1
|
Tang B, Liu Z, Xiong H, Zhang J, Dai J. IFN-λ: Unleashing Its Potential in Disease Therapies From Acute Inflammation Regulation to Cancer Immunotherapy. Immunology 2025. [PMID: 40421666 DOI: 10.1111/imm.13954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 04/21/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Type III interferons (IFN-λ), which include IFN-λ1 (or interleukin [IL]-29), IFN-λ2 (IL-28A), IFN-λ3 (IL-28B) and IFN-λ4, exert their effects through a unique receptor complex composed of interferon lambda receptor 1 (IFNLR1) and IL-10 receptor subunit beta (IL-10R2). Studies have highlighted their critical role in modulating immune response, particularly in the context of autoimmune diseases, viral infections and cancer. Unlike type I IFNs, which are broadly expressed, IFN-λ displays a more tissue-specific expression pattern, predominantly acting on epithelial cells and certain immune cell types, such as neutrophils and B cells. This specificity allows IFN-λ to play a pivotal role in mucosal immunity, particularly at barrier sites, such as the respiratory and gastrointestinal tracts. Emerging evidence suggests that IFN-λ has a dual role in both enhancing antiviral immunity and regulating inflammation, thus offering a promising therapeutic strategy for diseases like systemic lupus erythematosus, rheumatoid arthritis, asthma and various cancers. However, the precise mechanisms by which IFN-λ influence immune modulation and disease progression remain an area of active investigation. This review aims to provide an overview of the structure, function and signalling pathways of IFN-λ, exploring their role in immune-related diseases and discussing potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Benfeng Tang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, China
| | - Zhihong Liu
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
- School of Basic Medicine, Shandong First Medical University, Jinan, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Jun Dai
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| |
Collapse
|
2
|
Kohli AS, Sanyal S, Kaushal RS, Dwivedi M. An Insight into Immunological Therapeutic Approach against Cancer: Potential Anti-cancer Vaccines. Curr Genomics 2024; 26:175-190. [PMID: 40433416 PMCID: PMC12105320 DOI: 10.2174/0113892029319505240821063238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 05/29/2025] Open
Abstract
The development of a cancer vaccine comes with its complications and designing and developing a vaccine against foreign invaders such as bacterial and viral particles is not as complex and multi-faceted as the preparation of immunotherapy for host-infected cells which resemble our own body cells. The entire research and development framework of designing a vaccine for cancerous cells lies entirely on the remarkable aspect of notifying specific interactions and acclimatising the immune system. This review aims to compile the several fronts research-based methodology applies to in terms of developing a therapeutic, preventive or personalised vaccine for cancer. The approach lays focus on the identification and selection of targets for vaccine development which have come to light as immune biomarkers. Furthemore, significant aspects of personalised and precision vaccines and the fine line that runs between these approaches have also been discussed.
Collapse
Affiliation(s)
- Arjun Singh Kohli
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Gomtinagar Ext., Lucknow, 226028, India
| | - Somali Sanyal
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Gomtinagar Ext., Lucknow, 226028, India
| | | | - Manish Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Gomtinagar Ext., Lucknow, 226028, India
- Research Cell, Amity University Uttar Pradesh, Lucknow Campus, India
| |
Collapse
|
3
|
Al Shboul S, Boyle S, Singh A, Saleh T, Alrjoub M, Abu Al Karsaneh O, Mryyian A, Dawoud R, Gul S, Abu Baker S, Ball K, Hupp T, Brennan PM. FISH analysis reveals CDKN2A and IFNA14 co-deletion is heterogeneous and is a prominent feature of glioblastoma. Brain Tumor Pathol 2024; 41:4-17. [PMID: 38097874 DOI: 10.1007/s10014-023-00473-6] [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] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/06/2023] [Indexed: 01/09/2024]
Abstract
Deletion of CDKN2A occurs in 50% of glioblastomas (GBM), and IFNA locus deletion in 25%. These genes reside closely on chromosome 9. We investigated whether CDKN2A and IFNA were co-deleted within the same heterogeneous tumour and their prognostic implications. We assessed CDKN2A and IFNA14 deletions in 45 glioma samples using an in-house three-colour FISH probe. We examined the correlation between p16INK4a protein expression (via IHC) and CDKN2A deletion along with the impact of these genomic events on patient survival. FISH analyses demonstrated that grades II and III had either wildtype (wt) or amplified CDKN2A/IFNA14, whilst 44% of GBMs harboured homozygous deletions of both genes. Cores with CDKN2A homozygous deletion (n = 11) were negative for p16INK4a. Twenty p16INK4a positive samples lacked CDKN2A deletion with some of cells showing negative p16INK4a. There was heterogeneity in IFNA14/CDKN2A ploidy within each GBM. Survival analyses of primary GBMs suggested a positive association between increased p16INK4a and longer survival; this persisted when considering CDKN2A/IFNA14 status. Furthermore, wt (intact) CDKN2A/IFNA14 were found to be associated with longer survival in recurrent GBMs. Our data suggest that co-deletion of CDKN2A/IFNA14 in GBM negatively correlates with survival and CDKN2A-wt status correlated with longer survival, and with second surgery, itself a marker for improved patient outcomes.
Collapse
Affiliation(s)
- Sofian Al Shboul
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan.
| | - Shelagh Boyle
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Ashita Singh
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XR, Scotland, UK
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
| | - Moath Alrjoub
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ola Abu Al Karsaneh
- Department of Microbiology, Pathology, and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
| | - Amel Mryyian
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
| | - Rand Dawoud
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
| | - Sinem Gul
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XR, Scotland, UK
| | - Shaden Abu Baker
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Kathryn Ball
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XR, Scotland, UK
| | - Ted Hupp
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XR, Scotland, UK.
| | - Paul M Brennan
- Translational Neurosurgery, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
- Translational Neurosurgery, Department of Clinical Neurosciences, Royal Infirmary of Edinburgh, Edinburgh, EH16 4SB, Scotland, UK.
| |
Collapse
|
4
|
Heimberger AB, Tripathi S, Platanias LC. Targeting Cytokines and Their Pathways for the Treatment of Cancer. Cancers (Basel) 2023; 15:5224. [PMID: 37958397 PMCID: PMC10649760 DOI: 10.3390/cancers15215224] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
This Special Issue focuses on the evolving role of immune modulatory cytokines, from their initial use as monotherapeutic recombinant proteins to their more contemporaneous use as modifiers for adoptive cellular immunotherapy [...].
Collapse
Affiliation(s)
- Amy B. Heimberger
- Department of Neurological Surgery, Feinberg School of Medicine, Chicago, IL 60611, USA;
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA;
| | - Shashwat Tripathi
- Department of Neurological Surgery, Feinberg School of Medicine, Chicago, IL 60611, USA;
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA;
| | - Leonidas C. Platanias
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA;
- Division of Hematology Oncology, Department of Medicine, Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Medicine, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
5
|
Tarhini AA, Joshi I, Garner F. Sargramostim and immune checkpoint inhibitors: combinatorial therapeutic studies in metastatic melanoma. Immunotherapy 2021; 13:1011-1029. [PMID: 34157863 DOI: 10.2217/imt-2021-0119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The use of immune checkpoint inhibitors in patients with metastatic melanoma generates clinical benefit, including improved survival. Yet disease resistance and immune-related adverse events persist as unmet needs. Sargramostim, a yeast-derived recombinant human GM-CSF, has shown clinical activity against diverse solid tumors, including metastatic melanoma. Here we review the use of sargramostim for treatment of advanced melanoma. Potential sargramostim applications in melanoma draw on the unique ability of GM-CSF to link innate and adaptive immune responses. We review preclinical and translational data describing the mechanism of action of sargramostim and synergy with immune checkpoint inhibitors to enhance efficacy and reduce treatment-related toxicity.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- Cutaneous Oncology & Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Ila Joshi
- Pre-Clinical & Translational Research & Development, Partner Therapeutics, 19 Muzzey Street, Lexington, MA 02421, USA
| | - Fiona Garner
- Immuno-Oncology Clinical Development & Translational Medicine, Partner Therapeutics, 19 Muzzey Street, Lexington, MA 02421, USA
| |
Collapse
|
6
|
Araujo B de Lima V, Hansen M, Spanggaard I, Rohrberg K, Reker Hadrup S, Lassen U, Svane IM. Immune Cell Profiling of Peripheral Blood as Signature for Response During Checkpoint Inhibition Across Cancer Types. Front Oncol 2021; 11:558248. [PMID: 33842304 PMCID: PMC8027233 DOI: 10.3389/fonc.2021.558248] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Despite encouraging results with immune checkpoint inhibition (ICI), a large fraction of cancer patients still does not achieve clinical benefit. Finding predictive markers in the complexity of the tumor microenvironment is a challenging task and often requires invasive procedures. In our study, we looked for putative variables related to treatment benefit among immune cells in peripheral blood across different tumor types treated with ICIs. For that, we included 33 patients with different solid tumors referred to our clinical unit for ICI. Peripheral blood mononuclear cells were isolated at baseline, 6 and 20 weeks after treatment start. Characterization of immune cells was carried out by multi-color flow cytometry. Response to treatment was assessed radiologically by RECIST 1.1. Clinical outcome correlated with a shift towards an effector-like T cell phenotype, PD-1 expression by CD8+T cells, low levels of myeloid-derived suppressor cells and classical monocytes. Dendritic cells seemed also to play a role in terms of survival. From these findings, we hypothesized that patients responding to ICI had already at baseline an immune profile, here called ‘favorable immune periphery’, providing a higher chance of benefitting from ICI. We elaborated an index comprising cell types mentioned above. This signature correlated positively with the likelihood of benefiting from the treatment and ultimately with longer survival. Our study illustrates that patients responding to ICI seem to have a pre-existing immune profile in peripheral blood that favors good outcome. Exploring this signature can help to identify patients likely to achieve benefit from ICI.
Collapse
Affiliation(s)
| | - Morten Hansen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Iben Spanggaard
- Department of Oncology, Phase 1 Unit, Rigshospitalet, Copenhagen, Denmark
| | | | - Sine Reker Hadrup
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Ulrik Lassen
- Department of Oncology, Phase 1 Unit, Rigshospitalet, Copenhagen, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| |
Collapse
|
7
|
Zhou H, Jiang M, Yuan H, Ni W, Tai G. Dual roles of myeloid-derived suppressor cells induced by Toll-like receptor signaling in cancer. Oncol Lett 2020; 21:149. [PMID: 33552267 PMCID: PMC7798029 DOI: 10.3892/ol.2020.12410] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 12/03/2020] [Indexed: 12/16/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are one of the major components of the tumor microenvironment (TME), and are the main mediators of tumor-induced immunosuppression. Recent studies have reported that the survival, differentiation and immunosuppressive activity of MDSCs are affected by the Toll-like receptor (TLR) signaling pathway. However, the regulatory effect of TLR signaling on MDSCs remains controversial. TLR-induced MDSC can acquire different immunosuppressive activities to influence the immune response that can be either beneficial or detrimental to cancer immunotherapy. The present review summarizes the effects of TLR signals on the number, phenotype and inhibitory activity of MDSCs, and their role in cancer immunotherapy, which cannot be ignored if effective cancer immunotherapies are to be developed for the immunosuppression of the TME.
Collapse
Affiliation(s)
- Hongyue Zhou
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Mengyu Jiang
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hongyan Yuan
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Weihua Ni
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guixiang Tai
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
8
|
Wang F, Wang S, Zhou Q. The Resistance Mechanisms of Lung Cancer Immunotherapy. Front Oncol 2020; 10:568059. [PMID: 33194652 PMCID: PMC7606919 DOI: 10.3389/fonc.2020.568059] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022] Open
Abstract
Immunotherapy has revolutionized lung cancer treatment in the past decade. By reactivating the host’s immune system, immunotherapy significantly prolongs survival in some advanced lung cancer patients. However, resistance to immunotherapy is frequent, which manifests as a lack of initial response or clinical benefit to therapy (primary resistance) or tumor progression after the initial period of response (acquired resistance). Overcoming immunotherapy resistance is challenging owing to the complex and dynamic interplay among malignant cells and the defense system. This review aims to discuss the mechanisms that drive immunotherapy resistance and the innovative strategies implemented to overcome it in lung cancer.
Collapse
Affiliation(s)
- Fen Wang
- Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, Guangdong Lung Cancer Institute, South China University of Technology, Guangzhou, China.,Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Peking University Shenzhen Hospital, Shenzhen, China
| | - Shubin Wang
- Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qing Zhou
- Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, Guangdong Lung Cancer Institute, South China University of Technology, Guangzhou, China
| |
Collapse
|
9
|
Khadge S, Sharp JG, Thiele GM, McGuire TR, Talmadge JE. Fatty Acid Mediators in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1259:125-153. [PMID: 32578175 DOI: 10.1007/978-3-030-43093-1_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Patients with cancer frequently overexpress inflammatory cytokines with an associated neutrophilia both of which may be downregulated by diets with high omega-3 polyunsaturated fatty acids (ω-3 PUFA). The anti-inflammatory activity of dietary ω-3 PUFA has been suggested to have anticancer properties and to improve survival of cancer patients. Currently, the majority of dietary research efforts do not differentiate between obesity and dietary fatty acid consumption as mediators of inflammatory cell expansion and tumor microenvironmental infiltration, initiation, and progression. In this chapter, we discuss the relationships between dietary lipids, inflammation, neoplasia and strategies to regulate these relationships. We posit that dietary composition, notably the ratio of ω-3 vs. ω-6 PUFA, regulates tumor initiation and progression and the frequency and sites of metastasis that, together, impact overall survival (OS). We focus on three broad topics: first, the role of dietary lipids in chronic inflammation and tumor initiation, progression, and regression; second, lipid mediators linking inflammation and cancer; and third, dietary lipid regulation of murine and human tumor initiation, progression, and metastasis.
