1
|
Randerson-Moor J, Davies J, Harland M, Nsengimana J, Bigirumurame T, Walker C, Laye J, Appleton ES, Ball G, Cook GP, Bishop DT, Salmond RJ, Newton-Bishop J. Systemic Inflammation, the Peripheral Blood Transcriptome, and Primary Melanoma. J Invest Dermatol 2024; 144:2513-2529.e17. [PMID: 38583742 DOI: 10.1016/j.jid.2024.02.034] [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: 08/07/2023] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 04/09/2024]
Abstract
Peripheral blood transcriptomes from 383 patients with newly diagnosed melanoma were subjected to differential gene expression analysis. The hypotheses were that impaired systemic immunity is associated with poorer prognosis (thicker tumors and fewer tumor-infiltrating lymphocytes) and evidence of systemic inflammation (high-sensitivity CRP and fibrinogen levels). Higher fibrinogen levels were associated with thicker primary tumors. In single-gene analysis, high-sensitivity CRP levels were significantly associated with higher blood CD274 expression (coding for PD-L1), but each was independently prognostic, with high-sensitivity CRP associated with increased mortality and higher CD274 protective, independent of age. Pathway analysis identified downregulation of immune cell signaling pathways in the blood of people with thicker tumors and notable upregulation of signal transducer and activator of transcription 1 gene STAT1 in people with brisk tumor-infiltrating lymphocytes. Transcriptomic data provided evidence for increased NF-kB signaling with higher inflammatory markers but with reduction in expression of HLA class II molecules and higher CD274, suggesting that aberrant systemic inflammation is a significant mediator of reduced immune function in melanoma. In summary, transcriptomic data revealed evidence of reduced immune function in patients with thicker tumors and fewer tumor-infiltrating lymphocytes at diagnosis. Inflammatory markers were associated with thicker primaries and independently with death from melanoma, suggesting that systemic inflammation contributes to that reduced immune function.
Collapse
Affiliation(s)
- Juliette Randerson-Moor
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - John Davies
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom; Leeds Institute of Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Mark Harland
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Jérémie Nsengimana
- Population Health Sciences Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle, United Kingdom
| | - Theophile Bigirumurame
- Population Health Sciences Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle, United Kingdom
| | - Christopher Walker
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Jon Laye
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Elizabeth S Appleton
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom; Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Graham Ball
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Graham P Cook
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - D Timothy Bishop
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Robert J Salmond
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Julia Newton-Bishop
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
2
|
Kwok SJJ, Forward S, Fahlberg MD, Assita ER, Cosgriff S, Lee SH, Abbott GR, Zhu H, Minasian NH, Vote AS, Martino N, Yun SH. High-dimensional multi-pass flow cytometry via spectrally encoded cellular barcoding. Nat Biomed Eng 2024; 8:310-324. [PMID: 38036616 DOI: 10.1038/s41551-023-01144-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 10/21/2023] [Indexed: 12/02/2023]
Abstract
Advances in immunology, immuno-oncology, drug discovery and vaccine development demand improvements in the capabilities of flow cytometry to allow it to measure more protein markers per cell at multiple timepoints. However, the size of panels of fluorophore markers is limited by overlaps in fluorescence-emission spectra, and flow cytometers typically perform cell measurements at one timepoint. Here we describe multi-pass high-dimensional flow cytometry, a method leveraging cellular barcoding via microparticles emitting near-infrared laser light to track and repeatedly measure each cell using more markers and fewer colours. By using live human peripheral blood mononuclear cells, we show that the method enables the time-resolved characterization of the same cells before and after stimulation, their analysis via a 10-marker panel with minimal compensation for spectral spillover and their deep immunophenotyping via a 32-marker panel, where the same cells are analysed in 3 back-to-back cycles with 10-13 markers per cycle, reducing overall spillover and simplifying marker-panel design. Cellular barcoding in flow cytometry extends the utility of the technique for high-dimensional multi-pass single-cell analyses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Han Zhu
- LASE Innovation Inc., Woburn, MA, USA
| | | | | | - Nicola Martino
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Cambridge, MA, USA
| | - Seok-Hyun Yun
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Cambridge, MA, USA.
| |
Collapse
|
3
|
Kovacsovics-Bankowski M, Sweere JM, Healy CP, Sigal N, Cheng LC, Chronister WD, Evans SA, Marsiglio J, Gibson B, Swami U, Erickson-Wayman A, McPherson JP, Derose YS, Eliason AL, Medina CO, Srinivasan R, Spitzer MH, Nguyen N, Hyngstrom J, Hu-Lieskovan S. Lower frequencies of circulating suppressive regulatory T cells and higher frequencies of CD4 + naïve T cells at baseline are associated with severe immune-related adverse events in immune checkpoint inhibitor-treated melanoma. J Immunother Cancer 2024; 12:e008056. [PMID: 38233101 PMCID: PMC10806651 DOI: 10.1136/jitc-2023-008056] [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] [Accepted: 12/18/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Immune-related adverse events (irAEs) are major barriers of clinical management and further development of immune checkpoint inhibitors (ICIs) for cancer therapy. Therefore, biomarkers associated with the onset of severe irAEs are needed. In this study, we aimed to identify immune features detectable in peripheral blood and associated with the development of severe irAEs that required clinical intervention. METHODS We used a 43-marker mass cytometry panel to characterize peripheral blood mononuclear cells from 28 unique patients with melanoma across 29 lines of ICI therapy before treatment (baseline), before the onset of irAEs (pre-irAE) and at the peak of irAEs (irAE-max). In the 29 lines of ICI therapy, 18 resulted in severe irAEs and 11 did not. RESULTS Unsupervised and gated population analysis showed that patients with severe irAEs had a higher frequency of CD4+ naïve T cells and lower frequency of CD16+ natural killer (NK) cells at all time points. Gated population analysis additionally showed that patients with severe irAEs had fewer T cell immunoreceptor with Ig and ITIM domain (TIGIT+) regulatory T cells at baseline and more activated CD38+ CD4+ central memory T cells (TCM) and CD39+ and Human Leukocyte Antigen-DR Isotype (HLA-DR)+ CD8+ TCM at peak of irAEs. The differentiating immune features at baseline were predominantly seen in patients with gastrointestinal and cutaneous irAEs and type 1 diabetes. Higher frequencies of CD4+ naïve T cells and lower frequencies of CD16+ NK cells were also associated with clinical benefit to ICI therapy. CONCLUSIONS This study demonstrates that high-dimensional immune profiling can reveal novel blood-based immune signatures associated with risk and mechanism of severe irAEs. Development of severe irAEs in melanoma could be the result of reduced immune inhibitory capacity pre-ICI treatment, resulting in more activated TCM cells after treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - John Marsiglio
- The University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Berit Gibson
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Umang Swami
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Alyssa Erickson-Wayman
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jordan P McPherson
- Department of Pharmacy, Huntsman Cancer Institute Cancer Hospital, Salt Lake City, Utah, USA
| | - Yoko S Derose
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | | | | | | | - Matthew H Spitzer
- Teiko.bio Inc, Salt Lake City, Utah, USA
- Department of Otolaryngology-Head and Neck Cancer, University of California San Francisco, San Francisco, California, USA
| | | | - John Hyngstrom
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Siwen Hu-Lieskovan
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
4
|
Carbone ML, Capone A, Guercio M, Reddel S, Silvestris DA, Lulli D, Ramondino C, Peluso D, Quintarelli C, Volpe E, Failla CM. Insight into immune profile associated with vitiligo onset and anti-tumoral response in melanoma patients receiving anti-PD-1 immunotherapy. Front Immunol 2023; 14:1197630. [PMID: 37680638 PMCID: PMC10482109 DOI: 10.3389/fimmu.2023.1197630] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/04/2023] [Indexed: 09/09/2023] Open
Abstract
Introduction Immunotherapy with checkpoint inhibitors is an efficient treatment for metastatic melanoma. Development of vitiligo upon immunotherapy represents a specific immune-related adverse event (irAE) diagnosed in 15% of patients and associated with a positive clinical response. Therefore, a detailed characterization of immune cells during vitiligo onset in melanoma patients would give insight into the immune mechanisms mediating both the irAE and the anti-tumor response. Methods To better understand these aspects, we analyzed T cell subsets from peripheral blood of metastatic melanoma patients undergoing treatment with anti-programmed cell death protein (PD)-1 antibodies. To deeply characterize the antitumoral T cell response concomitant to vitiligo onset, we analyzed T cell content in skin biopsies collected from melanoma patients who developed vitiligo. Moreover, to further characterize T cells in vitiligo skin lesion of melanoma patients, we sequenced T cell receptor (TCR) of cells derived from biopsies of vitiligo and primary melanoma of the same patient. Results and discussion Stratification of patients for developing or not developing vitiligo during anti-PD-1 therapy revealed an association between blood reduction of CD8-mucosal associated invariant T (MAIT), T helper (h) 17, natural killer (NK) CD56bright, and T regulatory (T-reg) cells and vitiligo onset. Consistently with the observed blood reduction of Th17 cells in melanoma patients developing vitiligo during immunotherapy, we found high amount of IL-17A expressing cells in the vitiligo skin biopsy, suggesting a possible migration of Th17 cells from the blood into the autoimmune lesion. Interestingly, except for a few cases, we found different TCR sequences between vitiligo and primary melanoma lesions. In contrast, shared TCR sequences were identified between vitiligo and metastatic tissues of the same patient. These data indicate that T cell response against normal melanocytes, which is involved in vitiligo onset, is not typically mediated by reactivation of specific T cell clones infiltrating primary melanoma but may be elicited by T cell clones targeting metastatic tissues. Altogether, our data indicate that anti-PD-1 therapy induces a de novo immune response, stimulated by the presence of metastatic cells, and composed of different T cell subtypes, which may trigger the development of vitiligo and the response against metastatic tumor.
Collapse
Affiliation(s)
- Maria Luigia Carbone
- Laboratory of Experimental Immunology, Istituto Dermopatico dell’Immacolata (IDI)-IRCCS, Rome, Italy
| | - Alessia Capone
- Laboratory of Molecular Neuroimmunology, Santa Lucia Foundation-IRCCS, Rome, Italy
| | - Marika Guercio
- Department of Oncology-Hematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Sofia Reddel
- Department of Oncology-Hematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | | | - Daniela Lulli
- Laboratory of Experimental Immunology, Istituto Dermopatico dell’Immacolata (IDI)-IRCCS, Rome, Italy
| | - Carmela Ramondino
- Laboratory of Experimental Immunology, Istituto Dermopatico dell’Immacolata (IDI)-IRCCS, Rome, Italy
| | - Daniele Peluso
- Department of Biology, University “Tor Vergata”, Rome, Italy
| | - Concetta Quintarelli
- Department of Oncology-Hematology, and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Elisabetta Volpe
- Laboratory of Molecular Neuroimmunology, Santa Lucia Foundation-IRCCS, Rome, Italy
| | - Cristina Maria Failla
- Laboratory of Experimental Immunology, Istituto Dermopatico dell’Immacolata (IDI)-IRCCS, Rome, Italy
| |
Collapse
|
5
|
Chen J, Wang D, Chan S, Yang Q, Wang C, Wang X, Sun R, Gui Y, Yu S, Yang J, Zhang H, Zhang X, Tang K, Zhang H, Liu S. Development and validation of a novel T cell proliferation-related prognostic model for predicting survival and immunotherapy benefits in melanoma. Aging (Albany NY) 2023; 15:204748. [PMID: 37227816 DOI: 10.18632/aging.204748] [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: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND T cell plays a crucial role in the occurrence and progression of Skin cutaneous melanoma (SKCM). This research aims to identify the actions of T cell proliferation-related genes (TRGs) on the prognosis and immunotherapy response of tumor patients. METHOD The clinical manifestation and gene expression data of SKCM patients were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. T cell proliferation-related molecular subtypes were identified utilizing consensus clustering. Subsequently, Cox and Lasso regression analysis was conducted to identify six prognostic genes, and a prognostic signature was constructed. A series of experiments, such as qRT-PCR, Western blotting and CCK8 assay, were then conducted to verify the reliability of the six genes. RESULTS In this study, a grading system was established to forecast survival time and responses to immunotherapy, providing an overview of the tumoral immune landscape. Meanwhile, we identified six prognostic signature genes. Notably, we also found that C1RL protein may inhibit the growth of melanoma cell lines. CONCLUSION The scoring system depending on six prognostic genes showed great efficiency in predicting survival time. The system could help to forecast prognosis of SKCM patients, characterize SKCM immunological condition, assess patient immunotherapy response.
