1
|
de Pellegars-Malhortie A, Picque Lasorsa L, Mazard T, Granier F, Prévostel C. Why Is Wnt/β-Catenin Not Yet Targeted in Routine Cancer Care? Pharmaceuticals (Basel) 2024; 17:949. [PMID: 39065798 PMCID: PMC11279613 DOI: 10.3390/ph17070949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Despite significant progress in cancer prevention, screening, and treatment, the still limited number of therapeutic options is an obstacle towards increasing the cancer cure rate. In recent years, many efforts were put forth to develop therapeutics that selectively target different components of the oncogenic Wnt/β-catenin signaling pathway. These include small molecule inhibitors, antibodies, and more recently, gene-based approaches. Although some of them showed promising outcomes in clinical trials, the Wnt/β-catenin pathway is still not targeted in routine clinical practice for cancer management. As for most anticancer treatments, a critical limitation to the use of Wnt/β-catenin inhibitors is their therapeutic index, i.e., the difficulty of combining effective anticancer activity with acceptable toxicity. Protecting healthy tissues from the effects of Wnt/β-catenin inhibitors is a major issue due to the vital role of the Wnt/β-catenin signaling pathway in adult tissue homeostasis and regeneration. In this review, we provide an up-to-date summary of clinical trials on Wnt/β-catenin pathway inhibitors, examine their anti-tumor activity and associated adverse events, and explore strategies under development to improve the benefit/risk profile of this therapeutic approach.
Collapse
Affiliation(s)
- Auriane de Pellegars-Malhortie
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
| | - Laurence Picque Lasorsa
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
| | - Thibault Mazard
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
- Medical Oncology Department, ICM, University of Montpellier, CEDEX 5, 34298 Montpellier, France
| | | | - Corinne Prévostel
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
| |
Collapse
|
2
|
Zhang R, Gui Z, Zhao J, Zhao L. BCL9 is a Risk Factor of Neck Lymph Nodes Metastasis and Correlated with Immune Cell Infiltration in Papillary Thyroid Carcinoma. Int J Gen Med 2024; 17:1451-1466. [PMID: 38645401 PMCID: PMC11032164 DOI: 10.2147/ijgm.s455846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/04/2024] [Indexed: 04/23/2024] Open
Abstract
Purpose B-cell lymphoma 9 (BCL9), a key transcription co-activator of the Wnt pathway, contributed to tumor progression and metastasis in various tumors, whereas, the role of BCL9 in papillary thyroid cancer (PTC) has not been investigated. Methods We acquired PTC gene expression data from The Cancer Genome Atlas (TCGA) database. Fifty-nine PTC tissues were applied to validate the clinical significance of BCL9. Cell experiments were applied to investigate the role of BCL9. Bioinformatics analysis was employed to investigate the biological functions of BCL9. Results We found that BCL9 was higher expressed (P < 0.05) and an independent risk factor for lymph node metastasis (OR = 3.770, P = 0.025), as well as associated with poorer progression-free survival (PFS) (P = 0.049) in PTC. BCL9 knockdown inhibited proliferation and invasion of PTC cells. BCL9 was positively associated with the key genes of Wnt/β-catenin and MAPK pathway by co-expression analysis. GO, KEGG and GSEA analysis showed BCL9 might participated in PPAR, cAMP, and focal adhesion pathway. CIBERSORT analysis found BCL9 was negatively associated with CD8+ T cells and NK cell infiltration and positively with PD-L1 expression. Conclusion Therefore, BCL9 was associated with lymph node metastasis and shorter PFS of PTC, due to promotion of PTC cell proliferation and invasion, activation of Wnt/β-catenin and MAPK pathway, inhibition of CD8+ T and NK cell infiltration, and promotion of PD-L1 expression.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Thyroid and Breast Surgery, Wuhan No. 1 Hospital, Wuhan, 430030, People’s Republic of China
| | - Zhengwei Gui
- Department of Thyroid and Breast Surgery, Tongji Hospital Affiliated with Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Jianguo Zhao
- Department of Thyroid and Breast Surgery, Wuhan No. 1 Hospital, Wuhan, 430030, People’s Republic of China
| | - Lu Zhao
- Department of Thyroid and Breast Surgery, Tongji Hospital Affiliated with Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
3
|
Xian W, Asad M, Wu S, Bai Z, Li F, Lu J, Zu G, Brintnell E, Chen H, Mao Y, Zhou G, Liao B, Wu J, Wang E, You L. Distinct immune escape and microenvironment between RG-like and pri-OPC-like glioma revealed by single-cell RNA-seq analysis. Front Med 2024; 18:147-168. [PMID: 37955814 DOI: 10.1007/s11684-023-1017-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 06/24/2023] [Indexed: 11/14/2023]
Abstract