Collapse
Affiliation(s)
- Saraswoti Khadge
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.,Vanderbilt University, Nashville, TN, USA
| | - John Graham Sharp
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey M Thiele
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.,Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Timothy R McGuire
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE, USA
| | - James E Talmadge
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
10
|
Balancing cancer immunotherapy and immune-related adverse events: The emerging role of regulatory T cells. J Autoimmun 2019; 104:102310. [PMID: 31421963 DOI: 10.1016/j.jaut.2019.102310] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023]
Abstract
Advances in our understanding οf tumor immunity have prompted a paradigm shift in oncology, with the emergence of immunotherapy, where therapeutic agents are used to target immune cells rather than cancer cells. A real breakthrough in the field of immunotherapy came with the use of immune checkpoint inhibitors (ICI), namely antagonistic antibodies that block key immune regulatory molecules (checkpoint molecules), such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed cell death protein (PD-1) and its ligand PD-L1, that under physiologic conditions suppress T cell effector function. However, despite the enormous success, a significant proportion of patients do not respond, while responses are frequently accompanied by life-threatening autoimmune related adverse events (irAEs). A major impediment in the effectiveness of ICI immunotherapy is the tumoral resistance, which is dependent on the immunosuppressive nature of tumor microenvironment (TME). Regulatory T cells (Tregs) are among the most abundant suppressive cells in the TME and their presence has been correlated with tumor progression, invasiveness as well as metastasis. Tregs are characterized by the expression of the transcription factor Foxp3 and various mechanisms ranging from cell-to-cell contact to secretion of inhibitory molecules have been implicated in their function. Notably, Tregs amply express most of the checkpoint molecules such as CTLA4, PD1 and LAG3 and therefore represent a direct target of ICI immunotherapy. Taking into consideration the critical role of Tregs in maintenance of immune homeostasis and avoidance of autoimmunity it is plausible that targeting of Tregs by ICI immunotherapy results in the development of irAEs. Since the use of ICI becomes common, and new immune checkpoint molecules are currently under clinical trials for the treatment of cancer, the occurrence of irAEs is expected to dramatically rise. Herein we review the current literature focusing on the role of Tregs in cancer evolution, ICI response and development of irAEs. Unraveling the complex mechanisms that hinder the tumor immune surveillance and in particular how ICI immunotherapy imprint on Treg activities to promote cancer regression while avoid development of irAEs, will empower the design of novel immunotherapeutic modalities in cancer with increased efficacy and diminished adverse events.
Collapse
|
11
|
Wang YH, Tsai DY, Ko YA, Yang TT, Lin IY, Hung KH, Lin KI. Blimp-1 Contributes to the Development and Function of Regulatory B Cells. Front Immunol 2019; 10:1909. [PMID: 31474988 PMCID: PMC6702260 DOI: 10.3389/fimmu.2019.01909] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/29/2019] [Indexed: 01/09/2023] Open
Abstract
Regulatory B cells (Bregs) are a B cell subset that plays a suppressive role in immune responses. The CD19+CD1dhiCD5+ Bregs that can execute regulatory functions via secreting IL-10 are defined as B10 cells. Bregs suppress autoimmune and inflammatory diseases, whereas they exacerbate infectious diseases caused by bacteria, viruses, or parasites. Notably, the molecular mechanisms regulating the development and functions of Bregs are still largely unknown. Furthermore, the biological impact of Bregs in fungal infection has not yet been demonstrated. Here, we compared the gene expression profiles of IL-10-producing and -non-producing mouse splenic B cells stimulated with lipopolysaccharide (LPS) or anti-CD40 antibody. Blimp-1, a transcription factor known to be critical for plasma cell differentiation, was found to be enriched in the IL-10-producing B cells. The frequency of Blimp-1+ B10 cells was increased in LPS-treated mice and in isolated B10 cells that were stimulated with LPS. Surprisingly, B cell-specific Blimp-1 knockout (Cko) mice, generated by CD19 promoter driven Cre recombinase-dependent deletion of Prdm1 (gene encoding Blimp-1), showed higher frequencies of B10 cells both in the steady state and following injection with LPS, as compared with control littermates. However, B10 cells lacking Blimp-1 failed to efficiently suppress the proliferation of naïve CD4+ T cells primed with anti-CD3 and anti-CD28 antibodies. B10 cells can be stimulated for further differentiation into plasmablasts, and a subset of plasmablasts express IL-10. We found that B10 cells from Cko mice failed to generate both IL-10-non-producing and IL-10-producing plasmablasts. Mechanistically, we found that Blimp-1 can directly suppress Il-10, whereas, in the presence of activated STAT3, Blimp-1 works together with activated STAT3 to upregulate Il-10. Moreover, we also found that B10 cells improve the clearance of Candida albicans infection but worsen the infection mortality. Notably, a lack of Blimp-1 in B10 cells did not change these effects of adoptively transferred B10 cells on fungal infections. Together, our data show that Blimp-1 regulates the generation, differentiation, and IL-10 production of Bregs.
Collapse
Affiliation(s)
- Ying-Hsiu Wang
- National Defense Medical Center, Graduate Institute of Life Sciences, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Dong-Yan Tsai
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-An Ko
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Tsan-Tzu Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - I-Ying Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Kuo-Hsuan Hung
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Kuo-I Lin
- National Defense Medical Center, Graduate Institute of Life Sciences, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
12
|
Butterfield LH, Vujanovic L, Santos PM, Maurer DM, Gambotto A, Lohr J, Li C, Waldman J, Chandran U, Lin Y, Lin H, Tawbi HA, Tarhini AA, Kirkwood JM. Multiple antigen-engineered DC vaccines with or without IFNα to promote antitumor immunity in melanoma. J Immunother Cancer 2019; 7:113. [PMID: 31014399 PMCID: PMC6480917 DOI: 10.1186/s40425-019-0552-x] [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: 11/24/2018] [Accepted: 02/27/2019] [Indexed: 02/08/2023] Open
Abstract
Background Cancer vaccines are designed to promote systemic antitumor immunity and tumor eradication. Cancer vaccination may be more efficacious in combination with additional interventions that may build on or amplify their effects. Methods Based on our previous clinical and in vitro studies, we designed an antigen-engineered DC vaccine trial to promote a polyclonal CD8+ and CD4+ T cell response against three shared melanoma antigens. The 35 vaccine recipients were then randomized to receive one month of high-dose IFNα or observation. Results The resulting clinical outcomes were 2 partial responses, 8 stable disease and 14 progressive disease among patients with measurable disease using RECIST 1.1, and, of 11 surgically treated patients with no evidence of disease (NED), 4 remain NED at a median follow-up of 3 years. The majority of vaccinated patients showed an increase in vaccine antigen-specific CD8+ and CD4+ T cell responses. The addition of IFNα did not appear to improve immune or clinical responses in this trial. Examination of the DC vaccine profiles showed that IL-12p70 secretion did not correlate with immune or clinical responses. In depth immune biomarker studies support the importance of circulating Treg and MDSC for development of antigen-specific T cell responses, and of circulating CD8+ and CD4+ T cell subsets in clinical responses. Conclusions DC vaccines are a safe and reliable platform for promoting antitumor immunity. This combination with one month of high dose IFNα did not improve outcomes. Immune biomarker analysis in the blood identified several predictive and prognostic biomarkers for further analysis, including MDSC. Trial registration NCT01622933. Electronic supplementary material The online version of this article (10.1186/s40425-019-0552-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lisa H Butterfield
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA. .,Department of Surgery, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA. .,Department of Immunology, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA. .,UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.
| | - Lazar Vujanovic
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Patricia M Santos
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Deena M Maurer
- Department of Immunology, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Joel Lohr
- Department of Immunology, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| | - Chunlei Li
- UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,Present address: Tsinghua University School of Medicine, Beijing, China
| | - Jacob Waldman
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Uma Chandran
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yan Lin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Huang Lin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hussein A Tawbi
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,Present address: Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ahmad A Tarhini
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,Present address: Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - John M Kirkwood
- Department of Medicine, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA.,UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Avenue, Suite 1.27, Pittsburgh, PA, 15213, USA
| |
Collapse
|
13
|
Prelaj A, Tay R, Ferrara R, Chaput N, Besse B, Califano R. Predictive biomarkers of response for immune checkpoint inhibitors in non-small-cell lung cancer. Eur J Cancer 2018; 106:144-159. [PMID: 30528799 DOI: 10.1016/j.ejca.2018.11.002] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 10/10/2018] [Accepted: 11/01/2018] [Indexed: 12/17/2022]
Abstract
Immune checkpoint blockade has been a pivotal development in the management of advanced non-small-cell lung cancer (NSCLC). Although durable antitumour activity and improved survival have been observed in a subset of patients, there is a need for additional predictive biomarkers to improve patient selection and avoid toxicity in potential non-responders. This review will address the use and limitations of tumour programmed death-ligand 1 expression as a predictive biomarker and review emerging biomarker strategies specifically related to NSCLC including genetic alterations (tumour mutation burden, loss and gain activated mutations), tumour-related factors (tumour microenvironment) and factors related to the host immune system. Novel approaches in biomarker detection such as peripheral blood monitoring will also be reviewed.
Collapse
Affiliation(s)
- Arsela Prelaj
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Department of Radiological, Pathological and Oncological Science, Sapienza University of Rome, Italy.
| | - Rebecca Tay
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK.
| | - Roberto Ferrara
- Laboratory of Immunomonitoring in Oncology, UMS 3655 CNRS/US 23 INSERM, Gustave Roussy, Villejuif, France.
| | - Nathalie Chaput
- Laboratory of Immunomonitoring in Oncology, UMS 3655 CNRS/US 23 INSERM, Gustave Roussy, Villejuif, France; Faculty of Pharmacy, University Paris-Saclay, Chatenay-Malabry, France.
| | - Benjamin Besse
- Cancer Medicine Department, Gustave Roussy, Villejuif, France; Paris-Sud University, Orsay, France.
| | - Raffaele Califano
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Department of Medical Oncology, Manchester University NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
14
|
Immune regulation and anti-cancer activity by lipid inflammatory mediators. Int Immunopharmacol 2018; 65:580-592. [PMID: 30447537 DOI: 10.1016/j.intimp.2018.10.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 10/02/2018] [Accepted: 10/17/2018] [Indexed: 12/19/2022]
Abstract
Rodent and clinical studies have documented that myeloid cell infiltration of tumors is associated with poor outcomes, neutrophilia and lymphocytopenia. This contrasts with increased lymphocyte infiltration of tumors, which is correlated with improved outcomes. Lifestyle parameters, such as obesity and diets with high levels of saturated fat and/or omega (ω)-6 polyunsaturated fatty acids (PUFAs), can influence these inflammatory parameters, including an increase in extramedullary myelopoiesis (EMM). While tumor secretion of growth factors (GFs) and chemokines regulate tumor-immune-cell crosstalk, lifestyle choices also contribute to inflammation, abnormal pathology and leukocyte infiltration of tumors. A relationship between obesity and high-fat diets (notably saturated fats in Western diets) and inflammation, tumor incidence, metastasis and poor outcomes is generally accepted. However, the mechanisms of dietary promotion of an inflammatory microenvironment and targeted drugs to inhibit the clinical sequelae are poorly understood. Thus, modifications of obesity and dietary fat may provide preventative or therapeutic approaches to control tumor-associated inflammation and disease progression. Currently, the majority of basic and clinical research does not differentiate between obesity and fatty acid consumption as mediators of inflammatory and neoplastic processes. In this review, we discuss the relationships between dietary PUFAs, inflammation and neoplasia and experimental strategies to improve our understanding of these relationships. We conclude that dietary composition, notably the ratio of ω-3 vs ω-6 PUFA regulates tumor growth and the frequency and sites of metastasis that together, impact overall survival (OS) in mice.