Collapse
Affiliation(s)
- Jiajie Chen
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Daiyue Wang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Shixin Chan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Qingqing Yang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Chen Wang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Xu Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Rui Sun
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Yu Gui
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Shuling Yu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Jinwei Yang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Haoxue Zhang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Xiaomin Zhang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230022, China
| | - Kechao Tang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230022, China
| | - Huabing Zhang
- Affiliated Chuzhou Hospital of Anhui Medical University, The First People’s Hospital of Chuzhou, Chuzhou, Anhui 230022, China
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230022, China
| | - Shengxiu Liu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| |
Collapse
|
6
|
Seyhan AA, Carini C. Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates. Int J Mol Sci 2022; 24:ijms24010041. [PMID: 36613491 PMCID: PMC9820306 DOI: 10.3390/ijms24010041] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Despite the recent successes and durable responses with immune checkpoint inhibitors (ICI), many cancer patients, including those with melanoma, do not derive long-term benefits from ICI therapies. The lack of predictive biomarkers to stratify patients to targeted treatments has been the driver of primary treatment failure and represents an unmet medical need in melanoma and other cancers. Understanding genomic correlations with response and resistance to ICI will enhance cancer patients' benefits. Building on insights into interplay with the complex tumor microenvironment (TME), the ultimate goal should be assessing how the tumor 'instructs' the local immune system to create its privileged niche with a focus on genomic reprogramming within the TME. It is hypothesized that this genomic reprogramming determines the response to ICI. Furthermore, emerging genomic signatures of ICI response, including those related to neoantigens, antigen presentation, DNA repair, and oncogenic pathways, are gaining momentum. In addition, emerging data suggest a role for checkpoint regulators, T cell functionality, chromatin modifiers, and copy-number alterations in mediating the selective response to ICI. As such, efforts to contextualize genomic correlations with response into a more insightful understanding of tumor immune biology will help the development of novel biomarkers and therapeutic strategies to overcome ICI resistance.
Collapse
Affiliation(s)
- Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02912, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
- Correspondence:
| | - Claudio Carini
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, New Hunt’s House, Guy’s Campus, King’s College London, London SE1 1UL, UK
- Biomarkers Consortium, Foundation of the National Institute of Health, Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Advances in the Application of Nanomaterials to the Treatment of Melanoma. Pharmaceutics 2022; 14:pharmaceutics14102090. [PMID: 36297527 PMCID: PMC9610396 DOI: 10.3390/pharmaceutics14102090] [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: 08/03/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022] Open
Abstract
Melanoma can be divided into cutaneous melanoma, uveal melanoma, mucosal melanoma, etc. It is a very aggressive tumor that is prone to metastasis. Patients with metastatic melanoma have a poor prognosis and shorter survival. Although current melanoma treatments have been dramatically improved, there are still many problems such as systemic toxicity and the off-target effects of drugs. The use of nanoparticles may overcome some inadequacies of current melanoma treatments. In this review, we summarize the limitations of current therapies for cutaneous melanoma, uveal melanoma, and mucosal melanoma, as well as the adjunct role of nanoparticles in different treatment modalities. We suggest that nanomaterials may have an effective intervention in melanoma treatment in the future.
Collapse
|
8
|
HLA-DRB1: A new potential prognostic factor and therapeutic target of cutaneous melanoma and an indicator of tumor microenvironment remodeling. PLoS One 2022; 17:e0274897. [PMID: 36129956 PMCID: PMC9491554 DOI: 10.1371/journal.pone.0274897] [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: 02/16/2022] [Accepted: 08/26/2022] [Indexed: 11/19/2022] Open
Abstract
Cutaneous melanoma (CM) is the most common skin cancer and one of the most aggressive cancers and its incidence has risen dramatically over the past few decades. The tumor microenvironment (TME) plays a crucial role in the occurrence and development of cutaneous melanoma. Nevertheless, the dynamics modulation of the immune and stromal components in the TME is not fully understood. In this study, 471 CM samples were obtained from TCGA database, and the ratio of tumor-infiltrating immune cells (TICs) in the TME were estimated using the ESTIMATE algorithms and CIBERSORT computational method. The differently expressed genes (DEGs) were applied to GO and KEGG function enrichment analysis, establishment of protein-protein interaction (PPI) network and univariate Cox regression analysis. Subsequently, we identified a predictive factor: HLA-DRB1 (major histocompatibility complex, class II, DR beta 1) by the intersection analysis of the hub genes of PPI network and the genes associated with the prognosis of the CM patients obtained by univariate Cox regression analysis. Correlation analysis and survival analysis showed that the expression level of HLA-DRB1 was negatively correlated with the Stage of the patients while positively correlated with the survival, prognosis and TME of melanoma. The GEPIA web server and the representative immunohistochemical images of HLA-DRB1 in the normal skin tissue and melanoma tissue from the Human Protein Atlas (HPA) database were applied to validate the expression level of HLA-DRB1. CIBERSORT analysis for the ratio of TICs indicated that 9 types of TICs were positively correlated with the expression level of HLA-DRB1 and only 4 types of TICs were negatively correlated with the expression level of HLA-DRB1. These results suggested that the expression level of HLA-DRB1 may be related to the immune activity of the TME and may affect the prognosis of CM patients by changing the status of the TME.
Collapse
|
9
|
Bahri R, Kiss O, Prise I, Garcia-Rodriguez KM, Atmoko H, Martínez-Gómez JM, Levesque MP, Dummer R, Smith MP, Wellbrock C, Bulfone-Paus S. Human Melanoma-Associated Mast Cells Display a Distinct Transcriptional Signature Characterized by an Upregulation of the Complement Component 3 That Correlates With Poor Prognosis. Front Immunol 2022; 13:861545. [PMID: 35669782 PMCID: PMC9163391 DOI: 10.3389/fimmu.2022.861545] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cutaneous melanoma is one of the most aggressive human malignancies and shows increasing incidence. Mast cells (MCs), long-lived tissue-resident cells that are particularly abundant in human skin where they regulate both innate and adaptive immunity, are associated with melanoma stroma (MAMCs). Thus, MAMCs could impact melanoma development, progression, and metastasis by secreting proteases, pro-angiogenic factors, and both pro-inflammatory and immuno-inhibitory mediators. To interrogate the as-yet poorly characterized role of human MAMCs, we have purified MCs from melanoma skin biopsies and performed RNA-seq analysis. Here, we demonstrate that MAMCs display a unique transcriptome signature defined by the downregulation of the FcεRI signaling pathway, a distinct expression pattern of proteases and pro-angiogenic factors, and a profound upregulation of complement component C3. Furthermore, in melanoma tissue, we observe a significantly increased number of C3+ MCs in stage IV melanoma. Moreover, in patients, C3 expression significantly correlates with the MC-specific marker TPSAB1, and the high expression of both markers is linked with poorer melanoma survival. In vitro, we show that melanoma cell supernatants and tumor microenvironment (TME) mediators such as TGF-β, IL-33, and IL-1β induce some of the changes found in MAMCs and significantly modulate C3 expression and activity in MCs. Taken together, these data suggest that melanoma-secreted cytokines such as TGF-β and IL-1β contribute to the melanoma microenvironment by upregulating C3 expression in MAMCs, thus inducing an MC phenotype switch that negatively impacts melanoma prognosis.
Collapse
Affiliation(s)
- Rajia Bahri
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Orsolya Kiss
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Ian Prise
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Karen M. Garcia-Rodriguez
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Haris Atmoko
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Julia M. Martínez-Gómez
- Department of Dermatology, Skin Cancer Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Mitchell P. Levesque
- Department of Dermatology, Skin Cancer Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, Skin Cancer Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Michael P. Smith
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Claudia Wellbrock
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Silvia Bulfone-Paus
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
10
|
Azuma K, Xiang H, Tagami T, Kasajima R, Kato Y, Karakawa S, Kikuchi S, Imaizumi A, Matsuo N, Ishii H, Tokito T, Kawahara A, Murotani K, Sasada T, Miyagi Y, Hoshino T. Clinical significance of plasma-free amino acids and tryptophan metabolites in patients with non-small cell lung cancer receiving PD-1 inhibitor: a pilot cohort study for developing a prognostic multivariate model. J Immunother Cancer 2022; 10:jitc-2021-004420. [PMID: 35569917 PMCID: PMC9109096 DOI: 10.1136/jitc-2021-004420] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2022] [Indexed: 12/18/2022] Open
Abstract
Background Amino acid metabolism is essential for tumor cell proliferation and regulation of immune cell function. However, the clinical significance of free amino acids (plasma-free amino acids (PFAAs)) and tryptophan-related metabolites in plasma has not been fully understood in patients with non-small cell lung cancer (NSCLC) who receive immune checkpoint inhibitors. Methods We conducted a single cohort observational study. Peripheral blood samples were collected from 53 patients with NSCLC before treatment with PD-1 (Programmed cell death-1) inhibitors. The plasma concentrations of 21 PFAAs, 14 metabolites, and neopterin were measured by liquid chromatography–mass spectrometry. Using Cox hazard analysis with these variables, a multivariate model was established to stratify patient overall survival (OS). Gene expression in peripheral blood mononuclear cells (PBMCs) was compared between the high-risk and low-risk patients by this multivariate model. Results On Cox proportional hazard analysis, higher concentrations of seven PFAAs (glycine, histidine, threonine, alanine, citrulline, arginine, and tryptophan) as well as lower concentrations of three metabolites (3h-kynurenine, anthranilic acid, and quinolinic acid) and neopterin in plasma were significantly correlated with better OS (p<0.05). In particular, the multivariate model, composed of a combination of serine, glycine, arginine, and quinolinic acid, could most efficiently stratify patient OS (concordance index=0.775, HR=3.23, 95% CI 2.04 to 5.26). From the transcriptome analysis in PBMCs, this multivariate model was significantly correlated with the gene signatures related to immune responses, such as CD8 T-cell activation/proliferation and proinflammatory immune responses, and 12 amino acid-related genes were differentially expressed between the high-risk and low-risk groups. Conclusions The multivariate model with PFAAs and metabolites in plasma might be useful for stratifying patients who will benefit from PD-1 inhibitors.
