1
|
Dai SL, Pan JQ, Su ZR. Multi-omics features of immunogenic cell death in gastric cancer identified by combining single-cell sequencing analysis and machine learning. Sci Rep 2024; 14:21751. [PMID: 39294296 PMCID: PMC11410816 DOI: 10.1038/s41598-024-73071-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024] Open
Abstract
Gastric cancer (GC) is a prevalent malignancy with high mortality rates. Immunogenic cell death (ICD) is a unique form of programmed cell death that is closely linked to antitumor immunity and plays a critical role in modulating the tumor microenvironment (TME). Nevertheless, elucidating the precise effect of ICD on GC remains a challenging endeavour. ICD-related genes were identified in single-cell sequencing datasets and bulk transcriptome sequencing datasets via the AddModuleScore function, weighted gene co-expression network (WGCNA), and differential expression analysis. A robust signature associated with ICD was constructed using a machine learning computational framework incorporating 101 algorithms. Furthermore, multiomics analysis, including single-cell sequencing analysis, bulk transcriptomic analysis, and proteomics analysis, was conducted to verify the correlation of these hub genes with the immune microenvironment features of GC and with GC invasion and metastasis. We screened 59 genes associated with ICD and developed a robust ICD-related gene signature (ICDRS) via a machine learning computational framework that integrates 101 different algorithms. Furthermore, we identified five key hub genes (SMAP2, TNFAIP8, LBH, TXNIP, and PIK3IP1) from the ICDRS. Through single-cell analysis of GC tumor s, we confirmed the strong correlations of the hub genes with immune microenvironment features. Among these five genes, LBH exhibited the most significant associations with a poor prognosis and with the invasion and metastasis of GC. Finally, our findings were validated through immunohistochemical staining of a large clinical sample set, and the results further supported that LBH promotes GC cell invasion by activating the epithelial-mesenchymal transition (EMT) pathway.
Collapse
Affiliation(s)
- Shu-Long Dai
- Department of General Surgery, Deqing People's Hospital, Deqing Campus, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 120 South Yingxi Road, Deqing, 313200, Zhejiang, P. R. China.
| | - Jian-Qiang Pan
- Department of Pathology, Deqing People's Hospital, Deqing Campus, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 120 South Yingxi Road, Deqing, 313200, Zhejiang, P. R. China
| | - Zhen-Rong Su
- Department of General Surgery, Deqing People's Hospital, Deqing Campus, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 120 South Yingxi Road, Deqing, 313200, Zhejiang, P. R. China
| |
Collapse
|
2
|
Lee Y, Min J, Kim S, Park W, Ko J, Jeon NL. Recapitulating the Cancer-Immunity Cycle on a Chip. Adv Healthc Mater 2024:e2401927. [PMID: 39221688 DOI: 10.1002/adhm.202401927] [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: 05/24/2024] [Revised: 08/11/2024] [Indexed: 09/04/2024]
Abstract
The cancer-immunity cycle is a fundamental framework for understanding how the immune system interacts with cancer cells, balancing T cell recognition and elimination of tumors while avoiding autoimmune reactions. Despite advancements in immunotherapy, there remains a critical need to dissect each phase of the cycle, particularly the interactions among the tumor, vasculature, and immune system within the tumor microenvironment (TME). Innovative platforms such as organ-on-a-chip, organoids, and bioprinting within microphysiological systems (MPS) are increasingly utilized to enhance the understanding of these interactions. These systems meticulously replicate crucial aspects of the TME and immune responses, providing robust platforms to study cancer progression, immune evasion, and therapeutic interventions with greater physiological relevance. This review explores the latest advancements in MPS technologies for modeling various stages of the cancer-immune cycle, critically evaluating their applications and limitations in advancing the understanding of cancer-immune dynamics and guiding the development of next-generation immunotherapeutic strategies.
Collapse
Affiliation(s)
- Yujin Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jaehong Min
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Solbin Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Wooju Park
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Jihoon Ko
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Noo Li Jeon
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Mechanical Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Advanced Machines and Design, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
- Qureator, Inc., San Diego, CA, 92110, USA
| |
Collapse
|
3
|
Conibear J. Rationale for concurrent chemoradiotherapy for patients with stage III non-small-cell lung cancer. Br J Cancer 2020; 123:10-17. [PMID: 33293671 PMCID: PMC7735212 DOI: 10.1038/s41416-020-01070-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
When treating patients with unresectable stage III non-small-cell lung cancer (NSCLC), those with a good performance status and disease measured within a radical treatment volume should be considered for definitive concurrent chemoradiotherapy (cCRT). This guidance is based on key scientific rationale from two large Phase 3 randomised studies and meta-analyses demonstrating the superiority of cCRT over sequential (sCRT). However, the efficacy of cCRT comes at the cost of increased acute toxicity versus sequential treatment. Currently, there are several documented approaches that are addressing this drawback, which this paper outlines. At the point of diagnosis, a multidisciplinary team (MDT) approach can enable accurate assessment of patients, to determine the optimal treatment strategy to minimise risks. In addition, reviewing the Advisory Committee on Radiation Oncology Practice (ACROP) guidelines can provide clinical oncologists with additional recommendations for outlining target volume and organ-at-risk delineation for standard clinical scenarios in definitive cCRT (and adjuvant radiotherapy). Furthermore, modern advances in radiotherapy treatment planning software and treatment delivery mean that radiation oncologists can safely treat substantially larger lung tumours with higher radiotherapy doses, with greater accuracy, whilst minimising the radiotherapy dose to the surrounding healthy tissues. The combination of these advances in cCRT may assist in creating comprehensive strategies to allow patients to receive potentially curative benefits from treatments such as immunotherapy, as well as minimising treatment-related risks.