The association of neurogenesis and gliogenesis with glioma remains unclear. By conducting single-cell RNA-seq analyses on 26 gliomas, we reported their classification into primitive oligodendrocyte precursor cell (pri-OPC)-like and radial glia (RG)-like tumors and validated it in a public cohort and TCGA glioma. The RG-like tumors exhibited wild-type isocitrate dehydrogenase and tended to carry EGFR mutations, and the pri-OPC-like ones were prone to carrying TP53 mutations. Tumor subclones only in pri-OPC-like tumors showed substantially down-regulated MHC-I genes, suggesting their distinct immune evasion programs. Furthermore, the two subgroups appeared to extensively modulate glioma-infiltrating lymphocytes in distinct manners. Some specific genes not expressed in normal immune cells were found in glioma-infiltrating lymphocytes. For example, glial/glioma stem cell markers OLIG1/PTPRZ1 and B cell-specific receptors IGLC2/IGKC were expressed in pri-OPC-like and RG-like glioma-infiltrating lymphocytes, respectively. Their expression was positively correlated with those of immune checkpoint genes (e.g., LGALS33) and poor survivals as validated by the increased expression of LGALS3 upon IGKC overexpression in Jurkat cells. This finding indicated a potential inhibitory role in tumor-infiltrating lymphocytes and could provide a new way of cancer immune evasion.
Collapse
Affiliation(s)
- Weiwei Xian
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Mohammad Asad
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Shuai Wu
- Glioma Surgery Division, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zhixin Bai
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Fengjiao Li
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Junfeng Lu
- Glioma Surgery Division, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Gaoyu Zu
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Erin Brintnell
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Hong Chen
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Guomin Zhou
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention, Fudan University, Shanghai, 200040, China
| | - Bo Liao
- School of Mathematics and Statistics, Hainan Normal University, Haikou, 570100, China
| | - Jinsong Wu
- Glioma Surgery Division, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Edwin Wang
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada.
| | - Linya You
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
4
|
Onnis V. Special Issue "Novel Anti-Proliferative Agents". Pharmaceuticals (Basel) 2023; 16:1437. [PMID: 37895908 PMCID: PMC10610072 DOI: 10.3390/ph16101437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Cancer is a disease that can affect any organ and spread to other nearby or distant organs [...].
Collapse
Affiliation(s)
- Valentina Onnis
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, CA, Italy
| |
Collapse
|
5
|
Reed ER, Jankowski SA, Spinella AJ, Noonan V, Haddad R, Nomoto K, Matsui J, Bais MV, Varelas X, Kukuruzinska MA, Monti S. β-catenin/CBP activation of mTORC1 signaling promotes partial epithelial-mesenchymal states in head and neck cancer. Transl Res 2023; 260:46-60. [PMID: 37353110 PMCID: PMC10527608 DOI: 10.1016/j.trsl.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 04/27/2023] [Accepted: 05/20/2023] [Indexed: 06/25/2023]
Abstract
Head and neck cancers, which include oral squamous cell carcinoma (OSCC) as a major subsite, exhibit cellular plasticity that includes features of an epithelial-mesenchymal transition (EMT), referred to as partial-EMT (p-EMT). To identify molecular mechanisms contributing to OSCC plasticity, we performed a multiphase analysis of single cell RNA sequencing (scRNAseq) data from human OSCC. This included a multiresolution characterization of cancer cell subgroups to identify pathways and cell states that are heterogeneously represented, followed by casual inference analysis to elucidate activating and inhibitory relationships between these pathways and cell states. This approach revealed signaling networks associated with hierarchical cell state transitions, which notably included an association between β-catenin-driven CREB-binding protein (CBP) activity and mTORC1 signaling. This network was associated with subpopulations of cancer cells that were enriched for markers of the p-EMT state and poor patient survival. Functional analyses revealed that β-catenin/CBP induced mTORC1 activity in part through the transcriptional regulation of a raptor-interacting protein, chaperonin containing TCP1 subunit 5 (CCT5). Inhibition of β-catenin-CBP activity through the use of the orally active small molecule, E7386, reduced the expression of CCT5 and mTORC1 activity in vitro, and inhibited p-EMT-associated markers and tumor development in a murine model of OSCC. Our study highlights the use of multiresolution network analyses of scRNAseq data to identify targetable signals for therapeutic benefit, thus defining an underappreciated association between β-catenin/CBP and mTORC1 signaling in head and neck cancer plasticity.