Collapse
|
15
|
Retseck J, Nasr A, Lin Y, Lin H, Mendiratta P, Butterfield LH, Tarhini AA. Long term impact of CTLA4 blockade immunotherapy on regulatory and effector immune responses in patients with melanoma. J Transl Med 2018; 16:184. [PMID: 29973204 PMCID: PMC6033230 DOI: 10.1186/s12967-018-1563-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/28/2018] [Indexed: 12/21/2022] Open
Abstract
Background We previously reported early on-treatment significant modulation in circulating regulatory T cell (Treg), myeloid derived suppressor cells (MDSC) and antigen-specific type I CD4+ and CD8+ T cells that correlated with clinical outcome in regionally advanced melanoma patients treated with neoadjuvant ipilimumab. Here, we investigated the long term immunologic impact of CTLA4 blockade. Methods Patients were treated with ipilimumab given at 10 mg/kg IV every 3 weeks for 2 doses bracketing surgery. Blood specimens were collected at baseline and during treatment for up to 9 months. We tested immune responses at 3, 6, and 9 months utilizing multicolor flow cytometry. We compared frequencies of circulating Treg and MDSC on-study to baseline levels, as well as frequencies of CD4+ and CD8+ T cells specific to shared tumor-associated antigens (Gp-100, MART-1, NY-ESO-1). Results Levels of Treg significantly increased when measured at 6 weeks following ipilimumab but returned to baseline by 3 months, with no significant difference in Treg levels between relapsed and relapse-free groups at 3, 6 or 9 months. However, lower baseline levels of circulating Treg (CD4+CD25hi+CD39+) were significantly associated with better relapse free survival (RFS) (p = 0.04). Levels of circulating monocytic HLA-DR+/loCD14+ MDSC were lower at baseline in the relapse-free group and further decreased at 6 weeks, though the differences did not reach statistical significance including measurements at 3, 6 or 9 months. We detected evidence of type I (interferon-γ producing), activated (CD69+) CD4+ and CD8+ antigen-specific T cell immunity against cancer-testis (NY-ESO-1) as well as melanocytic lineage (MART-1, gp100) antigens in the absence of therapeutic vaccination. These responses were significantly boosted at 6 weeks and persisted at 3, 6 and 9 months following the initiation of ipilimumab. Conclusions Lower Treg levels at baseline are significantly associated with RFS and increased Treg frequency after CTLA4 blockade was only transient. Lower MDSC was also associated with RFS and MDSC levels were further decreased after ipilimumab. Tumor specific effector immune responses are boosted with CTLA4 blockade and tend to be durable. Trial registration ClinicalTrials.gov Identifier: NCT00972933
Collapse
Affiliation(s)
- Janet Retseck
- University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - Alexis Nasr
- University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - Yan Lin
- University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - Huang Lin
- University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - Prateek Mendiratta
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Ave CA6-157, Cleveland, OH, 44195, USA
| | - Lisa H Butterfield
- Departments of Medicine, Surgery and Immunology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, USA
| | - Ahmad A Tarhini
- University of Pittsburgh Cancer Institute, Pittsburgh, USA. .,Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Ave CA6-157, Cleveland, OH, 44195, USA. .,Case Comprehensive Cancer Center, Cleveland, USA.
| |
Collapse
|
16
|
Conry RM, Westbrook B, McKee S, Norwood TG. Talimogene laherparepvec: First in class oncolytic virotherapy. Hum Vaccin Immunother 2018; 14:839-846. [PMID: 29420123 PMCID: PMC5893211 DOI: 10.1080/21645515.2017.1412896] [Citation(s) in RCA: 237] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Oncolytic viruses represent a novel drug class in which native or modified viruses mediate tumor regression through selective replication within and lysis of tumor cells as well as induction of systemic antitumor immunity capable of eradicating tumor at distant, uninjected sites. Talimogene laherparepvec (TVEC) is a type I herpes simplex virus genetically modified to preferentially replicate in tumor cells, enhance antigen loading of MHC class I molecules and express granulocyte-macrophage colony-stimulating factor to increase tumor-antigen presentation by dendritic cells. It is presently the only oncolytic virus approved by the FDA with an indication for advanced melanoma based upon improved durable response rate in a randomized, phase III trial. Clinical trials are underway in melanoma investigating TVEC as neoadjuvant monotherapy and in combination with checkpoint inhibitors for unresectable disease as well as in an array of other malignancies. It is appropriate to review TVEC's biology mechanism of action, clinical indication and future directions as a prototype of the burgeoning class of oncolytic viruses.
Collapse
Affiliation(s)
- Robert M Conry
- a Medicine/Division of Hematology and Oncology, University of Alabama at Birmingham , Birmingham , AL , USA
| | - Brian Westbrook
- b Medicine, University of Alabama at Birmingham , Birmingham , AL , USA
| | - Svetlana McKee
- a Medicine/Division of Hematology and Oncology, University of Alabama at Birmingham , Birmingham , AL , USA
| | | |
Collapse
|
17
|
Pitt JM, Vétizou M, Daillère R, Roberti MP, Yamazaki T, Routy B, Lepage P, Boneca IG, Chamaillard M, Kroemer G, Zitvogel L. Resistance Mechanisms to Immune-Checkpoint Blockade in Cancer: Tumor-Intrinsic and -Extrinsic Factors. Immunity 2017; 44:1255-69. [PMID: 27332730 DOI: 10.1016/j.immuni.2016.06.001] [Citation(s) in RCA: 792] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Indexed: 12/11/2022]
Abstract
Inhibition of immune regulatory checkpoints, such as CTLA-4 and the PD-1-PD-L1 axis, is at the forefront of immunotherapy for cancers of various histological types. However, such immunotherapies fail to control neoplasia in a significant proportion of patients. Here, we review how a range of cancer-cell-autonomous cues, tumor-microenvironmental factors, and host-related influences might account for the heterogeneous responses and failures often encountered during therapies using immune-checkpoint blockade. Furthermore, we describe the emerging evidence of how the strong interrelationship between the immune system and the host microbiota can determine responses to cancer therapies, and we introduce a concept by which prior or concomitant modulation of the gut microbiome could optimize therapeutic outcomes upon immune-checkpoint blockade.
Collapse
Affiliation(s)
- Jonathan M Pitt
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France; Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, 94276 Le Kremlin Bicêtre, France
| | - Marie Vétizou
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France; Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, 94276 Le Kremlin Bicêtre, France
| | - Romain Daillère
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France; Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, 94276 Le Kremlin Bicêtre, France
| | - María Paula Roberti
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France
| | - Takahiro Yamazaki
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France
| | - Bertrand Routy
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France; Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, 94276 Le Kremlin Bicêtre, France
| | - Patricia Lepage
- Micalis UMR 1319, Institut National de la Recherche Agronomique, 78360 Jouy-en-Josas, France
| | - Ivo Gomperts Boneca
- Unit of Biology and Genetics of the Bacterial Cell Wall, Institut Pasteur, 75015 Paris, France; Equipe Avenir, INSERM, 75015 Paris, France
| | - Mathias Chamaillard
- Université de Lille, Centre National de la Recherche Scientifique, INSERM, Centre Hospitalier Universitaire Lille, Institut Pasteur de Lille, U1019, UMR 8204, Centre d'Infection et d'Immunité de Lille, 59000 Lille, France
| | - Guido Kroemer
- INSERM U848, 94800 Villejuif, France; Metabolomics Platform, Gustave Roussy Cancer Campus, 94800 Villejuif, France; Equipe 11 Labellisée Ligue contre le Cancer, INSERM U1138, Centre de Recherche des Cordeliers, 75006 Paris, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique - Hôpitaux de Paris, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France; Université Pierre et Marie Curie, 75005 Paris, France; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Laurence Zitvogel
- Institut de Cancérologie, Gustave Roussy Cancer Campus, 94800 Villejuif, France; INSERM U1015, 94800 Villejuif, France; Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, 94276 Le Kremlin Bicêtre, France; Center of Clinical Investigations CICBT1428, Gustave Roussy Cancer Campus, 94805 Villejuif Cedex 05, France.
| |
Collapse
|
18
|
Butterfield LH, Zhao F, Lee S, Tarhini AA, Margolin KA, White RL, Atkins MB, Cohen GI, Whiteside TL, Kirkwood JM, Lawson DH. Immune Correlates of GM-CSF and Melanoma Peptide Vaccination in a Randomized Trial for the Adjuvant Therapy of Resected High-Risk Melanoma (E4697). Clin Cancer Res 2017; 23:5034-5043. [PMID: 28536308 DOI: 10.1158/1078-0432.ccr-16-3016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/31/2017] [Accepted: 05/19/2017] [Indexed: 12/23/2022]
Abstract
Purpose: E4697 was a multicenter intergroup randomized placebo-controlled phase III trial of adjuvant GM-CSF and/or a multiepitope melanoma peptide vaccine for patients with completely resected, high-risk stage III/IV melanoma.Experimental Design: A total of 815 patients were enrolled from December 1999 to October 2006 into this six-arm study. GM-CSF was chosen to promote the numbers and functions of dendritic cells (DC). The melanoma antigen peptide vaccine (Tyrosinase368-376 (370D), gp100209-217 (210M), MART-127-35) in montanide was designed to promote melanoma-specific CD8+ T-cell responses.Results: Although the overall RFS and OS were not significantly improved with the vaccine or GM-CSF when compared with placebo, immunomodulatory effects were observed in peripheral blood and served as important correlates to this therapeutic study. Peripheral blood was examined to evaluate the impact of GM-CSF and/or the peptide vaccine on peripheral blood immunity and to investigate potential predictive or prognostic biomarkers. A total of 11.3% of unvaccinated patients and 27.1% of vaccinated patients developed peptide-specific CD8+ T-cell responses. HLA-A2+ patients who had any peptide-specific CD8+ T-cell response at day +43 tended to have poorer OS in univariate analysis. Patients receiving GM-CSF had significant reduction in percentages of circulating myeloid dendritic cells (mDC) and plasmacytoid DC (pDC) at day +43. In a subset of patients who received GM-CSF, circulating myeloid-derived suppressor cells (MDSC), and anti-GM-CSF-neutralizing antibodies (Nabs) were also modulated. The majority of patients developed anti-GM-CSF Nabs, which correlated with improved RFS and OS.Conclusions: The assessment of cellular and humoral responses identified counterintuitive immune system changes correlating with clinical outcome. Clin Cancer Res; 23(17); 5034-43. ©2017 AACR.
Collapse
Affiliation(s)
| | - Fengmin Zhao
- Dana Farber Cancer Institute - ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | - Sandra Lee
- Dana Farber Cancer Institute - ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | | | | | - Richard L White
- Levine Cancer Institute, Carolinas Healthcare System, Charlotte, North Carolina
| | | | - Gary I Cohen
- Greater Baltimore Medical Center, Baltimore, Maryland
| | | | | | - David H Lawson
- Winship Cancer institute of Emory University, Atlanta, Georgia
| |
Collapse
|
19
|
The prognostic value of cytotoxic T-lymphocyte antigen 4 in cancers: a systematic review and meta-analysis. Sci Rep 2017; 7:42913. [PMID: 28211499 PMCID: PMC5314410 DOI: 10.1038/srep42913] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 01/17/2017] [Indexed: 12/21/2022] Open
Abstract
The outcomes of studies analyzing the prognostic role of CTLA-4 in cancers are controversial. Therefore, the aim of our meta-analysis was to clarify the correlation between CTLA-4 expression and OS in different cancer cases. Relevant literature was searched using PubMed, EMBASE, Web of Science, and the Cochrane Library. The clinicopathological features, hazard ratio (HR) and 95% confidence intervals (CI) were collected from these studies and were analyzed using Stata version 12.0 software. The pooled HR values showed no significant correlation between CTLA-4 expression levels and OS in relation to tumors (HR: 1.24, 95% CI: 0.98–1.56, I2 = 71.7%, P = 0.000). Further subgroup analyses were conducted and categorized by experimental methods, CTLA-4 sources and cancer types. The survey showed a significant correlation (HR: 1.47, 95% CI: 1.14–1.89) between high expression of CTLA-4 and OS in the SNP subgroup, and subgroups analyzing by PCR (HR: 1.50, 95% CI: 1.20–1.86) and flow cytometry (HR: 2.76, 95% CI: 1.49–5.14). In addition, our analysis observed significant differences between patients and controls in inCTLA-4+CD4+ lymphocytes, surCTLA-4+CD4+ lymphocytes, inCTLA-4+CD8+ lymphocytes, and surCTLA-4+CD8+ lymphocytes. Knowledge of the effects of CTLA-4 could potentially be used to effectively guide appropriate prognosis and therapeutic strategies in cancer patients.
Collapse
|
20
|
Regulatory T Cells in the Tumor Microenvironment and Cancer Progression: Role and Therapeutic Targeting. Vaccines (Basel) 2016; 4:vaccines4030028. [PMID: 27509527 PMCID: PMC5041022 DOI: 10.3390/vaccines4030028] [Citation(s) in RCA: 352] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/27/2016] [Accepted: 08/01/2016] [Indexed: 02/07/2023] Open
Abstract
Recent years have seen significant efforts in understanding and modulating the immune response in cancer. In this context, immunosuppressive cells, including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), have come under intense investigation for their proposed roles in suppressing tumor-specific immune responses and establishing an immunosuppressive tumor microenvironment, thus enabling tumor immune evasion. Additionally, recent evidence indicates that Tregs comprise diverse and heterogeneous subsets; phenotypically and functionally distinct subsets of tumor-infiltrating Tregs could contribute differently to cancer prognosis and clinical outcomes. Understanding Treg biology in the setting of cancer, and specifically the tumor microenvironment, is important for designing effective cancer therapies. In this review, we critically examine the role of Tregs in the tumor microenvironment and in cancer progression focusing on human studies. We also discuss the impact of current therapeutic modalities on Treg biology and the therapeutic opportunities for targeting Tregs to enhance anti-tumor immune responses and clinical benefits.