Collapse
Affiliation(s)
- Koichi Azuma
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Huihui Xiang
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Tomoyuki Tagami
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co Inc, Kawasaki, Japan
| | - Rika Kasajima
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yumiko Kato
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co Inc, Kawasaki, Japan
| | - Sachise Karakawa
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co Inc, Kawasaki, Japan
| | - Shinya Kikuchi
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co Inc, Kawasaki, Japan
| | - Akira Imaizumi
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co Inc, Kawasaki, Japan
| | - Norikazu Matsuo
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hidenobu Ishii
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takaaki Tokito
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Akihiko Kawahara
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Japan
| | - Kenta Murotani
- Biostatistics Center, Kurume University School of Medicine, Kurume, Japan
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Tomoaki Hoshino
- Division of Respirology, Neurology, and Rheumatology Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
11
|
Ai D, Xing Y, Zhang Q, Wang Y, Liu X, Liu G, Xia LC. Joint Analysis of Microbial and Immune Cell Abundance in Liver Cancer Tissue Using a Gene Expression Profile Deconvolution Algorithm Combined With Foreign Read Remapping. Front Immunol 2022; 13:853213. [PMID: 35493464 PMCID: PMC9047545 DOI: 10.3389/fimmu.2022.853213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/07/2022] [Indexed: 11/21/2022] Open
Abstract
Recent transcriptomics and metagenomics studies showed that tissue-infiltrating immune cells and bacteria interact with cancer cells to shape oncogenesis. This interaction and its effects remain to be elucidated. However, it is technically difficult to co-quantify immune cells and bacteria in their respective microenvironments. To address this challenge, we herein report the development of a complete a bioinformatics pipeline, which accurately estimates the number of infiltrating immune cells using a novel Particle Swarming Optimized Support Vector Regression (PSO-SVR) algorithm, and the number of infiltrating bacterial using foreign read remapping and the GRAMMy algorithm. It also performs systematic differential abundance analyses between tumor-normal pairs. We applied the pipeline to a collection of paired liver cancer tumor and normal samples, and we identified bacteria and immune cell species that were significantly different between tissues in terms of health status. Our analysis showed that this dual model of microbial and immune cell abundance had a better differentiation (84%) between healthy and diseased tissue. Caldatribacterium sp., Acidaminococcaceae sp., Planctopirus sp., Desulfobulbaceae sp.,Nocardia farcinica as well as regulatory T cells (Tregs), resting mast cells, monocytes, M2 macrophases, neutrophils were identified as significantly different (Mann Whitney Test, FDR< 0.05). Our open-source software is freely available from GitHub at https://github.com/gutmicrobes/PSO-SVR.git.
Collapse
Affiliation(s)
- Dongmei Ai
- Basic Experimental Center of Natural Science, University of Science and Technology Beijing, Beijing, China
- *Correspondence: Dongmei Ai, ; Li C. Xia,
| | - Yonglian Xing
- School of Mathematics and Physics, University of Science and Technology Beijing, Beijing, China
| | - Qingchuan Zhang
- National Engineering Laboratory for Agri-Product Quality Traceability, Beijing Technology and Business University, Beijing, China
| | - Yishu Wang
- School of Mathematics and Physics, University of Science and Technology Beijing, Beijing, China
| | - Xiuqin Liu
- School of Mathematics and Physics, University of Science and Technology Beijing, Beijing, China
| | - Gang Liu
- School of Mathematics and Physics, University of Science and Technology Beijing, Beijing, China
| | - Li C. Xia
- School of Mathematics, South China University of Technology, Guangzhou, China
- *Correspondence: Dongmei Ai, ; Li C. Xia,
| |
Collapse
|
12
|
Loo K, Smithy JW, Postow MA, Betof Warner A. Factors Determining Long-Term Antitumor Responses to Immune Checkpoint Blockade Therapy in Melanoma. Front Immunol 2022; 12:810388. [PMID: 35087529 PMCID: PMC8787112 DOI: 10.3389/fimmu.2021.810388] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
With the increasing promise of long-term survival with immune checkpoint blockade (ICB) therapies, particularly for patients with advanced melanoma, clinicians and investigators are driven to identify prognostic and predictive factors that may help to identify individuals who are likely to experience durable benefit. Several ICB combinations are being actively developed to expand the armamentarium of treatments for patients who may not achieve long-term responses to ICB single therapies alone. Thus, negative predictive markers are also of great interest. This review seeks to deepen our understanding of the mechanisms underlying the durability of ICB treatments. We will discuss the currently available long-term data from the ICB clinical trials and real-world studies describing the survivorship of ICB-treated melanoma patients. Additionally, we explore the current treatment outcomes in patients rechallenged with ICB and the patterns of ICB resistance based on sites of disease, namely, liver or CNS metastases. Lastly, we discuss the landscape in melanoma in the context of prognostic or predictive factors as markers of long-term response to ICB.
Collapse
Affiliation(s)
- Kimberly Loo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Internal Medicine, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY, United States
| | - James W. Smithy
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Michael A. Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Allison Betof Warner
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
13
|
Safai B, Wu AG, Hamby CV. Prognostic Biomarkers in Melanoma: Tailoring Treatments to the Patient. THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2021; 14:44-48. [PMID: 35096254 PMCID: PMC8794494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
BACKGROUND It is often difficult to accurately predict how a melanoma will progress because melanomas can be so diverse in their genetic and histological makeup. OBJECTIVE We sought to characterize the current state and progression of biomedical markers towards their utilization as prognostic indicators for patients with melanoma. METHODS A literature search of the research repository databases PubMed and GoogleScholar was conducted using the following inclusion criteria: (1) published within the last 10 years, and (2) use of overall survival, disease progression, or clinical outcome as primary endpoints. Search terms included various permutations of "biomarkers," "prognostic," "immunologic," "serologic," "visual," and "melanoma." Results were evaluated for statistical power, results significance, and experimental design integrity. RESULTS The prognostic capabilities of clinical tests for malignant melanoma have made great strides in the last few years, with several serologic and immunohistochemical biomarkers being preliminarily linked to various measures of clinical prognosis. While clinical feasibility of a single sensitive and specific biomarker remains unfeasible, use of select combinations of tested biomarkers remain viable. CONCLUSION Diagnostic and prognostic genetic assays have begun to cross over from research to commercial application, giving physicians additional tools during the early stages of diagnosis to optimize and individualize treatments.
Collapse
Affiliation(s)
- Bijan Safai
- Dr. Safai is with the Department of Dermatology, Metropolitan Hospital in New York, New York
- Mr. Wu and Dr. Hamby are with New York Medical College School of Medicine in Valhalla, New York
| | - Albert G Wu
- Dr. Safai is with the Department of Dermatology, Metropolitan Hospital in New York, New York
- Mr. Wu and Dr. Hamby are with New York Medical College School of Medicine in Valhalla, New York
| | - Carl V Hamby
- Dr. Safai is with the Department of Dermatology, Metropolitan Hospital in New York, New York
- Mr. Wu and Dr. Hamby are with New York Medical College School of Medicine in Valhalla, New York
| |
Collapse
|
14
|
Di Pietro FR, Verkhovskaia S, Mastroeni S, Carbone ML, Abeni D, Di Rocco CZ, Samà N, Zappalà AR, Marchetti P, De Galitiis F, Failla CM, Fortes C. Clinical Predictors of Response to Anti-PD-1 First-Line Treatment in a Single-Centre Patient Cohort: A Real-World Study. Clin Oncol (R Coll Radiol) 2021; 34:e18-e24. [PMID: 34563446 DOI: 10.1016/j.clon.2021.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/04/2021] [Accepted: 09/07/2021] [Indexed: 01/25/2023]
Abstract
AIMS Cutaneous melanoma is one of the most immunogenic tumours. Immunotherapy with checkpoint inhibitors, such as anti-PD-1 antibodies, has significantly improved the prognosis in metastatic melanoma. However, only half of the patients respond to this therapy and have a favourable outcome. Identifying factors associated with treatment failure and early identification of responders are both important to select the best treatment approach for each patient. The aim of our study was to investigate clinical biomarkers of response to treatment with anti-PD-1 antibodies. MATERIALS AND METHODS We selected all patients with stage IV melanoma (n = 147), subjected to first-line treatment with anti-PD-1 in the last 10 years. We investigated the associations between patients' different clinical features and progression-free survival, using the Cox proportional hazards models. RESULTS In the multivariate analysis, an increased risk of disease progression was observed among patients with stage M1d metastases (hazard ratio 3.30; 95% confidence interval 1.58-6.91), compared with patients with stage M1a-M1b. Moreover, the risk of progression was greater in patients with the Eastern Cooperative Oncology Group Performance Status (ECOG PS) 1 (hazard ratio 2.04; 95% confidence interval 1.02-4.06) and in patients with ECOG PS ≥ 2 (hazard ratio 2.19; 95% confidence interval 1.05-4.55) compared with ECOG PS 0. High levels of lactate dehydrogenase (hazard ratio 2.06; 95% confidence interval 1.18-3.59) and the presence of respiratory diseases (hazard ratio 4.14; 95% confidence interval 1.42-12.0) at the beginning of anti-PD-1 treatment were also associated with an increased risk of disease progression. In a subgroup analysis, neutrophil count and neutrophil/lymphocyte ratio before anti-PD-1 treatment were higher in patients who underwent disease progression. CONCLUSION In our study population, independent predictors of disease progression among patients treated with first-line anti-PD-1 were as follows: ECOG PS, staging, lactate dehydrogenase and the presence of respiratory diseases.
Collapse
Affiliation(s)
- F R Di Pietro
- Department of Oncology and Dermatological Oncology, IDI-IRCCS, Rome, Italy
| | - S Verkhovskaia
- Department of Oncology and Dermatological Oncology, IDI-IRCCS, Rome, Italy
| | | | - M L Carbone
- Laboratory of Experimental Immunology, IDI-IRCCS, Rome, Italy
| | - D Abeni
- Epidemiology Unit, IDI-IRCCS, Rome, Italy
| | - C Z Di Rocco
- Department of Oncology and Dermatological Oncology, IDI-IRCCS, Rome, Italy
| | - N Samà
- Department of Oncology and Dermatological Oncology, IDI-IRCCS, Rome, Italy
| | - A R Zappalà
- Department of Oncology and Dermatological Oncology, IDI-IRCCS, Rome, Italy
| | - P Marchetti
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant'Andrea Hospital, Sapienza University, Rome, Italy; Medical Oncology Unit B, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - F De Galitiis
- Department of Oncology and Dermatological Oncology, IDI-IRCCS, Rome, Italy
| | - C M Failla
- Laboratory of Experimental Immunology, IDI-IRCCS, Rome, Italy.
| | - C Fortes
- Epidemiology Unit, IDI-IRCCS, Rome, Italy
| |
Collapse
|
15
|
Flaus A, Habouzit V, De Leiris N, Vuillez JP, Leccia MT, Perrot JL, Prevot N, Cachin F. FDG PET biomarkers for prediction of survival in metastatic melanoma prior to anti-PD1 immunotherapy. Sci Rep 2021; 11:18795. [PMID: 34552135 PMCID: PMC8458464 DOI: 10.1038/s41598-021-98310-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/31/2021] [Indexed: 12/22/2022] Open
Abstract
Our aim was to analyse whether biomarkers extracted from baseline 18F-FDG PET before anti-PD1 treatment contribute to prognostic survival information for early risk stratification in metastatic melanoma. Fifty-six patients, without prior systemic treatment, BRAF wild type, explored using 18F-FDG PET were included retrospectively. Our primary endpoint was overall survival (OS). Total metabolic tumoral volume (MTV) and forty-one IBSI compliant parameters were extracted from PET. Parameters associated with outcome were evaluated by a cox regression model and when significant helped build a prognostic score. Median follow-up was 22.1 months and 21 patients died. Total MTV and long zone emphasis (LZE) correlated with shorter OS and served to define three risk categories for the prognostic score. For low, intermediate and high risk groups, survival rates were respectively 91.1% (IC 95 80–1), 56.1% (IC 95 37.1–85) and 19% (IC 95 0.06–60.2) and hazard ratios were respectively 0.11 (IC 95 0.025–0.46), P = 0.0028, 1.2 (IC 95 0.48–2.8), P = 0.74 and 5.9 (IC 95 2.5–14), P < 0.0001. To conclude, a prognostic score based on total MTV and LZE separated metastatic melanoma patients in 3 categories with dramatically different outcomes. Innovative therapies should be tested in the group with the lowest prognosis score for future clinical trials.