Collapse
Affiliation(s)
- John Conibear
- Department of Clinical Oncology, St. Bartholomew's Hospital, London, UK.
| |
Collapse
|
4
|
Ishimwe N, Zhang W, Qian J, Zhang Y, Wen L. Autophagy regulation as a promising approach for improving cancer immunotherapy. Cancer Lett 2020; 475:34-42. [PMID: 32014460 DOI: 10.1016/j.canlet.2020.01.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023]
Abstract
Autophagy plays a critical role in intracellular metabolism and maintaining cellular homeostasis. Certain tumor cells present a higher basal autophagy rate and autophagy inhibition can lead to impaired metabolic dysfunction in autophagy-dependent tumor cells. Autophagy status in immune cells dictates their fate and response to antigen; however, autophagy in immune cells may be beneficial or detrimental depending on the developmental stage of the cell and more specifically its degree of differentiation. Autophagy-deficient hosts present variations in many metabolites, proteins and enzymes that may have tumor-promoting or -inhibiting effects. The centrality of autophagy in the metabolism of some cancers and immune cells poses as a critical target whose mechanisms must be further unraveled to optimize patient response and prevent tumor recurrence.
Collapse
Affiliation(s)
- Nestor Ishimwe
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, PR China; Guangzhou First People's Hospital, School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, PR China.
| | - Wenbin Zhang
- Guangzhou First People's Hospital, School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, PR China
| | - Jieying Qian
- Guangzhou First People's Hospital, School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, PR China
| | - Yunjiao Zhang
- Guangzhou First People's Hospital, School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, Guangdong, 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, And Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, Guangdong, 510006, PR China.
| | - Longping Wen
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, PR China; Guangzhou First People's Hospital, School of Medicine and Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, Guangdong, 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, And Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, Guangdong, 510006, PR China.
| |
Collapse
|
5
|
Subbiah V, Murthy R, Hong DS, Prins RM, Hosing C, Hendricks K, Kolli D, Noffsinger L, Brown R, McGuire M, Fu S, Piha-Paul S, Naing A, Conley AP, Benjamin RS, Kaur I, Bosch ML. Cytokines Produced by Dendritic Cells Administered Intratumorally Correlate with Clinical Outcome in Patients with Diverse Cancers. Clin Cancer Res 2018; 24:3845-3856. [PMID: 30018119 DOI: 10.1158/1078-0432.ccr-17-2707] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/05/2018] [Accepted: 05/07/2018] [Indexed: 01/08/2023]
Abstract
Purpose: Dendritic cells (DC) initiate adaptive immune responses through the uptake and presentation of antigenic material. In preclinical studies, intratumorally injected activated DCs (aDCs; DCVax-Direct) were superior to immature DCs in rejecting tumors from mice.Experimental Design: This single-arm, open-label phase I clinical trial evaluated the safety and efficacy of aDCs, administered intratumorally, in patients with solid tumors. Three dose levels (2 million, 6 million, and 15 million aDCs per injection) were tested using a standard 3 + 3 dose-escalation trial design. Feasibility, immunogenicity, changes to the tumor microenvironment after direct injection, and survival were evaluated. We also investigated cytokine production of aDCs prior to injection.Results: In total, 39 of the 40 enrolled patients were evaluable. The injections of aDCs were well tolerated with no dose-limiting toxicities. Increased lymphocyte infiltration was observed in 54% of assessed patients. Stable disease (SD; best response) at week 8 was associated with increased overall survival. Increased secretion of interleukin (IL)-8 and IL12p40 by aDCs was significantly associated with survival (P = 0.023 and 0.024, respectively). Increased TNFα levels correlated positively with SD at week 8 (P < 0.01).Conclusions: Intratumoral aDC injections were feasible and safe. Increased production of specific cytokines was correlated with SD and prolonged survival, demonstrating a link between the functional profile of aDCs prior to injection and patient outcomes. Clin Cancer Res; 24(16); 3845-56. ©2018 AACR.