Collapse
|
21
|
Hoeller C, Michielin O, Ascierto PA, Szabo Z, Blank CU. Systematic review of the use of granulocyte-macrophage colony-stimulating factor in patients with advanced melanoma. Cancer Immunol Immunother 2016; 65:1015-34. [PMID: 27372293 PMCID: PMC4995227 DOI: 10.1007/s00262-016-1860-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 06/15/2016] [Indexed: 12/24/2022]
Abstract
Several immunomodulatory checkpoint inhibitors have been approved for the treatment of patients with advanced melanoma, including ipilimumab, nivolumab and pembrolizumab. Talimogene laherparepvec is the first oncolytic virus to gain regulatory approval in the USA; it is also approved in Europe. Talimogene laherparepvec expresses granulocyte–macrophage colony-stimulating factor (GM-CSF), and with other GM-CSF-expressing oncolytic viruses in development, understanding the clinical relevance of this cytokine in treating advanced melanoma is important. Results of trials of GM-CSF in melanoma have been mixed, and while GM-CSF has the potential to promote anti-tumor responses, some preclinical data suggest that GM-CSF may sometimes promote tumor growth. GM-CSF has not been approved as a melanoma treatment. We undertook a systematic literature review of studies of GM-CSF in patients with advanced melanoma (stage IIIB–IV). Of the 503 articles identified, 26 studies met the eligibility criteria. Most studies investigated the use of GM-CSF in combination with another treatment, such as peptide vaccines or chemotherapy, or as an adjuvant to surgery. Some clinical benefit was reported in patients who received GM-CSF as an adjuvant to surgery, or in combination with other treatments. In general, outcomes for patients receiving peptide vaccines were not improved with the addition of GM-CSF. GM-CSF may be a valuable therapeutic adjuvant; however, further studies are needed, particularly head-to-head comparisons, to confirm the optimal dosing regimen and clinical effectiveness in patients with advanced melanoma.
Collapse
Affiliation(s)
- Christoph Hoeller
- Department of Dermatology, Medical University Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| | - Olivier Michielin
- Department of Oncology, Lausanne University Hospital, Champ de l'Air, Rue du Bugnon 21, 1011, Lausanne, Switzerland.,Ludwig Centre and Swiss Institute of Bioinformatics, Génopode Building, 1015, Lausanne, Switzerland
| | - Paolo A Ascierto
- Istituto Nazionale Tumori, Fondazione 'G. Pascale', Via Mariano Semmola, 52, 80131, Naples, Italy
| | - Zsolt Szabo
- Clinical Development, Amgen Europe GmbH, Dammstrasse 23, 6300, Zug, Switzerland
| | - Christian U Blank
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, Netherlands
| |
Collapse
|
22
|
Lasfar A, Gogas H, Zloza A, Kaufman HL, Kirkwood JM. IFN-λ cancer immunotherapy: new kid on the block. Immunotherapy 2016; 8:877-88. [PMID: 27381684 PMCID: PMC5619162 DOI: 10.2217/imt-2015-0021] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 03/21/2016] [Indexed: 02/08/2023] Open
Abstract
Interferon-lambda (IFN-λ) is a new IFN type, related to IFN-α, that is commonly used in the clinic. However, significant side effects accompanying IFN-α treatment limit enthusiasm for IFN-α. In this review, we discuss the current landscape of IFN-α use in oncology and describe the biologic characteristics of IFN-λ. IFN-λ offers unique advantages, including a more tumor cell selective targeting, lower off-target binding and an ability to generate both innate and adaptive immune responses. IFN-λ has also demonstrated therapeutic benefit in murine cancer models. IFN-λ may be used in clinic as a single agent or in combination with other immunotherapy agents, such as immune checkpoint inhibitors. Further clinical trials will be needed to fully elucidate the potential of this novel agent in oncology.
Collapse
Affiliation(s)
- Ahmed Lasfar
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - Helen Gogas
- First Department of Medicine, Medical School, University of Athens, Athens, Greece
| | - Andrew Zloza
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, NJ, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - Howard L Kaufman
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, NJ, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - John M Kirkwood
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh Medical Center, PA, USA
| |
Collapse
|
23
|
Retseck J, VanderWeele R, Lin HM, Lin Y, Butterfield LH, Tarhini AA. Phenotypic and functional testing of circulating regulatory T cells in advanced melanoma patients treated with neoadjuvant ipilimumab. J Immunother Cancer 2016; 4:38. [PMID: 27330811 PMCID: PMC4915044 DOI: 10.1186/s40425-016-0141-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/06/2016] [Indexed: 01/08/2023] Open
Abstract
Background We have previously investigated neoadjuvant ipilimumab (ipi) for patients with locally/regionally advanced melanoma. That initial assessment of peripheral blood mononuclear cells (PBMC) showed a significant increase in shared tumor associated antigen specific CD4+ and CD8+ T cell activation. We also observed a transient increase in circulating T regulatory cells (Treg) with a parallel increase in total CD4+ T cells, as well as a significant decrease in circulating myeloid derived suppressor cells (MDSC). The increase in circulating Treg frequency, as assessed at 6 weeks after initiation of ipilimumab, was significantly associated with improved progression free survival (PFS, p = 0.034; HR = 0.57) and returned to baseline levels by 12 weeks. To shed light on the unexpected positive correlation between increased Treg and PFS, we here investigated the suppressive activity of circulating Treg at baseline and 6 weeks. Methods Patients were treated with ipi (10 mg/kg intravenously every 3 weeks for 2 doses) bracketing definitive surgery. Treg (CD4+CD25+CD127dim/-) were isolated from pre-ipi (baseline) and post-ipi (6 weeks) PBMC samples. Treg were co-cultured with autologous responder CD4+ T cells that were stimulated with OKT3/IL-2/CD28 and CFSE-labeled T cells. 1:1, 1:2, and 1:5 ratios were tested. Flow cytometery was used to evaluate the degree of Treg proliferation suppression. Results Thirty-five patients were enrolled in the study; 18 patients had adequate PBMC samples with sufficient Treg isolated for Treg functional analysis. At 6 weeks following ipi, a decrease in percent of maximal inhibition of Th by Treg compared to baseline was seen for some patients. Scatter plot analysis showed no association between Treg frequency and function at any ratio or between circulating Treg frequency and function at baseline and at 6 weeks post-ipi. An increase in Treg suppressive function was significantly associated with a decrease in PFS (p = 0.02). Conclusions We find that Treg frequency measures do not correlate with suppressive activity measured ex vivo. Treg suppressive activity increases correlate with poorer patient outcomes. Electronic supplementary material The online version of this article (doi:10.1186/s40425-016-0141-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Janet Retseck
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Robert VanderWeele
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Hui-Min Lin
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Yan Lin
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Lisa H Butterfield
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Ahmad A Tarhini
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| |
Collapse
|
24
|
A Phase II Clinical Trial of TRC105 (Anti-Endoglin Antibody) in Adults With Advanced/Metastatic Urothelial Carcinoma. Clin Genitourin Cancer 2016; 15:77-85. [PMID: 27328856 DOI: 10.1016/j.clgc.2016.05.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/06/2016] [Accepted: 05/18/2016] [Indexed: 01/02/2023]
Abstract
BACKGROUND In this trial we assessed the efficacy and tolerability of TRC105, a chimeric monoclonal antibody that targets CD105 (endoglin) in patients with advanced, previously treated urothelial carcinoma (UC). PATIENTS AND METHODS Patients received TRC105 15 mg/kg every 2 weeks on days 1 and 15 of each 28-day cycle. The primary end point was progression-free survival (PFS) at 6 months. Secondary end points included safety, toxicity, and overall survival (OS). CD105 expression was evaluated using immunohistochemistry (IHC) in a separate cohort of 50 UC patients. Biomarker studies included immune subsets, circulating tumor cells (CTCs), circulating endothelial cells (CECs), circulating endothelial progenitor cells (CEPs), and osteopontin. RESULTS Of 13 patients enrolled, 12 were evaluable for OS and PFS. The 3-month PFS probability was 18.2% (median PFS, 1.9 months [95% confidence interval (CI), 1.8-2.1 months). This met the criterion for ending accrual on the basis of the 2-stage design. Median OS was 8.3 months (95% CI, 3.3-17.0 months). IHC for CD105 scores was not associated with T stage (P = .26) or presence of lymph nodes (P = .64). Baseline levels of regulatory T and B cells, CEPs, and changes in CEC level after TRC105 exhibited trends toward an association with PFS or OS. CTCs pre- and post-TRC105 were detected in 4 of 4 patients. CONCLUSION Although TRC105 was well tolerated, it did not improve 6-month PFS in heavily pretreated patients with advanced UC. CD105 staining was present in 50% of UC tumors at different intensities. Our observations on the pharmacodynamic significance of immune subsets, CECs, and CTCs warrant further study.
Collapse
|
25
|
Holmgaard RB, Brachfeld A, Gasmi B, Jones DR, Mattar M, Doman T, Murphy M, Schaer D, Wolchok JD, Merghoub T. Timing of CSF-1/CSF-1R signaling blockade is critical to improving responses to CTLA-4 based immunotherapy. Oncoimmunology 2016; 5:e1151595. [PMID: 27622016 DOI: 10.1080/2162402x.2016.1151595] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/03/2015] [Accepted: 02/03/2016] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED Colony stimulating factor-1 (CSF-1) is produced by a variety of cancers and recruits myeloid cells that suppress antitumor immunity, including myeloid-derived suppressor cells (MDSCs.) Here, we show that both CSF-1 and its receptor (CSF-1R) are frequently expressed in tumors from cancer patients, and that this expression correlates with tumor-infiltration of MDSCs. Furthermore, we demonstrate that these tumor-infiltrating MDSCs are highly immunosuppressive but can be reprogrammed toward an antitumor phenotype in vitro upon CSF-1/CSF-1R signaling blockade. Supporting these findings, we show that inhibition of CSF-1/CSF-1R signaling using an anti-CSF-1R antibody can regulate both the number and the function of MDSCs in murine tumors in vivo. We further find that treatment with anti-CSF-1R antibody induces antitumor T-cell responses and tumor regression in multiple tumor models when combined with CTLA-4 blockade therapy. However, this occurs only when administered after or concurrent with CTLA-4 blockade, indicating that timing of each therapeutic intervention is critical for optimal antitumor responses. Importantly, MDSCs present within murine tumors after CTLA-4 blockade showed increased expression of CSF-1R and were capable of suppressing T cell proliferation, and CSF-1/CSF-1R expression in the human tumors was not reduced after treatment with CTLA-4 blockade immunotherapy. Taken together, our findings suggest that CSF-1R-expressing MDSCs can be targeted to modulate the tumor microenvironment and that timing of CSF-1/CSF-1R signaling blockade is critical to improving responses to checkpoint based immunotherapy. SIGNIFICANCE Infiltration by immunosuppressive myeloid cells contributes to tumor immune escape and can render patients resistant or less responsive to therapeutic intervention with checkpoint blocking antibodies. Our data demonstrate that blocking CSF-1/CSF-1R signaling using a monoclonal antibody directed to CSF-1R can regulate both the number and function of tumor-infiltrating immunosuppressive myeloid cells. In addition, our findings suggest that reprogramming myeloid responses may be a key in effectively enhancing cancer immunotherapy, offering several new potential combination therapies for future clinical testing. More importantly for clinical trial design, the timing of these interventions is critical to achieving improved tumor protection.
Collapse
Affiliation(s)
- Rikke B Holmgaard
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Alexandra Brachfeld
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Billel Gasmi
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - David R Jones
- Department of Surgery, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Marissa Mattar
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | | | | | | | - Jedd D Wolchok
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College and Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Taha Merghoub
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| |
Collapse
|
26
|
Holmgaard RB, Zamarin D, Lesokhin A, Merghoub T, Wolchok JD. Targeting myeloid-derived suppressor cells with colony stimulating factor-1 receptor blockade can reverse immune resistance to immunotherapy in indoleamine 2,3-dioxygenase-expressing tumors. EBioMedicine 2016; 6:50-58. [PMID: 27211548 PMCID: PMC4856741 DOI: 10.1016/j.ebiom.2016.02.024] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/11/2016] [Accepted: 02/12/2016] [Indexed: 12/21/2022] Open
Abstract
Tumor indoleamine 2,3-dioxygenase (IDO) promotes immunosuppression by direct action on effector T cells and Tregs and through recruitment, expansion and activation of myeloid-derived suppressor cells (MDSCs). Targeting of MDSCs is clinically being explored as a therapeutic strategy, though optimal targeting strategies and biomarkers predictive of response are presently unknown. Maturation and tumor recruitment of MDSCs are dependent on signaling through the receptor tyrosine kinase CSF-1R on myeloid cells. Here, we show that MDSCs are the critical cell population in IDO-expressing B16 tumors in mediating accelerated tumor outgrowth and resistance to immunotherapy. Using a clinically relevant drug, we show that inhibition of CSF-1R signaling can functionally block tumor-infiltrating MDSCs and enhance anti-tumor T cell responses. Furthermore, inhibition of CSF-1R sensitizes IDO-expressing tumors to immunotherapy with T cell checkpoint blockade, and combination of CSF-1R blockade with IDO inhibitors potently elicits tumor regression. These findings provide evidence for a critical and functional role for MDSCs on the in vivo outcome of IDO-expressing tumors. Tumor-infiltrating MDSCs promote accelerated outgrowth of IDO-expressing tumors. MDSCs infiltrating IDO-expressing tumors mediate resistance to immunotherapy. Combination of CSF-1R blockade with IDO inhibitors potently elicits tumor regression. CSF-1R blockade sensitizes tumors to the effects of immune checkpoint blockade.
Our data demonstrate that therapy with CSF-1R-blocking agents offers therapeutic benefit as a single agent and potentiates the effect of immunotherapeutic agents in IDO-expressing tumors infiltrated with CSF-1R-expressing MDSCs. These findings provide important insights into basic mechanisms underlying IDO mediated immune suppression and resistance to immunotherapies. In addition, it provides a strong rationale for therapeutic combinations with CSF-1R inhibitors in tumors expressing elevated IDO and highly infiltrated with MDSCs as predictive biomarkers.
