51
|
Lequerica-Fernández P, Suárez-Canto J, Rodriguez-Santamarta T, Rodrigo JP, Suárez-Sánchez FJ, Blanco-Lorenzo V, Domínguez-Iglesias F, García-Pedrero JM, de Vicente JC. Prognostic Relevance of CD4 +, CD8 + and FOXP3 + TILs in Oral Squamous Cell Carcinoma and Correlations with PD-L1 and Cancer Stem Cell Markers. Biomedicines 2021; 9:biomedicines9060653. [PMID: 34201050 PMCID: PMC8227658 DOI: 10.3390/biomedicines9060653] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/27/2021] [Accepted: 05/31/2021] [Indexed: 12/22/2022] Open
Abstract
This study investigates the relevance of tumor-infiltrating lymphocytes (TILs) in oral squamous cell carcinoma (OSCC). Immunohistochemical analysis of stromal/tumoral CD4+, CD8+ and FOXP3+ TILs is performed in 125 OSCC patients. Potential relationships with the expression of tumoral PD-L1 and cancer stem cell (CSC) markers (NANOG, SOX2, OCT4, Nestin and Podoplanin (PDPN)) are assessed. CD4+ and CD8+ TILs are significantly associated with smoking and alcohol habits. CD4+ and CD8+ TILs show an inverse relationship with NANOG and SOX2 expression, and FOXP3+ TILs is significantly correlated with Nestin and PDPN expression. High infiltration of CD4+ and CD8+ TILs and a high tumoral CD8+/FOXP3+ ratio are significantly associated with tumors harboring positive PD-L1 expression. Infiltration of stromal/tumoral FOXP3+ TILs and a low stromal CD8+/FOXP3+ ratio are significantly associated with better disease-specific survival. Multivariate analysis reveals that the stromal CD8+/FOXP3+ TILs ratio is a significant independent prognostic factor. Regarding OSCC patient survival, the CD8+/FOXP3+ TILs ratio is an independent prognostic factor. TILs may act as biomarkers and potential therapeutic targets for OSCC.
Collapse
Affiliation(s)
- Paloma Lequerica-Fernández
- Department of Biochemistry, Hospital Universitario Central de Asturias (HUCA), C/Carretera de Rubín, s/n, 33011 Oviedo, Spain; (P.L.-F.); (F.J.S.-S.); (F.D.-I.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, C/Carretera de Rubín, s/n, 33011 Oviedo, Spain; (T.R.-S.); (J.P.R.)
| | - Julián Suárez-Canto
- Department of Pathology, Hospital Universitario de Cabueñes, 33394 Gijón, Spain;
| | - Tania Rodriguez-Santamarta
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, C/Carretera de Rubín, s/n, 33011 Oviedo, Spain; (T.R.-S.); (J.P.R.)
- Department of Oral and Maxillofacial Surgery, Hospital Universitario Central de Asturias (HUCA), C/Carretera de Rubín, s/n, 33011 Oviedo, Spain
| | - Juan Pablo Rodrigo
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, C/Carretera de Rubín, s/n, 33011 Oviedo, Spain; (T.R.-S.); (J.P.R.)
- Department of Otolaryngology, Hospital Universitario Central de Asturias (HUCA), C/Carretera de Rubín, s/n, 33011 Oviedo, Spain
- Department of Surgery, University of Oviedo, 33006 Oviedo, Spain
- Ciber de Cancer (CIBERONC), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Faustino Julián Suárez-Sánchez
- Department of Biochemistry, Hospital Universitario Central de Asturias (HUCA), C/Carretera de Rubín, s/n, 33011 Oviedo, Spain; (P.L.-F.); (F.J.S.-S.); (F.D.-I.)
| | - Verónica Blanco-Lorenzo
- Department of Pathology, Hospital Universitario Central de Asturias (HUCA), C/Carretera de Rubín, s/n, 33011 Oviedo, Spain;
| | - Francisco Domínguez-Iglesias
- Department of Biochemistry, Hospital Universitario Central de Asturias (HUCA), C/Carretera de Rubín, s/n, 33011 Oviedo, Spain; (P.L.-F.); (F.J.S.-S.); (F.D.-I.)
| | - Juana María García-Pedrero
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, C/Carretera de Rubín, s/n, 33011 Oviedo, Spain; (T.R.-S.); (J.P.R.)
- Ciber de Cancer (CIBERONC), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Correspondence: (J.M.G.-P.); (J.C.d.V.); Tel.: +34-985-107937 (J.M.G.-P.); +34-85-103638 (J.C.d.V.)
| | - Juan Carlos de Vicente
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, C/Carretera de Rubín, s/n, 33011 Oviedo, Spain; (T.R.-S.); (J.P.R.)
- Department of Oral and Maxillofacial Surgery, Hospital Universitario Central de Asturias (HUCA), C/Carretera de Rubín, s/n, 33011 Oviedo, Spain
- Department of Surgery, University of Oviedo, 33006 Oviedo, Spain
- Correspondence: (J.M.G.-P.); (J.C.d.V.); Tel.: +34-985-107937 (J.M.G.-P.); +34-85-103638 (J.C.d.V.)
| |
Collapse
|
52
|
Kostrzewa-Nowak D, Trzeciak-Ryczek A, Wityk P, Cembrowska-Lech D, Nowak R. Post-Effort Changes in Autophagy- and Inflammation-Related Gene Expression in White Blood Cells of Healthy Young Men. Cells 2021; 10:cells10061406. [PMID: 34204085 PMCID: PMC8229752 DOI: 10.3390/cells10061406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 01/18/2023] Open
Abstract
Acute, strenuous physical exertion requiring high levels of energy production induces the production of reactive oxygen species and metabolic disturbances that can damage the mitochondria. Thus, selective autophagic elimination of defective mitochondria may improve resistance to oxidative stress and potentially to inflammation. The main goal of this study was to evaluate the impacts of intense effort on changes in the expression of select genes related to post-effort inflammation and autophagy. Thirty-five men aged 16–21 years were recruited to the study. The impacts of both aerobic (endurance) and anaerobic (speed) efforts on selected genes encoding chemokines (CXCL5, 8–12) were analyzed. Significant increases in the expression of all studied genes excluding CXCL12 were observed. Moreover, both types of effort induced an increase in the expression of genes encoding IL-2, -4, -6, -10, IFN-γ and TNF-α, excluding IL-17A. Generally, these efforts caused a significant increase in the relative expression of apoptosis- (BCL2 and BAX) and autophagy- (BNIP3, BECN1, MAP1LC3B, ATG5, ATG7, ATG12, ATG16L1 and SQSTM1) related genes. It seems that the duration of physical activity and its bioenergetic cost has an important impact on the degree of increase in expression of this panel of autophagy-related genes. Anaerobic effort is more strenuous than aerobic effort and requires a higher bioenergetic investment. This may explain the stronger impact of anaerobic effort on the expression of the studied genes. This observation seems to support the protective role of autophagy proposed in prior studies.
Collapse
Affiliation(s)
- Dorota Kostrzewa-Nowak
- Centre for Human Structural and Functional Research, Institute of Physical Culture Sciences, University of Szczecin, 17C Narutowicza St., 70-240 Szczecin, Poland;
- Correspondence:
| | - Alicja Trzeciak-Ryczek
- Institute of Biology, University of Szczecin, 13 Wąska St., 71-415 Szczecin, Poland; (A.T.-R.); (D.C.-L.)
- The Centre for Molecular Biology and Biotechnology, University of Szczecin, 13 Wąska St., 71-415 Szczecin, Poland
| | - Paweł Wityk
- Faculty of Chemistry, Gdańsk University of Technology, 11/12 Narutowicza St., 80-233 Gdańsk, Poland;
| | - Danuta Cembrowska-Lech
- Institute of Biology, University of Szczecin, 13 Wąska St., 71-415 Szczecin, Poland; (A.T.-R.); (D.C.-L.)
- The Centre for Molecular Biology and Biotechnology, University of Szczecin, 13 Wąska St., 71-415 Szczecin, Poland
| | - Robert Nowak
- Centre for Human Structural and Functional Research, Institute of Physical Culture Sciences, University of Szczecin, 17C Narutowicza St., 70-240 Szczecin, Poland;
| |
Collapse
|
53
|
Mills JK, Henderson MA, Giuffrida L, Petrone P, Westwood JA, Darcy PK, Neeson PJ, Kershaw MH, Gyorki DE. Generating CAR T cells from tumor-infiltrating lymphocytes. Ther Adv Vaccines Immunother 2021; 9:25151355211017119. [PMID: 34159293 PMCID: PMC8186112 DOI: 10.1177/25151355211017119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 04/01/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T-cell therapies have demonstrated promising, though limited, efficacy against melanoma. Methods: We designed a model system to explore the efficacy of dual specific T cells derived from melanoma patient TILs by transduction with a Her2-specific CAR. Results: Metastatic melanoma cells in our biobank constitutively expressed Her2 antigen. CAR-TIL produced greater amounts of IFN compared with parental TIL, when co-cultured with Her2 expressing tumor lines, including autologous melanoma tumor lines, although no consistent increase in cytotoxicity by TIL was afforded by expression of a CAR. Results of an in vivo study in NSG mice demonstrated tumor shrinkage when CAR-TILs were used in an adoptive cell therapy protocol. Conclusion: Potential limitations of transduced TIL in our study included limited proliferative potential and a terminally differentiated phenotype, which would need addressing in further work before consideration of clinical translation.
Collapse
Affiliation(s)
- Jane K Mills
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Melissa A Henderson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Lauren Giuffrida
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Pasquale Petrone
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Jennifer A Westwood
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Paul J Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - David E Gyorki
- Department of Cancer Surgery, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia
| |
Collapse
|
54
|
Inoue T, Byrne T, Inoue M, Tait ME, Wall P, Wang A, Dermyer MR, Laklai H, Binder JJ, Lees C, Hollingsworth R, Maruri-Avidal L, Kirn DH, McDonald DM. Oncolytic Vaccinia Virus Gene Modification and Cytokine Expression Effects on Tumor Infection, Immune Response, and Killing. Mol Cancer Ther 2021; 20:1481-1494. [PMID: 34045231 DOI: 10.1158/1535-7163.mct-20-0863] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/04/2021] [Accepted: 05/25/2021] [Indexed: 12/27/2022]
Abstract
Oncolytic vaccinia viruses have promising efficacy and safety profiles in cancer therapy. Although antitumor activity can be increased by manipulating viral genes, the relative efficacy of individual modifications has been difficult to assess without side-by-side comparisons. This study sought to compare the initial antitumor activity after intravenous administration of five vaccinia virus variants of the same Western Reserve backbone and thymidine kinase gene deletion in RIP-Tag2 transgenic mice with spontaneous pancreatic neuroendocrine tumors. Tumors had focal regions of infection at 5 days after all viruses. Natural killer (NK) cells were restricted to these sites of infection, but CD8+ T cells and tumor cell apoptosis were widespread and varied among the viruses. Antitumor activity of virus VV-A34, bearing amino acid substitution A34K151E to increase viral spreading, and virus VV-IL2v, expressing a mouse IL2 variant (mIL2v) with attenuated IL2 receptor alpha subunit binding, was similar to control virus VV-GFP. However, antitumor activity was significantly greater after virus VV-A34/IL2v, which expressed mIL2v together with A34K151E mutation and viral B18R gene deletion, and virus VV-GMCSF that expressed mouse GM-CSF. Both viruses greatly increased expression of CD8 antigens Cd8a/Cd8b1 and cytotoxicity genes granzyme A, granzyme B, Fas ligand, and perforin-1 in tumors. VV-A34/IL2v led to higher serum IL2 and greater tumor expression of death receptor ligand TRAIL, but VV-GMCSF led to higher serum GM-CSF, greater expression of leukocyte chemokines and adhesion molecules, and more neutrophil recruitment. Together, the results show that antitumor activity is similarly increased by viral expression of GM-CSF or IL2v combined with additional genetic modifications.