Collapse
Affiliation(s)
- Vivek Subbiah
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ravi Murthy
- Department of Interventional Radiology, Division of Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David S Hong
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert M Prins
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Chitra Hosing
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | - Robert Brown
- Department of Pathology and Laboratory Medicine, UT Health, University of Texas Health Science Center, Houston, Texas
| | - Mary McGuire
- Department of Pathology and Laboratory Medicine, UT Health, University of Texas Health Science Center, Houston, Texas
| | - Siquing Fu
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarina Piha-Paul
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anthony P Conley
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert S Benjamin
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Indreshpal Kaur
- Cell Therapy Labs, GMP Laboratory, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
6
|
Sánchez-Paulete AR, Teijeira A, Cueto FJ, Garasa S, Pérez-Gracia JL, Sánchez-Arráez A, Sancho D, Melero I. Antigen cross-presentation and T-cell cross-priming in cancer immunology and immunotherapy. Ann Oncol 2018; 28:xii44-xii55. [PMID: 28945841 DOI: 10.1093/annonc/mdx237] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) are the main professional antigen-presenting cells for induction of T-cell adaptive responses. Cancer cells express tumor antigens, including neoantigens generated by nonsynonymous mutations, but are poor for antigen presentation and for providing costimulatory signals for T-cell priming. Mounting evidence suggests that antigen transfer to DCs and their surrogate presentation on major histocompatibility complex class I and II molecules together with costimulatory signals is paramount for induction of viral and cancer immunity. Of the great diversity of DCs, BATF3/IRF8-dependent conventional DCs type 1 (cDC1) excel at cross-presentation of tumor cell-associated antigens. Location of cDC1s in the tumor correlates with improved infiltration by CD8+ T cells and tumor-specific T-cell immunity. Indeed, cDC1s are crucial for antitumor efficacy using checkpoint inhibitors and anti-CD137 agonist monoclonal antibodies in mouse models. Enhancement and exploitation of T-cell cross-priming by cDC1s offer opportunities for improved cancer immunotherapy, including in vivo targeting of tumor antigens to internalizing receptors on cDC1s and strategies to increase their numbers, activation and priming capacity within tumors and tumor-draining lymph nodes.
Collapse
Affiliation(s)
- A R Sánchez-Paulete
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona
| | - A Teijeira
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona
| | - F J Cueto
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid.,Department of Biochemistry, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid
| | - S Garasa
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona
| | - J L Pérez-Gracia
- University Clinic, University of Navarra, Pamplona, Spain.,CIBERONC, Madrid, Spain
| | - A Sánchez-Arráez
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona
| | - D Sancho
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid
| | - I Melero
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona.,University Clinic, University of Navarra, Pamplona, Spain.,CIBERONC, Madrid, Spain
| |
Collapse
|
7
|
Yip K, Melcher A, Harrington K, Illidge T, Nobes J, Webster A, Smith D, Lorigan P, Nathan P, Larkin J. Pembrolizumab in Combination with Radiotherapy for Metastatic Melanoma - Introducing the PERM Trial. Clin Oncol (R Coll Radiol) 2018; 30:201-203. [PMID: 29402599 DOI: 10.1016/j.clon.2018.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 12/26/2022]
Affiliation(s)
- K Yip
- Ipswich Hospital, Ipswich, UK.
| | - A Melcher
- Institute of Cancer Research, Sutton, London, UK; The Royal Marsden Hospital, Chelsea, London, UK
| | - K Harrington
- Institute of Cancer Research, Sutton, London, UK; The Royal Marsden Hospital, Chelsea, London, UK
| | - T Illidge
- University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Withington, Manchester, UK
| | - J Nobes
- Norfolk and Norwich University Hospital, Norwich, UK
| | - A Webster
- Radiotherapy Trials Quality Assurance Group, Mount Vernon Cancer Centre, Northwood, UK
| | - D Smith
- The Royal Marsden Hospital, Chelsea, London, UK
| | - P Lorigan
- University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Withington, Manchester, UK
| | - P Nathan
- Mount Vernon Cancer Centre, Northwood, UK
| | - J Larkin
- The Royal Marsden Hospital, Chelsea, London, UK
| |
Collapse
|
8
|
Alfaro C, Sanmamed MF, Rodríguez-Ruiz ME, Teijeira Á, Oñate C, González Á, Ponz M, Schalper KA, Pérez-Gracia JL, Melero I. Interleukin-8 in cancer pathogenesis, treatment and follow-up. Cancer Treat Rev 2017; 60:24-31. [PMID: 28866366 DOI: 10.1016/j.ctrv.2017.08.004] [Citation(s) in RCA: 235] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/11/2017] [Accepted: 08/13/2017] [Indexed: 12/23/2022]
Abstract
Interleukin-8 (CXCL8) was originally described asa chemokine whose main function is the attraction of a polymorphonuclear inflammatory leukocyte infiltrate acting on CXCR1/2. Recently, it has been found that tumors very frequently coopt the production of this chemokine, which in this malignant context exerts different pro-tumoral functions. Reportedly, these include angiogenesis, survival signaling for cancer stem cells and attraction of myeloid cells endowed with the ability to immunosuppress and locally provide growth factors. Given the fact that in cancer patients IL-8 is mainly produced by tumor cells themselves, its serum concentration has been shown to correlate with tumor burden. Thus, IL-8 serum concentrations have been shown to be useful asa pharmacodynamic biomarker to early detect response to immunotherapy. Finally, because of the roles that IL-8 plays in favoring tumor progression, several therapeutic strategies are being developed to interfere with its functions. Such interventions hold promise, especially for therapeutic combinations in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Carlos Alfaro
- Immunology and Immunotherapy, Centre for Applied Medical Research (CIMA), Pamplona, Spain; Department of Oncology, University Clinic of Navarra, Pamplona, Spain; CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Spain.
