Marseille, Equipe Labellisée Fondation pour la Recherche Médicale, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Stéphanie O Morin
- Immunity and Cancer Team, Centre de Recherche en Cancérologie de Marseille, Equipe Labellisée Fondation pour la Recherche Médicale, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Berna Bou Tayeh
- Immunity and Cancer Team, Centre de Recherche en Cancérologie de Marseille, Equipe Labellisée Fondation pour la Recherche Médicale, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Armelle Goubard
- Centre de Recherche en Cancérologie de Marseille, Plateforme d'essai préclinique TrGET, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Emmanuelle Josselin
- Centre de Recherche en Cancérologie de Marseille, Plateforme d'essai préclinique TrGET, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Rémy Castellano
- Centre de Recherche en Cancérologie de Marseille, Plateforme d'essai préclinique TrGET, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Cyril Fauriat
- Immunity and Cancer Team, Centre de Recherche en Cancérologie de Marseille, Equipe Labellisée Fondation pour la Recherche Médicale, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Geoffrey Guittard
- Immunity and Cancer Team, Centre de Recherche en Cancérologie de Marseille, Equipe Labellisée Fondation pour la Recherche Médicale, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Daniel Olive
- Immunity and Cancer Team, Centre de Recherche en Cancérologie de Marseille, Equipe Labellisée Fondation pour la Recherche Médicale, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Jacques A Nunès
- Immunity and Cancer Team, Centre de Recherche en Cancérologie de Marseille, Equipe Labellisée Fondation pour la Recherche Médicale, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| |
Collapse
|
10
|
Mollica Poeta V, Massara M, Capucetti A, Bonecchi R. Chemokines and Chemokine Receptors: New Targets for Cancer Immunotherapy. Front Immunol 2019; 10:379. [PMID: 30894861 PMCID: PMC6414456 DOI: 10.3389/fimmu.2019.00379] [Citation(s) in RCA: 392] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/21/2018] [Accepted: 02/14/2019] [Indexed: 12/21/2022] Open
Abstract
Immunotherapy is a clinically validated treatment for many cancers to boost the immune system against tumor growth and dissemination. Several strategies are used to harness immune cells: monoclonal antibodies against tumor antigens, immune checkpoint inhibitors, vaccination, adoptive cell therapies (e.g., CAR-T cells) and cytokine administration. In the last decades, it is emerging that the chemokine system represents a potential target for immunotherapy. Chemokines, a large family of cytokines with chemotactic activity, and their cognate receptors are expressed by both cancer and stromal cells. Their altered expression in malignancies dictates leukocyte recruitment and activation, angiogenesis, cancer cell proliferation, and metastasis in all the stages of the disease. Here, we review first attempts to inhibit the chemokine system in cancer as a monotherapy or in combination with canonical or immuno-mediated therapies. We also provide recent findings about the role in cancer of atypical chemokine receptors that could become future targets for immunotherapy.