Collapse
Affiliation(s)
- Tomoyoshi Inoue
- UCSF Helen Diller Family Comprehensive Cancer Center, Cardiovascular Research Institute and Department of Anatomy, University of California, San Francisco, San Francisco, California
| | - Thomas Byrne
- UCSF Helen Diller Family Comprehensive Cancer Center, Cardiovascular Research Institute and Department of Anatomy, University of California, San Francisco, San Francisco, California
| | - Mitsuko Inoue
- UCSF Helen Diller Family Comprehensive Cancer Center, Cardiovascular Research Institute and Department of Anatomy, University of California, San Francisco, San Francisco, California
| | - Madeline E Tait
- UCSF Helen Diller Family Comprehensive Cancer Center, Cardiovascular Research Institute and Department of Anatomy, University of California, San Francisco, San Francisco, California
| | | | - Annabel Wang
- Cancer Vaccines & Immunotherapeutics, Oncology Research & Development, Pfizer, La Jolla, California
| | - Michael R Dermyer
- Cancer Vaccines & Immunotherapeutics, Oncology Research & Development, Pfizer, La Jolla, California
| | - Hanane Laklai
- Cancer Vaccines & Immunotherapeutics, Oncology Research & Development, Pfizer, La Jolla, California
| | - Joseph J Binder
- Cancer Vaccines & Immunotherapeutics, Oncology Research & Development, Pfizer, La Jolla, California
| | - Clare Lees
- Cancer Vaccines & Immunotherapeutics, Oncology Research & Development, Pfizer, La Jolla, California
| | - Robert Hollingsworth
- Cancer Vaccines & Immunotherapeutics, Oncology Research & Development, Pfizer, La Jolla, California
| | | | | | - Donald M McDonald
- UCSF Helen Diller Family Comprehensive Cancer Center, Cardiovascular Research Institute and Department of Anatomy, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
55
|
Johansson-Percival A, Ganss R. Therapeutic Induction of Tertiary Lymphoid Structures in Cancer Through Stromal Remodeling. Front Immunol 2021; 12:674375. [PMID: 34122434 PMCID: PMC8191417 DOI: 10.3389/fimmu.2021.674375] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/04/2021] [Indexed: 01/01/2023] Open
Abstract
Improving the effectiveness of anti-cancer immunotherapy remains a major clinical challenge. Cytotoxic T cell infiltration is crucial for immune-mediated tumor rejection, however, the suppressive tumor microenvironment impedes their recruitment, activation, maturation and function. Nevertheless, solid tumors can harbor specialized lymph node vasculature and immune cell clusters that are organized into tertiary lymphoid structures (TLS). These TLS support naïve T cell infiltration and intratumoral priming. In many human cancers, their presence is a positive prognostic factor, and importantly, predictive for responsiveness to immune checkpoint blockade. Thus, therapeutic induction of TLS is an attractive concept to boost anti-cancer immunotherapy. However, our understanding of how cancer-associated TLS could be initiated is rudimentary. Exciting new reagents which induce TLS in preclinical cancer models provide mechanistic insights into the exquisite stromal orchestration of TLS formation, a process often associated with a more functional or "normalized" tumor vasculature and fueled by LIGHT/LTα/LTβ, TNFα and CC/CXC chemokine signaling. These emerging insights provide innovative opportunities to induce and shape TLS in the tumor microenvironment to improve immunotherapies.
Collapse
Affiliation(s)
- Anna Johansson-Percival
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia
- Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia
| | - Ruth Ganss
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia
- Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
56
|
Yuan Y, Zhu Z, Lan Y, Duan S, Zhu Z, Zhang X, Li G, Qu H, Feng Y, Cai H, Song Z. Development and Validation of a CD8+ T Cell Infiltration-Related Signature for Melanoma Patients. Front Immunol 2021; 12:659444. [PMID: 34040608 PMCID: PMC8141567 DOI: 10.3389/fimmu.2021.659444] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
Aim Immunotherapy shows efficacy in only a subset of melanoma patients. Here, we intended to construct a risk score model to predict melanoma patients’ sensitivity to immunotherapy. Methods Integration analyses were performed on melanoma patients from high-dimensional public datasets. The CD8+ T cell infiltration related genes (TIRGs) were selected via TIMER and CIBERSORT algorithm. LASSO Cox regression was performed to screen for the crucial TIRGs. Single sample gene set enrichment analysis (ssGSEA) and ESTIMATE algorithm were used to evaluate the immune activity. The prognostic value of the risk score was determined by univariate and multivariate Cox regression analysis. Results 184 candidate TIRGs were identified in melanoma patients. Based on the candidate TIRGs, melanoma patients were classified into three clusters which were characterized by different immune activity. Six signature genes were further screened out of 184 TIRGs and a representative risk score for patient survival was constructed based on these six signature genes. The risk score served as an indicator for the level of CD8+ T cell infiltration and acted as an independent prognostic factor for the survival of melanoma patients. By using the risk score, we achieved a good predicting result for the response of cancer patients to immunotherapy. Moreover, pan-cancer analysis revealed the risk score could be used in a wide range of non-hematologic tumors. Conclusions Our results showed the potential of using signature gene-based risk score as an indicator to predict melanoma patients’ sensitivity to immunotherapy.
Collapse
Affiliation(s)
- Yuan Yuan
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States.,Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Zheng Zhu
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Ying Lan
- School of Nursing, Yueyang Vocational and Technical College, Yueyang, China
| | - Saili Duan
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China.,Xiangya School of Medicine of Central South University, Changsha, China
| | - Ziqing Zhu
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China.,Xiangya School of Medicine of Central South University, Changsha, China
| | - Xi Zhang
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Guoyin Li
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, China
| | - Hui Qu
- Department of Plastic Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Yanhui Feng
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Hui Cai
- Department of Orthopaedics, Loudi Central Hospital, Loudi, China
| | - Zewen Song
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
57
|
Picard M, Yonekura S, Slowicka K, Petta I, Rauber C, Routy B, Richard C, Iebba V, Tidjani Alou M, Becharef S, Ly P, Pizzato E, Lehmann CHK, Amon L, Klein C, Opolon P, Gomperts Boneca I, Scoazec JY, Hollebecque A, Malka D, Ghiringhelli F, Dudziak D, Berx G, Vereecke L, van Loo G, Kroemer G, Zitvogel L, Roberti MP. Ileal immune tonus is a prognosis marker of proximal colon cancer in mice and patients. Cell Death Differ 2021; 28:1532-1547. [PMID: 33262469 PMCID: PMC8167112 DOI: 10.1038/s41418-020-00684-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Ileal epithelial cell apoptosis and the local microbiota modulate the effects of oxaliplatin against proximal colon cancer by modulating tumor immunosurveillance. Here, we identified an ileal immune profile associated with the prognosis of colon cancer and responses to chemotherapy. The whole immune ileal transcriptome was upregulated in poor-prognosis patients with proximal colon cancer, while the colonic immunity of healthy and neoplastic areas was downregulated (except for the Th17 fingerprint) in such patients. Similar observations were made across experimental models of implanted and spontaneous murine colon cancer, showing a relationship between carcinogenesis and ileal inflammation. Conversely, oxaliplatin-based chemotherapy could restore a favorable, attenuated ileal immune fingerprint in responders. These results suggest that chemotherapy inversely shapes the immune profile of the ileum-tumor axis, influencing clinical outcome.
Collapse
Affiliation(s)
- Marion Picard
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Institut Pasteur, Unit Biology and Genetics of the Bacterial Cell Wall, Paris, France
- INSERM, Equipe Avenir, Paris, France
| | - Satoru Yonekura
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Karolina Slowicka
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ioanna Petta
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Conrad Rauber
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Bertrand Routy
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Corentin Richard
- Department of Medical Oncology, Center GF Leclerc, Dijon, France
- Plateform Transfer in Biological Oncology, Dijon, France
| | - Valerio Iebba
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Maryam Tidjani Alou
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Sonia Becharef
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Pierre Ly
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Eugenie Pizzato
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Christian H K Lehmann
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Medical Immunology Campus Erlangen, University Hospital of Erlangen, Friedrich-Alexander-University (FAU) of Erlangen-Nürnberg, Erlangen, Germany
| | - Lukas Amon
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Medical Immunology Campus Erlangen, University Hospital of Erlangen, Friedrich-Alexander-University (FAU) of Erlangen-Nürnberg, Erlangen, Germany
| | - Christophe Klein
- Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
| | - Paule Opolon
- Departement de Biologie et Pathologie Médicales, Gustave Roussy Cancer Campus, Villejuif, France
| | - Ivo Gomperts Boneca
- Institut Pasteur, Unit Biology and Genetics of the Bacterial Cell Wall, Paris, France
- INSERM, Equipe Avenir, Paris, France
- CNRS UMR2001, Paris, France
| | - Jean-Yves Scoazec
- Departement de Biologie et Pathologie Médicales, Gustave Roussy Cancer Campus, Villejuif, France
| | - Antoine Hollebecque
- Departement de Médicine Oncologique, Gustave Roussy Cancer Campus, Villejuif, France
| | - David Malka
- Departement de Médicine Oncologique, Gustave Roussy Cancer Campus, Villejuif, France
| | - François Ghiringhelli
- Department of Medical Oncology, Center GF Leclerc, Dijon, France
- Plateform Transfer in Biological Oncology, Dijon, France
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Medical Immunology Campus Erlangen, University Hospital of Erlangen, Friedrich-Alexander-University (FAU) of Erlangen-Nürnberg, Erlangen, Germany
| | - Geert Berx
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lars Vereecke
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Geert van Loo
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Guido Kroemer
- Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Cell Biology and Metabolomics Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de, Paris, France
- Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Maria Paula Roberti
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France.
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.
- Clinical Cooperation Unit Applied Tumor Immunity, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
58
|
Habal MV, Miller AM, Rao S, Lin S, Obradovic A, Khosravi-Maharlooei M, See S, Roy P, Ronzon S, Ho SH, Marboe C, Naka Y, Takeda K, Restaino S, Han A, Mancini D, Givertz M, Madsen JC, Sykes M, Addonizio L, Farr M, Zorn E. T cell repertoire analysis suggests a prominent bystander response in human cardiac allograft vasculopathy. Am J Transplant 2021; 21:1465-1476. [PMID: 33021057 PMCID: PMC8672660 DOI: 10.1111/ajt.16333] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/02/2020] [Accepted: 09/16/2020] [Indexed: 01/25/2023]
Abstract
T cells are implicated in the pathogenesis of cardiac allograft vasculopathy (CAV), yet their clonality, specificity, and function are incompletely defined. Here we used T cell receptor β chain (TCRB) sequencing to study the T cell repertoire in the coronary artery, endomyocardium, and peripheral blood at the time of retransplant in four cases of CAV and compared it to the immunoglobulin heavy chain variable region (IGHV) repertoire from the same samples. High-dimensional flow cytometry coupled with single-cell PCR was also used to define the T cell phenotype. Extensive overlap was observed between intragraft and blood TCRBs in all cases, a finding supported by robust quantitative diversity metrics. In contrast, blood and graft IGHV repertoires from the same samples showed minimal overlap. Coronary infiltrates included CD4+ and CD8+ memory T cells expressing inflammatory (IFNγ, TNFα) and profibrotic (TGFβ) cytokines. These were distinguishable from the peripheral blood based on memory, activation, and tissue residency markers (CD45RO, CTLA-4, and CD69). Importantly, high-frequency rearrangements were traced back to endomyocardial biopsies (2-6 years prior). Comparison with four HLA-mismatched blood donors revealed a repertoire of shared TCRBs, including a subset of recently described cross-reactive sequences. These findings provide supportive evidence for an active local intragraft bystander T cell response in late-stage CAV.
