1
|
Baumrucker CC, Harris N, Hoover S, Czerniecki BJ. Intratumoral Immunotherapy in Breast Cancer. Vaccines (Basel) 2025; 13:429. [PMID: 40333343 PMCID: PMC12031351 DOI: 10.3390/vaccines13040429] [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: 03/04/2025] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
Breast cancer remains the most frequently diagnosed cancer and the second highest cause of cancer death in females. Metastatic recurrence that is resistant to traditional therapies presents a major challenge, necessitating the development of an innovative treatment strategy. Immunotherapy has gained popularity in the treatment of cancer, particularly melanoma, lung cancer, and more recently breast cancer. Major developments in immunotherapy have been made with a better understanding of the tumor microenvironment and how the microenvironment can be manipulated to induce an anti-tumor immune response. Intratumorally delivered immunotherapy can be used to create a local immune response. This review provides a comprehensive overview of intratumoral immunotherapy for breast cancer and its resultant changes in the tumor microenvironment. The discussed immunotherapeutics include oncolytic viruses, nucleic acids, innate immune agonists, bacteria, chimeric antigen receptor T cells, and dendritic cells. The review also evaluates completed clinical trials using these therapies. Lastly, the review offers future perspectives in the development of breast cancer immunotherapy.
Collapse
Affiliation(s)
- Camille C. Baumrucker
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
| | - Nicole Harris
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Susan Hoover
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Brian J. Czerniecki
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
2
|
Montauti E, Oh DY, Fong L. CD4 + T cells in antitumor immunity. Trends Cancer 2024; 10:969-985. [PMID: 39242276 PMCID: PMC11464182 DOI: 10.1016/j.trecan.2024.07.009] [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: 06/04/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 09/09/2024]
Abstract
Advances in cancer immunotherapy have transformed cancer care and realized unprecedented responses in many patients. The growing arsenal of novel therapeutics - including immune checkpoint inhibition (ICI), adoptive T cell therapies (ACTs), and cancer vaccines - reflects the success of cancer immunotherapy. The therapeutic benefits of these treatment modalities are generally attributed to the enhanced quantity and quality of antitumor CD8+ T cell responses. Nevertheless, CD4+ T cells are now recognized to play key roles in both the priming and effector phases of the antitumor immune response. In addition to providing T cell help through co-stimulation and cytokine production, CD4+ T cells can also possess cytotoxicity either directly on MHC class II-expressing tumor cells or to other cells within the tumor microenvironment (TME). The presence of specific populations of CD4+ T cells, and their intrinsic plasticity, within the TME can represent an important determinant of clinical response to immune checkpoint inhibitors, vaccines, and chimeric antigen receptor (CAR) T cell therapies. Understanding how the antitumor functions of specific CD4+ T cell types are induced while limiting their protumorigenic attributes will enable more successful immunotherapies.
Collapse
Affiliation(s)
- Elena Montauti
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - David Y Oh
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA; Immunotherapy Integrated Research Center, Fred Hutchison Cancer Center, Seattle, WA, USA.
| |
Collapse
|
3
|
Gerlini G, Susini P, Sestini S, Brandani P, Giannotti V, Borgognoni L. Langerhans Cells in Sentinel Lymph Nodes from Melanoma Patients. Cancers (Basel) 2024; 16:1890. [PMID: 38791968 PMCID: PMC11119378 DOI: 10.3390/cancers16101890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/05/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Langerhans cells (LCs) are professional Dendritic Cells (DCs) involved in immunoregulatory functions. At the skin level, LCs are immature. In response to tissue injuries, they migrate to regional Lymph Nodes (LNs), reaching a full maturation state. Then, they become effective antigen-presenting cells (APCs) that induce anti-cancer responses. Notably, melanoma patients present several DC alterations in the Sentinel Lymph Node (SLN), where primary antitumoral immunity is generated. LCs are the most represented DCs subset in melanoma SLNs and are expected to play a key role in the anti-melanoma response. With this paper, we aim to review the current knowledge and future perspectives regarding LCs and melanoma. METHODS A systematic review was carried out according to the PRISMA statement using the PubMed (MEDLINE) library from January 2004 to January 2024, searching for original studies discussing LC in melanoma. RESULTS The final synthesis included 15 articles. Several papers revealed significant LCs-melanoma interactions. CONCLUSIONS Melanoma immune escape mechanisms include SLN LC alterations, favoring LN metastasis arrival/homing and melanoma proliferation. The SLN LCs of melanoma patients are defective but not irreversibly, and their function may be restored by appropriate stimuli. Thus, LCs represent a promising target for future immunotherapeutic strategies and cancer vaccines.
Collapse
Affiliation(s)
- Gianni Gerlini
- Plastic and Reconstructive Surgery Unit, Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Santa Maria Annunziata Hospital, 50012 Florence, Italy; (S.S.); (P.B.); (V.G.); (L.B.)
| | - Pietro Susini
- Plastic Surgery Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy;
| | - Serena Sestini
- Plastic and Reconstructive Surgery Unit, Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Santa Maria Annunziata Hospital, 50012 Florence, Italy; (S.S.); (P.B.); (V.G.); (L.B.)
| | - Paola Brandani
- Plastic and Reconstructive Surgery Unit, Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Santa Maria Annunziata Hospital, 50012 Florence, Italy; (S.S.); (P.B.); (V.G.); (L.B.)
| | - Vanni Giannotti
- Plastic and Reconstructive Surgery Unit, Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Santa Maria Annunziata Hospital, 50012 Florence, Italy; (S.S.); (P.B.); (V.G.); (L.B.)
| | - Lorenzo Borgognoni
- Plastic and Reconstructive Surgery Unit, Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Santa Maria Annunziata Hospital, 50012 Florence, Italy; (S.S.); (P.B.); (V.G.); (L.B.)
| |
Collapse
|
4
|
Reticker-Flynn NE, Engleman EG. Lymph nodes: at the intersection of cancer treatment and progression. Trends Cell Biol 2023; 33:1021-1034. [PMID: 37149414 PMCID: PMC10624650 DOI: 10.1016/j.tcb.2023.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 05/08/2023]
Abstract
Metastasis to lymph nodes (LNs) is a common feature of disease progression in most solid organ malignancies. Consequently, LN biopsy and lymphadenectomy are common clinical practices, not only because of their diagnostic utility but also as a means of deterring further metastatic spread. LN metastases have the potential to seed additional tissues and can induce metastatic tolerance, a process by which tumor-specific immune tolerance in LNs promotes further disease progression. Nonetheless, phylogenetic studies have revealed that distant metastases are not necessarily derived from nodal metastases. Furthermore, immunotherapy efficacy is increasingly being attributed to initiation of systemic immune responses within LNs. We argue that lymphadenectomy and nodal irradiation should be approached with caution, particularly in patients receiving immunotherapy.