Collapse
Affiliation(s)
- Valeria Mollica Poeta
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Matteo Massara
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy
| | - Arianna Capucetti
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Raffaella Bonecchi
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
11
|
Löfroos AB, Kadivar M, Resic Lindehammer S, Marsal J. Colorectal cancer-infiltrating T lymphocytes display a distinct chemokine receptor expression profile. Eur J Med Res 2017; 22:40. [PMID: 29020986 PMCID: PMC5637168 DOI: 10.1186/s40001-017-0283-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/28/2016] [Accepted: 10/02/2017] [Indexed: 02/08/2023] Open
Abstract
Background T lymphocytes exert important homeostatic functions in the healthy intestinal mucosa, whereas in case of colorectal cancer (CRC), infiltration of T lymphocytes into the tumor is crucial for an effective anti-tumor immune response. In both situations, the recruitment mechanisms of T lymphocytes into the tissues are essential for the immunological functions deciding the outcome. The recruitment of T lymphocytes is largely dependent on their expression of various chemokine receptors. The aim of this study was to identify potential chemokine receptors involved in the recruitment of T lymphocytes to normal human colonic mucosa and to CRC tissue, respectively, by examining the expression of 16 different chemokine receptors on T lymphocytes isolated from these tissues. Methods Tissues were collected from patients undergoing bowel resection for CRC. Lymphocytes were isolated through enzymatic tissue degradation of CRC tissue and nearby located unaffected mucosa, respectively. The expression of a broad panel of chemokine receptors on the freshly isolated T lymphocytes was examined by flow cytometry. Results In the normal colonic mucosa, the frequencies of cells expressing CCR2, CCR4, CXCR3, and CXCR6 differed significantly between CD4+ and CD8+ T lymphocytes, suggesting that the molecular mechanisms mediating T lymphocyte recruitment to the gut differ between CD4+ and CD8+ T lymphocytes. In CRC, the frequencies of cells expressing CCR2 and CXCR5 were significantly lower in both the CD4+ and CD8+ T lymphocyte populations compared to unaffected colonic mucosa, and the frequency of CCR9+ cytotoxic T lymphocytes was significantly decreased in CRC tissue. Conclusions With regard to the normal gut mucosa, the results suggest that the molecular mechanisms mediating T lymphocyte recruitment differ between CD4+ and CD8+ T lymphocytes, which are important for understanding gut homeostasis. Importantly, T lymphocytes from CRC compared to normal colonic tissue displayed a distinct chemokine receptor expression profile, suggesting that mechanisms for recruitment of T lymphocytes to CRC tissue are skewed compared to normal colonic mucosa. Understanding these mechanisms could help in developing new strategies in cancer immunotherapy and to optimize already available alternatives such as immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Ann-Britt Löfroos
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Immunology Section, Lund University, Lund, Sweden
| | | | - Sabina Resic Lindehammer
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Immunology Section, Lund University, Lund, Sweden
| | - Jan Marsal
- Department of Clinical Sciences, Lund University, Lund, Sweden. .,Immunology Section, Lund University, Lund, Sweden. .,Department of Gastroenterology, Skane University Hospital, 22185, Lund, Sweden.
| |
Collapse
|
12
|
Nardone V, Botta C, Caraglia M, Martino EC, Ambrosio MR, Carfagno T, Tini P, Semeraro L, Misso G, Grimaldi A, Boccellino M, Facchini G, Berretta M, Vischi G, Rocca BJ, Barone A, Tassone P, Tagliaferri P, Del Vecchio MT, Pirtoli L, Correale P. Tumor infiltrating T lymphocytes expressing FoxP3, CCR7 or PD-1 predict the outcome of prostate cancer patients subjected to salvage radiotherapy after biochemical relapse. Cancer Biol Ther 2017; 17:1213-1220. [PMID: 27791459 DOI: 10.1080/15384047.2016.1235666] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/22/2022] Open
Abstract
Tumor immunologic microenvironment is strongly involved in tumor progression and the presence of tumor infiltrating lymphocytes (TIL) with different phenotypes has been demonstrated to be of prognostic relevance in different malignancies. We investigated whether TIL infiltration of tumor tissues could also predict the outcome of prostate cancer patients. To this end, we carried out a retrospective analysis correlating the outcome of locally advanced prostate cancer patients undergone salvage radiotherapy upon relapse after radical surgery with the infiltration by different TIL populations. Twenty-two patients with resectable prostate cancer, with a mean age of 67 (+/-3.93) years, who received salvage radiotherapy with a mean of 69.66 (+/- 3.178) Gy in 8 weeks, between June 1999 and January 2009 and with a median follow up of 123 (+/- 55.82) months, were enrolled in this study. We evaluated, by immunohistochemistry, the intratumoral (t) and peripheral stroma (p) infiltration by CD45, CD3, CD4, CD8, CCR7, FoxP3 or PD-1-positive cells on tumor samples taken at the diagnosis (d) and relapse times (R). We correlated these variables with patients' biochemical progression free survival (bPFS), post-radiotherapy progression free survival (PFS), and overall survival (OS). Substantial changes in the rate of TIL subsets were found between the first and the second biopsy with progressive increase in CD4, CCR7, FoxP3, PD-1+ cells. Our analysis revealed that higher CD8p,R+ and lower PD-1R+ TIL scores correlated to a longer bPFS. Higher CD8p,R+ and CCR7t,R+ TIL scores and lower CD45p,R+ and FoxP3p,R+ TIL scores correlated to a prolonged PFS and OS. These results suggest that the immunological microenvironment of primary tumor is strictly correlated with patient outcome and provide the rationale for immunological treatment of prostate cancer.