| | - Miguel F Sanmamed
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Álvaro Teijeira
- Immunology and Immunotherapy, Centre for Applied Medical Research (CIMA), Pamplona, Spain; CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Spain
| | - Carmen Oñate
- Immunology and Immunotherapy, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Álvaro González
- CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Spain; Department of Biochemistry, University Clinic of Navarra, Pamplona, Spain
| | - Mariano Ponz
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain
| | - Kurt A Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - José L Pérez-Gracia
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain; CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Spain
| | - Ignacio Melero
- Immunology and Immunotherapy, Centre for Applied Medical Research (CIMA), Pamplona, Spain; Department of Oncology, University Clinic of Navarra, Pamplona, Spain; CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Spain.
| |
Collapse
|
9
|
Aznar MA, Tinari N, Rullán AJ, Sánchez-Paulete AR, Rodriguez-Ruiz ME, Melero I. Intratumoral Delivery of Immunotherapy-Act Locally, Think Globally. THE JOURNAL OF IMMUNOLOGY 2017; 198:31-39. [PMID: 27994166 DOI: 10.4049/jimmunol.1601145] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/31/2016] [Indexed: 12/16/2022]
Abstract
Immune mechanisms have evolved to cope with local entry of microbes acting in a confined fashion but eventually inducing systemic immune memory. Indeed, in situ delivery of a number of agents into tumors can mimic in the malignant tissue the phenomena that control intracellular infection leading to the killing of infected cells. Vascular endothelium activation and lymphocyte attraction, together with dendritic cell-mediated cross-priming, are the key elements. Intratumoral therapy with pathogen-associated molecular patterns or recombinant viruses is being tested in the clinic. Cell therapies can be also delivered intratumorally, including infusion of autologous dendritic cells and even tumor-reactive T lymphocytes. Intralesional virotherapy with an HSV vector expressing GM-CSF has been recently approved by the Food and Drug Administration for the treatment of unresectable melanoma. Immunomodulatory monoclonal Abs have also been successfully applied intratumorally in animal models. Local delivery means less systemic toxicity while focusing the immune response on the malignancy and the affected draining lymph nodes.
Collapse
Affiliation(s)
- M Angela Aznar
- Division of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona 31008, Spain
| | - Nicola Tinari
- Department of Experimental and Clinical Sciences, G. D'Annunzio University and Foundation, Chieti 66100, Italy
| | - Antonio J Rullán
- Department of Medical Oncology, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona 08908, Spain; and
| | - Alfonso R Sánchez-Paulete
- Division of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona 31008, Spain
| | - María E Rodriguez-Ruiz
- Division of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona 31008, Spain.,University Clinic, University of Navarra and Health Research Institute of Navarra, 31008 Pamplona, Spain
| | - Ignacio Melero
- Division of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona 31008, Spain; .,University Clinic, University of Navarra and Health Research Institute of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
10
|
Immunomodulatory Function of the Tumor Suppressor p53 in Host Immune Response and the Tumor Microenvironment. Int J Mol Sci 2016; 17:ijms17111942. [PMID: 27869779 PMCID: PMC5133937 DOI: 10.3390/ijms17111942] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/07/2016] [Accepted: 11/10/2016] [Indexed: 02/06/2023] Open
Abstract
The tumor suppressor p53 is the most frequently mutated gene in human cancers. Most of the mutations are missense leading to loss of p53 function in inducing apoptosis and senescence. In addition to these autonomous effects of p53 inactivation/dysfunction on tumorigenesis, compelling evidence suggests that p53 mutation/inactivation also leads to gain-of-function or activation of non-autonomous pathways, which either directly or indirectly promote tumorigenesis. Experimental and clinical results suggest that p53 dysfunction fuels pro-tumor inflammation and serves as an immunological gain-of-function driver of tumorigenesis via skewing immune landscape of the tumor microenvironment (TME). It is now increasingly appreciated that p53 dysfunction in various cellular compartments of the TME leads to immunosuppression and immune evasion. Although our understanding of the cellular and molecular processes that link p53 activity to host immune regulation is still incomplete, it is clear that activating/reactivating the p53 pathway in the TME also represents a compelling immunological strategy to reverse immunosuppression and enhance antitumor immunity. Here, we review our current understanding of the potential cellular and molecular mechanisms by which p53 participates in immune regulation and discuss how targeting the p53 pathway can be exploited to alter the immunological landscape of tumors for maximizing therapeutic outcome.