Collapse
Affiliation(s)
- A Flaus
- Nuclear Medecine Department, Saint-Etienne University Hospital, University of Saint-Etienne, Saint-Etienne, France. .,Nuclear Medicine Department, East Group Hospital, Hospices Civils de Lyon, Lyon, France. .,Service de Medecine Nucléaire, Hôpital Nord, CHU de Saint-Etienne, 42 055, Saint-Etienne, Cedex 2, France.
| | - V Habouzit
- Nuclear Medecine Department, Saint-Etienne University Hospital, University of Saint-Etienne, Saint-Etienne, France
| | - N De Leiris
- Nuclear Medecine Department, CHU Grenoble Alpes, University Grenoble Alpes, Grenoble, France.,Laboratoire Radiopharmaceutiques Biocliniques, University Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000, Grenoble, France
| | - J P Vuillez
- Nuclear Medecine Department, CHU Grenoble Alpes, University Grenoble Alpes, Grenoble, France.,Laboratoire Radiopharmaceutiques Biocliniques, University Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000, Grenoble, France
| | - M T Leccia
- Dermatology Department, CHU Grenoble Alpes, University Grenoble Alpes, Grenoble, France
| | - J L Perrot
- Dermatology Department, Saint-Etienne University Hospital, University of Saint-Etienne, Saint-Etienne, France
| | - N Prevot
- Nuclear Medecine Department, Saint-Etienne University Hospital, University of Saint-Etienne, Saint-Etienne, France
| | - F Cachin
- Nuclear Medicine Department, Jean Perrin Cancer Center of Clermont-Ferrand, Clermont-Ferrand, France
| |
Collapse
|
16
|
Buma AIG, Muller M, de Vries R, Sterk PJ, van der Noort V, Wolf-Lansdorf M, Farzan N, Baas P, van den Heuvel MM. eNose analysis for early immunotherapy response monitoring in non-small cell lung cancer. Lung Cancer 2021; 160:36-43. [PMID: 34399166 DOI: 10.1016/j.lungcan.2021.07.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/20/2021] [Accepted: 07/28/2021] [Indexed: 01/28/2023]
Abstract
OBJECTIVES Exhaled breath analysis by electronic nose (eNose) has shown to be a potential predictive biomarker before start of anti-PD-1 therapy in patients with non-small cell lung carcinoma (NSCLC). We hypothesized that the eNose could also be used as an early monitoring tool to identify responders more accurately at early stage of treatment when compared to baseline. In this proof-of-concept study we aimed to definitely discriminate responders from non-responders after six weeks of treatment. MATERIALS AND METHODS This was a prospective observational study in patients with advanced NSCLC eligible for anti-PD-1 treatment. The efficacy of treatment was assessed by the Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 at 3-month follow-up. We analyzed SpiroNose exhaled breath data of 94 patients (training cohort n = 62, validation cohort n = 32). Data analysis involved signal processing and statistics based on Independent Samples T-tests and Linear Discriminant Analysis (LDA) followed by Receiver Operating Characteristic (ROC) analysis. RESULTS In the training cohort, a specificity of 73% was obtained at a 100% sensitivity level to identify objective responders. The Area Under the Curve (AUC) was 0.95 (CI: 0.89-1.00). In the validation cohort, these results were confirmed with an AUC of 0.97 (CI: 0.91-1.00). CONCLUSION Exhaled breath analysis by eNose early during treatment allows for a highly accurate, non-invasive and low-cost identification of advanced NSCLC patients who benefit from anti-PD-1 therapy.
Collapse
Affiliation(s)
| | - Mirte Muller
- Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rianne de Vries
- Amsterdam University Medical Center, Amsterdam, the Netherlands; Breathomix B.V. (www.breathomix.com), Leiden, the Netherlands
| | - Peter J Sterk
- Amsterdam University Medical Center, Amsterdam, the Netherlands
| | | | | | - Niloufar Farzan
- Breathomix B.V. (www.breathomix.com), Leiden, the Netherlands
| | - Paul Baas
- Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | |
Collapse
|
17
|
Byun HK, Chang JS, Jung M, Koom WS, Chung KY, Oh BH, Roh MR, Kim KH, Lee CK, Shin SJ. Prediction of Immune-Checkpoint Blockade Monotherapy Response in Patients With Melanoma Based on Easily Accessible Clinical Indicators. Front Oncol 2021; 11:659754. [PMID: 34123816 PMCID: PMC8190329 DOI: 10.3389/fonc.2021.659754] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/27/2021] [Indexed: 01/26/2023] Open
Abstract
Background Immune checkpoint blocker (ICB) has shown significant clinical activity in melanoma. However, there are no clinically approved biomarkers to aid patient selection. We aimed to identify patients with advanced or metastatic melanoma who are likely to benefit from ICB monotherapy using easily accessible clinical indicators. Materials and Methods We retrospectively reviewed the records of 134 patients with advanced or metastatic melanoma who received ICB monotherapy between 2014 and 2018. Prognostic factors of overall survival (OS) and progression-free survival (PFS) were determined using Cox regression analysis. Results During the median follow-up of 13.7 months, the median OS and PFS were 18.4 and 3.4 months, respectively. Visceral/central nervous system (CNS) metastasis (OS: adjusted hazards ratio [HR], 1.82; p=.014; PFS: HR, 1.59; p=.024), lymphopenia (<1000 cells/µL) within 3 months (OS: HR, 1.89, p=.006; PFS: HR, 1.70; p=.010), and elevated baseline lactate dehydrogenase (LDH) level (OS: HR, 2.61; p<.001; PFS: HR, 2.66; p<.001) were independent prognostic factors for both poor OS and PFS. Development of immune-related adverse events (irAE; e.g., hypothyroidism or vitiligo) within 6 months showed a trend toward better OS in multivariable analysis (HR, 0.37; p=.058). Patients with normal LDH levels and no visceral/CNS metastasis had a substantially better OS than the others (median, 40.4 vs. 13.6 months; p<.001). Among others, patients who developed irAE within 6 months achieved long-term OS (median, 43.6 vs. 13.1 months; p=.008). A decision tree was suggested using four risk factors, and the risk stratification provided significant distinction between the survival curves. Conclusion The four easily accessible clinical indicators associated with better treatment outcomes after ICB monotherapy in patients with advanced or metastatic melanoma were LDH level, the extent of disease, lymphopenia, and irAE. The combined use of these indicators can be clinically useful in improving risk stratification of patients treated with ICB monotherapy.
Collapse
Affiliation(s)
- Hwa Kyung Byun
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Jee Suk Chang
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Minkyu Jung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Woong Sub Koom
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Kee Yang Chung
- Department of Dermatology, Yonsei University College of Medicine, Seoul, South Korea
| | - Byung Ho Oh
- Department of Dermatology, Yonsei University College of Medicine, Seoul, South Korea
| | - Mi Ryung Roh
- Department of Dermatology, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyung Hwan Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Choong-Kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Joon Shin
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
18
|
Li G, Zhu X, Liu C. Characterization of Immune Infiltration and Construction of a Prediction Model for Overall Survival in Melanoma Patients. Front Oncol 2021; 11:639059. [PMID: 33869027 PMCID: PMC8051586 DOI: 10.3389/fonc.2021.639059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/10/2021] [Indexed: 12/27/2022] Open
Abstract
Reports indicate that the use of anti-programmed cell death-1 (PD-1) and death ligand-1 (PD-L1) monoclonal antibodies for the treatment of patients diagnosed with melanoma has demonstrated promising efficacy. Nonetheless, this therapy is limited by the resistance induced by the tumor microenvironment (TME). As such, understanding the complexity of the TME is vital in enhancing the efficiency of immunotherapy. This study used four different methods to estimate the infiltrating level of immune cells. Besides, we analyzed their infiltration pattern in primary and metastatic melanoma obtained from The Cancer Genome Atlas (TCGA) database. As a consequence, we discovered a significantly higher infiltration of immune cells in metastatic melanoma compared to primary tumor. Consensus clustering identified four clusters in melanoma with different immune infiltration and clusters with higher immune infiltration demonstrated a better overall survival. To elucidate the underlying mechanisms of immune cell infiltration, the four clusters were subdivided into two subtypes denoted as hot and cold tumors based on immune infiltration and predicted immune response. Enrichment analysis of differentially expressed genes (DEGs) revealed different transcriptome alterations in two types of tumors. Additionally, we found tyrosinase-related protein1 (TYRP1) was negatively correlated with CD8A expression. In vitro experiments showed that knockdown TYRP1 promoted the expression of HLA-A, B, and C. Eventually, we constructed a prediction model which was validated in our external cohort. Notably, this model also performed effectively in predicting the survival of patients under immunotherapy. In summary, this work provides a deeper understanding of the state of immune infiltration in melanoma and a prediction model that might guide the clinical treatment of patients with melanoma.
Collapse
Affiliation(s)
- Gang Li
- Plastic Surgery, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | | |
Collapse
|
19
|
Malvehy J, Samoylenko I, Schadendorf D, Gutzmer R, Grob JJ, Sacco JJ, Gorski KS, Anderson A, Pickett CA, Liu K, Gogas H. Talimogene laherparepvec upregulates immune-cell populations in non-injected lesions: findings from a phase II, multicenter, open-label study in patients with stage IIIB-IVM1c melanoma. J Immunother Cancer 2021; 9:jitc-2020-001621. [PMID: 33785610 PMCID: PMC8011715 DOI: 10.1136/jitc-2020-001621] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2021] [Indexed: 11/20/2022] Open
Abstract
Background Talimogene laherparepvec (T-VEC), an oncolytic virus, was designed to selectively replicate in and lyse tumor cells, releasing tumor-derived antigen to stimulate a tumor-specific immune response. Methods In this phase II study in patients with unresectable stage IIIB–IV melanoma, we evaluated non-injected lesions to establish whether baseline or change in intratumoral CD8+ T-cell density (determined using immunohistochemistry) correlated with T-VEC clinical response. Results Of 112 enrolled patients, 111 received ≥1 dose of T-VEC. After a median follow-up of 108.0 weeks, objective/complete response rates were 28%/14% in the overall population and 32%/18% in patients with stage IIIB–IVM1a disease. No unexpected toxicity occurred. Baseline and week 6 change from baseline CD8+ T-cell density results were available for 91 and 65 patients, respectively. Neither baseline nor change in CD8+ T-cell density correlated with objective response rate, changes in tumor burden, duration of response or durable response rate. However, a 2.4-fold median increase in CD8+ T-cell density in non-injected lesions from baseline to week 6 was observed. In exploratory analyses, multiparameter immunofluorescence showed that after treatment there was an increase in the proportion of infiltrating CD8+ T-cells expressing granzyme B and checkpoint markers (programmed death-1, programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte antigen-4) in non-injected lesions, together with an increase in helper T-cells. Consistent with T-cell infiltrate, we observed an increase in the adaptive resistance marker PD-L1 in non-injected lesions. Conclusions This study indicates that T-VEC induces systemic immune activity and alters the tumor microenvironment in a way that will likely enhance the effects of other immunotherapy agents in combination therapy. Trial registration number NCT02366195.