Collapse
Affiliation(s)
- Nathan E Reticker-Flynn
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Edgar G Engleman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
5
|
Han B, Fang T, Zhang Y, Zhang Y, Gao J, Xue Y. Association of the TGFβ gene family with microenvironmental features of gastric cancer and prediction of response to immunotherapy. Front Oncol 2022; 12:920599. [PMID: 36119489 PMCID: PMC9478444 DOI: 10.3389/fonc.2022.920599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/01/2022] [Indexed: 11/26/2022] Open
Abstract
In the complex tumor microenvironment, TGFβ is a pleiotropic cytokine involved in regulating cellular processes such as cancer cell proliferation, apoptosis and metastasis. TGFβ defines three subtypes (TGFβ1, TGFβ2, and TGFβ3), of which TGFβ is highly expressed in many cancers, especially those showing high dissemination potential. In addition, increased expression of TGFβ in multiple cancers is usually positively correlated with epithelial mesenchymal transition (EMT) and coordinated with the expression of genes driving EMT-related genes. TGFβ signaling in the tumor microenvironment inhibits the antitumor function of multiple immune cell populations, including T cells and natural killer cells, and the resulting immunosuppression severely limits the efficacy of immune checkpoint inhibitors and other immunotherapeutic approaches. As a major pathway to enhance the efficacy of cancer immunotherapy effects, the role of TGFβ signaling inhibitors have been evaluated in many clinical trials. However, the potential functions and mechanisms of TGFβ1, TGFβ2 and TGFβ3 in gastric cancer progression and tumor immunology are unclear. In this study, we comprehensively analyzed TGFβ1, TGFβ2 and TGFβ3 and gastric cancer microenvironmental features, including immune cell infiltration, EMT, hypoxia, mutation, immunotherapy and drug treatment, based on HMUCH sequencing data (GSE184336) and public databases. We also validated the protein expression levels of TGFβ in gastric cancer tissues as well as the role of TGFβ factor in cytology experiments. This report reveals the important role of the TGFβ gene family in gastric cancer and provides possible relationships and potential mechanisms of TGFβ in gastric cancer.
Collapse
|
6
|
Zhu Y, Li X, Wang L, Hong X, Yang J. Metabolic reprogramming and crosstalk of cancer-related fibroblasts and immune cells in the tumor microenvironment. Front Endocrinol (Lausanne) 2022; 13:988295. [PMID: 36046791 PMCID: PMC9421293 DOI: 10.3389/fendo.2022.988295] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/25/2022] [Indexed: 12/13/2022] Open
Abstract
It is notorious that cancer cells alter their metabolism to adjust to harsh environments of hypoxia and nutritional starvation. Metabolic reprogramming most often occurs in the tumor microenvironment (TME). TME is defined as the cellular environment in which the tumor resides. This includes surrounding blood vessels, fibroblasts, immune cells, signaling molecules and the extracellular matrix (ECM). It is increasingly recognized that cancer cells, fibroblasts and immune cells within TME can regulate tumor progression through metabolic reprogramming. As the most significant proportion of cells among all the stromal cells that constitute TME, cancer-associated fibroblasts (CAFs) are closely associated with tumorigenesis and progression. Multitudinous studies have shown that CAFs participate in and promote tumor metabolic reprogramming and exert regulatory effects via the dysregulation of metabolic pathways. Previous studies have demonstrated that curbing the substance exchange between CAFs and tumor cells can dramatically restrain tumor growth. Emerging studies suggest that CAFs within the TME have emerged as important determinants of metabolic reprogramming. Metabolic reprogramming also occurs in the metabolic pattern of immune cells. In the meanwhile, immune cell phenotype and functions are metabolically regulated. Notably, immune cell functions influenced by metabolic programs may ultimately lead to alterations in tumor immunity. Despite the fact that multiple previous researches have been devoted to studying the interplays between different cells in the tumor microenvironment, the complicated relationship between CAFs and immune cells and implications of metabolic reprogramming remains unknown and requires further investigation. In this review, we discuss our current comprehension of metabolic reprogramming of CAFs and immune cells (mainly glucose, amino acid, and lipid metabolism) and crosstalk between them that induces immune responses, and we also highlight their contributions to tumorigenesis and progression. Furthermore, we underscore potential therapeutic opportunities arising from metabolism dysregulation and metabolic crosstalk, focusing on strategies targeting CAFs and immune cell metabolic crosstalk in cancer immunotherapy.
Collapse
Affiliation(s)
- Yifei Zhu
- School of Medicine, Southeast University, Nanjing, China
| | - Xinyan Li
- School of Medicine, Southeast University, Nanjing, China
| | - Lei Wang
- School of Medicine, Southeast University, Nanjing, China
| | - Xiwei Hong
- School of Medicine, Southeast University, Nanjing, China
| | - Jie Yang
- Department of General surgery, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| |
Collapse
|
7
|
van Pul KM, Notohardjo JCL, Fransen MF, Koster BD, Stam AGM, Chondronasiou D, Lougheed SM, Bakker J, Kandiah V, van den Tol MP, Jooss K, Vuylsteke RJCLM, van den Eertwegh AJM, de Gruijl TD. Local delivery of low-dose anti–CTLA-4 to the melanoma lymphatic basin leads to systemic T
reg
reduction and effector T cell activation. Sci Immunol 2022; 7:eabn8097. [DOI: 10.1126/sciimmunol.abn8097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Preclinical studies show that locoregional CTLA-4 blockade is equally effective in inducing tumor eradication as systemic delivery, without the added risk of immune-related side effects. This efficacy is related to access of the CTLA-4 blocking antibodies to tumor-draining lymph nodes (TDLNs). Local delivery of anti–CTLA-4 after surgical removal of primary melanoma, before sentinel lymph node biopsy (SLNB), provides a unique setting to clinically assess the role of TDLN in the biological efficacy of locoregional CTLA-4 blockade. Here, we have evaluated the safety, tolerability, and immunomodulatory effects in the SLN and peripheral blood of a single dose of tremelimumab [a fully human immunoglobulin gamma-2 (IgG2) mAb directed against CTLA-4] in a dose range of 2 to 20 mg, injected intradermally at the tumor excision site 1 week before SLNB in 13 patients with early-stage melanoma (phase 1 trial; NCT04274816). Intradermal delivery was safe and well tolerated and induced activation of migratory dendritic cell (DC) subsets in the SLN. It also induced profound and durable decreases in regulatory T cell (T
reg
) frequencies and activation of effector T cells in both SLN and peripheral blood. Moreover, systemic T cell responses against NY-ESO-1 or MART-1 were primed or boosted (
N
= 7), in association with T cell activation and central memory T cell differentiation. These findings indicate that local administration of anti–CTLA-4 may offer a safe and promising adjuvant treatment strategy for patients with early-stage melanoma. Moreover, our data demonstrate a central role for TDLN in the biological efficacy of CTLA-4 blockade and support TDLN-targeted delivery methods.