Collapse
Affiliation(s)
- Marlena V. Habal
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY,Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY
| | - April M.I Miller
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Samhita Rao
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Sijie Lin
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Aleksandar Obradovic
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | | | - Sarah See
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Poulomi Roy
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Shihab Ronzon
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Siu-hong Ho
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Charles Marboe
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Yoshifumi Naka
- Department of Surgery, Division of Cardiothoracic Surgery, Columbia University Irving Medical Center, New York, NY
| | - Koji Takeda
- Department of Surgery, Division of Cardiothoracic Surgery, Columbia University Irving Medical Center, New York, NY
| | - Susan Restaino
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY
| | - Arnold Han
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Donna Mancini
- Department of Medicine, Mount Sinai Hospital, Icahn School of Medicine, New York, NY
| | - Michael Givertz
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Joren C. Madsen
- Center for Transplantation Science, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| | - Linda Addonizio
- Department of Pediatrics, Division of Pediatric Cardiology, Columbia University Irving Medical Center, New York, NY
| | - Maryjane Farr
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY
| | - Emmanuel Zorn
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
59
|
Hu Q, Nonaka K, Wakiyama H, Miyashita Y, Fujimoto Y, Jogo T, Hokonohara K, Nakanishi R, Hisamatsu Y, Ando K, Kimura Y, Masuda T, Oki E, Mimori K, Oda Y, Mori M. Cytolytic activity score as a biomarker for antitumor immunity and clinical outcome in patients with gastric cancer. Cancer Med 2021; 10:3129-3138. [PMID: 33769705 PMCID: PMC8085935 DOI: 10.1002/cam4.3828] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND A simple measure of immune cytolytic activity (CYT) base on mRNA expression levels of two genes, GZMA and PRF1, was recently reported. Here, we aimed to evaluate the CYT score's potential as a measure of antitumor immunity and predictor of clinical outcome in gastric cancer (GC) patients. MATERIALS AND METHODS We evaluated the correlations between tumor-infiltrating immune cells and the CYT score in 238 GC samples from The Cancer Genome Atlas (TCGA). Next, we investigated CYT score associations with molecular subtypes, somatic mutation load, and immune checkpoint molecules in GC samples from TCGA and Asian Cancer Research Group (ACRG). Moreover, we evaluated the clinical significance of the CYT score calculated by reverse transcription (RT)-quantitative PCR (qPCR) data in 123 GC samples and the association of the CYT score with the response to anti-PD-1 therapy in 7 GC samples from Kyushu University Hospital. RESULTS The CYT score positively correlated with the proportions of tumor-infiltrating CD8+ T cells and macrophages and negatively correlated with the proportion of regulatory T cells in GC tissues. A high CYT score was associated with common immune checkpoint molecules, a high mutation, the Epstein-Barr virus subtype, and the microsatellite instability subtype in GC. Moreover, a low CYT score was a poor prognosis factor in patients with GC. Finally, the CYT score was higher in a responder to anti-PD-1 therapy compared to nonresponders. CONCLUSION The CYT score reflects antitumor immunity and predicts clinical outcome in GC patients.
Collapse
Affiliation(s)
- Qingjiang Hu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kentaro Nonaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroaki Wakiyama
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yu Miyashita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshiaki Fujimoto
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoko Jogo
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kentaro Hokonohara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryota Nakanishi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichi Hisamatsu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koji Ando
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasue Kimura
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takaaki Masuda
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
60
|
High endothelial venules are present in pharyngeal and laryngeal carcinomas and they are associated with better prognosis. Pathol Res Pract 2021; 220:153392. [PMID: 33647862 DOI: 10.1016/j.prp.2021.153392] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Tumors lymphocytic infiltration has prognostic and predictive value. However, the mechanisms involved in lymphocyte recruitment remain poorly characterized. High endothelial venules (HEV) are blood vessels specialized in lymphocyte recruitment, recently showing prognostic significance in some types of cancer. Their implications in laryngeal or pharyngeal cancer is largely unknown. AIM OF THE STUDY To investigate the possible presence of HEVs in head and neck cancer. MATERIAL AND METHODS Oropharyngeal (n = 61), hypopharyngeal (n = 53) and laryngeal (n = 21) squamous cell carcinomas were immunohistochemically studied with the MECA-79 antibody, which specifically recognizes HEVs. Histological and clinical factors were correlated with HEVs' presence. RESULTS HEVs were present in 34% of tumors, showing significant correlations with oropharyngeal localization, higher lymphocytic response, lower tumor budding, lower T status, absence of distant metastases and better overall and progression-free survival. CONCLUSION HEVs represent an important prognostic factor in head and neck cancer.
Collapse
|
61
|
Liu S, Ren J, Ten Dijke P. Targeting TGFβ signal transduction for cancer therapy. Signal Transduct Target Ther 2021; 6:8. [PMID: 33414388 PMCID: PMC7791126 DOI: 10.1038/s41392-020-00436-9] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 12/19/2022] Open
Abstract
Transforming growth factor-β (TGFβ) family members are structurally and functionally related cytokines that have diverse effects on the regulation of cell fate during embryonic development and in the maintenance of adult tissue homeostasis. Dysregulation of TGFβ family signaling can lead to a plethora of developmental disorders and diseases, including cancer, immune dysfunction, and fibrosis. In this review, we focus on TGFβ, a well-characterized family member that has a dichotomous role in cancer progression, acting in early stages as a tumor suppressor and in late stages as a tumor promoter. The functions of TGFβ are not limited to the regulation of proliferation, differentiation, apoptosis, epithelial-mesenchymal transition, and metastasis of cancer cells. Recent reports have related TGFβ to effects on cells that are present in the tumor microenvironment through the stimulation of extracellular matrix deposition, promotion of angiogenesis, and suppression of the anti-tumor immune reaction. The pro-oncogenic roles of TGFβ have attracted considerable attention because their intervention provides a therapeutic approach for cancer patients. However, the critical function of TGFβ in maintaining tissue homeostasis makes targeting TGFβ a challenge. Here, we review the pleiotropic functions of TGFβ in cancer initiation and progression, summarize the recent clinical advancements regarding TGFβ signaling interventions for cancer treatment, and discuss the remaining challenges and opportunities related to targeting this pathway. We provide a perspective on synergistic therapies that combine anti-TGFβ therapy with cytotoxic chemotherapy, targeted therapy, radiotherapy, or immunotherapy.
Collapse
Affiliation(s)
- Sijia Liu
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Jiang Ren
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Peter Ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
62
|
Abstract
Accessible prognostic tools are needed to individualize treatment of neuroendocrine tumors (NETs). Data suggest neutrophil/lymphocyte ratios (NLRs) have prognostic value in some solid tumors, including NETs. In the randomized double-blind CLARINET study (NCT00353496; EudraCT 2005-004904-35), the somatostatin analog lanreotide autogel/depot increased progression-free survival (PFS) compared with placebo in patients with inoperable or metastatic intestinal and pancreatic NETs (grades 1–2, Ki-67 < 10%). The exploratory post-hoc analyses presented here evaluated the prognostic value of NLR in the CLARINET study cohort, in the context of and independently from treatment. Kaplan–Meier PFS plots were generated for patients with available NLR data, in subgroups based on NLR values, and 24-month survival rates were calculated. P values and hazard ratios for prognostic effects were generated using Cox models. 31216222 Baseline characteristics were balanced between lanreotide autogel/depot 120 mg (n = 100) and placebo (n = 101) arms. Irrespective of treatment, raw 24-month PFS rates were comparable across subgroups based on NLR tertiles [37.3% (low), 38.8% (middle), 38.8% (high); n = 67 per group] and NLR cutoff of 4 [38.1% (NLR ≤ 4; n = 176), 40.0% (NLR > 4; n = 25)]. Furthermore, NLRs were not prognostic in Cox models, irrespective of subgroups used. The therapeutic effect of lanreotide autogel/depot 120 mg was independent of NLRs (P > 0.1). These exploratory post-hoc analyses in patients with advanced intestinal and pancreatic NETs contrast with previous data suggesting NLR has prognostic potential in NETs. This may reflect the inclusion of patients with lower-grade tumors or use of higher NLR cutoff values in the current analysis.
Collapse
|
63
|
Gogineni V, Morand S, Staats H, Royfman R, Devanaboyina M, Einloth K, Dever D, Stanbery L, Aaron P, Manning L, Walter A, Edelman G, Dworkin L, Nemunaitis J. Current Ovarian Cancer Maintenance Strategies and Promising New Developments. J Cancer 2021; 12:38-53. [PMID: 33391401 PMCID: PMC7738841 DOI: 10.7150/jca.49406] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/17/2020] [Indexed: 12/11/2022] Open
Abstract
While ovarian cancer typically responds well to front line treatment, many patients will relapse within 5 years. Treatment options are less effective at each recurrence highlighting the need for novel maintenance therapies. PolyADP-ribose polymerase (PARP) inhibitors have recently gained approval in ovarian cancer maintenance. Niraparib was approved regardless of BRCA mutation status, however impact on overall survival is limited. Oliparib was approved for BRCA mutant and BRCA wildtype/homologous recombination deficient patients. This review will focus on current frontline ovarian cancer treatment as well molecularly based approaches to ovarian cancer management.
Collapse
|
64
|
Pires A, Greenshields-Watson A, Jones E, Smart K, Lauder SN, Somerville M, Milutinovic S, Kendrick H, Hindley JP, French R, Smalley MJ, Watkins WJ, Andrews R, Godkin A, Gallimore A. Immune Remodeling of the Extracellular Matrix Drives Loss of Cancer Stem Cells and Tumor Rejection. Cancer Immunol Res 2020; 8:1520-1531. [PMID: 33023965 PMCID: PMC7611107 DOI: 10.1158/2326-6066.cir-20-0070] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/19/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022]
Abstract
The nature of the tumor microenvironment (TME) influences the ability of tumor-specific T cells to control tumor growth. In this study, we performed an unbiased comparison of the TME of regulatory T-cell (Treg)-replete and Treg-depleted carcinogen-induced tumors, including Treg-depleted responding (regressing) and non-responding (growing) tumors. This analysis revealed an inverse relationship between extracellular matrix (ECM) and T-cell infiltrates where responding tumors were T-cell rich and ECM poor, whereas the converse was observed in non-responder tumors. For this reason, we hypothesized that the ECM acted as a barrier to successful T-cell infiltration and tumor rejection. However, further experiments revealed that this was not the case but instead showed that an effective T-cell response dramatically altered the density of ECM in the TME. Along with loss of ECM and high numbers of infiltrating T cells, responder tumors were distinguished by the development of lymphatic and blood vessel networks with specialized immune function. ECM-rich tumors exhibited a stem cell-like gene expression profile and superior tumor-initiating capacity, whereas such features were absent in responder tumors. Overall, these findings define an extended role for an effective immune response, not just in direct killing of tumor cells but in widescale remodeling of the TME to favor loss of ECM, elimination of cancer stem cells, and propagation of adaptive immunity.