Collapse
Affiliation(s)
- Valerio Nardone
- a Radiotherapy Unit, Department of Oncology , Siena University School of Medicine , Italy.,b Tuscany Tumor Institute (ITT) , Firenze , Italy
| | - Cirino Botta
- c Medical Oncology Unit, Department of Clinical and Experimental Medicine , "Magna Graecia" University of Catanzaro , Catanzaro , Italy
| | - Michele Caraglia
- d Department of Biochemistry , Biophysics and General Pathology, Second University of Naples , Naples , Italy
| | - Elodia Claudia Martino
- a Radiotherapy Unit, Department of Oncology , Siena University School of Medicine , Italy
| | - Maria Raffaella Ambrosio
- e Section of Pathology, Department of Medical Biotechnology , University of Siena , Siena , Italy
| | - Tommaso Carfagno
- a Radiotherapy Unit, Department of Oncology , Siena University School of Medicine , Italy.,b Tuscany Tumor Institute (ITT) , Firenze , Italy
| | - Paolo Tini
- a Radiotherapy Unit, Department of Oncology , Siena University School of Medicine , Italy.,b Tuscany Tumor Institute (ITT) , Firenze , Italy
| | - Leonardo Semeraro
- a Radiotherapy Unit, Department of Oncology , Siena University School of Medicine , Italy.,b Tuscany Tumor Institute (ITT) , Firenze , Italy
| | - Gabriella Misso
- d Department of Biochemistry , Biophysics and General Pathology, Second University of Naples , Naples , Italy
| | - Anna Grimaldi
- d Department of Biochemistry , Biophysics and General Pathology, Second University of Naples , Naples , Italy
| | - Mariarosaria Boccellino
- d Department of Biochemistry , Biophysics and General Pathology, Second University of Naples , Naples , Italy
| | - Gaetano Facchini
- f Urogynechological Department , INT Fondazione "G. Pascale " Naples , Italy
| | | | - Gianluca Vischi
- a Radiotherapy Unit, Department of Oncology , Siena University School of Medicine , Italy.,b Tuscany Tumor Institute (ITT) , Firenze , Italy
| | - Bruno Jim Rocca
- e Section of Pathology, Department of Medical Biotechnology , University of Siena , Siena , Italy.,h Pathology Unit , Ospedale di Circolo di Busto Arsizio (VA) Italy
| | - Aurora Barone
- e Section of Pathology, Department of Medical Biotechnology , University of Siena , Siena , Italy
| | - Pierfrancesco Tassone
- c Medical Oncology Unit, Department of Clinical and Experimental Medicine , "Magna Graecia" University of Catanzaro , Catanzaro , Italy
| | - Pierosandro Tagliaferri
- c Medical Oncology Unit, Department of Clinical and Experimental Medicine , "Magna Graecia" University of Catanzaro , Catanzaro , Italy
| | - Maria Teresa Del Vecchio
- e Section of Pathology, Department of Medical Biotechnology , University of Siena , Siena , Italy
| | - Luigi Pirtoli
- a Radiotherapy Unit, Department of Oncology , Siena University School of Medicine , Italy.,b Tuscany Tumor Institute (ITT) , Firenze , Italy
| | - Pierpaolo Correale
- a Radiotherapy Unit, Department of Oncology , Siena University School of Medicine , Italy.,b Tuscany Tumor Institute (ITT) , Firenze , Italy
| |
Collapse
|
13
|
Botta C, Misso G, Martino EC, Pirtoli L, Cusi MG, Tassone P, Tagliaferri P, Caraglia M, Correale P. The route to solve the interplay between inflammation, angiogenesis and anti-cancer immune response. Cell Death Dis 2016; 7:e2299. [PMID: 27441651 PMCID: PMC4973351 DOI: 10.1038/cddis.2016.211] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/08/2022]