Collapse
Affiliation(s)
- Kim M. van Pul
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Amsterdam UMC location Vrije Universiteit, Surgical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Jessica C. L. Notohardjo
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Marieke F. Fransen
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam UMC location Vrije Universiteit, Pulmonary Diseases, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
| | - Bas D. Koster
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Anita G. M. Stam
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Dafni Chondronasiou
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Sinéad M. Lougheed
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Joyce Bakker
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Vinitha Kandiah
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - M. Petrousjka van den Tol
- Amsterdam UMC location Vrije Universiteit, Surgical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | | | | | - Alfons J. M. van den Eertwegh
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| | - Tanja D. de Gruijl
- Amsterdam UMC location Vrije Universiteit, Medical Oncology, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands
- Cancer Center Amsterdam, Cancer Immunology, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunology, Cancer Immunology, Amsterdam, Netherlands
| |
Collapse
|
8
|
NK and cells with NK-like activities in cancer immunotherapy-clinical perspectives. Med Oncol 2022; 39:131. [PMID: 35716327 DOI: 10.1007/s12032-022-01735-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/13/2022] [Indexed: 01/10/2023]
Abstract
Natural killer (NK) cells are lymphoid cells of innate immunity that take important roles in immune surveillance. NK cells are considered as a bridge between innate and adaptive immunity, and their infiltration into tumor area is related positively with prolonged patient survival. They are defined as CD16+ CD56+ CD3- cells in clinic. NK cells promote cytolytic effects on target cells and induce their apoptosis. Loss of NK cell cytotoxic activity and reduction in the number of activating receptors are the current issues for application of such cells in cellular immunotherapy, which resulted in the diminished long-term effects. The focus of this review is to discuss about the activity of NK cells and cells with NK-like activity including natural killer T (NKT), cytokine-induced killer (CIK) and lymphokine-activated killer (LAK) cells in immunotherapy of human solid cancers.
Collapse
|
9
|
Gruijs M, Ganzevles SH, Stigter-van Walsum M, van der Mast R, van Ostaijen-ten Dam MM, Tuk CW, Schilham MW, Leemans CR, Brakenhoff RH, van Egmond M, van de Ven R, Bakema JE. NK Cell-Dependent Antibody-Mediated Immunotherapy Is Improved In Vitro and In Vivo When Combined with Agonists for Toll-like Receptor 2 in Head and Neck Cancer Models. Int J Mol Sci 2021; 22:11057. [PMID: 34681717 PMCID: PMC8541276 DOI: 10.3390/ijms222011057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
The immunosuppressive character of head and neck cancers may explain the relatively low response rates to antibody therapy targeting a tumor antigen, such as cetuximab, and anti-PD-1 checkpoint inhibition. Immunostimulatory agents that overcome tumor-derived inhibitory signals could augment therapeutic efficacy, thereby enhancing tumor elimination and improving patient survival. Here, we demonstrate that cetuximab treatment combined with immunostimulatory agonists for Toll-like receptor (TLR) 2 induces profound immune responses. Natural killer (NK) cells, isolated from healthy individuals or patients with head and neck cancer, harbored enhanced cytotoxic capacity and increased tumor-killing potential in vitro. Additionally, combination treatment increased the release of several pro-inflammatory cytokines and chemokines by NK cells. Tumor-bearing mice that received cetuximab and the TLR2 ligand Pam3CSK4 showed increased infiltration of immune cells into the tumors compared to mice that received cetuximab monotherapy, resulting in a significant delay in tumor growth or even complete tumor regression. Moreover, combination treatment resulted in improved overall survival in vivo. In conclusion, combining tumor-targeting antibody-based immunotherapy with TLR stimulation represents a promising treatment strategy to improve the clinical outcomes of cancer patients. This treatment could well be applied together with other therapeutic strategies such as anti-PD-(L)1 checkpoint inhibition to further overcome immunosuppression.
Collapse
MESH Headings
- Animals
- Antibody-Dependent Cell Cytotoxicity/immunology
- Cell Line, Tumor
- Cetuximab/pharmacology
- Cetuximab/therapeutic use
- Cytokines/metabolism
- Drug Therapy, Combination
- Female
- Head and Neck Neoplasms/therapy
- Humans
- Immunotherapy
- Killer Cells, Natural/immunology
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Lipopeptides/pharmacology
- Lipopeptides/therapeutic use
- Mice
- Mice, Nude
- Receptors, IgG/agonists
- Receptors, IgG/metabolism
- Toll-Like Receptor 2/agonists
- Toll-Like Receptor 2/metabolism
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Mandy Gruijs
- Amsterdam UMC, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam—Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.G.); (R.v.d.M.); (C.W.T.); (M.v.E.)
| | - Sonja H. Ganzevles
- Amsterdam UMC, Department of Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam—Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (S.H.G.); (M.S.-v.W.); (C.R.L.); (R.H.B.); (J.E.B.)
| | - Marijke Stigter-van Walsum
- Amsterdam UMC, Department of Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam—Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (S.H.G.); (M.S.-v.W.); (C.R.L.); (R.H.B.); (J.E.B.)
| | - Richard van der Mast
- Amsterdam UMC, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam—Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.G.); (R.v.d.M.); (C.W.T.); (M.v.E.)
- Amsterdam UMC, Department of Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam—Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (S.H.G.); (M.S.-v.W.); (C.R.L.); (R.H.B.); (J.E.B.)
| | - Monique M. van Ostaijen-ten Dam
- Leiden University Medical Center, Department of Pediatrics, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (M.M.v.O.-t.D.); (M.W.S.)
| | - Cornelis W. Tuk
- Amsterdam UMC, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam—Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.G.); (R.v.d.M.); (C.W.T.); (M.v.E.)
| | - Marco W. Schilham
- Leiden University Medical Center, Department of Pediatrics, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (M.M.v.O.-t.D.); (M.W.S.)
| | - C. René Leemans
- Amsterdam UMC, Department of Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam—Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (S.H.G.); (M.S.-v.W.); (C.R.L.); (R.H.B.); (J.E.B.)
| | - Ruud H. Brakenhoff
- Amsterdam UMC, Department of Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam—Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (S.H.G.); (M.S.-v.W.); (C.R.L.); (R.H.B.); (J.E.B.)
| | - Marjolein van Egmond
- Amsterdam UMC, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam—Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.G.); (R.v.d.M.); (C.W.T.); (M.v.E.)