Collapse
|
11
|
D'Hooghe E, Buttiglieri S, Bisignano G, Brusa D, Camussi G, Matera L. Apoptic Renal Carcinoma Cells are Better Inducers of Cross-Presenting Activity than Their Primary Necrotic Counterpart. Int J Immunopathol Pharmacol 2016; 20:707-17. [DOI: 10.1177/039463200702000406] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Vaccination with tumor-loaded dendritic cells (DC) is a promising treatment strategy for patients with renal cell carcinoma (RCC). Cells undergoing cell death proved useful as a source of tumor antigen for DC loading. Both apoptotic and necrotic tumor cells have been shown to efficiently load RCC-tumor antigens on DC. However, no direct comparison of these two kinds of death has been attempted in the same RCC. We compared DC pulsed with apoptotic cells, whole cell lysates or their supernatants of the cell line K1, derived from a patient with clear cell RCC, to determine their ability to activate T cells. Monocyte-derived DCs were pulsed with the different sources of tumor antigen, matured and co-cultured with autologouos peripheral blood lymphocytes. After three weekly re-stimulations with DCs, generation of cytotoxic T lymphocytes CTL was assessed by IFN-γ release in an ELISpot assay in the presence of the sensitizing target. By comparison with lysate, apoptotic tumor cells induced a higher frequency of MHC class I-restricted IFN-γ releasing lymphocytes. A higher CTL response was induced by pulsing DCs with cell lysate supernatant compared with whole cell lysate. These results indicate that, although necrotic death has been regarded as highly permissive when compared to apoptotic death, the immunogenicity of the death treatment may vary from one tumor to another.
Collapse
Affiliation(s)
| | - S. Buttiglieri
- Centro per la Ricerca in Medicina Sperimentale (CeRMS), Turin
| | - G. Bisignano
- Laboratorio di Immunologia dei Tumori, Dipartimento di Medicina Interna, Turin University, Turin
| | - D. Brusa
- Laboratorio di Immunologia dei Tumori, Dipartimento di Medicina Interna, Turin University, Turin
| | - G. Camussi
- Dipartimento di Medicina Interna, Cattedra di Nefrologia, Turin University, Turin, Italy
| | - L. Matera
- Laboratorio di Immunologia dei Tumori, Dipartimento di Medicina Interna, Turin University, Turin
| |
Collapse
|
12
|
Guo G, Cui Y. New perspective on targeting the tumor suppressor p53 pathway in the tumor microenvironment to enhance the efficacy of immunotherapy. J Immunother Cancer 2015; 3:9. [PMID: 25806108 PMCID: PMC4372251 DOI: 10.1186/s40425-015-0053-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 02/26/2015] [Indexed: 12/17/2022] Open
Abstract
About 50% of human cancers harbor somatic mutations of the tumor suppressor p53 (p53 or Trp53) gene. Many of those mutations result in the inactivation of the p53 pathway and are often associated with the stabilization and accumulation of mutant p53 proteins. Therefore, increased p53 expression in tumors is frequently used as a surrogate marker for p53 mutation and inactivation. Moreover, this elevated p53 expression also makes it an ideal tumor associated antigen (TAA) for cancer vaccines. Recent advances in our understanding of p53 as a crucial transcription factor reveal that p53 is an important sensor of cellular stress under genotoxic, chemotoxic, pathological, and even normal physiological conditions. Experimental and clinical observations by our laboratory and others have demonstrated that p53 also participates in immune regulation as p53 dysfunction skews host immune responses towards pro-inflammation, which further promotes tumor progression. Furthermore, recent studies using a genetic approach revealed that p53-restoration or re-activation led to tumor regression and clearance, which were at least partially caused by the activation of innate antitumor immunity. Since many of the currently used cancer therapeutics, including radiotherapy and chemotherapy, disrupt tumor growth by inducing DNA damage via genotoxic or chemotoxic stress, which activates the p53 pathway in the tumor microenvironment, we postulate that some of those observed therapeutic benefits might also be partially mediated through their immune stimulatory effects. Here, we briefly review our current understanding of the potential cellular and molecular mechanisms by which p53 participates in immune regulation and, subsequently, extend our discussion to the immunostimulatory potential of existing and new approaches of targeting the p53-pathway to alter the immunological landscape of tumors for maximizing immunotherapy outcome.