Affiliation(s)
- C Botta
- Department of Clinical and Experimental Medicine, Magna Graecia University, Catanzaro, Italy
| | - G Misso
- Department of Biochemistry, Biophysics and General Pathology, Second Naples University, Naples, Italy
| | | | - L Pirtoli
- Department of Oncology, Siena, Italy
| | - M G Cusi
- Department of Medical Biotechnology University of Siena, Siena, Italy
| | - P Tassone
- Department of Clinical and Experimental Medicine, Magna Graecia University, Catanzaro, Italy
| | - P Tagliaferri
- Department of Clinical and Experimental Medicine, Magna Graecia University, Catanzaro, Italy
| | - M Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second Naples University, Naples, Italy
| | | |
Collapse
|
14
|
Correale P, Botta C, Martino EC, Ulivieri C, Battaglia G, Carfagno T, Rossetti MG, Fioravanti A, Guidelli GM, Cheleschi S, Gandolfo C, Carbone F, Baldari TC, Tassone P, Tagliaferri P, Pirtoli L, Cusi MG. Phase Ib study of poly-epitope peptide vaccination to thymidylate synthase (TSPP) and GOLFIG chemo-immunotherapy for treatment of metastatic colorectal cancer patients. Oncoimmunology 2015; 5:e1101205. [PMID: 27141384 DOI: 10.1080/2162402x.2015.1101205] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/05/2015] [Revised: 09/23/2015] [Accepted: 09/23/2015] [Indexed: 12/14/2022] Open
Abstract
Thymidylate synthase (TS) is a tumor-associated enzyme critical for DNA replication and main 5'-fluorouracil (5'-FU) target. TSPP/VAC1 is a multi-arm trial phase-Ib trial program aimed to investigate the toxicity and biomodulatory activity of a poly-epitope-peptide vaccine to TS (TSPP) in cancer patients (pts). Here, we present the results of the TSPP/VAC1/arm C trial aimed to evaluate TSPP in combination with chemo-immunotherapy in pretreated metastatic colo-rectal cancer (mCRC) pts. Twenty-nine pts, 14 males and 15 females, received poly-chemotherapy with gemcitabine [GEM; 1,000 mg/sqm, day-1], oxaliplatin [OX; 80 mg/sqm, day-2], levofolinate [100 mg/sqm, days 1-2], bolus/infusional 5'-FU [400 mg/800 mg/sqm, days 1-2], sargramostim [50 μg, days 3-7/q30], and interleukin-2 [sc. 0.5 MIU twice a day, days 8-14/18-30] [GOLFIG-regimen]. Seventeen pts received sc. TSPP injections at escalating dosage [3 pts, 100 µg (DL-1); 3 pts, 200 µg (DL-2) and 11pts, 300 µg (DL-3)] one week after each chemotherapy cycle (concomitant module), while 10 out 12 pts received TSPP (300 µg) after 12 GOLFIG courses [dose level (DL)-0] (sequential module). TSPP MTD was not achieved. Adverse events consisted in swelling/erythema at injection sites (17 cases), G1-2 haematological (16 cases) and gastro-enteric events (12), fever, rhinitis, conjunctivitis, and poly-arthralgia and rise in auto-antibodies [ANA, ENA, c-ANCA, p-ANCA in the DL1-3 pts]. Both treatment-modules showed immunomodulating and antitumor activity (disease-control-rate, DL1-3 and DL0 were 70.6% and 83.3%, respectively) with a better survival recorded in the second group [median OS DL1-3 vs. DL0 = 8 vs. 16 mo, p = 0.049]. The promising long-term survival produced by the sequential treatment module deserves further phase II evaluation.