- Amsterdam UMC, Department of Surgery, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Rieneke van de Ven
- Amsterdam UMC, Department of Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam—Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (S.H.G.); (M.S.-v.W.); (C.R.L.); (R.H.B.); (J.E.B.)
| | - Jantine E. Bakema
- Amsterdam UMC, Department of Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam—Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (S.H.G.); (M.S.-v.W.); (C.R.L.); (R.H.B.); (J.E.B.)
| |
Collapse
|
10
|
Sabree SA, Voigt AP, Blackwell SE, Vishwakarma A, Chimenti MS, Salem AK, Weiner GJ. Direct and indirect immune effects of CMP-001, a virus-like particle containing a TLR9 agonist. J Immunother Cancer 2021; 9:jitc-2021-002484. [PMID: 34083419 PMCID: PMC8183212 DOI: 10.1136/jitc-2021-002484] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 02/06/2023] Open
Abstract
Background CMP-001, also known as vidutolimod, is a virus-like particle containing a TLR9 agonist that is showing promise in early clinical trials. Our group previously demonstrated that the immunostimulatory effects of CMP-001 are dependent on an anti-Qβ antibody response which results in opsonization of CMP-001 and uptake by plasmacytoid dendritic cells (pDCs) that then produce interferon (IFN)-α. IFN-α then leads to an antitumor T-cell response that is responsible for the in vivo efficacy of CMP-001. Here, we explore mechanisms by which the initial effects of CMP-001 on pDCs activate other cells that can contribute to development of an antitumor T-cell response. Methods Uptake of CMP-001 by various peripheral blood mononuclear cell (PBMC) populations and response to anti-Qβ-coated CMP-001 were evaluated by flow cytometry and single-cell RNA sequencing. Purified monocytes were treated with anti-Qβ-coated CMP-001 or recombinant IFN-α to evaluate direct and secondary effects of anti-Qβ-coated CMP-001 on monocytes. Results Monocytes had the highest per cell uptake of anti-Qβ-coated CMP-001 with lower levels of uptake by pDCs and other cell types. Treatment of PBMCs with anti-Qβ-coated CMP-001 induced upregulation of IFN-responsive genes including CXCL10, PDL1, and indoleamine-2,3-dioxygenase (IDO) expression by monocytes. Most of the impact of anti-Qβ-coated CMP-001 on monocytes was indirect and mediated by IFN-α, but uptake of anti-Qβ-coated CMP-001 altered the monocytic response to IFN-α and resulted in enhanced expression of PDL1, IDO, and CD80 and suppressed expression of CXCL10. These changes included an enhanced ability to induce autologous CD4 T-cell proliferation. Conclusions Anti-Qβ-coated CMP-001 induces IFN-α production by pDCs which has secondary effects on a variety of cells including monocytes. Uptake of anti-Qβ-coated CMP-001 by monocytes alters their response to IFN-α, resulting in enhanced expression of PDL1, IDO and CD80 and suppressed expression of CXCL10. Despite aspects of an immunosuppressive phenotype, these monocytes demonstrated increased ability to augment autologous CD4 T-cell proliferation. These findings shed light on the complexity of the mechanism of action of anti-Qβ-coated CMP-001 and provide insight into pathways that may be targeted to further enhance the efficacy of this novel approach to immunotherapy.
Collapse
Affiliation(s)
- Shakoora A Sabree
- Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, IA, USA.,Medical Scientist Training Program, The University of Iowa Carver College of Medicine, Iowa City, IA, USA.,Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, USA
| | - Andrew P Voigt
- Medical Scientist Training Program, The University of Iowa Carver College of Medicine, Iowa City, IA, USA.,Department of Ophthalmology and Visual Sciences, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Sue E Blackwell
- Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, USA
| | - Ajaykumar Vishwakarma
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA.,Division of Pharmaceutics and Translational Therapeutics, The University of Iowa College of Pharmacy, Iowa City, IA, USA
| | - Michael S Chimenti
- Iowa Institute of Human Genetics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Aliasger K Salem
- Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, USA.,Division of Pharmaceutics and Translational Therapeutics, The University of Iowa College of Pharmacy, Iowa City, IA, USA
| | - George J Weiner
- Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, USA .,Division of Pharmaceutics and Translational Therapeutics, The University of Iowa College of Pharmacy, Iowa City, IA, USA.,Department of Internal Medicine, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| |
Collapse
|
11
|
van Pul KM, Fransen MF, van de Ven R, de Gruijl TD. Immunotherapy Goes Local: The Central Role of Lymph Nodes in Driving Tumor Infiltration and Efficacy. Front Immunol 2021; 12:643291. [PMID: 33732264 PMCID: PMC7956978 DOI: 10.3389/fimmu.2021.643291] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/09/2021] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint blockade (ICB) has changed the therapeutic landscape of oncology but its impact is limited by primary or secondary resistance. ICB resistance has been related to a lack of T cells infiltrating into the tumor. Strategies to overcome this hurdle have so far focused on the tumor microenvironment, but have mostly overlooked the role of tumor-draining lymph nodes (TDLN). Whereas for CTLA-4 blockade TDLN have long since been implicated due to its perceived mechanism-of-action involving T cell priming, only recently has evidence been emerging showing TDLN to be vital for the efficacy of PD-1 blockade as well. TDLN are targeted by developing tumors to create an immune suppressed pre-metastatic niche which can lead to priming of dysfunctional antitumor T cells. In this review, we will discuss the evidence that therapeutic targeting of TDLN may ensure sufficient antitumor T cell activation and subsequent tumor infiltration to facilitate effective ICB. Indeed, waves of tumor-specific, proliferating stem cell-like, or progenitor exhausted T cells, either newly primed or reinvigorated in TDLN, are vital for PD-1 blockade efficacy. Both tumor-derived migratory dendritic cell (DC) subsets and DC subsets residing in TDLN, and an interplay between them, have been implicated in the induction of these T cells, their imprinting for homing and subsequent tumor control. We propose that therapeutic approaches, involving local delivery of immune modulatory agents for optimal access to TDLN, aimed at overcoming hampered DC activation, will enable ICB by promoting T cell recruitment to the tumor, both in early and in advanced stages of cancer.