Collapse
Affiliation(s)
- Rikke B Holmgaard
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Dmitriy Zamarin
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States; Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, United States; Weill Cornell Medical College, New York, NY 10065, United States; Graduate School of Medical Sciences of Cornell University, New York, NY 10065, United States
| | - Alexander Lesokhin
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States; Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, United States; Weill Cornell Medical College, New York, NY 10065, United States; Graduate School of Medical Sciences of Cornell University, New York, NY 10065, United States
| | - Taha Merghoub
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jedd D Wolchok
- Swim Across America/Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States; Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, United States; Weill Cornell Medical College, New York, NY 10065, United States; Graduate School of Medical Sciences of Cornell University, New York, NY 10065, United States.
| |
Collapse
|
27
|
Ribas A, Shin DS, Zaretsky J, Frederiksen J, Cornish A, Avramis E, Seja E, Kivork C, Siebert J, Kaplan-Lefko P, Wang X, Chmielowski B, Glaspy JA, Tumeh PC, Chodon T, Pe'er D, Comin-Anduix B. PD-1 Blockade Expands Intratumoral Memory T Cells. Cancer Immunol Res 2016. [PMID: 26787823 DOI: 10.1158/2326-6066.cir-15-0210.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tumor responses to programmed cell death protein 1 (PD-1) blockade therapy are mediated by T cells, which we characterized in 102 tumor biopsies obtained from 53 patients treated with pembrolizumab, an antibody to PD-1. Biopsies were dissociated, and single-cell infiltrates were analyzed by multicolor flow cytometry using two computational approaches to resolve the leukocyte phenotypes at the single-cell level. There was a statistically significant increase in the frequency of T cells in patients who responded to therapy. The frequency of intratumoral B cells and monocytic myeloid-derived suppressor cells significantly increased in patients' biopsies taken on treatment. The percentage of cells with a regulatory T-cell phenotype, monocytes, and natural killer cells did not change while on PD-1 blockade therapy. CD8(+) memory T cells were the most prominent phenotype that expanded intratumorally on therapy. However, the frequency of CD4(+) effector memory T cells significantly decreased on treatment, whereas CD4(+) effector T cells significantly increased in nonresponding tumors on therapy. In peripheral blood, an unusual population of blood cells expressing CD56 was detected in two patients with regressing melanoma. In conclusion, PD-1 blockade increases the frequency of T cells, B cells, and myeloid-derived suppressor cells in tumors, with the CD8(+) effector memory T-cell subset being the major T-cell phenotype expanded in patients with a response to therapy.
Collapse
Affiliation(s)
- Antoni Ribas
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California. Division of Surgical-Oncology, Department of Surgery, University of California Los Angeles, Los Angeles, California. Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, Los Angeles, California.
| | - Daniel Sanghoon Shin
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Jesse Zaretsky
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Juliet Frederiksen
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Andrew Cornish
- Departments of Biological Sciences and Systems Biology, Columbia University, New York, New York
| | - Earl Avramis
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Elizabeth Seja
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Christine Kivork
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | | | - Paula Kaplan-Lefko
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Xiaoyan Wang
- Department of General Internal Medicine and Healthy Services Research, University of California Los Angeles, Los Angeles, California
| | - Bartosz Chmielowski
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - John A Glaspy
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Paul C Tumeh
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, Los Angeles, California. Department of Medicine, Division of Dermatology. University of California Los Angeles, Los Angeles, California
| | - Thinle Chodon
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, New York
| | - Dana Pe'er
- Departments of Biological Sciences and Systems Biology, Columbia University, New York, New York
| | - Begoña Comin-Anduix
- Division of Surgical-Oncology, Department of Surgery, University of California Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, Los Angeles, California.
| |
Collapse
|
28
|
Ribas A, Shin DS, Zaretsky J, Frederiksen J, Cornish A, Avramis E, Seja E, Kivork C, Siebert J, Kaplan-Lefko P, Wang X, Chmielowski B, Glaspy JA, Tumeh PC, Chodon T, Pe'er D, Comin-Anduix B. PD-1 Blockade Expands Intratumoral Memory T Cells. Cancer Immunol Res 2016; 4:194-203. [PMID: 26787823 DOI: 10.1158/2326-6066.cir-15-0210] [Citation(s) in RCA: 309] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 12/04/2015] [Indexed: 02/06/2023]
Abstract
Tumor responses to programmed cell death protein 1 (PD-1) blockade therapy are mediated by T cells, which we characterized in 102 tumor biopsies obtained from 53 patients treated with pembrolizumab, an antibody to PD-1. Biopsies were dissociated, and single-cell infiltrates were analyzed by multicolor flow cytometry using two computational approaches to resolve the leukocyte phenotypes at the single-cell level. There was a statistically significant increase in the frequency of T cells in patients who responded to therapy. The frequency of intratumoral B cells and monocytic myeloid-derived suppressor cells significantly increased in patients' biopsies taken on treatment. The percentage of cells with a regulatory T-cell phenotype, monocytes, and natural killer cells did not change while on PD-1 blockade therapy. CD8(+) memory T cells were the most prominent phenotype that expanded intratumorally on therapy. However, the frequency of CD4(+) effector memory T cells significantly decreased on treatment, whereas CD4(+) effector T cells significantly increased in nonresponding tumors on therapy. In peripheral blood, an unusual population of blood cells expressing CD56 was detected in two patients with regressing melanoma. In conclusion, PD-1 blockade increases the frequency of T cells, B cells, and myeloid-derived suppressor cells in tumors, with the CD8(+) effector memory T-cell subset being the major T-cell phenotype expanded in patients with a response to therapy.
Collapse
Affiliation(s)
- Antoni Ribas
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California. Division of Surgical-Oncology, Department of Surgery, University of California Los Angeles, Los Angeles, California. Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, Los Angeles, California.
| | - Daniel Sanghoon Shin
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Jesse Zaretsky
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Juliet Frederiksen
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Andrew Cornish
- Departments of Biological Sciences and Systems Biology, Columbia University, New York, New York
| | - Earl Avramis
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Elizabeth Seja
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Christine Kivork
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | | | - Paula Kaplan-Lefko
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Xiaoyan Wang
- Department of General Internal Medicine and Healthy Services Research, University of California Los Angeles, Los Angeles, California
| | - Bartosz Chmielowski
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - John A Glaspy
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, California
| | - Paul C Tumeh
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, Los Angeles, California. Department of Medicine, Division of Dermatology. University of California Los Angeles, Los Angeles, California
| | - Thinle Chodon
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, New York
| | - Dana Pe'er
- Departments of Biological Sciences and Systems Biology, Columbia University, New York, New York
| | - Begoña Comin-Anduix
- Division of Surgical-Oncology, Department of Surgery, University of California Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, Los Angeles, California.
| |
Collapse
|
29
|
Zhang H, Li ZL, Ye SB, Ouyang LY, Chen YS, He J, Huang HQ, Zeng YX, Zhang XS, Li J. Myeloid-derived suppressor cells inhibit T cell proliferation in human extranodal NK/T cell lymphoma: a novel prognostic indicator. Cancer Immunol Immunother 2015; 64:1587-99. [PMID: 26497849 PMCID: PMC4643115 DOI: 10.1007/s00262-015-1765-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 10/04/2015] [Indexed: 01/04/2023]
Abstract
The expansion of myeloid-derived suppressor cells (MDSCs) and its correlation with advanced disease stage have been shown in solid cancers. Here, we investigated the functional features and clinical significance of MDSCs in extranodal NK/T cell lymphoma (ENKL). A higher percentage of circulating HLA-DR−CD33+CD11b+ MDSCs was observed in ENKL patients than in healthy controls (P < 0.05, n = 32) by flow cytometry analysis. These MDSCs from ENKL patients (ENKL-MDSCs) consisted of CD14+ monocytic (Mo-MDSCs, >60 %) and CD15+ granulocytic (PMN-MDSCs, <20 %) MDSCs. Furthermore, these ENKL-MDSCs expressed higher levels of Arg-1, iNOS and IL-17 compared to the levels of MDSCs from healthy donors, and they expressed moderate levels of TGFβ and IL-10 but lower levels of CD66b. The ENKL-MDSCs strongly suppressed the anti-CD3-induced allogeneic and autologous CD4 T cell proliferation (P < 0.05), but they only slightly suppressed CD8 T cell proliferation (P > 0.05). Interestingly, ENKL-MDSCs inhibited the secretion of IFNγ but promoted IL-10, IL-17 and TGFβ secretion as well as Foxp3 expression in T cells. The administration of inhibitors of iNOS, Arg-1 and ROS significantly reversed the suppression of anti-CD3-induced T cell proliferation by MDSCs (P < 0.05). Importantly, based on multivariate Cox regression analysis, the HLA-DR−CD33+CD11b+ cells and CD14+ Mo-MDSCs were independent predictors for disease-free survival (DFS, P = 0.013 and 0.016) and overall survival (OS, P = 0.017 and 0.027). Overall, our results identified for the first time that ENKL-MDSCs (mainly Mo-MDSCs) have a prognostic value for patients and a suppressive function on T cell proliferation.
Collapse
Affiliation(s)
- Han Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Ze-Lei Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Shu-Biao Ye
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Li-Ying Ouyang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Intensive Care Unit Department, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Yu-Shan Chen
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Radiotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Jia He
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Hui-Qiang Huang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Yi-Xin Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Xiao-Shi Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
| | - Jiang Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
| |
Collapse
|
30
|
Tarhini AA, Zahoor H, Lin Y, Malhotra U, Sander C, Butterfield LH, Kirkwood JM. Baseline circulating IL-17 predicts toxicity while TGF-β1 and IL-10 are prognostic of relapse in ipilimumab neoadjuvant therapy of melanoma. J Immunother Cancer 2015; 3:39. [PMID: 26380086 PMCID: PMC4570556 DOI: 10.1186/s40425-015-0081-1] [Citation(s) in RCA: 302] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/10/2015] [Indexed: 01/31/2023] Open
Abstract
Background We evaluated candidate circulating serum cytokines, chemokines and growth factors in patients with locally/regionally advanced melanoma receiving neoadjuvant ipilimumab with toxicity and clinical outcome. Methods Patients were treated with ipilimumab (10 mg/kg IV every 3 weeks, 2 doses) before and after surgery. xMAP multiplex serum testing for 36 functionally selected cytokines and chemokines was performed at baseline and at six weeks (following ipilimumab). Based on our prior data, the association of IL-17 and immune related colitis was tested. Serum cytokines were divided into functional groups (Th1, Th2, Regulatory, Proinflammatory) and were assessed at baseline and week 6 using sparse-group Lasso modeling to assess the association of various cytokine groups with progression free survival (PFS). The linear combination of the cytokines/chemokines in this model was then used as a risk score and a Kaplan-Meier curve was generated to examine the association of the dichotomized score and PFS. Results Thirty-five patients were enrolled whose staging was: IIIB (3; N2b), IIIC (30; N2c, N3), IV (2). Median follow-up was 18 months. Among 33 evaluable patients, median PFS was 11 months (95 % CI = 6.2–19.2). IL-17 was found to correlate significantly with the incidence of grade 3 diarrhea/colitis when measured at baseline (p = 0.02) with a trend towards significance at 6 weeks (p = 0.06). In the modeling analysis, at baseline, the linear combination of 2 regulatory cytokines [TGF- β1 (ρ = 0.19) and IL-10 (ρ = -0.34)] was significantly associated with PFS (HR 2.66; p = 0.035). No significant correlations with clinical outcomes were found in examining the week 6 cytokines. Conclusions Baseline IL-17 level was significantly associated with the later development of severe diarrhea/colitis while the combination of baseline TGF- β1 and IL-10 levels were associated with therapeutic clinical outcome after neoadjuvant ipilimumab. These findings warrant further investigation and validation. Trial registration ClinicalTrials.gov Identifier NCT00972933.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Haris Zahoor
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Yan Lin
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Usha Malhotra
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Cindy Sander
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Lisa H Butterfield
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - John M Kirkwood
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| |
Collapse
|
31
|
Maccalli C, Giannarelli D, Capocefalo F, Pilla L, Fonsatti E, Di Giacomo AM, Parmiani G, Maio M. Immunological markers and clinical outcome of advanced melanoma patients receiving ipilimumab plus fotemustine in the NIBIT-M1 study. Oncoimmunology 2015; 5:e1071007. [PMID: 27057436 DOI: 10.1080/2162402x.2015.1071007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/04/2015] [Accepted: 07/05/2015] [Indexed: 12/30/2022] Open
Abstract
Clinical activity was observed in metastatic melanoma (MM) patients treated with ipilimumab (IPI) combined with fotemustine (FTM) in the phase II NIBIT-M1 study. Peripheral blood mononuclear cells (PBMCs) and serum were collected from MM patients at pre- and at weeks 12 and 24 post-treatment. A comprehensive phenotypic and functional immunomonitoring of circulating T cells, and the detection of soluble immunoregulatory molecules was carried out and correlated with clinical outcome. The frequency at baseline and along the treatment of circulating T central memory cells expressing activation/differentiation markers, such as CD3+CD4+CD45RO+BTLA+, CD3+CD4+4-1BB or Th17 lymphocytes correlated with the clinical outcome of MM patients. Moreover, either the absence or the presence of soluble NKG2D ligands (ULBP-1 or -2) at baseline in the serum of MM patients enabled to discriminate subjects with long-term survival (median overall survival, (OS) = 33.6 mo for ULBP-1 and -2) from poor survivors (OS = 9.8 or 6.6 mo, respectively). Conversely, no significant association between the levels of soluble MICA, MICB and ULBP-3 and the clinical outcome of patients was observed. An inverse correlation between circulating levels of these molecules at baseline and frequency of either CD3+CD4+CD45RO+BTLA+ or Th17 or CD3+CD4+4-1BB+ T cells occurred in patients with a favorable clinical outcome. The simultaneous monitoring of different immune parameters, though validation in a large cohort of patients is needed, allowed to identify an association between phenotypic and soluble markers representing a possible predictive immunological signature for the clinical activity of IPI plus FTM.