Collapse
Affiliation(s)
- Josep Malvehy
- Dermatology Department and IDIBAPS, Hospital Clinic of Barcelona, Barcelona, Catalunya, Spain
| | - Igor Samoylenko
- NN Blokhin Russian Cancer Research Center, Moscow, Russian Federation
| | - Dirk Schadendorf
- Department of Dermatology, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Ralf Gutzmer
- Department of Dermatology and Allergy, Skin Cancer Center Hannover, Hannover Medical School, Hannover, Germany
| | | | - Joseph J Sacco
- Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, Wirral, UK.,University of Liverpool, Liverpool, UK
| | | | | | | | - Kate Liu
- Amgen Inc, Thousand Oaks, California, USA
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
20
|
Willsmore ZN, Coumbe BGT, Crescioli S, Reci S, Gupta A, Harris RJ, Chenoweth A, Chauhan J, Bax HJ, McCraw A, Cheung A, Osborn G, Hoffmann RM, Nakamura M, Laddach R, Geh JLC, MacKenzie-Ross A, Healy C, Tsoka S, Spicer JF, Josephs DH, Papa S, Lacy KE, Karagiannis SN. Combined anti-PD-1 and anti-CTLA-4 checkpoint blockade: Treatment of melanoma and immune mechanisms of action. Eur J Immunol 2021; 51:544-556. [PMID: 33450785 DOI: 10.1002/eji.202048747] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/23/2020] [Indexed: 12/19/2022]
Abstract
Cytotoxic T-lymphocyte associated protein-4 (CTLA-4) and the Programmed Death Receptor 1 (PD-1) are immune checkpoint molecules that are well-established targets of antibody immunotherapies for the management of malignant melanoma. The monoclonal antibodies, Ipilimumab, Pembrolizumab, and Nivolumab, designed to interfere with T cell inhibitory signals to activate immune responses against tumors, were originally approved as monotherapy. Treatment with a combination of immune checkpoint inhibitors may improve outcomes compared to monotherapy in certain patient groups and these clinical benefits may be derived from unique immune mechanisms of action. However, treatment with checkpoint inhibitor combinations also present significant clinical challenges and increased rates of immune-related adverse events. In this review, we discuss the potential mechanisms attributed to single and combined checkpoint inhibitor immunotherapies and clinical experience with their use.
Collapse
Affiliation(s)
- Zena N Willsmore
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
| | - Ben G T Coumbe
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
| | - Silvia Crescioli
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
| | - Sara Reci
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
| | - Ayushi Gupta
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
| | - Robert J Harris
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
| | - Alicia Chenoweth
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Jitesh Chauhan
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
| | - Heather J Bax
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
- School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Alexa McCraw
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
| | - Anthony Cheung
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Gabriel Osborn
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
| | - Ricarda M Hoffmann
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
| | - Mano Nakamura
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
| | - Roman Laddach
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London, United Kingdom
| | - Jenny L C Geh
- Department of Plastic Surgery at Guy's, King's, and St. Thomas' Hospitals, London, United Kingdom
| | - Alastair MacKenzie-Ross
- Department of Plastic Surgery at Guy's, King's, and St. Thomas' Hospitals, London, United Kingdom
| | - Ciaran Healy
- Department of Plastic Surgery at Guy's, King's, and St. Thomas' Hospitals, London, United Kingdom
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London, United Kingdom
| | - James F Spicer
- School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Debra H Josephs
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
- School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Sophie Papa
- Department of Medical Oncology, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom
- ImmunoEngineering, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Katie E Lacy
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, United Kingdom
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| |
Collapse
|
21
|
Cancer Stem Cells-Key Players in Tumor Relapse. Cancers (Basel) 2021; 13:cancers13030376. [PMID: 33498502 PMCID: PMC7864187 DOI: 10.3390/cancers13030376] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/10/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
Tumor relapse and treatment failure are unfortunately common events for cancer patients, thus often rendering cancer an uncurable disease. Cancer stem cells (CSCs) are a subset of cancer cells endowed with tumor-initiating and self-renewal capacity, as well as with high adaptive abilities. Altogether, these features contribute to CSC survival after one or multiple therapeutic approaches, thus leading to treatment failure and tumor progression/relapse. Thus, elucidating the molecular mechanisms associated with stemness-driven resistance is crucial for the development of more effective drugs and durable responses. This review will highlight the mechanisms exploited by CSCs to overcome different therapeutic strategies, from chemo- and radiotherapies to targeted therapies and immunotherapies, shedding light on their plasticity as an insidious trait responsible for their adaptation/escape. Finally, novel CSC-specific approaches will be described, providing evidence of their preclinical and clinical applications.
Collapse
|
22
|
Meneveau MO, Sahli ZT, Lynch KT, Mauldin IS, Slingluff CL. Immunotyping and Quantification of Melanoma Tumor-Infiltrating Lymphocytes. Methods Mol Biol 2021; 2265:515-528. [PMID: 33704737 DOI: 10.1007/978-1-0716-1205-7_36] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The density of tumour-infiltrating lymphocytes (TILs) in melanoma is correlated with improved clinical prognosis; however, standardized TIL immunotyping and quantification protocols are lacking. Herein, we provide a review of the technologies being utilized for the immunotyping and quantification of melanoma TILs.
Collapse
Affiliation(s)
- Max O Meneveau
- Department of Surgery, The University of Virginia Health System, Charlottesville, VA, USA
| | - Zeyad T Sahli
- Department of Surgery, The University of Virginia Health System, Charlottesville, VA, USA
| | - Kevin T Lynch
- Department of Surgery, The University of Virginia Health System, Charlottesville, VA, USA
| | - Ileana S Mauldin
- Department of Surgery, The University of Virginia Health System, Charlottesville, VA, USA
| | - Craig L Slingluff
- Department of Surgery, The University of Virginia Health System, Charlottesville, VA, USA.
| |
Collapse
|
23
|
|
24
|
Poran A, Scherer J, Bushway ME, Besada R, Balogh KN, Wanamaker A, Williams RG, Prabhakara J, Ott PA, Hu-Lieskovan S, Khondker ZS, Gaynor RB, Rooney MS, Srinivasan L. Combined TCR Repertoire Profiles and Blood Cell Phenotypes Predict Melanoma Patient Response to Personalized Neoantigen Therapy plus Anti-PD-1. CELL REPORTS MEDICINE 2020; 1:100141. [PMID: 33294862 PMCID: PMC7691446 DOI: 10.1016/j.xcrm.2020.100141] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/02/2020] [Accepted: 10/22/2020] [Indexed: 01/15/2023]
Abstract
T cells use highly diverse receptors (TCRs) to identify tumor cells presenting neoantigens arising from genetic mutations and establish anti-tumor activity. Immunotherapy harnessing neoantigen-specific T cells to target tumors has emerged as a promising clinical approach. To assess whether a comprehensive peripheral mononuclear blood cell analysis predicts responses to a personalized neoantigen cancer vaccine combined with anti-PD-1 therapy, we characterize the TCR repertoires and T and B cell frequencies in 21 patients with metastatic melanoma who received this regimen. TCR-α/β-chain sequencing reveals that prolonged progression-free survival (PFS) is strongly associated with increased clonal baseline TCR repertoires and longitudinal repertoire stability. Furthermore, the frequencies of antigen-experienced T and B cells in the peripheral blood correlate with repertoire characteristics. Analysis of these baseline immune features enables prediction of PFS following treatment. This method offers a pragmatic clinical approach to assess patients’ immune state and to direct therapeutic decision making. Pre-treatment blood-based factors predict response to immunotherapy TCR repertoire clonality and stability associate with improved clinical outcomes Baseline T and B cell memory phenotypes associate with improved clinical outcomes Combined baseline TCR repertoire and PBMC phenotypes predict immunotherapy response
Collapse
Affiliation(s)
- Asaf Poran
- Neon Therapeutics/BioNTech US, Cambridge, MA, USA
- Corresponding author
| | | | | | - Rana Besada
- Neon Therapeutics/BioNTech US, Cambridge, MA, USA
| | | | | | | | | | - Patrick A. Ott
- Dana Farber Cancer Institute, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA, USA
| | - Siwen Hu-Lieskovan
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | | | | | | | |
Collapse
|
25
|
Arasu UT, Deen AJ, Pasonen-Seppänen S, Heikkinen S, Lalowski M, Kärnä R, Härkönen K, Mäkinen P, Lázaro-Ibáñez E, Siljander PRM, Oikari S, Levonen AL, Rilla K. HAS3-induced extracellular vesicles from melanoma cells stimulate IHH mediated c-Myc upregulation via the hedgehog signaling pathway in target cells. Cell Mol Life Sci 2020; 77:4093-4115. [PMID: 31820036 PMCID: PMC7532973 DOI: 10.1007/s00018-019-03399-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 11/11/2019] [Accepted: 11/26/2019] [Indexed: 12/17/2022]
Abstract
Intercellular communication is fundamental to the survival and maintenance of all multicellular systems, whereas dysregulation of communication pathways can drive cancer progression. Extracellular vesicles (EVs) are mediators of cell-to-cell communication that regulate a variety of cellular processes involved in tumor progression. Overexpression of a specific plasma membrane enzyme, hyaluronan synthase 3 (HAS3), is one of the factors that can induce EV shedding. HAS3, and particularly its product hyaluronan (HA), are carried by EVs and are known to be associated with the tumorigenic properties of cancer cells. To elucidate the specific effects of cancerous, HAS3-induced EVs on target cells, normal human keratinocytes and melanoma cells were treated with EVs derived from GFP-HAS3 expressing metastatic melanoma cells. We found that the HA receptor CD44 participated in the regulation of EV binding to target cells. Furthermore, GFP-HAS3-positive EVs induced HA secretion, proliferation and invasion of target cells. Our results suggest that HAS3-EVs contains increased quantities of IHH, which activates the target cell hedgehog signaling cascade and leads to the activation of c-Myc and regulation of claspin expression. This signaling of IHH in HAS3-EVs resulted in increased cell proliferation. Claspin immunostaining correlated with HA content in human cutaneous melanocytic lesions, supporting our in vitro findings and suggesting a reciprocal regulation between claspin expression and HA synthesis. This study shows for the first time that EVs originating from HAS3 overexpressing cells carry mitogenic signals that induce proliferation and epithelial-to-mesenchymal transition in target cells. The study also identifies a novel feedback regulation between the hedgehog signaling pathway and HA metabolism in melanoma, mediated by EVs carrying HA and IHH.
Collapse
Affiliation(s)
- Uma Thanigai Arasu
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.
| | - Ashik Jawahar Deen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | - Sami Heikkinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Maciej Lalowski
- Faculty of Medicine, Biochemistry and Developmental Biology, Meilahti Clinical Proteomics Core Facility, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Riikka Kärnä
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Kai Härkönen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Petri Mäkinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Elisa Lázaro-Ibáñez
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Centre for Drug Research, University of Helsinki, Helsinki, Finland
| | - Pia R-M Siljander
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Centre for Drug Research, University of Helsinki, Helsinki, Finland
- EV Group and EV Core, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Sanna Oikari
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kirsi Rilla
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
26
|
Shen H, Liu T, Cui J, Borole P, Benjamin A, Kording K, Issadore D. A web-based automated machine learning platform to analyze liquid biopsy data. LAB ON A CHIP 2020; 20:2166-2174. [PMID: 32420563 DOI: 10.1039/d0lc00096e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Liquid biopsy (LB) technologies continue to improve in sensitivity, specificity, and multiplexing and can measure an ever growing library of disease biomarkers. However, clinical interpretation of the increasingly large sets of data these technologies generate remains a challenge. Machine learning is a popular approach to discover and detect signatures of disease. However, limited machine learning expertise in the LB field has kept the discipline from fully leveraging these tools and risks improper analyses and irreproducible results. In this paper, we develop a web-based automated machine learning tool tailored specifically for LB, where machine learning models can be built without the user's input. We also incorporate a differential privacy algorithm, designed to limit the effects of overfitting that can arise from users iteratively developing a panel with feedback from our platform. We validate our approach by performing a meta-analysis on 11 published LB datasets, and found that we had similar or better performance compared to those reported in the literature. Moreover, we show that our platform's performance improved when incorporating information from prior LB datasets, suggesting that this approach can continue to improve with increased access to LB data. Finally, we show that by using our platform the results achieved in the literature can be matched using 40% of the number of subjects in the training set, potentially reducing study cost and time. This self-improving and overfitting-resistant automatic machine learning platform provides a new standard that can be used to validate machine learning works in the LB field.