Collapse
Affiliation(s)
- Pierpaolo Correale
- Unit of Radiotherapy, Department of Oncology, University of Siena , Italy
| | - Cirino Botta
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro , Italy
| | | | | | - Giuseppe Battaglia
- Unit of Radiotherapy, Department of Oncology, University of Siena , Italy
| | - Tommaso Carfagno
- Unit of Radiotherapy, Department of Oncology, University of Siena , Italy
| | | | - Antonella Fioravanti
- Unit of Rheumatology, Department of Clinical Medicine and Immunologic Sciences, University of Siena , Italy
| | - Giacomo Maria Guidelli
- Unit of Rheumatology, Department of Clinical Medicine and Immunologic Sciences, University of Siena , Italy
| | - Sara Cheleschi
- Unit of Rheumatology, Department of Clinical Medicine and Immunologic Sciences, University of Siena , Italy
| | - Claudia Gandolfo
- Microbiology and Virology Unit, Department of Medical Biotechnology, University of Siena , Italy
| | - Francesco Carbone
- Unit of Radiology, Department of Oncology, University of Siena , Italy
| | | | - Pierfrancesco Tassone
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro , Italy
| | - Pierosandro Tagliaferri
- Medical Oncology Unit, Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro , Italy
| | - Luigi Pirtoli
- Unit of Radiotherapy, Department of Oncology, University of Siena , Italy
| | - Maria Grazia Cusi
- Microbiology and Virology Unit, Department of Medical Biotechnology, University of Siena , Italy
| |
Collapse
|
15
|
Cusi MG, Botta C, Pastina P, Rossetti MG, Dreassi E, Guidelli GM, Fioravanti A, Martino EC, Gandolfo C, Pagliuchi M, Basile A, Carbone SF, Ricci V, Micheli L, Tassone P, Tagliaferri P, Pirtoli L, Correale P. Phase I trial of thymidylate synthase poly-epitope peptide (TSPP) vaccine in advanced cancer patients. Cancer Immunol Immunother 2015; 64:1159-73. [PMID: 26031574 PMCID: PMC11029252 DOI: 10.1007/s00262-015-1711-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/26/2014] [Accepted: 05/05/2015] [Indexed: 01/04/2023]
Abstract
Thymidylate synthase (TS) poly-epitope peptide (TSPP) is a 27-mer peptide vaccine containing the amino acidic sequences of three epitopes with HLA-A2.1-binding motifs of TS, an enzyme overexpressed in cancer cells, which plays a crucial role in DNA repair and replication. Based on the results of preclinical studies, we designed a phase Ib trial (TSPP/VAC1) to investigate, in a dose escalation setting, the safety and the biological activity of TSPP vaccination alone (arm A) or in combination with GM-CSF and IL-2 (arm B) in cancer patients. Twenty-one pretreated metastatic cancer patients, with a good performance status (ECOG ≤ 1) and no severe organ failure or immunological disease, were enrolled in the study (12 in arm A, nine in arm B) between April 2011 and January 2012, with a median follow-up of 28 months. TSPP resulted safe, and its maximal tolerated dose was not achieved. No grade 4 toxicity was observed. The most common adverse events were grade 2 dermatological reactions to the vaccine injection, cough, rhinitis, fever, poly-arthralgia, gastro-enteric symptoms and, to a lesser extent, moderate hypertension and hypothyroidism. We detected a significant rise in auto-antibodies and TS-epitope-specific CTL precursors. Furthermore, TSPP showed antitumor activity in this group of pretreated patients; indeed, we recorded one partial response and seven disease stabilizations (SD) in arm A, and three SD in arm B. Taken together, our findings provide the framework for the evaluation of the TSPP anti-tumor activity in further disease-oriented clinical trials.