Collapse
Affiliation(s)
- Eric R Reed
- Data Intensive Studies Center, Tufts University, Medford, Massachusetts; Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts; Bioinformatics Program, Boston University, Boston, Massachusetts.
| | - Stacy A Jankowski
- Department of Translational Dental Medicine, Boston University School of Dental Medicine, Boston, Massachusetts; Molecular and Translational Medicine Program, Boston University School of Medicine, Boston, Massachusetts
| | - Anthony J Spinella
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Vikki Noonan
- Division of Oral Pathology, Boston University School of Dental Medicine, Boston, Massachusetts
| | - Robert Haddad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Junji Matsui
- Eisai Inc, 200 Metro Blvd, Nutley, NJ, 07110, USA
| | - Manish V Bais
- Department of Translational Dental Medicine, Boston University School of Dental Medicine, Boston, Massachusetts
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts.
| | - Maria A Kukuruzinska
- Department of Translational Dental Medicine, Boston University School of Dental Medicine, Boston, Massachusetts.
| | - Stefano Monti
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts; Bioinformatics Program, Boston University, Boston, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts.
| |
Collapse
|
6
|
Yu X, Li W, Feng Y, Gao Z, Wu Q, Xia Y. The prognostic value of hedgehog signaling in bladder cancer by integrated bioinformatics. Sci Rep 2023; 13:6241. [PMID: 37069207 PMCID: PMC10110581 DOI: 10.1038/s41598-023-33140-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 04/07/2023] [Indexed: 04/19/2023] Open
Abstract
Bladder cancer is the second most prevalent urological malignancy. It's a big contributor to cancer-related deaths throughout the globe. Researchers discovered that the hedgehog signaling (HhS) pathway contributed to the onset and spread of many different kinds of cancer. Nevertheless, the present understanding of the function of HhS in the bladder cancer molecular landscape is incomplete. Raw data were gotten from the IMvigor210, the Gene Expression Omnibus, and The Cancer Genome Atlas databases. Bioinformatics was used to examine the HhS score of each sample, and the enrichment of differentially expressed genes (DEGs), differentiation characteristics, immunological infiltration, and metabolic activity. The HhS prognostic signature was developed with significant assistance from the least absolute shrinkage and selection operator regression and Cox regression. An HhS-related nomogram was developed to assist in the prediction of patients' survival probability. We found that HhS was linked to poor prognosis in bladder cancer, and its activation was linked to the Basal subtype of bladder cancer. Bladder cancer with high HhS activity has higher glycolysis, nucleotide metabolism, amino acid metabolism, and other cancer-promoting metabolic activities. Furthermore, HhS mediates an immunosuppressive microenvironment in bladder cancer on the basis that HhS negatively correlates with the CD8 + T cells and correlates positively with immune checkpoints and T cell exhaustion scores. Finally, an HhS-related signature was developed for predicting the prognosis of patients with bladder cancer. Targeting HhS may be a potential therapy choice for bladder cancer.
Collapse
Affiliation(s)
- Xin Yu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Wenge Li
- Department of Oncology, Shanghai Artemed Hospital, Shanghai, People's Republic of China
| | - Yanjun Feng
- Department of Oncology, Shanghai Artemed Hospital, Shanghai, People's Republic of China
| | - Zhijie Gao
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Qi Wu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, People's Republic of China.