Collapse
Affiliation(s)
- Hanfei Shen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Wistuba-Hamprecht K, Gouttefangeas C, Weide B, Pawelec G. Immune Signatures and Survival of Patients With Metastatic Melanoma, Renal Cancer, and Breast Cancer. Front Immunol 2020; 11:1152. [PMID: 32582215 PMCID: PMC7296133 DOI: 10.3389/fimmu.2020.01152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/11/2020] [Indexed: 12/27/2022] Open
Abstract
Despite remarkable recent progress in treating solid cancers, especially the success of immunomodulatory antibody therapies for numerous different cancer types, it remains the case that many patients fail to respond to treatment. It is therefore of immense importance to identify biomarkers predicting clinical responses to treatment and patient survival, which would not only assist in targeting treatments to patients most likely to benefit, but might also provide mechanistic insights into the reasons for success or failure of the therapy. Several peripheral blood or tumor tissue diagnostic and predictive biomarkers known to be informative for cancer patient survival may be applicable for this purpose. The use of peripheral blood ("liquid biopsy") offers numerous advantages not only for predicting treatment responses at baseline but also for monitoring patients on-therapy. Assessment of the tumor microenvironment and infiltrating immune cells also delivers important information on cancer-host interactions but the requirement for tumor tissues makes this more challenging, especially for monitoring sequential changes in the individual patient. In this contribution, we will review our findings on immune signatures potentially informative for clinical outcome in melanoma, breast cancer and renal cell carcinoma, particularly the outcome of checkpoint blockade, by applying multiparametric flow cytometry and mass cytometry, routine clinical monitoring and functional testing for predicting and following individual patient responses to therapy.
Collapse
Affiliation(s)
- Kilian Wistuba-Hamprecht
- Division of Dermatooncology, Department of Dermatology, University Medical Centre Tübingen, Tübingen, Germany
- Immunoguiding Workgroup of the Cancer Immunotherapy Association (CIP/CIMT), Mainz, Germany
| | - Cécile Gouttefangeas
- Immunoguiding Workgroup of the Cancer Immunotherapy Association (CIP/CIMT), Mainz, Germany
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
- Germany and German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Benjamin Weide
- Division of Dermatooncology, Department of Dermatology, University Medical Centre Tübingen, Tübingen, Germany
| | - Graham Pawelec
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
- Germany and German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Partner Site Tübingen, Tübingen, Germany
- Health Sciences North Research Institute, Sudbury, ON, Canada
| |
Collapse
|
28
|
Welsh SJ, Corrie PG. Biomarkers Predicting for Response and Relapse with Melanoma Systemic Therapy. Acta Derm Venereol 2020; 100:adv00142. [PMID: 32346748 PMCID: PMC9189745 DOI: 10.2340/00015555-3497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2020] [Indexed: 11/16/2022] Open
Abstract
For all primary cutaneous squamous cell carcinomas (cSCCs), physical examination should include full skin examination, recording of tumour diameter and regional lymph-node-basin status. Surgery is the treatment of choice, with a minimal 5-mm margin. For elderly patients with well-differentiated tumours, other surgical modalities can be explored. Surgery for organ-transplant recipients should not be delayed. The issue with cSCC is identifying high-risk tumours with staging, as this may alter treatment and follow-up schedules. Adjuvant radiation therapy should be considered for incomplete resection, when re-excision is impossible or there are poor-prognosis histological findings. Recommendations are at least biannual dermatological surveillance for 2 years, but in elderly patients with small, well-differentiated tumours long-term follow-up is not always necessary. In case of positive lymph nodes, radical dissection is needed, with regional postoperative adjuvant radiation. Advanced cSCCs are defined as unresectable local, regional or distant disease requiring systemic treatment. Their only approved treat-ment is the PD-1 inhibitor, cemiplimab. Trials evaluating adjuvant or neo-adjuvant anti-PD-1 are ongoing. Platin-based chemo or anti-epidermal growth-factor-receptor therapies are possible second-line treatments. For transplant patients, minimizing immunosuppression and switching to sirolimus must be considered at first appearance of cSCC.
Collapse
|
29
|
Liang J, Chen D, Chen L, She X, Zhang H, Xiao Y. The potentiality of immunotherapy for sarcomas: a summary of potential predictive biomarkers. Future Oncol 2020; 16:1211-1223. [PMID: 32396026 DOI: 10.2217/fon-2020-0118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Sarcomas are rare and heterogeneous malignant tumors of mesenchymal origin. A total of 25-50% of patients treated with initial curative intent will develop as recurrent and metastatic disease. In the recurrent and metastatic setting, effect of chemotherapy is limited; therefore, more effective therapies are urgently desired. As a brake for activation of T cell, PD-1/PD-L1 plays a crucial role in the progression of tumor by altering status of immune surveillance. Some success has been acquired recently in the use of PD-1/PD-L1 inhibitors for the treatment of several solid tumors, for examples, non-small-cell lung cancer and melanoma. Immunotherapeutic strategies based on PD-1/PD-L1 for sarcomas have also been explored these years. As in other cancers, major challenges are identification of biomarkers to predict response for immunotherapy, optimization of patient's benefit and minimization of side effects. Therefore, we focused on potential biomarkers of immunotherapy for treatment of sarcomas in this review.
Collapse
Affiliation(s)
- Jin Liang
- Department of Medical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan province 650032, PR China
| | - Dedian Chen
- Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan, Kunming, Yunnan 650118, PR China
| | - Liyao Chen
- Department of Radiotherapy, The First People's Hospital of Yuxi City. Yuxi, Yunnan province 653100, PR China
| | - Xueke She
- The Medical Department, 3D Medicines Inc., Shanghai, 201114, PR China
| | - Hushan Zhang
- The Medical Department, 3D Medicines Inc., Shanghai, 201114, PR China.,Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, PR China
| | - Yanbin Xiao
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan, Kunming, Yunnan province 650118, PR China
| |
Collapse
|
30
|
Basler L, Gabryś HS, Hogan SA, Pavic M, Bogowicz M, Vuong D, Tanadini-Lang S, Förster R, Kudura K, Huellner MW, Dummer R, Guckenberger M, Levesque MP. Radiomics, Tumor Volume, and Blood Biomarkers for Early Prediction of Pseudoprogression in Patients with Metastatic Melanoma Treated with Immune Checkpoint Inhibition. Clin Cancer Res 2020; 26:4414-4425. [PMID: 32253232 DOI: 10.1158/1078-0432.ccr-20-0020] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 03/09/2020] [Accepted: 04/01/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE We assessed the predictive potential of positron emission tomography (PET)/CT-based radiomics, lesion volume, and routine blood markers for early differentiation of pseudoprogression from true progression at 3 months. EXPERIMENTAL DESIGN 112 patients with metastatic melanoma treated with immune checkpoint inhibition were included in our study. Median follow-up duration was 22 months. 716 metastases were segmented individually on CT and 2[18F]fluoro-2-deoxy-D-glucose (FDG)-PET imaging at three timepoints: baseline (TP0), 3 months (TP1), and 6 months (TP2). Response was defined on a lesion-individual level (RECIST 1.1) and retrospectively correlated with FDG-PET/CT radiomic features and the blood markers LDH/S100. Seven multivariate prediction model classes were generated. RESULTS Two-year (median) overall survival, progression-free survival, and immune progression-free survival were 69% (not reached), 24% (6 months), and 42% (16 months), respectively. At 3 months, 106 (16%) lesions had progressed, of which 30 (5%) were identified as pseudoprogression at 6 months. Patients with pseudoprogressive lesions and without true progressive lesions had a similar outcome to responding patients and a significantly better 2-year overall survival of 100% (30 months), compared with 15% (10 months) in patients with true progressions/without pseudoprogression (P = 0.002). Patients with mixed progressive/pseudoprogressive lesions were in between at 53% (25 months). The blood prediction model (LDH+S100) achieved an AUC = 0.71. Higher LDH/S100 values indicated a low chance of pseudoprogression. Volume-based models: AUC = 0.72 (TP1) and AUC = 0.80 (delta-volume between TP0/TP1). Radiomics models (including/excluding volume-related features): AUC = 0.79/0.78. Combined blood/volume model: AUC = 0.79. Combined blood/radiomics model (including volume-related features): AUC = 0.78. The combined blood/radiomics model (excluding volume-related features) performed best: AUC = 0.82. CONCLUSIONS Noninvasive PET/CT-based radiomics, especially in combination with blood parameters, are promising biomarkers for early differentiation of pseudoprogression, potentially avoiding added toxicity or delayed treatment switch.
Collapse
Affiliation(s)
- Lucas Basler
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Hubert S Gabryś
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sabrina A Hogan
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matea Pavic
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marta Bogowicz
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Diem Vuong
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Robert Förster
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ken Kudura
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Martin W Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
31
|
Li CB, Song LR, Li D, Weng JC, Zhang LW, Zhang JT, Wu Z. Primary intracranial malignant melanoma: proposed treatment protocol and overall survival in a single-institution series of 15 cases combined with 100 cases from the literature. J Neurosurg 2020; 132:902-913. [PMID: 30835686 DOI: 10.3171/2018.11.jns181872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/06/2018] [Indexed: 01/04/2023]
Abstract
OBJECTIVE The overall survival and pertinent adverse factors for primary intracranial malignant melanoma (PIMM) have not been previously determined. This aim of this study was to determine the rates of progression-free survival (PFS) and overall survival (OS) and identify the adverse factors for PIMM. METHODS This study included 15 cases from the authors' own series and 100 cases with detailed clinical data that were obtained from the literature from 1914 to 2018 using the Ovid Medline, EMBASE, PubMed, Cochrane, and EBSCO databases. Patient demographics, treatment (surgery, chemotherapy, and radiotherapy [RT]), PFS, and OS were reviewed. Data from prior publications were processed and used according to PRISMA guidelines. RESULTS Diffuse lesions were identified in 24 (20.9%) patients, who had a younger age (p < 0.001). The mean follow-up time was 16.6 months, and 76 (66.1%) deaths occurred. The 6-month, 1-year, 3-year, and 5-year OS rates of the whole cohort were 62.8%, 49.9%, 28.9%, and 17.2%, respectively, with an estimated median survival time (EMST) of 12.0 months. The multivariate analysis revealed that gross-total resection (GTR) (HR 0.299, 95% CI 0.180-0.497, p < 0.001), radiotherapy (HR 0.577, 95% CI 0.359-0.929, p = 0.024), and chemotherapy (HR 0.420, 95% CI 0.240-0.735, p = 0.002) predicted a better OS. The EMST was 5.0 months in patients with diffuse-type PIMM and 13.0 months in patients with the solitary type. Patients receiving GTR with adjuvant RT and/or chemotherapy (GTR + [RT and/or chemo]) had significantly higher 1-year and 5-year OS rates (73.0% and 40.1%, respectively) and a longer EMST (53 months) than patients who underwent GTR alone (20.5 months) or RT and/or chemotherapy without GTR (13.0 months). CONCLUSIONS Optimal outcomes could be achieved by radical resection plus postoperative radiotherapy and/or chemotherapy. Patients with diffuse PIMM have a more severe clinical spectrum and poorer survival than patients with solitary PIMM. Immunotherapy and targeted therapy show promise as treatment options for PIMM based on results in patients with brain metastases from extracranial melanoma.