Collapse
Affiliation(s)
- Cristina Maccalli
- Italian Network for Biotherapy of Tumors-(NIBIT)-Laboratory, Siena, Italy; Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy; Unit of Immuno-biotherapy of Melanoma and Solid Tumors, San Raffaele Foundation Center, Milan, Italy
| | - Diana Giannarelli
- Unit of Statistics, Regina Elena National Cancer Institute , Rome, Italy
| | - Filippo Capocefalo
- Unit of Immuno-biotherapy of Melanoma and Solid Tumors, San Raffaele Foundation Center , Milan, Italy
| | - Lorenzo Pilla
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy; Unit of Immuno-biotherapy of Melanoma and Solid Tumors, San Raffaele Foundation Center, Milan, Italy
| | - Ester Fonsatti
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori , Siena, Italy
| | - Anna Maria Di Giacomo
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori , Siena, Italy
| | - Giorgio Parmiani
- Italian Network for Biotherapy of Tumors-(NIBIT)-Laboratory, Siena, Italy; Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy; Unit of Immuno-biotherapy of Melanoma and Solid Tumors, San Raffaele Foundation Center, Milan, Italy
| | - Michele Maio
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori , Siena, Italy
| |
Collapse
|
32
|
Ward FJ, Dahal LN, Khanolkar RC, Shankar SP, Barker RN. Targeting the alternatively spliced soluble isoform of CTLA-4: prospects for immunotherapy? Immunotherapy 2015; 6:1073-84. [PMID: 25428646 DOI: 10.2217/imt.14.73] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
CTLA-4 is an inhibitory protein that contributes to immune homeostasis and tolerance, a role that has led to its exploitation as a therapeutic in several clinical settings including cancer and autoimmune disease. Development of CTLA-4 therapies focused largely on the full-length receptor isoform but other CTLA-4 isoforms are also expressed, including a secretable form of CTLA-4 (soluble CTLA-4 [sCTLA-4]). The contribution of sCTLA-4 to immune regulation has been less well studied, primarily because it was identified some years after the original description of CTLA-4. Here, we examine how sCTLA-4 might contribute to immune regulation and ask whether it might be a biomarker to inform current CTLA-4 therapies or represent a novel CTLA-4 target for future therapeutics.
Collapse
Affiliation(s)
- Frank J Ward
- Section of Immunology & Infection, Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| | | | | | | | | |
Collapse
|
33
|
Tye GJ, Ioannou K, Amofah E, Quartey-Papafio R, Westrop SJ, Krishnamurthy P, Noble A, Harrison PM, Gaensler KML, Barber LD, Farzaneh F. The combined molecular adjuvant CASAC enhances the CD8+ T cell response to a tumor-associated self-antigen in aged, immunosenescent mice. IMMUNITY & AGEING 2015; 12:6. [PMID: 26157468 PMCID: PMC4495856 DOI: 10.1186/s12979-015-0033-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/08/2015] [Indexed: 02/03/2023]
Abstract
Background Ineffective induction of T cell mediated immunity in older individuals remains a persistent challenge for vaccine development. Thus, there is a need for more efficient and sophisticated adjuvants that will complement novel vaccine strategies for the elderly. To this end, we have investigated a previously optimized, combined molecular adjuvant, CASAC (Combined Adjuvant for Synergistic Activation of Cellular immunity), incorporating two complementary Toll-like receptor agonists, CpG and polyI:C, a class-II epitope, and interferon (IFN)-γ in aged mice. Findings In aged mice with typical features of immunosenescence, antigen specific CD8+ T cell responses were stimulated after serial vaccinations with CASAC or Complete/Incomplete Freund’s Adjuvant (CFA/IFA) and a class I epitope, deriving either from ovalbumin (SIINFEKL, SIL) or the melanoma-associated self-antigen, tyrosinase-related protein-2 (SVYDFFVWL, SVL). Pentamer analysis revealed that aged, CASAC/SIL-vaccinated animals had substantially higher frequencies of H-2Kb/SIL-specific CD8+ T cells compared to the CFA/IFA-vaccinated groups. Similarly, higher frequencies of H-2Kb/SVL-pentamer+ and IFN-γ+ CD8+ T cells were detected in the aged, CASAC + SVL-vaccinated mice than in their CFA/IFA-vaccinated counterparts. In both antigen settings, CASAC promoted significantly better functional CD8+ T cell activity. Conclusion These studies demonstrate that functional CD8+ T cells, specific for both foreign and tumour-associated self-antigens, can be effectively induced in aged immunosenescent mice using the novel multi-factorial adjuvant CASAC.
Collapse
Affiliation(s)
- Gee Jun Tye
- Department of Haematological Medicine, King's College London, Rayne Institute, London, UK.,Institute for Research in Molecular Medicine, Universiti Sains Malaysia, George town, Malaysia
| | - Kyriaki Ioannou
- Department of Haematological Medicine, King's College London, Rayne Institute, London, UK
| | - Eunice Amofah
- Department of Haematological Medicine, King's College London, Rayne Institute, London, UK
| | - Ruby Quartey-Papafio
- Department of Haematological Medicine, King's College London, Rayne Institute, London, UK
| | - Samantha J Westrop
- Department of Haematological Medicine, King's College London, Rayne Institute, London, UK
| | - Pramila Krishnamurthy
- Department of Haematological Medicine, King's College London, Rayne Institute, London, UK
| | - Alistair Noble
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, Guy's Hospital, London, UK
| | - Phillip M Harrison
- Division of Transplant and Mucosal Cell Biology, King's College London, London, UK
| | - Karin M L Gaensler
- Department of Medicine, University of California, San Francisco School of Medicine, San Francisco, USA
| | - Linda D Barber
- Department of Haematological Medicine, King's College London, Rayne Institute, London, UK
| | - Farzin Farzaneh
- Department of Haematological Medicine, King's College London, Rayne Institute, London, UK
| |
Collapse
|
34
|
Johnson DB, Puzanov I, Kelley MC. Talimogene laherparepvec (T-VEC) for the treatment of advanced melanoma. Immunotherapy 2015; 7:611-9. [PMID: 26098919 DOI: 10.2217/imt.15.35] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Melanoma often spreads to cutaneous or subcutaneous sites that are amenable to direct, intralesional injection. As such, developing effective injectable agents has been of considerable interest. Talimogene laherperepvec (T-VEC) is an injectable modified oncolytic herpes virus being developed for the treatment of advanced melanoma. Pre-clinical studies have shown that T-VEC preferentially infects melanoma cells and exerts antitumor activity through directly mediating cell death and by augmenting local and even distant immune responses. T-VEC has now been assessed in Phase II and III clinical trials and has demonstrated a tolerable side-effect profile and promising efficacy, showing an improved durable response rate and a trend toward superior overall survival compared to granulocyte-macrophage colony-stimulating factor. Despite these promising results, responses have been uncommon in patients with visceral metastases. T-VEC is currently being evaluated in combination with other immune therapies (ipilimumab and pembrolizumab) with early signs of activity. In this review, we discuss the preclinical rationale, the clinical experience, and future directions for T-VEC in advanced melanoma.
Collapse
Affiliation(s)
- Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, 777 PRB, 2220 Pierce Ave, Nashville, TN 37232, USA
| | - Igor Puzanov
- Department of Medicine, Vanderbilt University Medical Center, 777 PRB, 2220 Pierce Ave, Nashville, TN 37232, USA
| | - Mark C Kelley
- Department of Surgery, Vanderbilt University Medical Center, TN, USA
| |
Collapse
|
35
|
Floros T, Tarhini AA. Anticancer Cytokines: Biology and Clinical Effects of Interferon-α2, Interleukin (IL)-2, IL-15, IL-21, and IL-12. Semin Oncol 2015; 42:539-48. [PMID: 26320059 DOI: 10.1053/j.seminoncol.2015.05.015] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Efforts over nearly four decades have focused on ways to use cytokines to manipulate the host immune response towards cancer cell recognition and eradication. Significant advances were achieved with interleukin-2 (IL-2) and interferon-α (IFN-α), primarily in the treatment of patients with melanoma and renal cell carcinoma. However, the utility of other cytokines showing promise in the preclinical setting has not been established largely because of toxicity, the complex functionality of each cytokine and the difficulty mimicking in preclinical models the human environment. Here, we review the basic biology and the clinical experiences with IFN-α, IL-2, IL-15, IL-21, and IL-12. We will also review ongoing clinical trials and discuss future directions including potential use of cytokines in combination with other effective immunotherapy approaches that have come of age in recent years.
Collapse
Affiliation(s)
- Theofanis Floros
- University of Pittsburgh Cancer Institute, Pittsburgh, PA; Athens Naval and Veterans Hospital, Pittsburgh, PA
| | - Ahmad A Tarhini
- University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Pittsburgh School of Medicine, Pittsburgh, PA.
| |
Collapse
|
36
|
Mok S, Tsoi J, Koya RC, Hu-Lieskovan S, West BL, Bollag G, Graeber TG, Ribas A. Inhibition of colony stimulating factor-1 receptor improves antitumor efficacy of BRAF inhibition. BMC Cancer 2015; 15:356. [PMID: 25939769 PMCID: PMC4432503 DOI: 10.1186/s12885-015-1377-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 04/27/2015] [Indexed: 12/22/2022] Open
Abstract
Background Malignant melanoma is an aggressive tumor type that often develops drug resistance to targeted therapeutics. The production of colony stimulating factor 1 (CSF-1) in tumors recruits myeloid cells such as M2-polarized macrophages and myeloid derived suppressor cells (MDSC), leading to an immune suppressive tumor milieu. Methods We used the syngeneic mouse model of BRAFV600E-driven melanoma SM1, which secretes CSF-1, to evaluate the ability of the CSF-1 receptor (CSF-1R) inhibitor PLX3397 to improve the antitumor efficacy of the oncogenic BRAF inhibitor vemurafenib. Results Combined BRAF and CSF-1R inhibition resulted in superior antitumor responses compared with either therapy alone. In mice receiving PLX3397 treatment, a dramatic reduction of tumor-infiltrating myeloid cells (TIM) was observed. In this model, we could not detect a direct effect of TIMs or pro-survival cytokines produced by TIMs that could confer resistance to PLX4032 (vemurafenib). However, the macrophage inhibitory effects of PLX3397 treatment in combination with the paradoxical activation of wild type BRAF-expressing immune cells mediated by PLX4032 resulted in more tumor-infiltrating lymphocytes (TIL). Depletion of CD8+ T-cells abrogated the antitumor response to the combination therapy. Furthermore, TILs isolated from SM1 tumors treated with PLX3397 and PLX4032 displayed higher immune potentiating activity. Conclusions The combination of BRAF-targeted therapy with CSF-1R blockade resulted in increased CD8 T-cell responses in the SM1 melanoma model, supporting the ongoing evaluation of this therapeutic combination in patients with BRAFV600 mutant metastatic melanoma. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1377-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stephen Mok
- Department of Molecular and Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, USA. .,MD Anderson Cancer Center, Houston, Texas, USA.
| | - Jennifer Tsoi
- Department of Molecular and Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, USA.
| | - Richard C Koya
- Department of Surgery, Division of Surgical Oncology, University of California Los Angeles (UCLA), Los Angeles, CA, USA. .,Roswell Park Cancer Institute, Buffalo, New York, USA.
| | - Siwen Hu-Lieskovan
- Department of Medicine, Division of Hematology/Oncology, UCLA, University of California Los Angeles (UCLA), 11-934 Factor Building, 10833 Le Conte Avenue, Los Angeles, CA, 90095-1782, USA.
| | | | | | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, USA. .,Crump Institute for Molecular Imaging, UCLA, University of California Los Angeles (UCLA), Los Angeles, CA, USA. .,Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles (UCLA), Los Angeles, CA, USA.
| | - Antoni Ribas
- Department of Molecular and Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, USA. .,Department of Surgery, Division of Surgical Oncology, University of California Los Angeles (UCLA), Los Angeles, CA, USA. .,Department of Medicine, Division of Hematology/Oncology, UCLA, University of California Los Angeles (UCLA), 11-934 Factor Building, 10833 Le Conte Avenue, Los Angeles, CA, 90095-1782, USA. .,Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles (UCLA), Los Angeles, CA, USA.
| |
Collapse
|
37
|
Abstract
Cancer vaccines are designed to promote tumor specific immune responses, particularly cytotoxic CD8 positive T cells that are specific to tumor antigens. The earliest vaccines, which were developed in 1994-95, tested non-mutated, shared tumor associated antigens that had been shown to be immunogenic and capable of inducing clinical responses in a minority of people with late stage cancer. Technological developments in the past few years have enabled the investigation of vaccines that target mutated antigens that are patient specific. Several platforms for cancer vaccination are being tested, including peptides, proteins, antigen presenting cells, tumor cells, and viral vectors. Standard of care treatments, such as surgery and ablation, chemotherapy, and radiotherapy, can also induce antitumor immunity, thereby having cancer vaccine effects. The monitoring of patients' immune responses at baseline and after standard of care treatment is shedding light on immune biomarkers. Combination therapies are being tested in clinical trials and are likely to be the best approach to improving patient outcomes.