Collapse
Affiliation(s)
- Gang Guo
- Department of Biochemistry and Molecular Biology, Cancer Immunology, Inflammation & Tolerance Program, Georgia Regents University Cancer Center, Augusta, GA 30912 USA
| | - Yan Cui
- Department of Biochemistry and Molecular Biology, Cancer Immunology, Inflammation & Tolerance Program, Georgia Regents University Cancer Center, Augusta, GA 30912 USA
| |
Collapse
|
13
|
Carcinoma-derived interleukin-8 disorients dendritic cell migration without impairing T-cell stimulation. PLoS One 2011; 6:e17922. [PMID: 21423807 PMCID: PMC3056721 DOI: 10.1371/journal.pone.0017922] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 02/17/2011] [Indexed: 01/14/2023] Open
Abstract
Background Interleukin-8 (IL-8, CXCL8) is readily produced by human malignant cells.
Dendritic cells (DC) both produce IL-8 and express the IL-8 functional
receptors CXCR1 and CXCR2. Most human colon carcinomas produce IL-8. IL-8
importance in malignancies has been ascribed to angiogeneis promotion. Principal Findings IL-8 effects on human monocyte-derived DC biology were explored upon DC
exposure to recombinant IL-8 and with the help of an IL-8 neutralizing mAb.
In vivo experiments were performed in immunodeficient
mice xenografted with IL-8-producing human colon carcinomas and
comparatively with cell lines that do not produce IL-8. Allogenic T
lymphocyte stimulation by DC was explored under the influence of IL-8. DC
and neutrophil chemotaxis were measured by transwell-migration assays. Sera
from tumor-xenografted mice contained increasing concentrations of IL-8 as
the tumors progress. IL-8 production by carcinoma cells can be modulated by
low doses of cyclophosphamide at the transcription level. If human DC are
injected into HT29 or CaCo2 xenografted tumors, DC are retained
intratumorally in an IL-8-dependent fashion. However, IL-8 did not modify
the ability of DC to stimulate T cells. Interestingly, pre-exposure of DC to
IL-8 desensitizes such cells for IL-8-mediated in vitro or
in vivo chemoattraction. Thereby DC become disoriented
to subsequently follow IL-8 chemotactic gradients towards malignant or
inflamed tissue. Conclusions IL-8 as produced by carcinoma cells changes DC migration cues, without
directly interfering with DC-mediated T-cell stimulation.
Collapse
|
14
|
Perez-Gracia JL, Berraondo P, Martinez-Forero I, Alfaro C, Suarez N, Gurpide A, Sangro B, Hervas-Stubbs S, Ochoa C, Melero JA, Melero I. Clinical development of combination strategies in immunotherapy: are we ready for more than one investigational product in an early clinical trial? Immunotherapy 2011; 1:845-53. [PMID: 20636027 DOI: 10.2217/imt.09.51] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Stimulating the innate and adaptive immunity against cancer necessitates the tricking of a system evolved to fight microbial pathogens and directing its activity towards transformed self-tissue. Efficacious interventions to start and sustain the response will probably require a number of agents to tamper simultaneously or sequentially with several immune mechanisms. Although master switches controlling various functions may exist, the goal of a curative immune response will probably demand the combined actions of several therapeutic components. Synergy occurs when drugs interact in ways that enhance or magnify one or more effects or side effects. In cancer immunotherapy, two agents that have minor or no therapeutic effects as single agents can be powerful when combined. Mouse experimentation provides multiple examples of synergistic combinations. Elements to be combined include chiefly: tumor vaccines, adoptive T-cell therapies, cytokines, costimulatory molecules, molecular deactivation of immunosuppressive or tolerogenic pathways and immunostimulatory monoclonal antibodies. These novel therapies, even as single agents, are extremely complex products to be developed owing to the associated biomolecules, cell therapies or gene therapies. At present, drug-development programs are run individually for each immunotherapeutic agent and combinations are considered only at a later stage in clinical development, even in the absence of formal compulsory regulations to prevent clinical trials with combinations. As a result, instead of the search for maximal efficacy, ease of combination with standard treatments, intellectual property management, regulations and business-based decisions often guide the way. Even though the maximal effort must be made in order to prevent adverse effects in patients, it seems reasonable that combination pilot trials should be performed at an early stage, following safe completion of Phase I trials. These trials should be performed based on evidence for synergy in animal models and be simplified in terms of regulatory requirements. Such 'short-cut' combination immunotherapy trials can bring much needed efficacy earlier to the bedside.