Collapse
Affiliation(s)
- Kim M. van Pul
- Department of Medical Oncology, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Marieke F. Fransen
- Deparment of Pulmonary Diseases Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Rieneke van de Ven
- Department of Otolaryngology/Head-Neck Surgery, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Tanja D. de Gruijl
- Department of Medical Oncology, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
12
|
van Pul KM, Vuylsteke RJCLM, de Beijer MTA, van de Ven R, van den Tol MP, Stockmann HBAC, de Gruijl TD. Breast cancer-induced immune suppression in the sentinel lymph node is effectively countered by CpG-B in conjunction with inhibition of the JAK2/STAT3 pathway. J Immunother Cancer 2020; 8:jitc-2020-000761. [PMID: 33046620 PMCID: PMC7552844 DOI: 10.1136/jitc-2020-000761] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND We previously showed selectively hampered activation of lymph node-resident (LNR) dendritic cell (DC) subsets in the breast cancer (BrC) sentinel lymph node (SLN) to precede a state of profound T cell anergy. Reactivating these DC subsets by intratumoral delivery of the Toll-like receptor-9 (TLR9) agonist CpG-B could potentially offer a promising immune therapeutic strategy to combat this immune suppression and prevent disease spread. Unfortunately, CpG-B can limit its own immune stimulatory activity through direct TLR9-mediated activation of signal transducer and activator of transcription 3 (STAT3), pinpointed as a key regulator of immune suppression in the tumor microenvironment. Here, we have investigated whether in vitro exposure to CpG-B, with or without simultaneous inhibition of STAT3 signaling, could overcome immune suppression in BrC SLN. METHODS Immune modulatory effects of CpG-B (CPG7909) with or without the JAK2/STAT3 inhibitor (STAT3i) AG490 were assessed in ex vivo cultured BrC SLN-derived single-cell suspensions (N=29). Multiparameter flow cytometric analyses were conducted for DC and T cell subset characterization and assessment of (intracellular) cytokine profiles. T cell reactivity against the BrC-associated antigen Mammaglobin-A was determined by means of interferon-γ ELISPOT assay. RESULTS Although CpG-B alone induced activation of all DC subsets, combined inhibition of the JAK2/STAT3 pathway resulted in superior DC maturation (ie, increased CD83 expression), with most profound activation and maturation of LNR DC subsets. Furthermore, combined CpG-B and JAK2/STAT3 inhibition promoted Th1 skewing by counterbalancing the CpG-induced Th2/regulatory T cell response and significantly enhanced Mammaglobin-A specific T cell reactivity. CONCLUSION Ex vivo immune modulation of the SLN by CpG-B and simultaneous JAK2/STAT3 inhibition can effectively overcome BrC-induced immune suppression by preferential activation of LNR DC, ultimately restoring type 1-mediated antitumor immunity, thereby securing a BrC-specific T cell response. These findings provide a clear rationale for clinical exploration of SLN-immune potentiation through local CpG/STAT3i administration in patients with BrC.
Collapse
Affiliation(s)
- Kim M van Pul
- Medical Oncology-Cancer Center Amsterdam, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands.,Surgical Oncology, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands
| | | | - Monique T A de Beijer
- Medical Oncology-Cancer Center Amsterdam, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands
| | - Rieneke van de Ven
- Medical Oncology and Otolaryngology-Head and Neck Surgery-Cancer Center Amsterdam, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands
| | | | | | - Tanja D de Gruijl
- Medical Oncology-Cancer Center Amsterdam, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Cancel JC, Crozat K, Dalod M, Mattiuz R. Are Conventional Type 1 Dendritic Cells Critical for Protective Antitumor Immunity and How? Front Immunol 2019; 10:9. [PMID: 30809220 PMCID: PMC6379659 DOI: 10.3389/fimmu.2019.00009] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/04/2019] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) are endowed with a unique potency to prime T cells, as well as to orchestrate their expansion, functional polarization and effector activity in non-lymphoid tissues or in their draining lymph nodes. The concept of harnessing DC immunogenicity to induce protective responses in cancer patients was put forward about 25 years ago and has led to a multitude of DC-based vaccine trials. However, until very recently, objective clinical responses were below expectations. Conventional type 1 DCs (cDC1) excel in the activation of cytotoxic lymphocytes including CD8+ T cells (CTLs), natural killer (NK) cells, and NKT cells, which are all critical effector cell types in antitumor immunity. Efforts to investigate whether cDC1 might orchestrate immune defenses against cancer are ongoing, thanks to the recent blossoming of tools allowing their manipulation in vivo. Here we are reporting on these studies. We discuss the mouse models used to genetically deplete or manipulate cDC1, and their main caveats. We present current knowledge on the role of cDC1 in the spontaneous immune rejection of tumors engrafted in syngeneic mouse recipients, as a surrogate model to cancer immunosurveillance, and how this process is promoted by type I interferon (IFN-I) effects on cDC1. We also discuss cDC1 implication in promoting the protective effects of immunotherapies in mouse preclinical models, especially for adoptive cell transfer (ACT) and immune checkpoint blockers (ICB). We elaborate on how to improve this process by in vivo reprogramming of certain cDC1 functions with off-the-shelf compounds. We also summarize and discuss basic research and clinical data supporting the hypothesis that the protective antitumor functions of cDC1 inferred from mouse preclinical models are conserved in humans. This analysis supports potential applicability to cancer patients of the cDC1-targeting adjuvant immunotherapies showing promising results in mouse models. Nonetheless, further investigations on cDC1 and their implications in anti-cancer mechanisms are needed to determine whether they are the missing key that will ultimately help switching cold tumors into therapeutically responsive hot tumors, and how precisely they mediate their protective effects.
Collapse
Affiliation(s)
- Jean-Charles Cancel
- CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Aix Marseille University, Marseille, France
| | - Karine Crozat
- CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Aix Marseille University, Marseille, France
| | - Marc Dalod
- CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Aix Marseille University, Marseille, France
| | - Raphaël Mattiuz
- CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Aix Marseille University, Marseille, France
| |
Collapse
|
14
|
Bai R, Chen N, Cui J. [Study Progression on Non-small Cell Lung Cancer with EGFR Mutation
Treated by Immune Checkpoint Inhibitors]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2018; 21:641-648. [PMID: 30172273 PMCID: PMC6105349 DOI: 10.3779/j.issn.1009-3419.2018.08.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In recent years, epidermal growth factor receptor tyrosine kinase inhibitors have been recommended by many guidelines as first-line drugs for advanced non-small cell lung cancer (NSCLC) with EGFR gene mutations and no resistance. However, with the prolongation of medication time, most appear acquired resistance. In recent years, breakthroughs in inhibitors of programmed death-1 (PD-1) and its ligand (PD1 ligand, PD-L1) have rapidly changed the therapeutic model of NSCLC. Recent studies have shown that the efficacy of immune checkpoint inhibitors in EGFR-mutant NSCLC patients is not satisfactory, which might be caused by low PD-L1 expression, inhibitory immune microenvironment and low tumor mutation load. This review will elaborate the immune microenvironment of NSCLC patients with EGFR mutation, the latest study progression of immune checkpoint inhibitors and its combined with TKI, expecting to bring new hopes for the treatment of EGFR-mutant NSCLC patients.
.