Collapse
|
32
|
Vetma V, Guttà C, Peters N, Praetorius C, Hutt M, Seifert O, Meier F, Kontermann R, Kulms D, Rehm M. Convergence of pathway analysis and pattern recognition predicts sensitization to latest generation TRAIL therapeutics by IAP antagonism. Cell Death Differ 2020; 27:2417-2432. [PMID: 32081986 PMCID: PMC7370234 DOI: 10.1038/s41418-020-0512-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 12/28/2022] Open
Abstract
Second generation TRAIL-based therapeutics, combined with sensitising co-treatments, have recently entered clinical trials. However, reliable response predictors for optimal patient selection are not yet available. Here, we demonstrate that a novel and translationally relevant hexavalent TRAIL receptor agonist, IZI1551, in combination with Birinapant, a clinically tested IAP antagonist, efficiently induces cell death in various melanoma models, and that responsiveness can be predicted by combining pathway analysis, data-driven modelling and pattern recognition. Across a panel of 16 melanoma cell lines, responsiveness to IZI1551/Birinapant was heterogeneous, with complete resistance and pronounced synergies observed. Expression patterns of TRAIL pathway regulators allowed us to develop a combinatorial marker that predicts potent cell killing with high accuracy. IZI1551/Birinapant responsiveness could be predicted not only for cell lines, but also for 3D tumour cell spheroids and for cells directly isolated from patient melanoma metastases (80–100% prediction accuracies). Mathematical parameter reduction identified 11 proteins crucial to ensure prediction accuracy, with x-linked inhibitor of apoptosis protein (XIAP) and procaspase-3 scoring highest, and Bcl-2 family members strongly represented. Applied to expression data of a cohort of n = 365 metastatic melanoma patients in a proof of concept in silico trial, the predictor suggested that IZI1551/Birinapant responsiveness could be expected for up to 30% of patient tumours. Overall, response frequencies in melanoma models were very encouraging, and the capability to predict melanoma sensitivity to combinations of latest generation TRAIL-based therapeutics and IAP antagonists can address the need for patient selection strategies in clinical trials based on these novel drugs.
Collapse
Affiliation(s)
- Vesna Vetma
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Cristiano Guttà
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Nathalie Peters
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Christian Praetorius
- Center for Regenerative Therapies, Technical University Dresden, Dresden, Germany.,Skin Cancer Center at the University Cancer Centre, Department of Dermatology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Meike Hutt
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Oliver Seifert
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Friedegund Meier
- Skin Cancer Center at the University Cancer Centre, Department of Dermatology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Roland Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Dagmar Kulms
- Center for Regenerative Therapies, Technical University Dresden, Dresden, Germany.,Skin Cancer Center at the University Cancer Centre, Department of Dermatology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany.,Experimental Dermatology, Department of Dermatology, Technical University Dresden, Dresden, Germany
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany. .,Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland. .,Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany. .,Stuttgart Centre for Simulation Science (SC SimTech), University of Stuttgart, Stuttgart, Germany. .,Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.
| |
Collapse
|
33
|
Detection of Gene Mutations in Liquid Biopsy of Melanoma Patients: Overview and Future Perspectives. Curr Treat Options Oncol 2020; 21:19. [PMID: 32048063 DOI: 10.1007/s11864-020-0708-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OPINION STATEMENT Liquid biopsies are still far from widely implanted in the clinical arena. Issues related to the added sensitivity of this test beyond conventional methods have not been fully resolved. Additionally, issues related to the specificity of these results especially as many cancers may share common mutation need further investigations. One way to resolve this may include the development and testing of large gene panels to add higher specificity. On the other hand, further studies are needed to support the idea that ctDNA or circulating tumor cells may constitute a better representation of the tumor subpopulation that is capable of metastasizing, which will strongly support its clinical value. Finally, survival studies showing a positive impact of this technology will also justify its widespread implementation in clinical practice.
Collapse
|
34
|
Pruessmann W, Rytlewski J, Wilmott J, Mihm MC, Attrill GH, Dyring-Andersen B, Fields P, Zhan Q, Colebatch AJ, Ferguson PM, Thompson JF, Kallenbach K, Yusko E, Clark RA, Robins H, Scolyer RA, Kupper TS. Molecular analysis of primary melanoma T cells identifies patients at risk for metastatic recurrence. NATURE CANCER 2020; 1:197-209. [PMID: 33305293 PMCID: PMC7725220 DOI: 10.1038/s43018-019-0019-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/10/2019] [Indexed: 12/25/2022]
Abstract
Primary melanomas >1 mm thickness are potentially curable by resection, but can recur metastatically. We assessed the prognostic value of T cell fraction (TCFr) and repertoire T cell clonality, measured by high-throughput-sequencing of the T cell receptor beta chain (TRB) in T2-T4 primary melanomas (n=199). TCFr accurately predicted progression-free survival (PFS) and was independent of thickness, ulceration, mitotic rate, or age. TCFr was second only to tumor thickness in its predictive value, using a gradient boosted model. For accurate PFS prediction, adding TCFr to tumor thickness was superior to adding any other histopathological variable. Furthermore, a TCFr >20% was protective regardless of tumor ulceration status, mitotic rate or presence of nodal disease. TCFr is a quantitative molecular assessment that predicts metastatic recurrence in primary melanoma patients whose disease has been resected surgically. This study suggests that a successful T cell-mediated antitumor response can be present in primary melanomas.
Collapse
Affiliation(s)
- Wiebke Pruessmann
- Department of Dermatology and Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
- Department of Dermatology, University of Luebeck, Luebeck, Germany
| | | | - James Wilmott
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
| | - Martin C Mihm
- Department of Dermatology and Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Grace H Attrill
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
| | - Beatrice Dyring-Andersen
- Department of Dermatology and Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
- Novo Nordisk Foundation Centre for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Qian Zhan
- Department of Dermatology and Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Andrew J Colebatch
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Peter M Ferguson
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - John F Thompson
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Klaus Kallenbach
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Erik Yusko
- Adaptive Biotechnologies, Seattle, WA, USA
| | - Rachael A Clark
- Department of Dermatology and Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Harlan Robins
- Adaptive Biotechnologies, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Richard A Scolyer
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Thomas S Kupper
- Department of Dermatology and Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
35
|
Kaina B. Temozolomide in Glioblastoma Therapy: Role of Apoptosis, Senescence and Autophagy. Comment on Strobel et al., Temozolomide and Other Alkylating Agents in Glioblastoma Therapy. Biomedicines 2019, 7, 69. Biomedicines 2019; 7:biomedicines7040090. [PMID: 31717973 PMCID: PMC6966492 DOI: 10.3390/biomedicines7040090] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/02/2019] [Accepted: 11/05/2019] [Indexed: 01/16/2023] Open
Abstract
Temozolomide, a DNA methylating drug, is currently being used first-line in glioblastoma therapy. Although the mode of action of this so-called SN1 alkylating agent is well described, including the types of induced DNA damage triggering the DNA damage response and survival and death pathways, some researchers expressed doubt that data mostly obtained by in vitro models can be translated into the in vivo situation. In experimental settings, high doses of the agent are often used, which are likely to activate responses triggered by base N-alkylations instead of O6-methylguanine (O6MeG), which is the primary cytotoxic lesion induced by low doses of temozolomide and other methylating drugs in O6-methylguanine-DNA methyltransferase (MGMT) repair incompetent cells. However, numerous studies provided compelling evidence that O6MeG is not only a mutagenic, but also a powerful toxic lesion inducing DNA double-strand breaks, apoptosis, autophagy and cellular senescence. MGMT, repairing the lesion through methyl group transfer, is a key node in protecting cells against all these effects and has a significant impact on patient’s survival following temozolomide therapy, supporting the notion that findings obtained on a molecular and cellular level can be translated to the therapeutic setting in vivo. This comment summarizes the current knowledge on O6MeG-triggered pathways, including dose dependence and the question of thresholds, and comes up with the conclusion that data obtained on cell lines using low dose protocols are relevant and apoptosis, autophagy and senescence are therapeutically important endpoints.
Collapse
Affiliation(s)
- Bernd Kaina
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 65, D-55130 Mainz, Germany
| |
Collapse
|
36
|
Abolhassani AR, Schuler G, Kirchberger MC, Heinzerling L. C-reactive protein as an early marker of immune-related adverse events. J Cancer Res Clin Oncol 2019; 145:2625-2631. [PMID: 31492984 DOI: 10.1007/s00432-019-03002-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 08/09/2019] [Indexed: 01/08/2023]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) are effective against a wide variety of cancers. However, they also induce a plethora of unique immune-related adverse events (irAEs). Since for many organ systems symptoms can be unspecific, differential diagnosis with progression of disease or infection may be difficult. C-reactive protein (CRP) has been suggested as a marker for infection. The purpose of this study was to evaluate the diagnostic value of CRP in differentiating infectious causes from autoimmune side effects induced by ICIs. METHODS In order to investigate the role of CRP in irAEs, we screened our patient data base. Only events with full infectious workup were included. In total 88 events of irAEs in 37 melanoma patients were analyzed. CRP levels before and during irAEs were evaluated. Statistical analyses were conducted using the Chi-square test for categorical variables. RESULTS At the onset of irAE, CRP rose in 93% of cases to a mean of 52.7 mg/L (CI 35.1-70.3) from 8.4 mg/L at baseline (normal < 5 mg/L) (P < 0.0001). Other causes of CRP elevation including infectious diseases were excluded, and procalcitonin (PCT) levels were normal in 92% of events. Importantly, in 42% of cases CRP elevations preceded clinical symptoms. CONCLUSION CRP elevation can predict the onset of irAEs in patients treated with ICIs in the absence of infectious disease.
Collapse
Affiliation(s)
- Amir-Reza Abolhassani
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Ulmenweg 18, 91054, Erlangen, Germany
| | - Gerold Schuler
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Ulmenweg 18, 91054, Erlangen, Germany
| | - Michael Constantin Kirchberger
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Ulmenweg 18, 91054, Erlangen, Germany
| | - Lucie Heinzerling
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Ulmenweg 18, 91054, Erlangen, Germany.
| |
Collapse
|
37
|
de Assis LVM, Moraes MN, Castrucci AMDL. The molecular clock in the skin, its functionality, and how it is disrupted in cutaneous melanoma: a new pharmacological target? Cell Mol Life Sci 2019; 76:3801-3826. [PMID: 31222374 PMCID: PMC11105295 DOI: 10.1007/s00018-019-03183-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/13/2019] [Accepted: 06/05/2019] [Indexed: 02/07/2023]
Abstract
The skin is the interface between the organism and the external environment, acting as its first barrier. Thus, this organ is constantly challenged by physical stimuli such as UV and infrared radiation, visible light, and temperature as well as chemicals and pathogens. To counteract the deleterious effects of the above-mentioned stimuli, the skin has complex defense mechanisms such as: immune and neuroendocrine systems; shedding of epidermal squamous layers and apoptosis of damaged cells; DNA repair; and pigmentary system. Here we have reviewed the current knowledge regarding which stimuli affect the molecular clock of the skin, the consequences to skin-related biological processes and, based on such knowledge, we suggest some therapeutic targets. We also explored the recent advances regarding the molecular clock disruption in melanoma, its impact on the carcinogenic process, and its therapeutic value in melanoma treatment.