Collapse
Affiliation(s)
- Ana Pires
- Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, University Hospital of Wales, Cardiff, United Kingdom.
| | - Alexander Greenshields-Watson
- Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, University Hospital of Wales, Cardiff, United Kingdom
| | - Emma Jones
- Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, University Hospital of Wales, Cardiff, United Kingdom
| | - Kathryn Smart
- Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, University Hospital of Wales, Cardiff, United Kingdom
| | - Sarah N Lauder
- Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, University Hospital of Wales, Cardiff, United Kingdom
| | - Michelle Somerville
- Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, University Hospital of Wales, Cardiff, United Kingdom
| | - Stefan Milutinovic
- Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, University Hospital of Wales, Cardiff, United Kingdom
| | - Howard Kendrick
- European Cancer Stem Cell Research Institute, Cardiff University, Hadyn Ellis Building, Cardiff, United Kingdom
| | - James P Hindley
- Indoor Biotechnologies, Vision Court, Cardiff, United Kingdom
| | - Rhiannon French
- European Cancer Stem Cell Research Institute, Cardiff University, Hadyn Ellis Building, Cardiff, United Kingdom
| | - Matthew J Smalley
- European Cancer Stem Cell Research Institute, Cardiff University, Hadyn Ellis Building, Cardiff, United Kingdom
| | - William J Watkins
- Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, University Hospital of Wales, Cardiff, United Kingdom
| | - Robert Andrews
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Andrew Godkin
- Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, University Hospital of Wales, Cardiff, United Kingdom
| | - Awen Gallimore
- Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, University Hospital of Wales, Cardiff, United Kingdom
| |
Collapse
|
65
|
Wang T, Shen Y, Luyten S, Yang Y, Jiang X. Tissue-resident memory CD8+ T cells in cancer immunology and immunotherapy. Pharmacol Res 2020; 159:104876. [DOI: 10.1016/j.phrs.2020.104876] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023]
|
66
|
High Endothelial Venule with Concomitant High CD8+ Tumor-Infiltrating Lymphocytes Is Associated with a Favorable Prognosis in Resected Gastric Cancer. J Clin Med 2020; 9:jcm9082628. [PMID: 32823631 PMCID: PMC7464373 DOI: 10.3390/jcm9082628] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/24/2022] Open
Abstract
CD8+ tumor-infiltrating lymphocytes (TILs) play a major role in antitumor immunity. High endothelial venules (HEVs) are related to diverse immune cells in solid tumors. We analyzed CD8+ and Foxp3+ TILs in combination with HEVs to determine their prognostic role in advanced gastric cancer (AGC). We enrolled 157 patients with AGC in this study. The densities of CD8+ TILs and Foxp3+ TILs were calculated using immunohistochemical staining. HEVs were evaluated by MECA-79 expression. HEVs were identified in 60 (38.2%) cases and was significantly associated with an increased number of CD8+ TILs (p = 0.027) but not of Foxp3+ TILs (p = 0.455) and CD20+ TILs (p = 0.163). A high CD8+/HEV+ level was significantly associated with nodal metastasis (p = 0.048). In survival analysis, patients with high CD8+/HEV+ levels demonstrated the longest overall survival (OS) (p = 0.015). Furthermore, a high CD8+/HEV+ level was an independent prognostic factor in AGC (p = 0.011; hazard ratio (HR) = 0.435; 95% confidence interval (CI) = 0.245–0.837). HEVs were found to play an important role in antitumor immunity associated with CD8+ TILs in AGC. This analysis of HEVs and CD8+ TILs helps stratify patients with AGC and sheds light on tumor immunity.
Collapse
|
67
|
Like a Rolling Stone: Sting-Cgas Pathway and Cell-Free DNA as Biomarkers for Combinatorial Immunotherapy. Pharmaceutics 2020; 12:pharmaceutics12080758. [PMID: 32796670 PMCID: PMC7464249 DOI: 10.3390/pharmaceutics12080758] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 12/19/2022] Open
Abstract
Combining immune checkpoint inhibitors with other treatments likely to harness tumor immunity is a rising strategy in oncology. The exact modalities of such a combinatorial regimen are yet to be defined, and most attempts have relied so far on concomitant dosing, rather than sequential or phased administration. Because immunomodulating features are likely to be time-, dose-, and-schedule dependent, the need for biomarkers providing real-time information is critical to better define the optimal time-window to combine immune checkpoint inhibitors with other drugs. In this review, we present the various putative markers that have been investigated as predictive tools with immune checkpoint inhibitors and could be used to help further combining treatments. Whereas none of the current biomarkers, such as the PDL1 expression of a tumor mutational burden, is suitable to identify the best way to combine treatments, monitoring circulating tumor DNA is a promising strategy, in particular to check whether the STING-cGAS pathway has been activated by cytotoxics. As such, circulating tumor DNA could help defining the best time-window to administrate immune checkpoint inhibitors after that cytotoxics have been given.
Collapse
|
68
|
Combes F, Meyer E, Sanders NN. Immune cells as tumor drug delivery vehicles. J Control Release 2020; 327:70-87. [PMID: 32735878 DOI: 10.1016/j.jconrel.2020.07.043] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022]
Abstract
This review article describes the use of immune cells as potential candidates to deliver anti-cancer drugs deep within the tumor microenvironment. First, the rationale of using drug carriers to target tumors and potentially decrease drug-related side effects is discussed. We further explain some of the current limitations when using nanoparticles for this purpose. Next, a comprehensive step-by-step description of the migration cascade of immune cells is provided as well as arguments on why immune cells can be used to address some of the limitations associated with nanoparticle-mediated drug delivery. We then describe the benefits and drawbacks of using red blood cells, platelets, granulocytes, monocytes, macrophages, myeloid-derived suppressor cells, T cells and NK cells for tumor-targeted drug delivery. An additional section discusses the versatility of nanoparticles to load anti-cancer drugs into immune cells. Lastly, we propose increasing the circulatory half-life and development of conditional release strategies as the two main future pillars to improve the efficacy of immune cell-mediated drug delivery to tumors.
Collapse
Affiliation(s)
- Francis Combes
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Evelyne Meyer
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium.
| |
Collapse
|
69
|
Derynck R, Turley SJ, Akhurst RJ. TGFβ biology in cancer progression and immunotherapy. Nat Rev Clin Oncol 2020; 18:9-34. [DOI: 10.1038/s41571-020-0403-1] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2020] [Indexed: 02/07/2023]
|
70
|
Chen J, Meng X, Zhou Q, Feng J, Zheng W, Wang Z, Wang J, Wang Y. Effect of CXCR5-Positive Cell Infiltration on the Immune Contexture and Patient Prognosis in Head and Neck Squamous Cell Carcinoma. Onco Targets Ther 2020; 13:5869-5877. [PMID: 32606797 PMCID: PMC7319516 DOI: 10.2147/ott.s248958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/22/2020] [Indexed: 11/23/2022] Open
Abstract
Purpose CXCR5-positive (CXCR5+) tumor cell infiltration has different prognostic values in different types of cancer. The objective was to evaluate the effect of CXCR5+ cell infiltration in head and neck squamous cell carcinoma (HNSCC). Patients and Methods The study included two patient cohorts: The Cancer Genome Atlas cohort (TCGA, n = 472) and the Renji Hospital cohort (RJHC, n = 201). The TCGA and RJHC cohorts were analyzed for CXCR5-related mRNAs and CXCR5+ cell infiltration, respectively. We then evaluated the correlation between CXCR5 mRNA and CXCR5+ cell infiltration in terms of overall survival and the immune contexture. Results The 5-year overall survival rate was significantly correlated with high CXCR5 mRNA expression and CXCR5+ cell infiltration in the TCGA and RJHC cohorts, respectively (p < 0.01), even after adjusting for confounders. Moreover, high CXCR5 mRNA expression was associated with more CD4+ T cells, CD8+ T cells, plasma cells, and less dendritic cells. A high CXCR5 mRNA expression was also correlated with increased expression of cytotoxic IFNG, TNFSF11 (RANKL), GZMA, GZMB, GZMK, GZMM, and PRF1 and increased expression of the immunosuppressive gene PDCD1 (PD-1), CD274 (PD-L1), CTLA4, LAG3, HAVCR2 (TIM-3), BTLA, and TIGIT. Conclusion HNSCC patients with a high intratumoral CXCR5 expression had a better prognosis than those with low intratumoral CXCR5 expression. Moreover, CXCR5+ cell infiltration could be used as an independent prognostic biomarker or as a potential therapeutic target. The presence of CXCR5+ cells affects the infiltration of immunocytes in head and neck cancer, differently from what was reported in other cancer types. Further randomized controlled trials or studies with more patients are needed to validate our results.
Collapse
Affiliation(s)
- Jun Chen
- Head and Neck Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Xiangchao Meng
- Bone and Joint Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Qinyi Zhou
- Head and Neck Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jialin Feng
- Head and Neck Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Wenjie Zheng
- Head and Neck Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Zhuoying Wang
- Head and Neck Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jiadong Wang
- Head and Neck Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - You Wang
- Bone and Joint Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
71
|
Avagliano A, Fiume G, Pelagalli A, Sanità G, Ruocco MR, Montagnani S, Arcucci A. Metabolic Plasticity of Melanoma Cells and Their Crosstalk With Tumor Microenvironment. Front Oncol 2020; 10:722. [PMID: 32528879 PMCID: PMC7256186 DOI: 10.3389/fonc.2020.00722] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/16/2020] [Indexed: 12/21/2022] Open
Abstract
Cutaneous melanoma (CM) is a highly aggressive and drug resistant solid tumor, showing an impressive metabolic plasticity modulated by oncogenic activation. In particular, melanoma cells can generate adenosine triphosphate (ATP) during cancer progression by both cytosolic and mitochondrial compartments, although CM energetic request mostly relies on glycolysis. The upregulation of glycolysis is associated with constitutive activation of BRAF/MAPK signaling sustained by BRAFV600E kinase mutant. In this scenario, the growth and progression of CM are strongly affected by melanoma metabolic changes and interplay with tumor microenvironment (TME) that sustain tumor development and immune escape. Furthermore, CM metabolic plasticity can induce a metabolic adaptive response to BRAF/MEK inhibitors (BRAFi/MEKi), associated with the shift from glycolysis toward oxidative phosphorylation (OXPHOS). Therefore, in this review article we survey the metabolic alterations and plasticity of CM, its crosstalk with TME that regulates melanoma progression, drug resistance and immunosurveillance. Finally, we describe hallmarks of melanoma therapeutic strategies targeting the shift from glycolysis toward OXPHOS.
Collapse
Affiliation(s)
- Angelica Avagliano
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Alessandra Pelagalli
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy.,Institute of Biostructures and Bioimages, National Research Council, Naples, Italy
| | - Gennaro Sanità
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Stefania Montagnani
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
72
|
Chen M, Qiao G, Hylander BL, Mohammadpour H, Wang XY, Subjeck JR, Singh AK, Repasky EA. Adrenergic stress constrains the development of anti-tumor immunity and abscopal responses following local radiation. Nat Commun 2020; 11:1821. [PMID: 32286326 PMCID: PMC7156731 DOI: 10.1038/s41467-020-15676-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
The abscopal effect following ionizing radiation therapy (RT) is considered to be a rare event. This effect does occur more frequently when combined with other therapies, including immunotherapy. Here we demonstrate that the frequency of abscopal events following RT alone is highly dependent upon the degree of adrenergic stress in the tumor-bearing host. Using a combination of physiologic, pharmacologic and genetic strategies, we observe improvements in the control of both irradiated and non-irradiated distant tumors, including metastatic tumors, when adrenergic stress or signaling through β-adrenergic receptor is reduced. Further, we observe cellular and molecular evidence of improved, antigen-specific, anti-tumor immune responses which also depend upon T cell egress from draining lymph nodes. These data suggest that blockade of β2 adrenergic stress signaling could be a useful, safe, and feasible strategy to improve efficacy in cancer patients undergoing radiation therapy.