Collapse
Affiliation(s)
- Rilan Bai
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China
| | - Naifei Chen
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China
| | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
15
|
Koster BD, van den Hout MFCM, Sluijter BJR, Molenkamp BG, Vuylsteke RJCLM, Baars A, van Leeuwen PAM, Scheper RJ, Petrousjka van den Tol M, van den Eertwegh AJM, de Gruijl TD. Local Adjuvant Treatment with Low-Dose CpG-B Offers Durable Protection against Disease Recurrence in Clinical Stage I-II Melanoma: Data from Two Randomized Phase II Trials. Clin Cancer Res 2018; 23:5679-5686. [PMID: 28972083 DOI: 10.1158/1078-0432.ccr-17-0944] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/12/2017] [Accepted: 07/18/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Although risk of recurrence after surgical removal of clinical stage I-II melanoma is considerable, there is no adjuvant therapy with proven efficacy. Here, we provide clinical evidence that a local conditioning regimen, aimed at immunologic arming of the tumor-draining lymph nodes, may provide durable protection against disease recurrence (median follow-up, 88.8 months).Experimental Design: In two randomized phase II trials, patients, diagnosed with stage I-II melanoma after excision of the primary tumor, received local injections at the primary tumor excision site within 7 days preceding re-excision and sentinel lymph node (SLN) biopsy of either a saline placebo (n = 22) or low-dose CpG type B (CpG-B) with (n = 9) or without (n = 21) low-dose GM-CSF.Results: CpG-B treatment was shown to be safe, to boost locoregional and systemic immunity, to be associated with lower rates of tumor-involved SLN (10% vs. 36% in controls, P = 0.04), and, at a median follow-up of 88.8 months, to profoundly improve recurrence-free survival (P = 0.008), even for patients with histologically confirmed (i.e., pathologic) stage I-II disease (P = 0.02).Conclusions: Potentially offering durable protection, local low-dose CpG-B administration in early-stage melanoma provides an adjuvant treatment option for a large group of patients currently going untreated despite being at considerable risk for disease recurrence. Once validated in a larger randomized phase III trial, this nontoxic immunopotentiating regimen may prove clinically transformative. Clin Cancer Res; 23(19); 5679-86. ©2017 AACR.
Collapse
Affiliation(s)
- Bas D Koster
- Department of Medical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Mari F C M van den Hout
- Department of Pathology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Berbel J R Sluijter
- Department of Surgical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Barbara G Molenkamp
- Department of Surgical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Ronald J C L M Vuylsteke
- Department of Surgical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Arnold Baars
- Department of Pathology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Paul A M van Leeuwen
- Department of Surgical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Rik J Scheper
- Department of Pathology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - M Petrousjka van den Tol
- Department of Surgical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Alfons J M van den Eertwegh
- Department of Medical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
16
|
Yuan S, Xu H, Xie J. [New Progress in the Relationship between Immune Cells, PD-1
in Tumor Microenvironment and the Efficacy of EGFR-TKIs]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2018; 20:775-780. [PMID: 29167008 PMCID: PMC5973281 DOI: 10.3779/j.issn.1009-3419.2017.11.09] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In recent years, targeted therapy of epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKIs) is the leading treatment modality for patients with advanced non-small cell lung cancer (NSCLC) and EGFR gene mutation. However, with the prolongation of the medication time, most of the patients appeared drug resistance. Tumor microenvironment is the internal environment for the survival and development of tumor cells. The immune response which mediated by immune cells, like regulatory T (Treg), dendritic cells, macrophages, fibroblasts, etc. And the programmed cell death receptor 1 (PD-1) with its ligand PD-1L/PD-2L may participate in the drug resistance of EGFR-TKIs. This review will elaborate the possible mechanism of the interaction of immune cells on EGFR-TKIs in the tumor microenvironment, in order to seek new targets, and further improve the anti-tumor efficacy and prolong the effective time of EGFR-TKIs.
Collapse
Affiliation(s)
- Shiyang Yuan
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Nanchang University,
Nanchang 330006, China
| | - Hui Xu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Nanchang University,
Nanchang 330006, China
| | - Junping Xie
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Nanchang University,
Nanchang 330006, China
| |
Collapse
|
17
|
Di Gennaro P, Gerlini G, Caporale R, Sestini S, Brandani P, Urso C, Pimpinelli N, Borgognoni L. T regulatory cells mediate immunosuppresion by adenosine in peripheral blood, sentinel lymph node and TILs from melanoma patients. Cancer Lett 2018; 417:124-130. [PMID: 29306022 DOI: 10.1016/j.canlet.2017.12.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/18/2017] [Accepted: 12/21/2017] [Indexed: 01/23/2023]
Abstract
T regulatory cells (Tregs), involved in tumour tolerance, can generate Adenosine by CD39/CD73 surface enzymes, which identify four Tregs subsets: CD39+CD73- nTregs, CD39+CD73+ iTregs, CD39-CD73+ oTregs and CD39-CD73- xTregs. In melanoma patients, increased Tregs levels are detected in peripheral blood (PB), sentinel lymph node (SLN) and tumour infiltrating lymphocytes (TILs), but Adenosine role was not investigated yet. We examined total Tregs and Adenosine subsets in PB, SLN and TILs from melanoma patients (n = 32) and PB from healthy donors (HD; n = 10) by flow cytometry. Total Tregs significantly increased in stage III-IV patients PB, in SLN and TILs, as compared to HD/stage I-II patients. Tregs subsets analyses showed that: 1) PB nTregs significantly increased in SLN and decreased in TILs; 2) iTregs significantly increased in stage III-IV patients PB and further significantly increased in SLN and TILs; 3) PB oTregs and xTregs significantly decreased in SLN and TILs. Patients clinical features did not significantly influence total Tregs, except SLN excision order. Results confirmed Tregs role in melanoma progression and indicate Adenosine generation as a novel escape mechanism, being nTregs and iTregs increased in PB/SLN/TILs.
Collapse
Affiliation(s)
- P Di Gennaro
- Plastic and Reconstructive Surgery Unit - Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Tuscan Tumour Institute (ITT) - Santa Maria Annunziata Hospital, Florence, Italy.
| | - G Gerlini
- Plastic and Reconstructive Surgery Unit - Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Tuscan Tumour Institute (ITT) - Santa Maria Annunziata Hospital, Florence, Italy
| | - R Caporale
- Central Laboratory, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - S Sestini
- Plastic and Reconstructive Surgery Unit - Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Tuscan Tumour Institute (ITT) - Santa Maria Annunziata Hospital, Florence, Italy
| | - P Brandani
- Plastic and Reconstructive Surgery Unit - Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Tuscan Tumour Institute (ITT) - Santa Maria Annunziata Hospital, Florence, Italy
| | - C Urso
- Dept. Anatomic Pathology - Dermatopathology Section, Santa Maria Annunziata Hospital, Florence, Italy
| | - N Pimpinelli
- Dept. Surgery and Translational Medicine, Dermatology Section, University of Florence, Italy
| | - L Borgognoni
- Plastic and Reconstructive Surgery Unit - Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Tuscan Tumour Institute (ITT) - Santa Maria Annunziata Hospital, Florence, Italy
| |
Collapse
|
18
|
Hargadon KM. Strategies to Improve the Efficacy of Dendritic Cell-Based Immunotherapy for Melanoma. Front Immunol 2017; 8:1594. [PMID: 29209327 PMCID: PMC5702020 DOI: 10.3389/fimmu.2017.01594] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022] Open
Abstract
Melanoma is a highly aggressive form of skin cancer that frequently metastasizes to vital organs, where it is often difficult to treat with traditional therapies such as surgery and radiation. In such cases of metastatic disease, immunotherapy has emerged in recent years as an exciting treatment option for melanoma patients. Despite unprecedented successes with immune therapy in the clinic, many patients still experience disease relapse, and others fail to respond at all, thus highlighting the need to better understand factors that influence the efficacy of antitumor immune responses. At the heart of antitumor immunity are dendritic cells (DCs), an innate population of cells that function as critical regulators of immune tolerance and activation. As such, DCs have the potential to serve as important targets and delivery agents of cancer immunotherapies. Even immunotherapies that do not directly target or employ DCs, such as checkpoint blockade therapy and adoptive cell transfer therapy, are likely to rely on DCs that shape the quality of therapy-associated antitumor immunity. Therefore, understanding factors that regulate the function of tumor-associated DCs is critical for optimizing both current and future immunotherapeutic strategies for treating melanoma. To this end, this review focuses on advances in our understanding of DC function in the context of melanoma, with particular emphasis on (1) the role of immunogenic cell death in eliciting tumor-associated DC activation, (2) immunosuppression of DC function by melanoma-associated factors in the tumor microenvironment, (3) metabolic constraints on the activation of tumor-associated DCs, and (4) the role of the microbiome in shaping the immunogenicity of DCs and the overall quality of anti-melanoma immune responses they mediate. Additionally, this review highlights novel DC-based immunotherapies for melanoma that are emerging from recent progress in each of these areas of investigation, and it discusses current issues and questions that will need to be addressed in future studies aimed at optimizing the function of melanoma-associated DCs and the antitumor immune responses they direct against this cancer.