Collapse
Affiliation(s)
- Leonardo Vinícius Monteiro de Assis
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, R. do Matão, Trav. 14, No. 101, São Paulo, 05508-090, Brazil
| | - Maria Nathalia Moraes
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, R. do Matão, Trav. 14, No. 101, São Paulo, 05508-090, Brazil
- School of Health Science, University Anhembi Morumbi, São Paulo, Brazil
| | - Ana Maria de Lauro Castrucci
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, R. do Matão, Trav. 14, No. 101, São Paulo, 05508-090, Brazil.
| |
Collapse
|
38
|
Bobrowicz M, Zagozdzon R, Domagala J, Vasconcelos-Berg R, Guenova E, Winiarska M. Monoclonal Antibodies in Dermatooncology-State of the Art and Future Perspectives. Cancers (Basel) 2019; 11:E1420. [PMID: 31554169 PMCID: PMC6826541 DOI: 10.3390/cancers11101420] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/08/2019] [Accepted: 09/17/2019] [Indexed: 12/21/2022] Open
Abstract
Monoclonal antibodies (mAbs) targeting specific proteins are currently the most popular form of immunotherapy used in the treatment of cancer and other non-malignant diseases. Since the first approval of anti-CD20 mAb rituximab in 1997 for the treatment of B-cell malignancies, the market is continuously booming and the clinically used mAbs have undergone a remarkable evolution. Novel molecular targets are constantly emerging and the development of genetic engineering have facilitated the introduction of modified mAbs with improved safety and increased capabilities to activate the effector mechanisms of the immune system. Next to their remarkable success in hematooncology, mAbs have also an already established role in the treatment of solid malignancies. The recent development of mAbs targeting the immune checkpoints has opened new avenues for the use of this form of immunotherapy, also in the immune-rich milieu of the skin. In this review we aim at presenting a comprehensive view of mAbs' application in the modern treatment of skin cancer. We present the characteristics and efficacy of mAbs currently used in dermatooncology and summarize the recent clinical trials in the field. We discuss the side effects and strategies for their managing.
Collapse
Affiliation(s)
| | - Radoslaw Zagozdzon
- Department of Clinical Immunology, Medical University of Warsaw, 02-006 Warsaw, Poland.
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, 02-006 Warsaw, Poland.
| | - Joanna Domagala
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland.
- Postgraduate School of Molecular Medicine, 02-091 Warsaw, Poland.
| | - Roberta Vasconcelos-Berg
- Department of Dermatology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland.
| | - Emmanuella Guenova
- Department of Dermatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland.
- Department of Dermatology, University of Lausanne, 1011 Lausanne, Switzerland.
| | - Magdalena Winiarska
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland.
| |
Collapse
|
39
|
Lee EY, Kulkarni RP. Circulating biomarkers predictive of tumor response to cancer immunotherapy. Expert Rev Mol Diagn 2019; 19:895-904. [PMID: 31469965 DOI: 10.1080/14737159.2019.1659728] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction: The advent of checkpoint blockade immunotherapy has revolutionized cancer treatment, but clinical response to immunotherapies is highly heterogeneous among individual patients and between cancer types. This represents a challenge to oncologists when choosing specific immunotherapies for personalized medicine. Thus, biomarkers that can predict tumor responsiveness to immunotherapies before and during treatment are invaluable. Areas covered: We review the latest advances in 'liquid biopsy' biomarkers for noninvasive prediction and in-treatment monitoring of tumor response to immunotherapy, focusing primarily on melanoma and non-small cell lung cancer. We concentrate on high-quality studies published within the last five years on checkpoint blockade immunotherapies, and highlight significant breakthroughs, identify key areas for improvement, and provide recommendations for how these diagnostic tools can be translated into clinical practice. Expert opinion: The first biomarkers proposed to predict tumor response to immunotherapy were based on PD1/PDL1 expression, but their predictive value is limited to specific cancers or patient populations. Recent advances in single-cell molecular profiling of circulating tumor cells and host cells using next-generation sequencing has dramatically expanded the pool of potentially useful predictive biomarkers. As immunotherapy moves toward personalized medicine, a composite panel of both genomic and proteomic biomarkers will have enormous utility in therapeutic decision-making.
Collapse
Affiliation(s)
- Ernest Y Lee
- Department of Bioengineering, UCLA , Los Angeles , CA , USA.,Department of Dermatology, UCLA , Los Angeles , CA , USA.,UCLA-Caltech Medical Scientist Training Program, David Geffen School of Medicine at UCLA , Los Angeles , CA , USA
| | - Rajan P Kulkarni
- Department of Dermatology, OHSU , Portland , OR , USA.,Cancer Early Detection and Advanced Research Center (CEDAR), Knight Cancer Institute (KCI), OHSU , Portland , OR , USA.,Division of Operative Care, Portland VA Medical Center (PVAMC) , Portland , OR , USA
| |
Collapse
|
40
|
Benitez MLR, Bender CB, Oliveira TL, Schachtschneider KM, Collares T, Seixas FK. Mycobacterium bovis BCG in metastatic melanoma therapy. Appl Microbiol Biotechnol 2019; 103:7903-7916. [PMID: 31402426 DOI: 10.1007/s00253-019-10057-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/22/2019] [Accepted: 07/26/2019] [Indexed: 12/13/2022]
Abstract
Melanoma is the most aggressive form of skin cancer, with a high mortality rate and with 96,480 new cases expected in 2019 in the USS. BRAFV600E, the most common driver mutation, is found in around 50% of melanomas, contributing to tumor growth, angiogenesis, and metastatic progression. Dacarbazine (DTIC), an alkylate agent, was the first chemotherapeutic agent approved by the US Food and Drug Administration (FDA) used as a standard treatment. Since then, immunotherapies have been approved for metastatic melanoma (MM) including ipilimumab and pembrolizumab checkpoint inhibitors that help decrease the risk of progression. Moreover, Mycobacterium bovis Bacillus Calmette-Guerin (BCG) serves as an adjuvant therapy that induces the recruitment of natural killer NK, CD4+, and CD8+ T cells and contributes to antitumor immunity. BCG can be administered in combination with chemotherapeutic and immunotherapeutic agents and can be genetically manipulated to produce recombinant BCG (rBCG) strains that express heterologous proteins or overexpress immunogenic proteins, increasing the immune response and improving patient survival. In this review, we highlight several studies utilizing rBCG immunotherapy for MM in combination with other therapeutic agents.
Collapse
Affiliation(s)
- Martha Lucia Ruiz Benitez
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Camila Bonnemann Bender
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Thaís Larré Oliveira
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA.,National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Tiago Collares
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Fabiana Kömmling Seixas
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil.
| |
Collapse
|
41
|
Unraveling the crosstalk between melanoma and immune cells in the tumor microenvironment. Semin Cancer Biol 2019; 59:236-250. [PMID: 31404607 DOI: 10.1016/j.semcancer.2019.08.002] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/10/2019] [Accepted: 08/04/2019] [Indexed: 12/12/2022]
Abstract
Cutaneous melanoma is the most common skin cancer with an incidence that has been rapidly increasing in the past decades. Melanomas are among the most immunogenic tumors and, as such, have the greatest potential to respond favorably to immunotherapy. However, like many cancers, melanomas acquire various suppressive mechanisms, which generally act in concert, to escape innate and adaptive immune detection and destruction. Intense research into the cellular and molecular events associated with melanomagenesis, which ultimately lead to immune suppression, has resulted in the discovery of new therapeutic targets and synergistic combinations of immunotherapy, targeted therapy and chemotherapy. Tremendous effort to determine efficacy of single and combination therapies in pre-clinical and clinical phase I-III trials has led to FDA-approval of several immunotherapeutic agents that could potentially be beneficial for aggressive, highly refractory, advanced and metastatic melanomas. The increasing availability of approved combination therapies for melanoma and more rapid assessment of patient tumors has increased the feasibility of personalized treatment to overcome patient and tumor heterogeneity and to achieve greater clinical benefit. Here, we review the evolution of the immune system during melanomagenesis, mechanisms exploited by melanoma to suppress anti-tumor immunity and methods that have been developed to restore immunity. We emphasize that an effective therapeutic strategy will require coordinate activation of tumor-specific immunity as well as increased recognition and accessibility of melanoma cells in primary tumors and distal metastases. This review integrates available knowledge on melanoma-specific immunity, molecular signaling pathways and molecular targeting strategies that could be utilized to envision therapeutics with broader application and greater efficacy for early stage and advanced metastatic melanoma.
Collapse
|
42
|
Dummer R, Mangana J, Frauchiger AL, Lang C, Micaletto S, Barysch MJ. How I treat metastatic melanoma. ESMO Open 2019; 4:e000509. [PMID: 31423341 PMCID: PMC6677980 DOI: 10.1136/esmoopen-2019-000509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 01/08/2023] Open
Abstract
Tremendous progress in basic and clinical research has completely revolutionised the management of advanced melanoma, and this dramatic development is still ongoing. In this environment, state-of-the-art patient care is a major challenge. We describe how patient-centred medicine is organised in a leading referral centre that is also involved in early and late clinical trials and is part of a worldwide network for translational research.
Collapse
Affiliation(s)
- Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.
| | - Joanna Mangana
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | | | - Claudia Lang
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Sara Micaletto
- Department of Dermatology, University Hospital, Zurich, Switzerland
| | - Marjam J Barysch
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
43
|
|
44
|
Abstract
Genetic material derived from tumours is constantly shed into the circulation of cancer patients both in the form of circulating free nucleic acids and within circulating cells or extracellular vesicles. Monitoring cancer-specific genomic alterations, particularly mutant allele frequencies, in circulating nucleic acids allows for a non-invasive liquid biopsy for detecting residual disease and response to therapy. The advent of molecular targeted treatments and immunotherapies with increasing effectiveness requires corresponding effective molecular biology methods for the detection of biomarkers such as circulating nucleic acid to monitor and ultimately personalise therapy. The use of polymerase chain reaction (PCR)-based methods, such as droplet digital PCR, allows for a very sensitive analysis of circulating tumour DNA, but typically only a limited number of gene mutations can be detected in parallel. In contrast, next-generation sequencing allows for parallel analysis of multiple mutations in many genes. The development of targeted next-generation sequencing cancer gene panels optimised for the detection of circulating free DNA now provides both the flexibility of multiple mutation analysis coupled with a sensitivity that approaches or even matches droplet digital PCR. In this review, we discuss the advantages and disadvantages of these current molecular technologies in conjunction with how this field is evolving in the context of melanoma diagnosis, prognosis, and monitoring of response to therapy.
Collapse
|
45
|
Proof of concept study of mass cytometry in septic shock patients reveals novel immune alterations. Sci Rep 2018; 8:17296. [PMID: 30470767 PMCID: PMC6251894 DOI: 10.1038/s41598-018-35932-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/13/2018] [Indexed: 12/29/2022] Open
Abstract
Innovative single cell technologies such as mass cytometry (CyTOF) widen possibilities to deeply improve characterisation of immune alterations mechanisms in human diseases. So far, CyTOF has not been used in sepsis – a condition characterized by complex immune disorders. Here, we evaluated feasibility of CyTOF analysis in patients with septic shock. We designed a mass cytometry panel of 25 extracellular markers to study mononuclear cells from 5 septic shock patients and 5 healthy donors. We explored single-cell data with global and specific unsupervised approaches such as heatmaps, SPADE and viSNE. We first validated relevance of our CyTOF results by highlighting established immune hallmarks of sepsis, such as decreased monocyte HLA-DR expression and increased expressions of PD1 and PD-L1 on CD4 T cells and monocytes. We then showed that CyTOF analysis reveals novel aspects of sepsis-induced immune alterations, e.g. B cell shift towards plasma cell differentiation and uniform response of several monocyte markers defining an immune signature in septic patients. This proof of concept study demonstrates CyTOF suitability to analyse immune features of septic patients. Mass cytometry could thus represent a powerful tool to identify novel pathophysiological mechanisms and therapeutic targets for immunotherapy in septic shock patients.
Collapse
|