Collapse
Affiliation(s)
- Lisa H Butterfield
- Departments of Medicine, Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
38
|
Gibney GT, Kudchadkar RR, DeConti RC, Thebeau MS, Czupryn MP, Tetteh L, Eysmans C, Richards A, Schell MJ, Fisher KJ, Horak CE, Inzunza HD, Yu B, Martinez AJ, Younos I, Weber JS. Safety, correlative markers, and clinical results of adjuvant nivolumab in combination with vaccine in resected high-risk metastatic melanoma. Clin Cancer Res 2015; 21:712-20. [PMID: 25524312 PMCID: PMC4620684 DOI: 10.1158/1078-0432.ccr-14-2468] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE The anti-programmed death-1 (PD-1) antibody nivolumab (BMS-936558) has clinical activity in patients with metastatic melanoma. Nivolumab plus vaccine was investigated as adjuvant therapy in resected stage IIIC and IV melanoma patients. EXPERIMENTAL DESIGN HLA-A*0201 positive patients with HMB-45, NY-ESO-1, and/or MART-1 positive resected tumors received nivolumab (1 mg/kg, 3 mg/kg, or 10 mg/kg i.v.) with a multi-peptide vaccine (gp100, MART-1, and NY-ESO-1 with Montanide ISA 51 VG) every 2 weeks for 12 doses followed by nivolumab maintenance every 12 weeks for 8 doses. Primary objective was safety and determination of a maximum tolerated dose (MTD). Secondary objectives included relapse-free survival (RFS), overall survival (OS), and immunologic correlative studies. RESULTS Thirty-three patients were enrolled. Median age was 47 years; 55% were male. Two patients had stage IIIC disease; 31 patients had stage IV disease. Median follow-up was 32.1 months. MTD was not reached. Most common related adverse events (>40%) were vaccine injection site reaction, fatigue, rash, pruritus, nausea, and arthralgias. Five related grade 3 adverse events [hypokalemia (1), rash (1), enteritis (1), and colitis (2)] were observed. Ten of 33 patients relapsed. Estimated median RFS was 47.1 months; median OS was not reached. Increases in CTLA-4(+)/CD4(+), CD25(+)Treg/CD4(+), and tetramer specific CD8(+) T-cell populations were observed with treatment (P < 0.05). Trends for lower baseline myeloid-derived suppressor cell and CD25(+)Treg/CD4(+) populations were seen in nonrelapsing patients; PD-L1 tumor status was not significantly associated with RFS. CONCLUSIONS Nivolumab with vaccine is well tolerated as adjuvant therapy and demonstrates immunologic activity with promising survival in high-risk resected melanoma, justifying further study.
Collapse
Affiliation(s)
- Geoffrey T Gibney
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida. Department of Oncologic Sciences, University of South Florida, Tampa, Florida.
| | - Ragini R Kudchadkar
- Division of Oncology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Ronald C DeConti
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida. Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| | - Melissa S Thebeau
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| | - Maria P Czupryn
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| | - Leticia Tetteh
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| | - Cabell Eysmans
- Division of Oncology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Allison Richards
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| | - Michael J Schell
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Kate J Fisher
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Christine E Horak
- Bristol-Myers Squibb, Early Clinical and Translational Research, Princeton, New Jersey
| | - H David Inzunza
- Bristol-Myers Squibb, Early Clinical and Translational Research, Princeton, New Jersey
| | - Bin Yu
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| | - Alberto J Martinez
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| | - Ibrahim Younos
- Department of Clinical Pharmacology, College of Medicine, Minufiya University, Minufiya, Egypt. Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Jeffrey S Weber
- Department of Cutaneous Oncology and Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida. Department of Oncologic Sciences, University of South Florida, Tampa, Florida
| |
Collapse
|
39
|
Abstract
The quality of the host immune response in patients with advanced melanoma is compromised with a bias towards Th2-type polarization and a tumor microenvironment that facilitates disease progression. Overcoming tumor-induced immune suppression through strategies that build upon the immunomodulatory qualities and clinical activity of interferon-α as demonstrated in the melanoma adjuvant setting is a major clinical need. The recent advances in the field of immune checkpoint modulation and the unprecedented clinical activity in advanced melanoma opens the door on novel combinations that may overcome tumor tolerogenic mechanisms that are known to suppress the potent anti-tumor impact of interferon (IFN)-α. Promising preliminary data suggest that such combinations may move the clinical management of advanced melanoma into the next level, beyond what is currently seen with immune checkpoint blockers alone.
Collapse
Affiliation(s)
- Imran Rafique
- University of Pittsburgh Medical Center, Pittsburgh, PA
| | - John M Kirkwood
- University of Pittsburgh Medical Center, Pittsburgh, PA; University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | - Ahmad A Tarhini
- University of Pittsburgh Medical Center, Pittsburgh, PA; University of Pittsburgh Cancer Institute, Pittsburgh, PA.
| |
Collapse
|
40
|
Messmer MN, Netherby CS, Banik D, Abrams SI. Tumor-induced myeloid dysfunction and its implications for cancer immunotherapy. Cancer Immunol Immunother 2014; 64:1-13. [PMID: 25432147 DOI: 10.1007/s00262-014-1639-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 11/19/2014] [Indexed: 01/27/2023]
Abstract
Immune function relies on an appropriate balance of the lymphoid and myeloid responses. In the case of neoplasia, this balance is readily perturbed by the dramatic expansion of immature or dysfunctional myeloid cells accompanied by a reciprocal decline in the quantity/quality of the lymphoid response. In this review, we seek to: (1) define the nature of the atypical myelopoiesis observed in cancer patients and the impact of this perturbation on clinical outcomes; (2) examine the potential mechanisms underlying these clinical manifestations; and (3) explore potential strategies to restore normal myeloid cell differentiation to improve activation of the host antitumor immune response. We posit that fundamental alterations in myeloid homeostasis triggered by the neoplastic process represent critical checkpoints that govern therapeutic efficacy, as well as offer novel cellular-based biomarkers for tracking changes in disease status or relapse.
Collapse
Affiliation(s)
- Michelle N Messmer
- Department of Immunology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | | | | | | |
Collapse
|
41
|
Rudolph BM, Loquai C, Gerwe A, Bacher N, Steinbrink K, Grabbe S, Tuettenberg A. Increased frequencies of CD11b(+) CD33(+) CD14(+) HLA-DR(low) myeloid-derived suppressor cells are an early event in melanoma patients. Exp Dermatol 2014; 23:202-4. [PMID: 24495013 DOI: 10.1111/exd.12336] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2014] [Indexed: 11/28/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous cell population characterized by immunosuppressive activity. Elevated levels of MDSC in peripheral blood are found in inflammatory diseases as well as in malignant tumors where they are supposed to be major contributors to mechanisms of tumor-associated tolerance. We investigated the frequency and function of MDSC in peripheral blood of melanoma patients and observed an accumulation of CD11b(+) CD33(+) CD14(+) HLA-DR(low) MDSC in all stages of disease (I-IV), including early stage I patients. Disease progression and enhanced tumor burden did not result in a further increase in frequencies or change in phenotype of MDSC. By investigation of specific MDSC-associated cytokines in patients' sera, we found an accumulation of IL-8 in all stages of disease. T-cell proliferation assays revealed that MDSC critically contribute to suppressed antigen-specific T-cell reactivity and thus might explain the frequently observed transient effects of immunotherapeutic strategies in melanoma patients.
Collapse
Affiliation(s)
- Berenice M Rudolph
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
42
|
Solito S, Marigo I, Pinton L, Damuzzo V, Mandruzzato S, Bronte V. Myeloid-derived suppressor cell heterogeneity in human cancers. Ann N Y Acad Sci 2014; 1319:47-65. [DOI: 10.1111/nyas.12469] [Citation(s) in RCA: 302] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Samantha Solito
- Department of Surgery; Oncology and Gastroenterology; Oncology and Immunology Section; University of Padova; Padova Italy
| | | | - Laura Pinton
- Department of Surgery; Oncology and Gastroenterology; Oncology and Immunology Section; University of Padova; Padova Italy
| | - Vera Damuzzo
- Department of Surgery; Oncology and Gastroenterology; Oncology and Immunology Section; University of Padova; Padova Italy
| | - Susanna Mandruzzato
- Department of Surgery; Oncology and Gastroenterology; Oncology and Immunology Section; University of Padova; Padova Italy
- Istituto Oncologico Veneto; IOV-IRCCS; Padova Italy
| | - Vincenzo Bronte
- Pathology and Diagnostics; Verona University Hospital; Verona Italy
| |
Collapse
|
43
|
Saenger Y, Magidson J, Liaw B, de Moll E, Harcharik S, Fu Y, Wassmann K, Fisher D, Kirkwood J, Oh WK, Friedlander P. Blood mRNA expression profiling predicts survival in patients treated with tremelimumab. Clin Cancer Res 2014; 20:3310-8. [PMID: 24721645 DOI: 10.1158/1078-0432.ccr-13-2906] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Tremelimumab (ticilimumab, Pfizer), is a monoclonal antibody (mAb) targeting cytotoxic T lymphocyte-associated antigen-4 (CTLA-4). Ipilimumab (Yervoy, BMS), another anti-CTLA-4 antibody, is approved by the U.S. Federal Drug Administration (FDA). Biomarkers are needed to identify the subset of patients who will achieve tumor control with CTLA-4 blockade. EXPERIMENTAL DESIGN Pretreatment peripheral blood samples from 218 patients with melanoma who were refractory to prior therapy and receiving tremelimumab in a multicenter phase II study were measured for 169 mRNA transcripts using reverse transcription polymerase chain reaction (RT-PCR). A two-class latent model yielded a risk score based on four genes that were highly predictive of survival (P < 0.001). This signature was validated in an independent population of 260 treatment-naïve patients with melanoma enrolled in a multicenter phase III study of tremelimumab. RESULTS Median follow-up was 297 days for the training population and 386 days for the test population. Expression levels of the 169 genes were closely correlated across the two populations (r = 0.9939). A four-gene model, including cathepsin D (CTSD), phopholipase A2 group VII (PLA2G7), thioredoxin reductase 1 (TXNRD1), and interleukin 1 receptor-associated kinase 3 (IRAK3), predicted survival in the test population (P = 0.001 by log-rank test). This four-gene model added to the predictive value of clinical predictors (P < 0.0001). CONCLUSIONS Expression levels of CTSD, PLA2G7, TXNRD1, and IRAK3 in peripheral blood are predictive of survival in patients with melanoma treated with tremelimumab. Blood mRNA signatures should be further explored to define patient subsets likely to benefit from immunotherapy.
Collapse
Affiliation(s)
- Yvonne Saenger
- Authors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, CanadaAuthors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, Canada
| | - Jay Magidson
- Authors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, Canada
| | - Bobby Liaw
- Authors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, Canada
| | - Ellen de Moll
- Authors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, Canada
| | - Sara Harcharik
- Authors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, Canada
| | - Yichun Fu
- Authors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, Canada
| | - Karl Wassmann
- Authors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, Canada
| | - David Fisher
- Authors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, Canada
| | - John Kirkwood
- Authors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, Canada
| | - William K Oh
- Authors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, Canada
| | - Philip Friedlander
- Authors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, CanadaAuthors' Affiliations: Division of Hematology and Oncology, Tisch Cancer Institute, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York; Statistical Innovations, Belmont; Department of Dermatology, Harvard Medical School, Boston, Massachusetts; Departments of Medicine, Dermatology and Translational Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and Gene News, Ontario, Canada
| |
Collapse
|
44
|
Karakhanova S, Mosl B, Harig S, von Ahn K, Fritz J, Schmidt J, Jäger D, Werner J, Bazhin AV. Influence of interferon-alpha combined with chemo (radio) therapy on immunological parameters in pancreatic adenocarcinoma. Int J Mol Sci 2014; 15:4104-25. [PMID: 24608924 PMCID: PMC3975387 DOI: 10.3390/ijms15034104] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/06/2014] [Accepted: 02/14/2014] [Indexed: 01/01/2023] Open
Abstract
Prognosis of patients with carcinoma of the exocrine pancreas is particularly poor. A combination of chemotherapy with immunotherapy could be an option for treatment of pancreatic cancer. The aim of this study was to perform an immunomonitoring of 17 patients with pancreatic cancer from the CapRI-2 study, and tumor-bearing mice treated with combination of chemo (radio) therapies with interferon-2α. Low doses of interferon-2α led to a decrease in total leukocyte and an increase in monocyte counts. Furthermore, we observed a positive effect of interferon-2α therapy on the dendritic cells and NK (natural killer) cell activation immediately after the first injection. In addition, we recorded an increased amount of interferon-γ and IL-10 in the serum following the interferon-2α therapy. These data clearly demonstrate that pancreatic carcinoma patients also show an immunomodulatory response to interferon-2α therapy. Analysis of immunosuppressive cells in the Panc02 orthotopic mouse model of pancreatic cancer revealed an accumulation of the myeloid-derived suppressor cells in spleens and tumors of the mice treated with interferon-2α and 5-fluorouracil. The direct effect of the drugs on myeloid-derived suppressor cells was also registered in vitro. These data expose the importance of immunosuppressive mechanisms induced by combined chemo-immunotherapy.