Collapse
Affiliation(s)
- Kristian M. Hargadon
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Hampden-Sydney, VA, United States
| |
Collapse
|
19
|
Inoue S, Setoyama Y, Odaka A, Kitagawa D, Beck Y. Chemoimmunotherapeutic effect of combined treatment with ex vivo generated antigen-presenting immune cells and conventional antitumor agents in a mouse neuroblastoma model. J Pediatr Surg 2017; 52:1642-1650. [PMID: 28457491 DOI: 10.1016/j.jpedsurg.2017.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/14/2017] [Accepted: 04/12/2017] [Indexed: 01/21/2023]
Abstract
PURPOSE Combining antitumor immunotherapy with conventional intensive multimodal therapy may be considered for advanced neuroblastoma. We investigated combination therapy with ex vivo generated immunostimulatory cells and intraperitoneal doxorubicin. METHODS Immunogenic death of neuro-2a neuroblastoma cells was induced by doxorubicin or cisplatin (negative control). Mouse bone marrow cells were cultured with granulocyte-macrophage colony-stimulating factor, followed by addition of doxorubicin-killed neuro-2a cells with or without interleukin-4 and/or CpG-oligodeoxynucleotide to induce immunostimulatory cells. CD8α+ lymphocytes were cocultured with killed neuro-2a cells and immunostimulatory cells, and interferon-γ was measured in the supernatant. Furthermore, female A/J mice were injected with viable neuro-2a cells, followed by immunostimulatory cells and doxorubicin. Then intraabdominal tumor nodules were evaluated. RESULTS Bone marrow-derived immunostimulatory cells only promoted interferon-γ production by CD8α+ lymphocytes after first being stimulated by doxorubicin-killed neuro-2a cells and interleukin-4, followed by CpG-oligodeoxynucleotide. These cells had a surface antigen expression profile compatible with activated dendritic cells and suppressed tumors in mice intravenously injected with neuro-2a cells. Despite a similar surface antigen profile, the in vivo antitumor effect was stronger after injection of immunostimulatory cells induced by doxorubicin-killed neuro-2a cells compared with cells induced by cisplatin-killed neuro-2a cells. Moreover, interferon-γ production was greater when CD8α+ lymphocytes were cocultured with doxorubicin-killed neuro-2a cells and immunostimulatory cells rather than with cisplatin-killed cells. CONCLUSION Cells with antitumor activity can be induced from bone marrow cells. Combining such cells with doxorubicin may activate antitumor immunity in tumor-bearing mice. Interactions between induced immunostimulatory cells and conventional chemotherapy might be important for antitumor immunity.
Collapse
Affiliation(s)
- Seiichiro Inoue
- Department of Hepato-Biliary-Pancreatic and Pediatric Surgery, Saitama Medical Center, Saitama Medical University, Kamoda 1981, Kawagoe, Saitama, 3508550, Japan.
| | - Yumiko Setoyama
- Department of Medical Research, Saitama Medical Center, Saitama Medical University, Japan
| | - Akio Odaka
- Department of Hepato-Biliary-Pancreatic and Pediatric Surgery, Saitama Medical Center, Saitama Medical University, Kamoda 1981, Kawagoe, Saitama, 3508550, Japan
| | - Daiki Kitagawa
- Department of Hepato-Biliary-Pancreatic and Pediatric Surgery, Saitama Medical Center, Saitama Medical University, Kamoda 1981, Kawagoe, Saitama, 3508550, Japan
| | - Yoshifumi Beck
- Department of Hepato-Biliary-Pancreatic and Pediatric Surgery, Saitama Medical Center, Saitama Medical University, Kamoda 1981, Kawagoe, Saitama, 3508550, Japan
| |
Collapse
|
20
|
van Dinther D, Stolk DA, van de Ven R, van Kooyk Y, de Gruijl TD, den Haan JMM. Targeting C-type lectin receptors: a high-carbohydrate diet for dendritic cells to improve cancer vaccines. J Leukoc Biol 2017; 102:1017-1034. [PMID: 28729358 PMCID: PMC5597514 DOI: 10.1189/jlb.5mr0217-059rr] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/13/2017] [Accepted: 06/16/2017] [Indexed: 12/23/2022] Open
Abstract
There is a growing understanding of why certain patients do or do not respond to checkpoint inhibition therapy. This opens new opportunities to reconsider and redevelop vaccine strategies to prime an anticancer immune response. Combination of such vaccines with checkpoint inhibitors will both provide the fuel and release the brake for an efficient anticancer response. Here, we discuss vaccine strategies that use C-type lectin receptor (CLR) targeting of APCs, such as dendritic cells and macrophages. APCs are a necessity for the priming of antigen-specific cytotoxic and helper T cells. Because CLRs are natural carbohydrate-recognition receptors highly expressed by multiple subsets of APCs and involved in uptake and processing of Ags for presentation, these receptors seem particularly interesting for targeting purposes.
Collapse
Affiliation(s)
- Dieke van Dinther
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Dorian A Stolk
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Rieneke van de Ven
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Tanja D de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Joke M M den Haan
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| |
Collapse
|
21
|
Abstract
Cancer immunotherapy can successfully promote long-term anticancer immune responses, although there is still only a limited number of patients who benefit from such treatment, and it can sometimes have severe treatment-associated adverse events. Compared with systemic immunomodulation, local immunomodulation may enable more effective treatment at lower doses and, at the same time, prevent systemic toxicity. Local delivery of engineered three-dimensional scaffolds may fulfil this role by acting as synthetic immune niches that boost anticancer immunity. In this Opinion article, we highlight the potential of scaffold-based adoptive cell transfer and scaffold-based cancer vaccines that, although applied locally, can promote systemic antitumour immunity. Furthermore, we discuss how scaffold-based cancer immunotherapy may contribute to the development of the next generation of cancer treatments.