Collapse
MESH Headings
- Adrenergic Agents/pharmacology
- Adrenergic beta-Antagonists/pharmacology
- Animals
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Disease Models, Animal
- Gene Expression Regulation, Neoplastic/drug effects
- Immunity
- Lymph Nodes/pathology
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/radiotherapy
- Radiation, Ionizing
- Receptors, Adrenergic, beta-2/metabolism
- Signal Transduction/drug effects
- Stress, Physiological
Collapse
Affiliation(s)
- Minhui Chen
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Guanxi Qiao
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Bonnie L Hylander
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Hemn Mohammadpour
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Xiang-Yang Wang
- Department of Genetics, Virginia Commonwealth University, Richmond, VI, 23298, USA
| | - John R Subjeck
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Anurag K Singh
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
- Department of Radiation Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
73
|
Spagnuolo A, Palazzolo G, Sementa C, Gridelli C. Vascular endothelial growth factor receptor tyrosine kinase inhibitors for the treatment of advanced non-small cell lung cancer. Expert Opin Pharmacother 2020; 21:491-506. [DOI: 10.1080/14656566.2020.1713092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- A Spagnuolo
- Division of Medical Oncology, ‘S. G. Moscati’ Hospital, Avellino, Italy
| | - G Palazzolo
- Division of Medical Oncology, “ULSS 15 Cittadella”, Cittadella, Padova, Italy
| | - C Sementa
- Division of Legal Medicine, ‘S. G. Moscati’ Hospital, Avellino, Italy
| | - C Gridelli
- Division of Medical Oncology, ‘S. G. Moscati’ Hospital, Avellino, Italy
| |
Collapse
|
74
|
Zhang J, Endres S, Kobold S. Enhancing tumor T cell infiltration to enable cancer immunotherapy. Immunotherapy 2020; 11:201-213. [PMID: 30730277 DOI: 10.2217/imt-2018-0111] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Cancer immunotherapy has changed the treatment landscape for cancer patients, especially for those with metastatic spread. While the immunotherapeutic armamentarium is constantly growing, as exemplified by approved compounds, clinical outcome remains variable both within and across entities. A sufficient infiltration into the tumor microenvironment and successful activation of effector T lymphocytes against tumor cells have been identified as predictors for responses to T cell-based immunotherapies. However, tumor cells have developed a variety of mechanisms to reduce T cell homing and access to the tumor tissue to prevent activity of anticancer immunity. As a consequence, investigations have interrogated strategies to improve the efficacy of cancer immunotherapies by enhancing T cell infiltration into tumor tissues. In this review, we summarize mechanisms of how tumor tissue shapes immune suppressive microenvironment to prevent T cell access to the tumor site. We focus on current strategies to improve cancer immunotherapies through enhancing T cell infiltration.
Collapse
Affiliation(s)
- Jin Zhang
- Center of Integrated Protein Science Munich (CIPS-M) & Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337 Munich, Germany, Member of the German Center of Lung Research
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) & Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337 Munich, Germany, Member of the German Center of Lung Research
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) & Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337 Munich, Germany, Member of the German Center of Lung Research
| |
Collapse
|
75
|
Wang K, He H. Pancreatic Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1296:243-257. [PMID: 34185297 DOI: 10.1007/978-3-030-59038-3_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The pancreatic ductal adenocarcinoma (PDAC) microenvironment is a diverse and complex milieu of immune, stromal, and tumor cells and is characterized by a dense stroma, which mediates the interaction between the tumor and the immune system within the tumor microenvironment (TME). The interaction between stromal and tumor cells signals and shapes the immune infiltration of TME. The desmoplastic compartment contains infiltrated immune cells including tumor-associated macrophages (TAMs) and large numbers of fibroblasts/myofibroblasts dominated by pancreatic stellate cells (PSCs) which contribute to fibrosis. The highly fibrotic stroma with its extensive infiltration of immunosuppressive cells forms the major component of the pro-tumorigenic microenvironment (Laklai et al. Nat Med 22:497-505, 2016, Zhu et al. Cancer Res 74:5057-5069, 2014) provides a barrier to the delivery of cytotoxic agents and limits T-cell access to tumor cells (Feig et al. Proc Natl Acad Sci USA 110:20212-20217, 2013, Provenzano et al Cancer Cell 21:418-429, 2012). Activated PSCs reduced infiltration of cytotoxic T cells to the juxtatumoral stroma (immediately adjacent to the tumor epithelial cells) of PDAC (Ene-Obong et al. Gastroenterology 145:1121-1132, 2013). M1 macrophages activate an immune response against tumor, but M2 macrophages are involved in immunosuppression promoting tumor progression (Noy and Pollard Immunity 41:49-61, 2014, Ruffell et al. Trends Immunol 33:119-126, 2012). The desmoplastic stroma is reported to protect tumor cells against chemotherapies, promoting their proliferation and migration. However, experimental depletion of the desmoplastic stroma has led to more aggressive cancers in animal studies (Nielsen et al. World J Gastroenterol 22:2678-2700, 2016). Hence reprogramming rather than simple depletion of the PDAC stroma has the potential for developing new therapeutic strategies for PC treatment. Modulation of PSCs/fibrosis and immune infiltration/inflammation composes the major aspects of TME reprogramming.
Collapse
Affiliation(s)
- Kai Wang
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, VIC, Australia
| | - Hong He
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, VIC, Australia.
| |
Collapse
|
76
|
Ye J, Mills BN, Zhao T, Han BJ, Murphy JD, Patel AP, Johnston CJ, Lord EM, Belt BA, Linehan DC, Gerber SA. Assessing the Magnitude of Immunogenic Cell Death Following Chemotherapy and Irradiation Reveals a New Strategy to Treat Pancreatic Cancer. Cancer Immunol Res 2020; 8:94-107. [PMID: 31719057 PMCID: PMC6946873 DOI: 10.1158/2326-6066.cir-19-0373] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/18/2019] [Accepted: 11/07/2019] [Indexed: 12/22/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) continues to have a dismal prognosis, in part, due to ineffective treatment strategies. The efficacy of some chemotherapies and especially radiotherapy is mediated partially by the immune system. Therefore, we hypothesized that profiling the immune response following chemotherapy and/or irradiation can be used as a readout for treatment efficacy but also to help identify optimal therapeutic schedules for PDAC. Using murine models of PDAC, we demonstrated that concurrent administration of stereotactic body radiotherapy (SBRT) and a modified dose of FOLFIRINOX (mFX) resulted in superior tumor control when compared with single or sequential treatment groups. Importantly, this combined treatment schedule enhanced the magnitude of immunogenic cell death, which in turn amplified tumor antigen presentation by dendritic cells and intratumoral CD8+ T-cell infiltration. Concurrent therapy also resulted in systemic immunity contributing to the control of established metastases. These findings provide a rationale for pursuing concurrent treatment schedules of SBRT with mFX in PDAC.
Collapse
Affiliation(s)
- Jian Ye
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Bradley N Mills
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Tony Zhao
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Booyeon J Han
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Joseph D Murphy
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Ankit P Patel
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Carl J Johnston
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Edith M Lord
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Brian A Belt
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - David C Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Scott A Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, New York.
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
77
|
Chen H, Cong X, Wu C, Wu X, Wang J, Mao K, Li J, Zhu G, Liu F, Meng X, Song J, Sun X, Wang X, Liu S, Zhang S, Yang X, Song Y, Yang YG, Sun T. Intratumoral delivery of CCL25 enhances immunotherapy against triple-negative breast cancer by recruiting CCR9 + T cells. SCIENCE ADVANCES 2020; 6:eaax4690. [PMID: 32064335 PMCID: PMC6989134 DOI: 10.1126/sciadv.aax4690] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 11/20/2019] [Indexed: 05/22/2023]
Abstract
CCR9+ T cells have an increased potential to be activated and therefore may mediate strong antitumor responses. Here, we found, however, that CCL25, the only chemokine for CCR9+ cells, is not expressed in human or murine triple-negative breast cancers (TNBCs), raising a hypothesis that intratumoral delivery of CCL25 may enhance antitumor immunotherapy in TNBCs. We first determined whether this approach can enhance CD47-targeted immunotherapy using a tumor acidity-responsive nanoparticle delivery system (NP-siCD47/CCL25) to sequentially release CCL25 protein and CD47 small interfering RNA in tumor. NP-siCD47/CCL25 significantly increased infiltration of CCR9+CD8+ T cells and down-regulated CD47 expression in tumor, resulting in inhibition of tumor growth and metastasis through a T cell-dependent immunity. Furthermore, the antitumor effect of NP-siCD47/CCL25 was synergistically enhanced when used in combination with programmed cell death protein-1/programmed death ligand-1 blockades. This study offers a strategy to enhance immunotherapy by promoting CCR9+CD8+ T cell tumor infiltration.
Collapse
Affiliation(s)
- Hongmei Chen
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Xiuxiu Cong
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Chenxi Wu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Xuan Wu
- Institute of Translational Medicine, China Medical University, Liaoning, China
| | - Jialiang Wang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- International Center of Future Science, Jilin University, Changchun, Jilin, China
| | - Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- International Center of Future Science, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Jie Li
- Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Ge Zhu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Feiqi Liu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Jia Song
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Xu Sun
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Xin Wang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Shi Zhang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Xianzhu Yang
- Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yanqiu Song
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- Corresponding author. (T.S.); (Y.S.); (Y.-G.Y.)
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- International Center of Future Science, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
- Corresponding author. (T.S.); (Y.S.); (Y.-G.Y.)
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- International Center of Future Science, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China
- Corresponding author. (T.S.); (Y.S.); (Y.-G.Y.)
| |
Collapse
|
78
|
Strazza M, Adam K, Smrcka AV, Lerrer S, Mor A. PLCε1 suppresses tumor growth by regulating murine T cell mobilization. Clin Exp Immunol 2019; 200:53-60. [PMID: 31867717 DOI: 10.1111/cei.13409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2019] [Indexed: 01/01/2023] Open
Abstract
Phospholipase C epsilon 1 (PLCε1) is a unique member of the phospholipase family, in that it also functions as a guanine nucleotide exchange factor (GEF) for the small GTPase Rap1. It is this function as a Rap1 GEF that gives PLCε1 an essential role in chemokine-mediated T cell adhesion. We have utilized a syngeneic tumor model, MC38 cells in C57BL/6 mice, and observed that tumors grow larger and more quickly in the absence of PLCε1. Single-cell analysis revealed an increased CD4+ /CD8+ ratio in the spleens, lymph nodes and tumors of PLCε1 knock-out tumor-bearing mice. T cells isolated from PLCε1 knock-out mice were less activated by multiple phenotypical parameters than those from wild-type mice. We additionally noted a decrease in expression of the chemokine receptors C-X-C chemokine receptor type 4 (CXCR4) and C-C motif chemokine receptor 4 (CCR4) on CD4+ T cells from the spleens, lymph nodes and tumors of PLCε1 knock-out mice compared to wild-type mice, and diminished migration of PLCε1-depleted CD3+ T cells towards stromal cell-derived factor (SDF)-1α. Based on these results, we conclude that PLCε1 is a potential regulator of tumor-infiltrating lymphocytes, functioning, at least in part, at the level of T cell trafficking and recruitment.
Collapse
Affiliation(s)
- M Strazza
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - K Adam
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - A V Smrcka
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - S Lerrer
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - A Mor
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
79
|
Cui X, Song P, Zhang L. [New Advances in the Treatment for Small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2019; 22:355-362. [PMID: 31196369 PMCID: PMC6580078 DOI: 10.3779/j.issn.1009-3419.2019.06.05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Small cell lung cancer (SCLC) is a refractory cancer with high degree of malignancy, rapid disease progression, poor prognosis and easy recurrence. In the past 30 years, the traditional treatment of SCLC, mainly chemotherapy and radiotherapy, has not changed significantly, and the effective treatment method for clinical needs is extremely urgent. The rapid development of precision medicine has revealed the molecular biological characteristics of SCLC, so its diagnosis and treatment will into a new era. At present, some studies have shown that anti-angiogenic drugs, immunotherapy and so on have improved the efficacy of SCLC treatment to some extent, and there are more studies on the diagnosis and treatment of SCLC, so a new field of SCLC treatment are coming and bringing more survival benefits to patients. New studies on targeted therapy, anti-angiogenesis drugs and immunotherapy of molecular pathology of SCLC are emerging. This paper reviews the new diagnosis and treatment methods of SCLC to provide new guidance for its clinical treatment.
.
Collapse
Affiliation(s)
- Xiaoxia Cui
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Peng Song
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
80
|
Shah AB, Sommerer KR, Almhanna K. Immune checkpoint inhibitors in gastrointestinal malignancies: what can we learn from experience with other tumors? Transl Gastroenterol Hepatol 2019; 4:73. [PMID: 31728430 PMCID: PMC6851451 DOI: 10.21037/tgh.2019.09.04] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/12/2019] [Indexed: 12/31/2022] Open
Abstract
Gastrointestinal (GI) malignancies are some of the most common cancers worldwide with high rates of morbidity and mortality. Immune checkpoint inhibitors have afforded additional treatment options for patients, but their success has been limited. Conversely, in other tumor types such as lung cancer, melanoma and renal cell carcinoma, treatment strategies with immune checkpoint inhibitors have propelled those agents into the front lines of treatment. Strategies utilized include combining immune checkpoint inhibitors with chemotherapy, other checkpoint inhibitors, and targeted therapy. In this review, we analyze combination strategies employed in other tumor types to help identify current and future approaches toward improving outcomes with immunotherapy in GI malignancies.