Collapse
Affiliation(s)
- Svetlana Karakhanova
- Department of General Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Beate Mosl
- Department of General Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Sabine Harig
- Department of General Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Katharina von Ahn
- Department of General Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Jasmin Fritz
- Department of General Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Jan Schmidt
- Department of General Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Dirk Jäger
- National Centre for Tumor Disease, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University, 81377 Munich, Germany.
| | - Alexandr V Bazhin
- Department of General Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
45
|
Meyer C, Cagnon L, Costa-Nunes CM, Baumgaertner P, Montandon N, Leyvraz L, Michielin O, Romano E, Speiser DE. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol Immunother 2014; 63:247-57. [PMID: 24357148 PMCID: PMC11029062 DOI: 10.1007/s00262-013-1508-5] [Citation(s) in RCA: 455] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 12/05/2013] [Indexed: 12/18/2022]
Abstract
Metastatic melanoma has a poor prognosis with high resistance to chemotherapy and radiation. Recently, the anti-CTLA-4 antibody ipilimumab has demonstrated clinical efficacy, being the first agent to significantly prolong the overall survival of inoperable stage III/IV melanoma patients. A major aim of patient immune monitoring is the identification of biomarkers that predict clinical outcome. We studied circulating myeloid-derived suppressor cells (MDSC) in ipilimumab-treated patients to detect alterations in the myeloid cell compartment and possible correlations with clinical outcome. Lin(-) CD14(+) HLA-DR(-) monocytic MDSC were enriched in peripheral blood of melanoma patients compared to healthy donors (HD). Tumor resection did not significantly alter MDSC frequencies. During ipilimumab treatment, MDSC frequencies did not change significantly compared to baseline levels. We observed high inter-patient differences. MDSC frequencies in ipilimumab-treated patients were independent of baseline serum lactate dehydrogenase levels but tended to increase in patients with severe metastatic disease (M1c) compared to patients with metastases in skin or lymph nodes only (M1a), who had frequencies comparable to HD. Interestingly, clinical responders to ipilimumab therapy showed significantly less lin(-) CD14(+) HLA-DR(-) cells as compared to non-responders. The data suggest that the frequency of monocytic MDSC may be used as predictive marker of response, as low frequencies identify patients more likely benefitting from ipilimumab treatment. Prospective clinical trials assessing MDSC frequencies as potential biomarkers are warranted to validate these observations.
Collapse
Affiliation(s)
- Christiane Meyer
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
| | - Laurène Cagnon
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
| | - Carla M. Costa-Nunes
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
| | - Petra Baumgaertner
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
| | - Nicole Montandon
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
| | - Loredana Leyvraz
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
| | - Olivier Michielin
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
- Service of Medical Oncology, Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Emanuela Romano
- Service of Medical Oncology, Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Daniel E. Speiser
- Ludwig Center for Cancer Research of the University of Lausanne, Lausanne, Switzerland
- Service of Medical Oncology, Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| |
Collapse
|
46
|
Tarhini AA, Edington H, Butterfield LH, Lin Y, Shuai Y, Tawbi H, Sander C, Yin Y, Holtzman M, Johnson J, Rao UNM, Kirkwood JM. Immune monitoring of the circulation and the tumor microenvironment in patients with regionally advanced melanoma receiving neoadjuvant ipilimumab. PLoS One 2014; 9:e87705. [PMID: 24498358 PMCID: PMC3912016 DOI: 10.1371/journal.pone.0087705] [Citation(s) in RCA: 249] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/02/2014] [Indexed: 11/21/2022] Open
Abstract
We evaluated neoadjuvant ipilimumab in patients with surgically operable regionally advanced melanoma in order to define markers of activity in the blood and tumor as assessed at baseline (before ipilimumab) and early on-treatment. Patients were treated with ipilimumab (10 mg/kg intravenously every 3 weeks ×2 doses) bracketing surgery. Tumor and blood biospecimens were obtained at baseline and at surgery. Flow cytometry and immunohistochemistry for select biomarkers were performed. Thirty five patients were enrolled; IIIB (3; N2b), IIIC (32; N2c, N3), IV (2). Worst toxicities included Grade 3 diarrhea/colitis (5; 14%), hepatitis (2; 6%), rash (1; 3%), elevated lipase (3; 9%). Median follow up was 18 months: among 33 evaluable patients, median progression free survival (PFS) was 11 months, 95% CI (6.2–19.2). There was a significant decrease in circulating myeloid derived suppressor cells (MDSC). Greater decrease in circulating monocyte gate MDSC Lin1−/HLA-DR−/CD33+/CD11b+ was associated with improved PFS (p = 0.03). There was a significant increase in circulating regulatory T cells (Treg; CD4+CD25hi+Foxp3+) that, unexpectedly, was associated with improved PFS (HR = 0.57; p = 0.034). Baseline evidence of fully activated type I CD4+ and CD8+ antigen-specific T cell immunity against cancer-testis (NY-ESO-1) and melanocytic lineage (MART-1, gp100) antigens was detected and was significantly potentiated after ipilimumab. In tumor, there was a significant increase in CD8+ T cells after ipilimumab (p = 0.02). Ipilimumab induced increased tumor infiltration by fully activated (CD69+) CD3+/CD4+ and CD3+/CD8+ T cells with evidence of induction/potentiation of memory T cells (CD45RO+). The change in Treg observed within the tumor showed an inverse relationship with clinical benefit and greater decrease in tumor MDSC subset Lin1−/HLA-DR−/CD33+/CD11b+ was associated with improved PFS at one year. Neoadjuvant evaluation revealed a significant immunomodulating role for ipilimumab on Treg, MDSC and effector T cells in the circulation and tumor microenvironment that warrants further pursuit in the quest for optimizing melanoma immunotherapy.
Collapse
Affiliation(s)
- Ahmad A. Tarhini
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Howard Edington
- Department of Surgery, West Penn Allegheny Health System, Pittsburgh, Pennsylvania, United States of America
| | - Lisa H. Butterfield
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Yan Lin
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Yongli Shuai
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Hussein Tawbi
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Cindy Sander
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Yan Yin
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Matthew Holtzman
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Jonas Johnson
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Uma N. M. Rao
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - John M. Kirkwood
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
47
|
Tarhini AA, Lin Y, Yeku O, LaFramboise WA, Ashraf M, Sander C, Lee S, Kirkwood JM. A four-marker signature of TNF-RII, TGF-α, TIMP-1 and CRP is prognostic of worse survival in high-risk surgically resected melanoma. J Transl Med 2014; 12:19. [PMID: 24457057 PMCID: PMC3909384 DOI: 10.1186/1479-5876-12-19] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/18/2014] [Indexed: 11/18/2022] Open
Abstract
Background E1694 tested GM2-KLH-QS21 vaccine versus high-dose interferon-α2b (HDI) as adjuvant therapy for operable stage IIB-III melanoma. We tested banked serum specimens from patients in the vaccine arm of E1694 for prognostic biomarkers. Methods Aushon Multiplex Platform was used to quantitate baseline serum levels of 115 analytes from 40 patients. Least absolute shrinkage and selection operator proportional hazard regression (Lasso PH) was used to select markers that are most informative for relapse-free survival (RFS) and overall survival (OS). Regular Cox PH models were then fit with the markers selected by the Lasso PH. Survival receiver operating characteristic (ROC) analysis was used to evaluate the ability of the models to predict 1-year RFS and 5-year OS. Results Four markers that include Tumor Necrosis Factor alpha Receptor II (TNF-RII), Transforming Growth Factor alpha (TGF-α), Tissue Inhibitor of Metalloproteinases 1 (TIMP-1), and C-reactive protein (CRP) were found to be most informative for the prediction of OS (high levels correlate with worse prognosis). The dichotomized risk score based on the four markers could significantly separate the OS curves (p = 0.0005). When using the four-marker PH model to predict 5-year OS, we achieved an area under the curve (AUC) of 89% (cross validated AUC = 72%). High baseline TNF-RII was also significantly associated with worse RFS. The RFS with high (above median) TNF-RII was significantly lower than low TNF-RII (p = 0.01). Conclusions The biomarker signature consisting of TNFR-II, TGF-α, TIMP-1 and CRP is significantly prognostic of survival in patients with high-risk melanoma and warrants further investigation.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue, 5th Floor, Suite 555, Pittsburgh, PA 15232, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Mok S, Koya RC, Tsui C, Xu J, Robert L, Wu L, Graeber T, West BL, Bollag G, Ribas A. Inhibition of CSF-1 receptor improves the antitumor efficacy of adoptive cell transfer immunotherapy. Cancer Res 2014; 74:153-161. [PMID: 24247719 PMCID: PMC3947337 DOI: 10.1158/0008-5472.can-13-1816] [Citation(s) in RCA: 259] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Colony stimulating factor 1 (CSF-1) recruits tumor-infiltrating myeloid cells (TIM) that suppress tumor immunity, including M2 macrophages and myeloid-derived suppressor cells (MDSC). The CSF-1 receptor (CSF-1R) is a tyrosine kinase that is targetable by small molecule inhibitors such as PLX3397. In this study, we used a syngeneic mouse model of BRAF(V600E)-driven melanoma to evaluate the ability of PLX3397 to improve the efficacy of adoptive cell therapy (ACT). In this model, we found that combined treatment produced superior antitumor responses compared with single treatments. In mice receiving the combined treatment, a dramatic reduction of TIMs and a skewing of MHCII(low) to MHCII(hi) macrophages were observed. Furthermore, mice receiving the combined treatment exhibited an increase in tumor-infiltrating lymphocytes (TIL) and T cells, as revealed by real-time imaging in vivo. In support of these observations, TILs from these mice released higher levels of IFN-γ. In conclusion, CSF-1R blockade with PLX3397 improved the efficacy of ACT immunotherapy by inhibiting the intratumoral accumulation of immunosuppressive macrophages.
Collapse
Affiliation(s)
- Stephen Mok
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095 (UCLA)
| | - Richard C. Koya
- Plexxikon Inc., Berkeley, California 94710, U.S.A; Roswell Park Cancer Institute, Buffalo, New York 14263
| | | | - Jingying Xu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095 (UCLA)
| | - Lídia Robert
- Department of Medicine, Division of Hematology/Oncology, UCLA
| | - Lily Wu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095 (UCLA)
- Institute for Molecular Medicine, UCLA
- Department of Urology, UCLA
- Department of Pediatrics, UCLA
| | - Thomas Graeber
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095 (UCLA)
- the Jonsson Comprehensive Cancer Center (JCCC) at UCLA
- Institute for Molecular Medicine, UCLA
- Crump Institute for Molecular Imaging, UCLA
| | - Brian L. West
- Plexxikon Inc., Berkeley, California 94710, U.S.A; Roswell Park Cancer Institute, Buffalo, New York 14263
| | - Gideon Bollag
- Plexxikon Inc., Berkeley, California 94710, U.S.A; Roswell Park Cancer Institute, Buffalo, New York 14263
| | - Antoni Ribas
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095 (UCLA)
- the Jonsson Comprehensive Cancer Center (JCCC) at UCLA
- Surgery, Division of Surgical Oncology, UCLA
- Institute for Molecular Medicine, UCLA
- Department of Medicine, Division of Hematology/Oncology, UCLA
| |
Collapse
|
49
|
Sasseville VG, Mansfield KG, Brees DJ. Safety biomarkers in preclinical development: translational potential. Vet Pathol 2013; 51:281-91. [PMID: 24091814 DOI: 10.1177/0300985813505117] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The identification, application, and qualification of safety biomarkers are becoming increasingly critical to successful drug discovery and development as companies are striving to develop drugs for difficult targets and for novel disease indications in a risk-adverse environment. Translational safety biomarkers that are minimally invasive and monitor drug-induced toxicity during human clinical trials are urgently needed to assess whether toxicities observed in preclinical toxicology studies are relevant to humans at therapeutic doses. The interpretation of data during the biomarker qualification phase should include careful consideration of the analytic method used, the biology, pharmacokinetic and pharmacodynamic properties of the biomarker, and the pathophysiology of the process studied. The purpose of this review is to summarize commonly employed technologies in the development of fluid- and tissue-based safety biomarkers in drug discovery and development and to highlight areas of ongoing novel assay development.
Collapse
Affiliation(s)
- V G Sasseville
- Discovery and Investigative Safety, Preclinical Safety, Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA.
| | | | | |
Collapse
|
50
|
Myeloid-derived suppressor cells are associated with disease progression and decreased overall survival in advanced-stage melanoma patients. Cancer Immunol Immunother 2013; 62:1711-22. [PMID: 24072401 DOI: 10.1007/s00262-013-1475-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/12/2013] [Indexed: 12/15/2022]
Abstract
Myeloid-derived suppressor cells are increased in the peripheral blood of advanced-stage cancer patients; however, no studies have shown a correlation of these immunosuppressive cells with clinical outcomes in melanoma patients. We characterized the frequency and suppressive function of multiple subsets of myeloid-derived suppressor cells in the peripheral blood of 34 patients with Stage IV melanoma, 20 patients with Stage I melanoma, and 15 healthy donors. The frequency of CD14+ MDSCs (Lin- CD11b+ HLA-DR- CD14+ CD33+) and CD14- MDSCs (Lin- CD11b+ HLA-DR- CD14- CD33+) was increased in the peripheral blood of Stage IV melanoma patients relative to healthy donors. The frequency of CD14+ and CD14- MDSCs correlated with each other and with the increased frequency of regulatory T cells, but not with classically defined monocytes. CD14- MDSCs isolated from the peripheral blood of Stage IV melanoma patients suppressed T cell activation more than those isolated from healthy donors, and the frequency of these cells correlated with disease progression and decreased overall survival. Our study provides the first evidence that the frequency of CD14- MDSCs negatively correlates with clinical outcomes in advanced-stage melanoma patients. These data indicate that suppressive MDSCs should be considered as targets for future immunotherapies.
Collapse
|