Collapse
Affiliation(s)
- Maria Grazia Cusi
- Department of Medical Biotechnologies, Siena University, Siena, Italy
| | - Cirino Botta
- Department of Experimental and Clinical Medicine, Catanzaro “Magna Graecia” University and Medical Oncology Unit, Catanzaro, Italy
| | - Pierpaolo Pastina
- Unit of Radiotherapy, Department of Medical, Surgical Sciences and Neurosciences, Siena University, Viale Bracci 11, 53100 Siena, Italy
| | | | - Elena Dreassi
- Department of Biotechnology, Chemistry and Pharmacy, Siena University, Siena, Italy
| | | | | | - Elodia Claudia Martino
- Unit of Radiotherapy, Department of Medical, Surgical Sciences and Neurosciences, Siena University, Viale Bracci 11, 53100 Siena, Italy
| | - Claudia Gandolfo
- Department of Medical Biotechnologies, Siena University, Siena, Italy
| | | | - Assunta Basile
- Unit of Psychology, Siena University Hospital, Siena, Italy
| | | | - Veronica Ricci
- Unit of Radiology, Siena University Hospital, Siena, Italy
| | - Lucia Micheli
- Department of Medical, Surgical Sciences, Neurosciences Siena University, Siena, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Catanzaro “Magna Graecia” University and Medical Oncology Unit, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Catanzaro “Magna Graecia” University and Medical Oncology Unit, Catanzaro, Italy
| | - Luigi Pirtoli
- Unit of Radiotherapy, Department of Medical, Surgical Sciences and Neurosciences, Siena University, Viale Bracci 11, 53100 Siena, Italy
| | - Pierpaolo Correale
- Unit of Radiotherapy, Department of Medical, Surgical Sciences and Neurosciences, Siena University, Viale Bracci 11, 53100 Siena, Italy
| |
Collapse
|
16
|
Zumwalt TJ, Goel A. Immunotherapy of Metastatic Colorectal Cancer: Prevailing Challenges and New Perspectives. CURRENT COLORECTAL CANCER REPORTS 2015; 11:125-140. [PMID: 26441489 PMCID: PMC4591512 DOI: 10.1007/s11888-015-0269-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022]
Abstract
Patients with recurring or metastatic colorectal cancer (mCRC) have strikingly low long-term survival, while conventional treatments such as chemotherapeutic intervention and radiation therapy marginally improve longevity. Although, many factors involving immunosurveillance and immunosuppression were recently validated as important for patient prognosis and care, a multitude of experimental immunotherapies designed to combat unresectable mCRC have, in few cases, successfully mobilized antitumor immune cells against malignancies, nor conclusively or consistently granted protection, complete remission, and/or stable disease from immunotherapy - of which benefit less than 10% of those receiving therapy. After decades of progress, however, new insights into the mechanisms of immunosuppression, tolerance, and mutation profiling established novel therapies that circumvent these immunological barriers. This review underlines the most exciting methods to date that manipulate immune cells to curb mCRC, including adoptive cell therapy, dendritic cell vaccines, and checkpoint inhibitor antibodies - of which hint at effective and enduring protection against disease progression and undetected micrometastases.
Collapse
Affiliation(s)
- Timothy J Zumwalt
- Center for Gastrointestinal Research; Center for Epigenetics, Cancer Prevention and Cancer Genomics, Baylor Research Institute and Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas, USA
| | - Ajay Goel
- Center for Gastrointestinal Research; Center for Epigenetics, Cancer Prevention and Cancer Genomics, Baylor Research Institute and Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas, USA
| |
Collapse
|
17
|
Botta C, Gullà A, Correale P, Tagliaferri P, Tassone P. Myeloid-derived suppressor cells in multiple myeloma: pre-clinical research and translational opportunities. Front Oncol 2014; 4:348. [PMID: 25538892 PMCID: PMC4258997 DOI: 10.3389/fonc.2014.00348] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/13/2014] [Accepted: 11/23/2014] [Indexed: 12/31/2022] Open
Abstract
Immunosuppressive cells have been reported to play an important role in tumor-progression mainly because of their capability to promote immune-escape, angiogenesis, and metastasis. Among them, myeloid-derived suppressor cells (MDSCs) have been recently identified as immature myeloid cells, induced by tumor-associated inflammation, able to impair both innate and adaptive immunity. While murine MDSCs are usually identified by the expression of CD11b and Gr1, human MDSCs represent a more heterogeneous population characterized by the expression of CD33 and CD11b, low or no HLA-DR, and variable CD14 and CD15. In particular, the last two may alternatively identify monocyte-like or granulocyte-like MDSC subsets with different immunosuppressive properties. Recently, a substantial increase of MDSCs has been found in peripheral blood and bone marrow (BM) of multiple myeloma (MM) patients with a role in disease progression and/or drug resistance. Pre-clinical models recapitulating the complexity of the MM-related BM microenvironment (BMM) are major tools for the study of the interactions between MM cells and cells of the BMM (including MDSCs) and for the development of new agents targeting MM-associated immune-suppressive cells. This review will focus on current strategies for human MDSCs generation and investigation of their immunosuppressive function in vitro and in vivo, taking into account the relevant relationship occurring within the MM–BMM. We will then provide trends in MDSC-associated research and suggest potential application for the treatment of MM.