| | - Yue Xia
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
7
|
Ziogas DC, Theocharopoulos C, Koutouratsas T, Haanen J, Gogas H. Mechanisms of resistance to immune checkpoint inhibitors in melanoma: What we have to overcome? Cancer Treat Rev 2023; 113:102499. [PMID: 36542945 DOI: 10.1016/j.ctrv.2022.102499] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Marching into the second decade after the approval of ipilimumab, it is clear that immune checkpoint inhibitors (ICIs) have dramatically improved the prognosis of melanoma. Although the current edge is already high, with a 4-year OS% of 77.9% for adjuvant nivolumab and a 6.5-year OS% of 49% for nivolumab/ipilimumab combination in the metastatic setting, a high proportion of patients with advanced melanoma have no benefit from immunotherapy, or experience an early disease relapse/progression in the first few months of treatment, surviving much less. Reasonably, the primary and acquired resistance to ICIs has entered into the focus of clinical research with positive (e.g., nivolumab and relatlimab combination) and negative feedbacks (e.g., nivolumab with pegylated-IL2, pembrolizumab with T-VEC, nivolumab with epacadostat, and combinatorial triplets of BRAF/MEK inhibitors with immunotherapy). Many intrinsic (intracellular or intra-tumoral) but also extrinsic (systematic) events are considered to be involved in the development of this resistance to ICIs: i) melanoma cell immunogenicity (e.g., tumor mutational burden, antigen-processing machinery and immunogenic cell death, neoantigen affinity and heterogeneity, genomic instability, melanoma dedifferentiation and phenotypic plasticity), ii) immune cell trafficking, T-cell priming, and cell death evasion, iii) melanoma neovascularization, cellular TME components(e.g., Tregs, CAFs) and extracellular matrix modulation, iv) metabolic antagonism in the TME(highly glycolytic status, upregulated CD39/CD73/adenosine pathway, iDO-dependent tryptophan catabolism), v) T-cell exhaustion and negative immune checkpoints, and vi) gut microbiota. In the present overview, we discuss how these parameters compromise the efficacy of ICIs, with an emphasis on the lessons learned by the latest melanoma studies; and in parallel, we describe the main ongoing approaches to overcome the resistance to immunotherapy. Summarizing this information will improve the understanding of how these complicated dynamics contribute to immune escape and will help to develop more effective strategies on how anti-tumor immunity can surpass existing barriers of ICI-refractory melanoma.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Charalampos Theocharopoulos
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Tilemachos Koutouratsas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - John Haanen
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| |
Collapse
|
8
|
Di Bartolomeo L, Vaccaro F, Irrera N, Borgia F, Li Pomi F, Squadrito F, Vaccaro M. Wnt Signaling Pathways: From Inflammation to Non-Melanoma Skin Cancers. Int J Mol Sci 2023; 24:ijms24021575. [PMID: 36675086 PMCID: PMC9867176 DOI: 10.3390/ijms24021575] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Canonical and non-canonical Wnt signaling pathways are involved in cell differentiation and homeostasis, but also in tumorigenesis. In fact, an exaggerated activation of Wnt signaling may promote tumor growth and invasion. We summarize the most intriguing evidence about the role of Wnt signaling in cutaneous carcinogenesis, in particular in the pathogenesis of non-melanoma skin cancer (NMSC). Wnt signaling is involved in several ways in the development of skin tumors: it may modulate the inflammatory tumor microenvironment, synergize with Sonic Hedgehog pathway in the onset of basal cell carcinoma, and contribute to the progression from precancerous to malignant lesions and promote the epithelial-mesenchymal transition in squamous cell carcinoma. Targeting Wnt pathways may represent an additional efficient approach in the management of patients with NMSC.