Collapse
Affiliation(s)
- Jose L Perez-Gracia
- Centro de Investigación Médica Aplicada y Clinica Universitaria. Universidad de Navarra, Avenida de Pio XII 55, 31008 Pamplona, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Dubrot J, Palazón A, Alfaro C, Azpilikueta A, Ochoa MC, Rouzaut A, Martinez-Forero I, Teijeira A, Berraondo P, Le Bon A, Hervás-Stubbs S, Melero I. Intratumoral injection of interferon-α and systemic delivery of agonist anti-CD137 monoclonal antibodies synergize for immunotherapy. Int J Cancer 2010; 128:105-18. [PMID: 20309938 DOI: 10.1002/ijc.25333] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
CD137 artificial costimulation results in complete tumor rejection in several mouse models. Type I interferons (IFN) exert antitumor effects through an array of molecular functions on malignant cells, tumor stroma and immune system cells. The fact that agonist anti-CD137 mAb induce tumor regressions in mice deficient in the unique receptor for Type I IFNs (IFNAR(-/-) ) indicated potential for treatment combinations. Indeed, combination of intratumor injections of mouse IFN-α and intraperitoneal injections of anti-CD137 mAb synergized as seen on subcutaneous lesions derived from the MC38 colon carcinoma, which is resistant to each treatment if given separately. Therapeutic activity was achieved both against lesions directly injected with IFN-α and against distant concomitant tumors. Experiments in bone marrow chimeras prepared with IFNAR(-/-) and WT mice concluded that expression of the receptor for Type I interferons is mainly required on cells of the hematopoietic compartment. Synergistic effects correlated with a remarkable cellular hyperplasia of the tumor draining lymph nodes (TDLNs). Enlarged TDLNs contained more plasmacytoid and conventional dendritic cells (DC) that more readily cross-presented. Importantly, numbers of both DC subtypes inversely correlated with the tumor size. Numbers of CD8 T cells specific for a dominant tumor antigen were increased at TDLNs by each separate treatment but only with slight augments due to the combination. Combined antitumor effects of the therapeutic strategy were also seen on subcutaneous TC-1 tumors established for 24 days before treatment onset. The described strategy is realistic because (i) agents of each kind are clinically available and (ii) equivalent procedures in humans are feasible.
Collapse
Affiliation(s)
- Juan Dubrot
- CIMA and Clinica Universitaria, Universidad de Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Naing A, Kurzrock R. Chemotherapy resistance and retreatment: a dogma revisited. Clin Colorectal Cancer 2010; 9:E1-4. [PMID: 20378496 DOI: 10.3816/ccc.2010.n.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The prevailing belief in cancer therapy is that retreatment with a given drug after the emergence of resistance is ineffective. Herein, we report several cases in which retreatment with drugs that had failed earlier in the disease trajectory caused renewed tumor regression. The possible biologic underpinnings that might explain this phenomenon are discussed.
Collapse
Affiliation(s)
- Aung Naing
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas M. D. Anderson Cancer Center, Houston
| | | |
Collapse
|
17
|
Murillo O, Dubrot J, Palazón A, Arina A, Azpilikueta A, Alfaro C, Solano S, Ochoa MC, Berasain C, Gabari I, Pérez-Gracia JL, Berraondo P, Hervás-Stubbs S, Melero I. In vivo depletion of DC impairs the anti-tumor effect of agonistic anti-CD137 mAb. Eur J Immunol 2009; 39:2424-36. [PMID: 19662633 DOI: 10.1002/eji.200838958] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Anti-CD137 mAb are capable of inducing tumor rejection in several syngeneic murine tumor models and are undergoing clinical trials for cancer. The anti-tumor effect involves co-stimulation of tumor-specific CD8(+) T cells. Whether antigen cross-presenting DC are required for the efficacy of anti-CD137 mAb treatment has never been examined. Here we show that the administration of anti-CD137 mAb eradicates EG7-OVA tumors by a strictly CD8beta(+) T-cell-dependent mechanism that correlates with increased CTL activity. Ex vivo analyses to determine the identity of the draining lymph node cell type responsible for tumor antigen cross-presentation revealed that CD11c(+) cells, most likely DC, are the main players in this tumor model. A minute number of tumor cells, revealed by the presence of OVA cDNA, reach tumor-draining lymph nodes. Direct antigen presentation by tumor cells themselves also participates in anti-OVA CTL induction. Using CD11c diphtheria toxin receptor-green fluorescent protein-->C57BL/6 BM chimeric mice, which allow for sustained ablation of DC with diphtheria toxin, we confirmed the involvement of DC in tumor antigen cross-presentation in CTL induction against OVA(257-264) epitope and in the antitumor efficacy induced by anti-CD137 mAb.