Collapse
Affiliation(s)
- Anand B. Shah
- Department of Pharmacy, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Khaldoun Almhanna
- Division of Hematology/Oncology, The Warren Alpert Medical School of Brown University, Lifespan Cancer Institute, Rhode Island Hospital, Providence, RI, USA
| |
Collapse
|
81
|
Weishaupt C, Steinert M, Brunner G, Schulze HJ, Fuhlbrigge RC, Goerge T, Loser K. Activation of human vascular endothelium in melanoma metastases induces ICAM-1 and E-selectin expression and results in increased infiltration with effector lymphocytes. Exp Dermatol 2019; 28:1258-1269. [PMID: 31444891 DOI: 10.1111/exd.14023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/18/2019] [Accepted: 08/15/2019] [Indexed: 12/12/2022]
Abstract
Lymphocytic infiltration into melanoma tissue is an important prerequisite for effective antitumoral immunity. However, analysis of human metastatic melanoma has shown that leucocyte adhesion receptor expression on melanoma blood vessels is very low or absent, thereby impairing the entry of cytotoxic lymphocytes into tumor tissue. We hypothesized that adhesion molecules can be induced on melanoma vasculature allowing better infiltration of cytotoxic lymphocytes. Quantitative real-time PCR and immunofluorescence staining indicated that the adhesion molecules ICAM-1 (CD54) and E-selectin (CD62E) can be significantly induced by intralesional application of TNF alpha in tissue from human melanoma metastases either in vitro or in vivo when grafted onto immunodeficient NSG (NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ) mice that preserved human vessels. Furthermore, activated human autologous CD3+ lymphocytes were injected intravenously into mice bearing melanoma xenografts treated with TNF-α or PBS in addition to the leucocyte chemoattractant TARC (CCL17). Significantly increased numbers of CD8+ cells were detected in TNF-α-treated melanoma metastases compared with PBS-treated controls. In addition, tumor cell apoptosis was enhanced and melanoma cell proliferation reduced as shown by TUNEL assay and KI-67 staining. We conclude that adhesion molecules can be induced on human melanoma vasculature resulting in significantly improved homing of activated autologous cytotoxic T cells to melanoma tissue and inhibition of melanoma cell proliferation. These observations should be considered when designing protocols for immunotherapy of malignant melanoma.
Collapse
Affiliation(s)
- Carsten Weishaupt
- Department of Dermatology, University Hospital of Muenster, Muenster, Germany
| | - Meike Steinert
- Department of Dermatology, University Hospital of Muenster, Muenster, Germany
| | - Georg Brunner
- Department of Cancer Research, Fachklinik Hornheide, Münster, Germany
| | | | - Robert C Fuhlbrigge
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tobias Goerge
- Department of Dermatology, University Hospital of Muenster, Muenster, Germany
| | - Karin Loser
- Department of Dermatology, University Hospital of Muenster, Muenster, Germany.,CRC1009, CRC-TR128 and Interdisciplinary Center of Clinical Research (IZKF), University of Münster, Münster, Germany
| |
Collapse
|
82
|
Li T, Li H, Li S, Xu S, Zhang W, Gao H, Xu H, Wu C, Wang W, Yu X, Liu L. Research progress and design optimization of CAR-T therapy for pancreatic ductal adenocarcinoma. Cancer Med 2019; 8:5223-5231. [PMID: 31339230 PMCID: PMC6718528 DOI: 10.1002/cam4.2430] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant cancer with limited treatment options. Chimeric antigen receptor T cells (CAR-T) are genetically engineered T cells that can specifically kill tumor cells without major histocompatibility complex restriction. Encouraging progress in CAR-T therapy for PDAC has been made in preclinical and early phase clinical trials. Challenges in CAR-T therapy for solid tumors still exist, including immunosuppressive microenvironment, interstitial barrier, poor chemotaxis, and the "on-target, off-tumor" effect. Applying neoantigens of PDAC as targets for CAR-T therapy, recognizing the CAR-T subgroup with better antitumor effect, and designing a CAR-T system targeting stroma of PDAC may contribute to develop a powerful CAR-T therapy for PDAC in the future.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm
- Biomarkers, Tumor
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/therapy
- Chemotaxis/immunology
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Research
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Microenvironment/immunology
- Pancreatic Neoplasms
Collapse
Affiliation(s)
- Tianjiao Li
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Hao Li
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Shuo Li
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Shuaishuai Xu
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Wuhu Zhang
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Heli Gao
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Huaxiang Xu
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Chuntao Wu
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Wenquan Wang
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Xianjun Yu
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| | - Liang Liu
- Department of Pancreatic Surgery, Shanghai Cancer CentreFudan UniversityShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Pancreatic Cancer InstituteShanghaiChina
- Pancreatic Cancer InstituteFudan UniversityShanghaiChina
| |
Collapse
|
83
|
Patterns of immune-cell infiltration in murine models of melanoma: roles of antigen and tissue site in creating inflamed tumors. Cancer Immunol Immunother 2019; 68:1121-1132. [PMID: 31134297 DOI: 10.1007/s00262-019-02345-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 05/08/2019] [Indexed: 12/22/2022]
Abstract
Immune-cell infiltration is associated with improved survival in melanoma. Human melanoma metastases may be grouped into immunotypes representing patterns of immune-cell infiltration: A (sparse), B (perivascular cuffing), and C (diffuse). Immunotypes have not been defined for murine melanomas, but may provide opportunities to understand mechanism-driving immunotype differences. We performed immunohistochemistry with immune-cell enumeration, immunotyping, and vascular density scoring in genetically engineered (Braf/Pten and Braf/Pten/β-catenin) and transplantable (B16-F1, B16-OVA, and B16-AAD) murine melanomas. The transplantable tumors were grown in subcutaneous (s.c.) or intraperitoneal (i.p.) locations. Braf/Pten and Braf/Pten/β-catenin tumors had low immune-cell densities, defining them as Immunotype A, as did B16-F1 tumors. B16-OVA (s.c. and i.p.) and B16-AAD s.c. tumors were Immunotype B, while B16-AAD i.p. tumors were primarily Immunotype C. Interestingly, the i.p. location was characterized by higher immune-cell counts in B16-OVA tumors, with counts that trended higher for B16-F1 and B16-AAD. The i.p. location was also characterized by higher vascularity in B16-F1 and B16-AAD tumors. These findings demonstrate that spontaneously mutated neoantigens in B16 melanomas were insufficient to induce robust intratumoral immune-cell infiltrates, but instead were Immunotype A tumors. The addition of model neoantigens (OVA or AAD) to B16 enhanced infiltration, but this most often resulted in Immunotype B. We find that tumor location may be an important element in enabling Immunotype C tumors. In aggregate, these data suggest important roles both for the antigen type and for the tumor location in defining immunotypes.
Collapse
|
84
|
Harjunpää H, Llort Asens M, Guenther C, Fagerholm SC. Cell Adhesion Molecules and Their Roles and Regulation in the Immune and Tumor Microenvironment. Front Immunol 2019; 10:1078. [PMID: 31231358 PMCID: PMC6558418 DOI: 10.3389/fimmu.2019.01078] [Citation(s) in RCA: 457] [Impact Index Per Article: 76.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/29/2019] [Indexed: 12/14/2022] Open
Abstract
The immune system and cancer have a complex relationship with the immune system playing a dual role in tumor development. The effector cells of the immune system can recognize and kill malignant cells while immune system-mediated inflammation can also promote tumor growth and regulatory cells suppress the anti-tumor responses. In the center of all anti-tumor responses is the ability of the immune cells to migrate to the tumor site and to interact with each other and with the malignant cells. Cell adhesion molecules including receptors of the immunoglobulin superfamily and integrins are of crucial importance in mediating these processes. Particularly integrins play a vital role in regulating all aspects of immune cell function including immune cell trafficking into tissues, effector cell activation and proliferation and the formation of the immunological synapse between immune cells or between immune cell and the target cell both during homeostasis and during inflammation and cancer. In this review we discuss the molecular mechanisms regulating integrin function and the role of integrins and other cell adhesion molecules in immune responses and in the tumor microenvironment. We also describe how malignant cells can utilize cell adhesion molecules to promote tumor growth and metastases and how these molecules could be targeted in cancer immunotherapy.
Collapse
Affiliation(s)
- Heidi Harjunpää
- Research Program of Molecular and Integrative Biosciences, Faculty of Bio- and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Marc Llort Asens
- Research Program of Molecular and Integrative Biosciences, Faculty of Bio- and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Carla Guenther
- Research Program of Molecular and Integrative Biosciences, Faculty of Bio- and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Susanna C Fagerholm
- Research Program of Molecular and Integrative Biosciences, Faculty of Bio- and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
85
|
Dong X, Liang J, Yang A, Qian Z, Kong D, Lv F. Fluorescence imaging guided CpG nanoparticles-loaded IR820-hydrogel for synergistic photothermal immunotherapy. Biomaterials 2019; 209:111-125. [PMID: 31034980 DOI: 10.1016/j.biomaterials.2019.04.024] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/11/2019] [Accepted: 04/21/2019] [Indexed: 02/05/2023]
Abstract
As synergistic photothermal immunotherapy has developed as one of the most attractive strategies for cancer therapy, it is crucial to design an effective photothermal immunotherapy system to enhance the synergistic anti-tumor effect and reveal the essential role of each treatment. In this study, we designed CpG self-crosslinked nanoparticles-loaded IR820-conjugated hydrogel with dual self-fluorescence to exert the combined photothermal-immunotherapy. IR820-hydrogel can be effective for hyperthermia to eliminate the primary tumor based on its comprehensive coverage and generated photothermal-induced tumor antigens for assisted immunotherapy. CpG self-crosslinked nanoparticles improved the immune response of adjuvant against melanoma without extra nano-carriers. The synergistic photothermal immunotherapy was achieved by the merging of CpG self-crosslinked nanoparticles and IR820-hydrogel. A possible mechanism of combined antitumor effect was further revealed by analyzing immune cells including CD8 +T cells, DCs, B cells, Treg and MDSC in tumor microenvironment. The specific antitumor immunity was provoked to remove the tumor residues and ultimately the combined treatment mode achieved more effective systemic therapeutic effect than either photothermal therapy or immunotherapy alone. Furthermore, self-fluorescent IR820-hydrogel and CpG nanoparticles exerted the imaging-guided combined photothermal-immunotherapy by the dual fluorescence imaging method without additional fluorescent labeling. This visible combined photothermal-immunotherapy offers a potential for precise cancer treatment.
Collapse
Affiliation(s)
- Xia Dong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China
| | - Jie Liang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China
| | - Afeng Yang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy, West China Hospital, And Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Deling Kong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China
| | - Feng Lv
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China.
| |
Collapse
|
86
|
Dong X, Liang J, Yang A, Qian Z, Kong D, Lv F. A Visible Codelivery Nanovaccine of Antigen and Adjuvant with Self-Carrier for Cancer Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:4876-4888. [PMID: 30628437 DOI: 10.1021/acsami.8b20364] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Codelivery nanovaccines of antigens and adjuvants have achieved positive therapy for cancer immunotherapy. The insufficient immunogenicity of these vaccines leads to the difficulty of eliciting robust immune effects for immune clearance due to the inadequate loading efficiency, complex preparation processes, low safety concerns, and weak immune responses. Herein, a visible codelivery nanovaccine of an antigen and adjuvant based on self-cross-linked antigen nanoparticles (ovalbumin nanoparticles (ONPs)) combined with the adjuvant (CpG) for cancer immunotherapy was prepared using antigens themselves as carriers. ONPs not only provide sufficient antigens for continuous simulation of the immune response with high antigen loading efficiency but also serve as natural carriers of CpG. In vitro and in vivo experiments proved that ONPs-CpG can elicit a robust immune response including DC maturity, T cell activation, and IFN-γ production. ONPs-CpG induced strong tumor-specific immunity and exhibited remarkable antitumor immunotherapy effects in vivo using mouse models of lymphoma. Furthermore, to perform the precise vaccine delivery, the dual fluorescent codelivery nanovaccine was monitored in real time in vivo by the visible imaging method. With regard to migration tracking, fluorescence imaging allowed for both high resolution and sensitivity of visible detection based on the fluorescence of ONPs and CpG. The multifunctional nanovaccine could function as a robust platform for cancer immunotherapy and a visible system for antigen-adjuvant tracking.