Collapse
Affiliation(s)
- Luca Di Bartolomeo
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, 98125 Messina, Italy
| | - Federico Vaccaro
- Department of Dermatology, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, Section of Pharmacology, University of Messina, 98125 Messina, Italy
| | - Francesco Borgia
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, 98125 Messina, Italy
| | - Federica Li Pomi
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, 98125 Messina, Italy
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, Section of Pharmacology, University of Messina, 98125 Messina, Italy
| | - Mario Vaccaro
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, 98125 Messina, Italy
- Correspondence:
| |
Collapse
|
9
|
Du W, Menjivar RE, Donahue KL, Kadiyala P, Velez-Delgado A, Brown KL, Watkoske HR, He X, Carpenter ES, Angeles CV, Zhang Y, Pasca di Magliano M. WNT signaling in the tumor microenvironment promotes immunosuppression in murine pancreatic cancer. J Exp Med 2023; 220:e20220503. [PMID: 36239683 PMCID: PMC9577101 DOI: 10.1084/jem.20220503] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/06/2022] [Accepted: 09/07/2022] [Indexed: 01/16/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is associated with activation of WNT signaling. Whether this signaling pathway regulates the tumor microenvironment has remained unexplored. Through single-cell RNA sequencing of human pancreatic cancer, we discovered that tumor-infiltrating CD4+ T cells express TCF7, encoding for the transcription factor TCF1. We conditionally inactivated Tcf7 in CD4 expressing T cells in a mouse model of pancreatic cancer and observed changes in the tumor immune microenvironment, including more CD8+ T cells and fewer regulatory T cells, but also compensatory upregulation of PD-L1. We then used a clinically available inhibitor of Porcupine, a key component of WNT signaling, and observed similar reprogramming of the immune response. WNT signaling inhibition has limited therapeutic window due to toxicity, and PD-L1 blockade has been ineffective in PDA. Here, we show that combination targeting reduces pancreatic cancer growth in an experimental model and might benefit the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Wenting Du
- Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Rosa E. Menjivar
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI
| | | | - Padma Kadiyala
- Immunology Program, University of Michigan, Ann Arbor, MI
| | - Ashley Velez-Delgado
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI
| | | | | | - Xi He
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI
| | - Eileen S. Carpenter
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI
| | - Christina V. Angeles
- Department of Surgery, University of Michigan, Ann Arbor, MI
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, MI
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, MI
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
10
|
Jiang M, Qin B, Li X, Liu Y, Guan G, You J. New advances in pharmaceutical strategies for sensitizing anti-PD-1 immunotherapy and clinical research. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1837. [PMID: 35929522 DOI: 10.1002/wnan.1837] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/30/2022] [Accepted: 07/14/2022] [Indexed: 01/31/2023]
Abstract
Attempts have been made continuously to use nano-drug delivery system (NDDS) to improve the effect of antitumor therapy. In recent years, especially in the application of immunotherapy represented by antiprogrammed death receptor 1 (anti-PD-1), it has been vigorously developed. Nanodelivery systems are significantly superior in a number of aspects including increasing the solubility of insoluble drugs, enhancing their targeting ability, prolonging their half-life, and reducing side effects. It can not only directly improve the efficacy of anti-PD-1 immunotherapy, but also indirectly enhance the antineoplastic efficacy of immunotherapy by boosting the effectiveness of therapeutic modalities such as chemotherapy, radiotherapy, photothermal, and photodynamic therapy (PTT/PDT). Here, we summarize the studies published in recent years on the use of nanotechnology in pharmaceutics to improve the efficacy of anti-PD-1 antibodies, analyze their characteristics and shortcomings, and combine with the current clinical research on anti-PD-1 antibodies to provide a reference for the design of future nanocarriers, so as to further expand the clinical application prospects of NDDSs. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Mengshi Jiang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bing Qin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiang Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yu Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Guannan Guan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Differentially Expressed Genes and Signaling Pathways Potentially Involved in Primary Resistance to Chemo-Immunotherapy in Advanced-Stage Gastric Cancer Patients. Int J Mol Sci 2022; 24:ijms24010001. [PMID: 36613445 PMCID: PMC9820415 DOI: 10.3390/ijms24010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/24/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Recently, the combination of chemotherapy plus nivolumab (chemo-immunotherapy) has become the standard of care for advanced-stage gastric cancer (GC) patients. However, despite its efficacy, up to 40% of patients do not respond to these treatments. Our study sought to identify variations in gene expression associated with primary resistance to chemo-immunotherapy. Diagnostic endoscopic biopsies were retrospectively obtained from advanced GC patients previously categorized as responders (R) or non-responders (NR). Thirty-four tumor biopsies (R: n = 16, NR: n = 18) were analyzed by 3′ massive analysis of cDNA ends (3′MACE). We found >30 differentially expressed genes between R and NRs. Subsequent pathway enrichment analyses demonstrated that angiogenesis and the Wnt-β-catenin signaling pathway were enriched in NRs. Concomitantly, we performed next generation sequencing (NGS) analyses in a subset of four NR patients that confirmed alterations in genes that belonged to the Wnt/β-catenin and the phosphoinositide 3-kinase (PI3K) pathways. We speculate that angiogenesis, the Wnt, and the PI3K pathways might offer actionable targets. We also discuss therapeutic alternatives for chemo-immunotherapy-resistant advanced-stage GC patients.