Collapse
Affiliation(s)
- Oihana Murillo
- Gene Therapy Unit, Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
It has long been established that inflammation and immunity play critical roles in the pathogenesis, control and eventual metastasis of cancers. With the advent of more sophisticated animal models and immunohistochemical techniques a greater understanding of the immune system and its interactions has occurred. Individual immune cells are dynamic structures that have variable behaviour controlled by complex interactions in the tumour microenvironment. In the setting of colorectal cancer it was first observed that peritumoral inflammatory infiltrates were associated with improved prognosis. Immunohistochemistry has shown the individual cells types within these infiltrates. It now appears that an adaptive immune response, differentiated along the T-helper 1 pathway controls tumour invasion and metastasis. Furthermore, the immune system exerts selection pressure leading to the evolution of tumour cell variants that can induce tolerance and disable adaptive immunity. These tumour cells then use the mechanisms of innate immunity to facilitate further growth, angiogenesis, invasion and eventual metastasis. These issues are investigated with particular relevance to colorectal cancer. Using the immune response to defeat CRC has been under intense investigation but has so far been unsuccessful. Nevertheless, researchers remain optimistic that immunotherapy will play an important role in the treatment of this common disease.
Collapse
Affiliation(s)
- Paul Salama
- School of Surgery and Pathology, University of Western Australia, Perth, Western Australia, Australia.
| | | |
Collapse
|
19
|
Santini SM, Lapenta C, Santodonato L, D'Agostino G, Belardelli F, Ferrantini M. IFN-alpha in the generation of dendritic cells for cancer immunotherapy. Handb Exp Pharmacol 2008:295-317. [PMID: 19031032 DOI: 10.1007/978-3-540-71029-5_14] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Dendritic cells (DCs) play a crucial role in linking innate and adaptive immunity, by virtue of their unique ability to take up and process antigens in the peripheral blood and tissues and, upon migration to draining lymph nodes, to present antigen to resting lymphocytes. Notably, these DC functions are modulated by cytokines and chemokines controlling the activation and maturation of these cells, thus shaping the response towards either immunity or tolerance.An ensemble of recent studies have emphasized an important role of type I IFNs in the DC differentiation/activation, suggesting the existence of a natural alliance between these cytokines and DCs in linking innate and adaptive immunity. Herein, we will review how type I IFNs can promote the ex vivo differentiation of human DCs and orient DC functions towards the priming and expansion of protective antitumor immune responses. We will also discuss how the knowledge on type I IFN-DC interactions could be exploited for the design of more selective and effective strategies of cancer immunotherapy.
Collapse
Affiliation(s)
- Stefano Maria Santini
- Section of Experimental Immunotherapy, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena, Rome, 299, 00161 Italy.
| | | | | | | | | | | |
Collapse
|
20
|
Dunbar E, Alatery A, Basta S. Cross-Priming of a Single Viral Protein from Lymphocytic Choriomeningitis Virus Alters Immunodominance Hierarchies of CD8+ T Cells during Subsequent Viral Infections. Viral Immunol 2007; 20:585-98. [DOI: 10.1089/vim.2007.0062] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Erin Dunbar
- Department of Microbiology and Immunology, Queen's University, Kingston, Ontario, Canada
| | - Attiya Alatery
- Department of Microbiology and Immunology, Queen's University, Kingston, Ontario, Canada
| | - Sameh Basta
- Department of Microbiology and Immunology, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
21
|
Arina A, Murillo O, Dubrot J, Azpilikueta A, Alfaro C, Pérez-Gracia JL, Bendandi M, Palencia B, Hervás-Stubbs S, Melero I. Cellular liaisons of natural killer lymphocytes in immunology and immunotherapy of cancer. Expert Opin Biol Ther 2007; 7:599-615. [PMID: 17477799 DOI: 10.1517/14712598.7.5.599] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
There is compelling evidence for the role of natural killer (NK) cells in tumor immunosurveillance and their beneficial effects on many experimentally successful immunotherapy strategies. NK cells mediate cell contact-dependent cellular cytotoxicity and produce pro-inflammatory cytokines, but do not rearrange antigen receptors. Their activation depends on various germline-encoded receptors, including CD16, which mediates recognition of antibody-coated target cells. NK cytotoxicity is checked by a repertoire of inhibitory receptors that scan adequate expression of major histocompatibility complex class I molecules on the potential target cell. Functional cross-talk of NK and dendritic cells suggests a critical role for NK cells in the initiation and regulation of cellular immunity. Considerable knowledge on the molecular basis of NK recognition/activation contrasts with a lack of successful translational research on these matters. However, there is plenty of opportunity for targeted intervention of inhibitory/activatory surface receptors and for adoptive cell therapy with autologous or allogeneic NK cells.
Collapse
Affiliation(s)
- Ainhoa Arina
- University of Navarra, Centro de Investigación Médica Aplicada and Clinica Universitaria, Gene Therapy Unit, Avda. Pio XII 55, 31008, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|