Collapse
Affiliation(s)
- Xia Dong
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , PR China
| | - Jie Liang
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , PR China
| | - Afeng Yang
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , PR China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy, West China Hospital, and Collaborative Innovation Center of Biotherapy , Sichuan University , Chengdu 610041 , Sichuan , PR China
| | - Deling Kong
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , PR China
| | | |
Collapse
|
87
|
Zhang G, Liu C, Bai H, Cao G, Cui R, Zhang Z. Combinatorial therapy of immune checkpoint and cancer pathways provides a novel perspective on ovarian cancer treatment. Oncol Lett 2019; 17:2583-2591. [PMID: 30854033 DOI: 10.3892/ol.2019.9902] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 11/21/2018] [Indexed: 12/14/2022] Open
Abstract
An increasing number of studies have reported that immunotherapy serves a significant role in ovarian cancer treatment. In recent years, blockade of checkpoint pathways, including programmed death-ligand 1 (PD-L1)/programmed death-1 and cytotoxic T-lymphocyte-associated protein 4, has demonstrated significant clinical and preclinical benefits in the treatment of ovarian cancer. Additionally, tumor-associated angiogenesis and homologous recombination deficiency frequently occurs in patients with high-grade ovarian cancer, which makes cancer cells more susceptible to targeted therapies, including therapies targeting poly (ADP-ribose) polymerase inhibitor, and anti-angiogenic approaches. Additionally, targeted therapy has been associated with elevated PD-L1 expression in tumor cells, increased T-cell infiltration in tumors and dendritic cell stimulation. This synergistic effect provides the rationale for the joint application of targeted therapy and immunotherapy. Checkpoint blockades are able to elicit durable antitumor immune reactions and complement the transient antitumor effect of targeted therapies. The current review discusses the underlying mechanism of these therapies and novel developments in combined therapy for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Guyu Zhang
- Department of Gynecology and Obstetrics, Beijing Chaoyang Hospital, Capital Medical University, Beijing 10000, P.R. China
| | - Chongdong Liu
- Department of Gynecology and Obstetrics, Beijing Chaoyang Hospital, Capital Medical University, Beijing 10000, P.R. China
| | - Huiming Bai
- Department of Gynecology and Obstetrics, Beijing Chaoyang Hospital, Capital Medical University, Beijing 10000, P.R. China
| | - Guangming Cao
- Department of Gynecology and Obstetrics, Beijing Chaoyang Hospital, Capital Medical University, Beijing 10000, P.R. China
| | - Ran Cui
- Department of Gynecology and Obstetrics, Beijing Chaoyang Hospital, Capital Medical University, Beijing 10000, P.R. China
| | - Zhengyu Zhang
- Department of Gynecology and Obstetrics, Beijing Chaoyang Hospital, Capital Medical University, Beijing 10000, P.R. China
| |
Collapse
|
88
|
Kaur A, Ecker BL, Douglass SM, Kugel CH, Webster MR, Almeida FV, Somasundaram R, Hayden J, Ban E, Ahmadzadeh H, Franco-Barraza J, Shah N, Mellis IA, Keeney F, Kossenkov A, Tang HY, Yin X, Liu Q, Xu X, Fane M, Brafford P, Herlyn M, Speicher DW, Wargo JA, Tetzlaff MT, Haydu LE, Raj A, Shenoy V, Cukierman E, Weeraratna AT. Remodeling of the Collagen Matrix in Aging Skin Promotes Melanoma Metastasis and Affects Immune Cell Motility. Cancer Discov 2019; 9:64-81. [PMID: 30279173 PMCID: PMC6328333 DOI: 10.1158/2159-8290.cd-18-0193] [Citation(s) in RCA: 278] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/23/2018] [Accepted: 09/19/2018] [Indexed: 01/30/2023]
Abstract
Physical changes in skin are among the most visible signs of aging. We found that young dermal fibroblasts secrete high levels of extracellular matrix (ECM) constituents, including proteoglycans, glycoproteins, and cartilage-linking proteins. The most abundantly secreted was HAPLN1, a hyaluronic and proteoglycan link protein. HAPLN1 was lost in aged fibroblasts, resulting in a more aligned ECM that promoted metastasis of melanoma cells. Reconstituting HAPLN1 inhibited metastasis in an aged microenvironment, in 3-D skin reconstruction models, and in vivo. Intriguingly, aged fibroblast-derived matrices had the opposite effect on the migration of T cells, inhibiting their motility. HAPLN1 treatment of aged fibroblasts restored motility of mononuclear immune cells, while impeding that of polymorphonuclear immune cells, which in turn affected regulatory T-cell recruitment. These data suggest that although age-related physical changes in the ECM can promote tumor cell motility, they may adversely affect the motility of some immune cells, resulting in an overall change in the immune microenvironment. Understanding the physical changes in aging skin may provide avenues for more effective therapy for older patients with melanoma. SIGNIFICANCE: These data shed light on the mechanochemical interactions that occur between aged skin, tumor, and immune cell populations, which may affect tumor metastasis and immune cell infiltration, with implications for the efficacy of current therapies for melanoma.See related commentary by Marie and Merlino, p. 19.This article is highlighted in the In This Issue feature, p. 1.
Collapse
Affiliation(s)
- Amanpreet Kaur
- Department of Biological Sciences, University of the Sciences, Philadelphia, Pennsylvania
- The Wistar Institute, Philadelphia, Pennsylvania
- School of Engineering and Applied Science, Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | - James Hayden
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Ehsan Ban
- School of Engineering and Applied Science, Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hossein Ahmadzadeh
- School of Engineering and Applied Science, Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Neelima Shah
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Ian A Mellis
- School of Engineering and Applied Science, Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | - Xiangfan Yin
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Qin Liu
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Xiaowei Xu
- Department of Pathology, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | - Jennifer A Wargo
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Lauren E Haydu
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Arjun Raj
- School of Engineering and Applied Science, Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vivek Shenoy
- School of Engineering and Applied Science, Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edna Cukierman
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | |
Collapse
|
89
|
|
90
|
Georganaki M, van Hooren L, Dimberg A. Vascular Targeting to Increase the Efficiency of Immune Checkpoint Blockade in Cancer. Front Immunol 2018; 9:3081. [PMID: 30627131 PMCID: PMC6309238 DOI: 10.3389/fimmu.2018.03081] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/12/2018] [Indexed: 12/14/2022] Open
Abstract
Boosting natural immunity against malignant cells has had a major breakthrough in clinical cancer therapy. This is mainly due to the successful development of immune checkpoint blocking antibodies, which release a break on cytolytic anti-tumor-directed T-lymphocytes. However, immune checkpoint blockade is only effective for a proportion of cancer patients, and a major challenge in the field is to understand and overcome treatment resistance. Immune checkpoint blockade relies on successful trafficking of tumor-targeted T-lymphocytes from the secondary lymphoid organs, through the blood stream and into the tumor tissue. Resistance to therapy is often associated with a low density of T-lymphocytes residing within the tumor tissue prior to treatment. The recruitment of leukocytes to the tumor tissue relies on up-regulation of adhesion molecules and chemokines by the tumor vasculature, which is denoted as endothelial activation. Tumor vessels are often poorly activated due to constitutive pro-angiogenic signaling in the tumor microenvironment, and therefore constitute barriers to efficient leukocyte recruitment. An emerging possibility to enhance the efficiency of cancer immunotherapy is to combine pro-inflammatory drugs with anti-angiogenic therapy, which can enable tumor-targeted T-lymphocytes to access the tumor tissue by relieving endothelial anergy and increasing adhesion molecule expression. This would pave the way for efficient immune checkpoint blockade. Here, we review the current understanding of the biological basis of endothelial anergy within the tumor microenvironment, and discuss the challenges and opportunities of combining vascular targeting with immunotherapeutic drugs as suggested by data from key pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Maria Georganaki
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, Uppsala, Sweden
| | - Luuk van Hooren
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, Uppsala, Sweden
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, Uppsala, Sweden
| |
Collapse
|
91
|
Petrillo M, Patella F, Pesapane F, Suter MB, Ierardi AM, Angileri SA, Floridi C, de Filippo M, Carrafiello G. Hypoxia and tumor angiogenesis in the era of hepatocellular carcinoma transarterial loco-regional treatments. Future Oncol 2018; 14:2957-2967. [DOI: 10.2217/fon-2017-0739] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
This review focuses upon interactions and potential therapeutic targets in the ‘vicious cycle’ between hypoxia and neoangiogenesis following treatment of hepatocellular carcinoma with transarterial loco-regional therapies. Biomarkers correlated with angiogenesis have been studied by many authors as prognostic determinants following transarterial intrahepatic therapy. According to these results future therapies directed toward specific factors related to angiogenesis could play a significant role in preventing local tumor recurrence and remote metastasis.
Collapse
Affiliation(s)
- Mario Petrillo
- Diagnostic & Interventional Radiology Service, San Paolo Hospital, Milan, Italy
| | - Francesca Patella
- Postgraduation School of Radiodiagnostic of Milan, Department of Health Sciences, Milan, Italy
| | - Filippo Pesapane
- Postgraduation School of Radiodiagnostic of Milan, Department of Health Sciences, Milan, Italy
| | - Matteo B Suter
- Department of Medical Oncology, ASST Sette laghi, Varese, Italy
| | - Anna M Ierardi
- Diagnostic & Interventional Radiology Service, San Paolo Hospital, Milan, Italy
| | | | - Chiara Floridi
- Department of Diagnostic & Interventional Radiology Fatebenefratelli Hospital, Milan, Italy
| | - Massimo de Filippo
- Department of Medicine & Surgery Via Gramsci Azienda Ospedaliero Universitaria di Parma, 14 Parma, Italy
| | | |
Collapse
|
92
|
Triulzi T, Forte L, Regondi V, Di Modica M, Ghirelli C, Carcangiu ML, Sfondrini L, Balsari A, Tagliabue E. HER2 signaling regulates the tumor immune microenvironment and trastuzumab efficacy. Oncoimmunology 2018; 8:e1512942. [PMID: 30546951 PMCID: PMC6287794 DOI: 10.1080/2162402x.2018.1512942] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 07/30/2018] [Accepted: 08/13/2018] [Indexed: 12/16/2022] Open
Abstract
Through whole-transcriptome profiling of HER2+ breast carcinomas (BCs), we previously showed that those sensitive to trastuzumab are addicted to this oncoprotein and are enriched in immune pathways, raising the hypothesis that HER2 itself regulates immune cell recruitment. In the present study we investigated the relationship between HER2 activity and the pro-trastuzumab tumor immune milieu. Gene expression profiling and immunohistochemistry analysis of 53 HER2+ BCs showed that trastuzumab-sensitive tumors expressed significantly higher levels of chemokines involved in immune cell recruitment, with higher infiltration of T cells and monocytes, and higher levels of PD-1 ligands than tumors that do not benefit from trastuzumab. In vitro analysis in HER2+ BC cells revealed that CCL2 production was induced by HER2 stimulation with EGF/HRG via the PI3K-NF-kB axis, and down-modulated by HER2 inhibition with trastuzumab. CCL2 expression was higher in HER2+/ER- than HER2+/ER+ BC cell lines, and degradation of ER by fulvestrant induced an enhancement in NF-κB transcriptional activity and consequent CCL2 expression. Trastuzumab efficacy relied on CCL2 levels and monocytes present in the tumor microenvironment in FVB mice bearing HER2+ mammary carcinoma cells. HER2 signals were also found to sustain the expression of PD-1 ligands in tumor cells via the MEK pathway. Overall, our results support the concept that the activated HER2 oncogene regulates recruitment and activation of tumor infiltrating immune cells and trastuzumab activity by inducing CCL2 and PD-1 ligands and that ER activity negatively controls the HER2-driven pro-trastuzumab tumor microenvironment.