Collapse
Affiliation(s)
- Cirino Botta
- Department of Experimental and Clinical Medicine, "Magna Graecia" University and Medical Oncology Unit, T. Campanella Cancer Center, "Salvatore Venuta" University Campus , Catanzaro , Italy
| | - Annamaria Gullà
- Department of Experimental and Clinical Medicine, "Magna Graecia" University and Medical Oncology Unit, T. Campanella Cancer Center, "Salvatore Venuta" University Campus , Catanzaro , Italy
| | | | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, "Magna Graecia" University and Medical Oncology Unit, T. Campanella Cancer Center, "Salvatore Venuta" University Campus , Catanzaro , Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, "Magna Graecia" University and Medical Oncology Unit, T. Campanella Cancer Center, "Salvatore Venuta" University Campus , Catanzaro , Italy ; Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University , Philadelphia, PA , USA
| |
Collapse
|
18
|
Paschos KA, Majeed AW, Bird NC. Natural history of hepatic metastases from colorectal cancer - pathobiological pathways with clinical significance. World J Gastroenterol 2014; 20:3719-3737. [PMID: 24744570 PMCID: PMC3983432 DOI: 10.3748/wjg.v20.i14.3719] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/28/2013] [Revised: 11/12/2013] [Accepted: 01/06/2014] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer hepatic metastases represent the final stage of a multi-step biological process. This process starts with a series of mutations in colonic epithelial cells, continues with their detachment from the large intestine, dissemination through the blood and/or lymphatic circulation, attachment to the hepatic sinusoids and interactions with the sinusoidal cells, such as sinusoidal endothelial cells, Kupffer cells, stellate cells and pit cells. The metastatic sequence terminates with colorectal cancer cell invasion, adaptation and colonisation of the hepatic parenchyma. All these events, termed the colorectal cancer invasion-metastasis cascade, include multiple molecular pathways, intercellular interactions and expression of a plethora of chemokines and growth factors, and adhesion molecules, such as the selectins, the integrins or the cadherins, as well as enzymes including matrix metalloproteinases. This review aims to present recent advances that provide insights into these cell-biological events and emphasizes those that may be amenable to therapeutic targeting.
Collapse
|
19
|
Botta C, Barbieri V, Ciliberto D, Rossi A, Rocco D, Addeo R, Staropoli N, Pastina P, Marvaso G, Martellucci I, Guglielmo A, Pirtoli L, Sperlongano P, Gridelli C, Caraglia M, Tassone P, Tagliaferri P, Correale P. Systemic inflammatory status at baseline predicts bevacizumab benefit in advanced non-small cell lung cancer patients. Cancer Biol Ther 2014; 14:469-75. [PMID: 23760488 DOI: 10.4161/cbt.24425] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022] Open
Abstract
Bevacizumab is a humanized anti-VEGF monoclonal antibody able to produce clinical benefit in advanced non-squamous non-small-cell lung cancer (NSCLC) patients when combined to chemotherapy. At present, while there is a rising attention to bevacizumab-related adverse events and costs, no clinical or biological markers have been identified and validated for baseline patient selection. Preclinical findings suggest an important role for myeloid-derived inflammatory cells, such as neutrophils and monocytes, in the development of VEGF-independent angiogenesis. We conducted a retrospective analysis to investigate the role of peripheral blood cells count and of an inflammatory index, the neutrophil-to-lymphocyte ratio (NLR), as predictors of clinical outcome in NSCLC patients treated with bevacizumab plus chemotherapy. One hundred and twelve NSCLC patients treated with chemotherapy ± bevacizumab were retrospectively evaluated for the predictive value of clinical or laboratory parameters correlated with inflammatory status. Univariate analysis revealed that a high number of circulating neutrophils and monocytes as well as a high NLR were associated with shorter progression-free survival (PFS) and overall survival (OS) in bevacizumab-treated patients only. We have thus developed a model based on the absence or the presence of at least one of the above-mentioned inflammatory parameters. We found that the absence of all variables strongly correlated with longer PFS and OS (9.0 vs. 7.0 mo, HR: 0.39, p = 0.002; and 20.0 vs. 12.0 mo, HR: 0.29, p < 0.001 respectively) only in NSCLC patients treated with bevacizumab plus chemotherapy. Our results suggest that a baseline systemic inflammatory status is marker of resistance to bevacizumab treatment in NSCLC patients.