Collapse
|
22
|
Burcelin R. [Gut microbiota and immune crosstalk in metabolic disease]. Biol Aujourdhui 2017; 211:1-18. [PMID: 28682223 DOI: 10.1051/jbio/2017008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Indexed: 05/28/2023]
Abstract
The aim of the review is to discuss about the role played by the defence crosstalk between the gut microbiota and the intestinal immune system, in the development of metabolic disease focusing on obesity and diabetes. Starting from physiological and pathological stand points and based on the latest published data, this review is addressing how the concept of the hologenome theory of evolution can drive the fate of metabolic disease. The notion of "metabolic infection" to explain the "metabolic inflammation" is discussed. This imply comments about the process of bacterial translocation and impaired intestinal immune defense against commensals. Eventually this review sets the soil for personalized medicine. The monthly increase in the number of publications on the gut microbiota to intestinal immune defense and the control of metabolism demonstrate the importance of this field of investigation. The notion of commensal as "self or non-self" has to be reevaluated in the light of the current data. Furthermore, data demonstrate the major role played by short chain fatty acids, secondary bile acids, LPS, peptidoglycans, indole derivatives, and other bacteria-related molecules on the shaping of cells involved in the intestinal protection against commensals is now becoming a central player in the incidence of metabolic diseases. The literature demonstrates that the onset of metabolic diseases and some specific co-morbidities can be explained by a gut microbiota to intestinal immune system crosstalk. Therefore, one should now consider this avenue of investigation as a putative source of biomarkers and therapeutic targets to personalize the treatment of metabolic disease and its co-morbidities. Gut microbiota is considered as a major regulator of metabolic disease. This reconciles the notion of metabolic inflammation and the epidemic development of the disease. In addition to evidence showing that a specific gut microbiota characterizes patients with obesity, type 2 diabetes, and hepatic steatosis, the mechanisms causal to the disease could be related to the translocation of microbiota from the gut to the tissues, which induces inflammation. The mechanisms regulating such a process are based on the crosstalk between the gut microbiota and the host immune system. The hologenome theory of evolution supports this concept and implies that therapeutic strategies aiming to control glycemia should take into account both the gut microbiota and the host immune system. This review discusses the latest evidence regarding the bidirectional impact of the gut microbiota on host immune system crosstalk for the control of metabolic disease, hyperglycemia, and obesity. To avoid redundancies with the literature, we will focus our attention on the intestinal immune system, identifying evidence for the generation of novel therapeutic strategies, which could be based on the control of the translocation of gut bacteria to tissues. Such novel strategies should hamper the role played by gut microbiota dysbiosis on the development of metabolic inflammation. Recent evidence in rodents allows us to conclude that an impaired intestinal immune system characterizes and could be causal in the development of metabolic disease. The fine understanding of the molecular mechanisms should allow for the development of a first line of treatment for metabolic disease and its co-morbidities.
Collapse
Affiliation(s)
- Rémy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), 31024 Toulouse, France - Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1048, Hôpital Rangueil, 31400 Toulouse, France - Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), 31432 Toulouse Cedex 4, France
| |
Collapse
|
23
|
Garg AD, Vara Perez M, Schaaf M, Agostinis P, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Dendritic cell-based anticancer immunotherapy. Oncoimmunology 2017; 6:e1328341. [PMID: 28811970 DOI: 10.1080/2162402x.2017.1328341] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 05/05/2017] [Indexed: 12/11/2022] Open
Abstract
Dendritic cell (DC)-based vaccines against cancer have been extensively developed over the past two decades. Typically DC-based cancer immunotherapy entails loading patient-derived DCs with an appropriate source of tumor-associated antigens (TAAs) and efficient DC stimulation through a so-called "maturation cocktail" (typically a combination of pro-inflammatory cytokines and Toll-like receptor agonists), followed by DC reintroduction into patients. DC vaccines have been documented to (re)activate tumor-specific T cells in both preclinical and clinical settings. There is considerable clinical interest in combining DC-based anticancer vaccines with T cell-targeting immunotherapies. This reflects the established capacity of DC-based vaccines to generate a pool of TAA-specific effector T cells and facilitate their infiltration into the tumor bed. In this Trial Watch, we survey the latest trends in the preclinical and clinical development of DC-based anticancer therapeutics. We also highlight how the emergence of immune checkpoint blockers and adoptive T-cell transfer-based approaches has modified the clinical niche for DC-based vaccines within the wide cancer immunotherapy landscape.
Collapse
Affiliation(s)
- Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Monica Vara Perez
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Marco Schaaf
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Paris, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
24
|
Re: Detection of Micrometastases by Flow Cytometry in Sentinel Lymph Nodes from Patients with Renal Tumours. Eur Urol 2017; 71:691-692. [DOI: 10.1016/j.eururo.2017.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/04/2017] [Indexed: 12/19/2022]
|
25
|
Burcelin R. Gut microbiota and immune crosstalk in metabolic disease. Mol Metab 2016; 5:771-81. [PMID: 27617200 PMCID: PMC5004167 DOI: 10.1016/j.molmet.2016.05.016] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 05/24/2016] [Accepted: 05/31/2016] [Indexed: 12/29/2022] Open
Abstract
Background Gut microbiota is considered as a major regulator of metabolic disease. This reconciles the notion of metabolic inflammation and the epidemic development of the disease. In addition to evidence showing that a specific gut microbiota characterizes patients with obesity, type 2 diabetes, and hepatic steatosis, the mechanisms causal to the disease could be related to the translocation of microbiota from the gut to the tissues, inducing inflammation. The mechanisms regulating such a process are based on the crosstalk between the gut microbiota and the host immune system. The hologenome theory of evolution supports this concept and implies that therapeutic strategies aiming to control glycemia should take into account both the gut microbiota and the host immune system. Scope of review This review discusses the latest evidence regarding the bidirectional impact of the gut microbiota on host immune system crosstalk for the control of metabolic disease, hyperglycemia, and obesity. To avoid redundancies with the literature, we will focus our attention on the intestinal immune system, identifying evidence for the generation of novel therapeutic strategies, which could be based on the control of the translocation of gut bacteria to tissues. Such novel strategies should hamper the role played by gut microbiota dysbiosis on the development of metabolic inflammation. Major conclusions Recent evidence in rodents allows us to conclude that an impaired intestinal immune system characterizes and could be causal in the development of metabolic disease. The fine understanding of the molecular mechanisms should allow for the development of a first line of treatment for metabolic disease and its co-morbidities. This article is part of a special issue on microbiota.
Collapse
Affiliation(s)
- Rémy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), F-31432 Toulouse Cedex 4, France
- Inserm 1048, Hôpital Rangueil, 31400 Toulouse, France. Tel.: +33 561 32 56 14; fax: +33 561 32 56 21.Inserm 1048Hôpital RangueilToulouse31400France
| |
Collapse
|