Collapse
Affiliation(s)
- Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori of Milan, Milan, Italy
| | - Luca Forte
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori of Milan, Milan, Italy
| | - Viola Regondi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori of Milan, Milan, Italy
| | - Martina Di Modica
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori of Milan, Milan, Italy
| | - Cristina Ghirelli
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori of Milan, Milan, Italy
| | - Maria Luisa Carcangiu
- Anatomic Pathology A Unit, Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori of Milan, Milan, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Andrea Balsari
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori of Milan, Milan, Italy.,Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori of Milan, Milan, Italy
| |
Collapse
|
93
|
Stasikowska-Kanicka O, Wągrowska-Danilewicz M, Danilewicz M. CD8+ and CD163+ infiltrating cells and PD-L1 immunoexpression in oral leukoplakia and oral carcinoma. APMIS 2018; 126:732-738. [PMID: 30160018 DOI: 10.1111/apm.12881] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 07/02/2018] [Indexed: 12/29/2022]
Abstract
Overexpression of inhibitory checkpoint PD1/PD-L1 plays an important role in carcinogenesis and patients prognosis. 70 cases of oral squamous cell carcinoma (OSCC), 23 cases of oral leukoplakia (OLK), and 19 control cases were immunohistochemically stained with anti-PD-L1, -CD8, and -CD163 antibodies. PD-L1 was expressed on dysplastic and subepithelial infiltrating cells of OLK as well as on cancer and tumor-infiltrating cells of OSCC. In OSCC, PD-L1 immunoexpression was significantly increased in comparison to OLK, and control groups. The correlative study showed significant correlations between the immunoexpression of PD-L1 and the number of CD8+, CD163+ cells in both OLK and OSCC groups. We found also significant negative correlation between the number of PD-L1+ infiltrating cells and the number of CD8+ cells in OSCC, and positive correlation between the number of PD-L1+ infiltrating cells and CD163+ cells in OLK and OSCC groups. In conclusion, our study indicate that CD163+ and CD8+ infiltrating cells influence the early and subsequent stages of oral carcinogenesis. We demonstrated also that studied tumors may evade the host immune system by PD-L1 immunoexpression not only on epithelial cells but on infiltrating cells as well.
Collapse
Affiliation(s)
| | | | - Marian Danilewicz
- Department of Pathomorphology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
94
|
Drakes ML, Stiff PJ. Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018; 10:E302. [PMID: 30200478 PMCID: PMC6162424 DOI: 10.3390/cancers10090302] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/20/2022] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| |
Collapse
|
95
|
Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018. [PMID: 30200478 DOI: 10.3390/cancers10090302]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
|
96
|
Drakes ML, Stiff PJ. Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018. [PMID: 30200478 DOI: 10.3390/cancers10090302] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| |
Collapse
|
97
|
Idorn M, Thor Straten P. Chemokine Receptors and Exercise to Tackle the Inadequacy of T Cell Homing to the Tumor Site. Cells 2018; 7:E108. [PMID: 30126117 PMCID: PMC6115859 DOI: 10.3390/cells7080108] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 01/05/2023] Open
Abstract
While cancer immune therapy has revolutionized the treatment of metastatic disease across a wide range of cancer diagnoses, a major limiting factor remains with regard to relying on adequate homing of anti-tumor effector cells to the tumor site both prior to and after therapy. Adoptive cell transfer (ACT) of autologous T cells have improved the outlook of patients with metastatic melanoma. Prior to the approval of checkpoint inhibitors, this strategy was the most promising. However, while response rates of up to 50% have been reported, this strategy is still rather crude. Thus, improvements are needed and within reach. A hallmark of the developing tumor is the evasion of immune destruction. Achieved through the recruitment of immune suppressive cell subsets, upregulation of inhibitory receptors and the development of physical and chemical barriers (such as poor vascularization and hypoxia) leaves the microenvironment a hostile destination for anti-tumor T cells. In this paper, we review the emerging strategies of improving the homing of effector T cells (TILs, CARs, TCR engineered T cells, etc.) through genetic engineering with chemokine receptors matching the chemokines of the tumor microenvironment. While this strategy has proven successful in several preclinical models of cancer and the strategy has moved into the first phase I/II clinical trial in humans, most of these studies show a modest (doubling) increase in tumor infiltration of effector cells, which raises the question of whether road blocks must be tackled for efficient homing. We propose a role for physical exercise in modulating the tumor microenvironment and preparing the platform for infiltration of anti-tumor immune cells. In a time of personalized medicine and genetic engineering, this "old tool" may be a way to augment efficacy and the depth of response to immune therapy.
Collapse
Affiliation(s)
- Manja Idorn
- Center for Cancer Immune Therapy, Herlev Gentofte University Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark.
| | - Per Thor Straten
- Center for Cancer Immune Therapy, Herlev Gentofte University Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark.
- Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| |
Collapse
|
98
|
Deng X, Luo S, Luo X, Hu M, Ma F, Wang Y, Lai X, Zhou L. Polysaccharides from Chinese Herbal Lycium barbarum Induced Systemic and Local Immune Responses in H22 Tumor-Bearing Mice. J Immunol Res 2018; 2018:3431782. [PMID: 29967800 PMCID: PMC6008830 DOI: 10.1155/2018/3431782] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/18/2018] [Accepted: 03/29/2018] [Indexed: 12/12/2022] Open
Abstract
Lycium barbarum polysaccharide (LBP) is isolated from the fruit of Chinese herbal Lycium barbarum. Previous studies had demonstrated that LBP could inhibit tumor growth and enhance the immunity in mice. However, the effect of LBP on systemic and local immune responses in vivo, especially on phenotypic and functional changes of T cells, is still largely unknown. In the present study, we investigated the effects of LBP on systemic and local T cell-dependent antitumor immune responses in H22 tumor-bearing mice. The results showed that LBP could inhibit the solid tumor growth in mice, but showed little effect on the body weight or spleen index. Furthermore, LBP could maintain high levels of T cells in peripheral blood (PB), tumor draining lymph node (TDLN), and tumor tissue, prevent the increase of Tregs while promote infiltration of CD8+ T cells in tumor tissue, inhibit the production of TGF-β1 and IL-10 in serum, decrease the exhaustion phenotype of T cells, and maintain cytotoxicity of lymphocytes. Taken together, our results demonstrated that LBP simultaneously induced systemic and local immune responses in H22 tumor-bearing mice by alleviating immunosuppression and maintaining antitumor immune responses in mice.
Collapse
Affiliation(s)
- Xiangliang Deng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Infinitus Chinese Herbal Immunity Research Centre, Guangzhou 510600, China
- Dongguan Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan 523000, China
| | - Shuang Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xia Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Minghua Hu
- Infinitus Chinese Herbal Immunity Research Centre, Guangzhou 510600, China
| | - Fangli Ma
- Infinitus Chinese Herbal Immunity Research Centre, Guangzhou 510600, China
| | - Yuanyuan Wang
- Infinitus Chinese Herbal Immunity Research Centre, Guangzhou 510600, China
| | - Xiaoping Lai
- Dongguan Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan 523000, China
| | - Lian Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
99
|
Stowman AM, Hickman AW, Mauldin IS, Mahmutovic A, Gru AA, Slingluff CL. Lymphoid aggregates in desmoplastic melanoma have features of tertiary lymphoid structures. Melanoma Res 2018; 28:237-245. [PMID: 29538091 PMCID: PMC5912978 DOI: 10.1097/cmr.0000000000000439] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Desmoplastic melanomas (DM) have unique and challenging clinical presentations and histomorphology. A characteristic feature is the presence of scattered lymphoid aggregates. However, the nature of these aggregates is not defined. We hypothesized that they may be tertiary lymphoid structures (TLS), and may be associated with programmed death ligand 1 (PD-L1) expression. We searched our tissue database for 'pure' DMs and for scars as control tissues, collected clinical information, and reviewed H&E histology. We performed multispectral imaging after staining for CD8, CD20, PNAd, FoxP3, CD83, and Ki67, and assessed PD-L1 expression by immunohistochemistry. Pure DM samples were evaluable in 11 patients. All had desmoplastic stroma and lymphoid aggregates on H&E. The lymphoid aggregates of eight of the 11 (72%) DM samples and only three of the 11 scars contained features of TLS, defined as distinct clusters of B cells and CD8 T cells, CD83 dendritic cells in T-cell zones, and PNAd vasculature resembling high endothelial venules. PD-L1 was expressed by at least 1% of melanoma cells in six and by at least 5% of immune cells in 10 of the 11 DM samples. We found that most lymphoid aggregates in DM are organized, classical TLS. PD-L1 expression was detected in most cases and was highest in two cases of DM with TLS. However, low PD-L1 expression in some cases suggests that some DM cells may be unresponsive to interferon-γ. TLS support antigen presentation and T-cell responses in chronic inflammation and cancer. Their presence in DM likely reflects an adaptive immune response, which may be enhanced with immune therapies.
Collapse
Affiliation(s)
- Anne M. Stowman
- University of Vermont Medical Center, Department of Pathology, Burlington, Vermont
| | | | - Ileana S. Mauldin
- University of Virginia Health System, Department of Surgery, Charlottesville, Virginia
| | - Adela Mahmutovic
- University of Virginia Health System, Department of Surgery, Charlottesville, Virginia
| | - Alejandro A. Gru
- University of Virginia Health System, Department of Pathology, Charlottesville, Virginia
| | - Craig L. Slingluff
- University of Virginia Health System, Department of Surgery, Charlottesville, Virginia
| |
Collapse
|
100
|
Arneth B. Comparison of Burnet's clonal selection theory with tumor cell-clone development. Theranostics 2018; 8:3392-3399. [PMID: 29930737 PMCID: PMC6010991 DOI: 10.7150/thno.24083] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/10/2018] [Indexed: 01/06/2023] Open
Abstract
Increasing evidence has shown that Darwin's theory of evolution provides vital insights into the emergence and etiology of different types of cancer. On a microscopic scale, cancer stem cells meet the conditions for the Darwinian process of natural selection. In particular, cancer stem cells undergo cell reproduction characterized by the emergence of heritable variability that promotes replication and cell survival. Methods: Evidence from previous studies was gathered to compare Burnet's clonal selection theory with the tumor evolution theory. Results: The findings show that the Darwinian theory offers a general framework for understanding fundamental aspects of cancer. As fundamental theoretical frameworks, Burnet's clonal selection theory and the tumor evolution theory can be used to explain cancer cell evolution and identify the beneficial adaptations that contribute to cell survival in tissue landscapes and tissue ecosystems. Conclusions: In conclusion, this study shows that both Burnet's clonal selection theory and the tumor evolution theory postulate that cancer cells in tissue ecosystems evolve through reiterative processes, such as clonal expansion, clonal selection, and genetic diversification. Therefore, both theories provide insights into the complexities and dynamics of cancer, including its development and progression. Finally, we take into account the occurrence of biologic variation in both tumor cells and lymphocytes. It is important to note that the presence of lymphocyte variations appears to be advantageous in the framework of tumor defense but also dangerous within the framework of autoimmune disease development.
Collapse
Affiliation(s)
- Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, University Hospital of the Universities of Giessen and Marburg UKGM, Justus Liebig University, Giessen, Giessen Germany
| |
Collapse
|