Collapse
Affiliation(s)
- Cirino Botta
- Medical Oncology Unit, Campus Salvatore Venuta, Department of Experimental and Clinical Medicine, Magna Graecia University and Tommaso Campanella Cancer Center, Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Ondondo B, Jones E, Godkin A, Gallimore A. Home sweet home: the tumor microenvironment as a haven for regulatory T cells. Front Immunol 2013; 4:197. [PMID: 23874342 PMCID: PMC3712544 DOI: 10.3389/fimmu.2013.00197] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/24/2013] [Accepted: 07/03/2013] [Indexed: 01/28/2023] Open
Abstract
CD4+Foxp3+ regulatory T cells (Tregs) have a fundamental role in maintaining immune balance by preventing autoreactivity and immune-mediated pathology. However this role of Tregs extends to suppression of anti-tumor immune responses and remains a major obstacle in the development of anti-cancer vaccines and immunotherapies. This feature of Treg activity is exacerbated by the discovery that Treg frequencies are not only elevated in the blood of cancer patients, but are also significantly enriched within tumors in comparison to other sites. These observations have sparked off the quest to understand the processes through which Tregs become elevated in cancer-bearing hosts and to identify the specific mechanisms leading to their accumulation within the tumor microenvironment. This manuscript reviews the evidence for specific mechanisms of intra-tumoral Treg enrichment and will discuss how this information may be utilized for the purpose of manipulating the balance of tumor-infiltrating T cells in favor of anti-tumor effector cells.
Collapse
Affiliation(s)
- Beatrice Ondondo
- Nuffield Department of Medicine, The Jenner Institute (ORCRB), University of Oxford , Oxford , UK
| | | | | | | |
Collapse
|
21
|
Rossi M, Botta C, Correale P, Tassone P, Tagliaferri P. Immunologic microenvironment and personalized treatment in multiple myeloma. Expert Opin Biol Ther 2013; 13 Suppl 1:S83-93. [PMID: 23692463 DOI: 10.1517/14712598.2013.799130] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Multiple myeloma (MM) is characterized by generalized immune suppression and increased susceptibility to infections and secondary malignancies. Malignant plasma cells (PCs) modulate the bone marrow microenvironment to favor their own survival and proliferation. These events lead to a severe deregulation of immune effectors. Extensive studies have been conducted to unveil the mechanisms through which MM cells negatively modulate immunity and to develop therapeutical approaches for restoring an efficient anti-MM immune response. AREAS COVERED This review article covers both the immunosuppressive effects exerted by MM and the immunomodulatory potential of novel anti-MM agents. A brief overview on the most promising immunotherapeutic approaches in the field is also provided. EXPERT OPINION MM leads to a progressive impairment of the immune system. Different approaches have been evaluated or are currently under investigation to boost a specific anti-MM immune response. The discovery that anti-MM agents like bortezomib also retain immunomodulatory properties provides evidence to support the development of combined treatment modalities. In the next future, immunotherapy will be likely included in selective treatments in early stages or in the post-transplantation setting with non toxic modalities that control or clear the neoplastic clone.
Collapse
Affiliation(s)
- Marco Rossi
- Magna Graecia University and T. Campanella Cancer Center, Department of Experimental and Clinical Medicine, Medical Oncology Unit, Viale Europa, 88100 Catanzaro, Italy
| | | | | | | | | |
Collapse
|