Collapse
|
12
|
Hua Y, Vella G, Rambow F, Allen E, Antoranz Martinez A, Duhamel M, Takeda A, Jalkanen S, Junius S, Smeets A, Nittner D, Dimmeler S, Hehlgans T, Liston A, Bosisio FM, Floris G, Laoui D, Hollmén M, Lambrechts D, Merchiers P, Marine JC, Schlenner S, Bergers G. Cancer immunotherapies transition endothelial cells into HEVs that generate TCF1 + T lymphocyte niches through a feed-forward loop. Cancer Cell 2022; 40:1600-1618.e10. [PMID: 36423635 PMCID: PMC9899876 DOI: 10.1016/j.ccell.2022.11.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 07/20/2022] [Accepted: 11/04/2022] [Indexed: 11/24/2022]
Abstract
The lack of T cell infiltrates is a major obstacle to effective immunotherapy in cancer. Conversely, the formation of tumor-associated tertiary-lymphoid-like structures (TA-TLLSs), which are the local site of humoral and cellular immune responses against cancers, is associated with good prognosis, and they have recently been detected in immune checkpoint blockade (ICB)-responding patients. However, how these lymphoid aggregates develop remains poorly understood. By employing single-cell transcriptomics, endothelial fate mapping, and functional multiplex immune profiling, we demonstrate that antiangiogenic immune-modulating therapies evoke transdifferentiation of postcapillary venules into inflamed high-endothelial venules (HEVs) via lymphotoxin/lymphotoxin beta receptor (LT/LTβR) signaling. In turn, tumor HEVs boost intratumoral lymphocyte influx and foster permissive lymphocyte niches for PD1- and PD1+TCF1+ CD8 T cell progenitors that differentiate into GrzB+PD1+ CD8 T effector cells. Tumor-HEVs require continuous CD8 and NK cell-derived signals revealing that tumor HEV maintenance is actively sculpted by the adaptive immune system through a feed-forward loop.
Collapse
Affiliation(s)
- Yichao Hua
- VIB Center for Cancer Biology, Leuven, Belgium; Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB Center for Cancer Biology, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Gerlanda Vella
- VIB Center for Cancer Biology, Leuven, Belgium; Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB Center for Cancer Biology, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Florian Rambow
- VIB Center for Cancer Biology, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium; Laboratory of Molecular Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium; Department of Applied Computational Cancer Research, Institute for AI in Medicine, University Hospital Essen, Essen, Germany; University of Duisburg-Essen, Essen, Germany
| | | | - Asier Antoranz Martinez
- Department of Imaging & Pathology, Laboratory of Translational Cell & Tissue Research and Department of Pathology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Marie Duhamel
- VIB Center for Cancer Biology, Leuven, Belgium; Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB Center for Cancer Biology, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Akira Takeda
- MediCity, Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Sirpa Jalkanen
- MediCity, Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Steffie Junius
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium; VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Ann Smeets
- Department of Surgical Oncology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - David Nittner
- VIB Center for Cancer Biology, Leuven, Belgium; Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Goethe-University, Frankfurt am Main, Germany
| | - Thomas Hehlgans
- Department of Immunology, University of Regensburg, Regensburg, Germany
| | - Adrian Liston
- VIB Center for Brain and Disease Research, Leuven, Belgium; Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge, UK
| | - Francesca Maria Bosisio
- Department of Imaging & Pathology, Laboratory of Translational Cell & Tissue Research and Department of Pathology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Giuseppe Floris
- Department of Imaging & Pathology, Laboratory of Translational Cell & Tissue Research and Department of Pathology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Damya Laoui
- Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium; Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Maija Hollmén
- MediCity, Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Diether Lambrechts
- VIB Center for Cancer Biology, Leuven, Belgium; Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | | | - Jean-Christophe Marine
- VIB Center for Cancer Biology, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium; Laboratory of Molecular Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
| | - Susan Schlenner
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| | - Gabriele Bergers
- VIB Center for Cancer Biology, Leuven, Belgium; Laboratory of Tumor Microenvironment and Therapeutic Resistance, VIB Center for Cancer Biology, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
13
|
Liu M, Yan W, Chen D, Luo J, Dai L, Chen H, Chen KN. IGFBP1 hiWNT3A lo Subtype in Esophageal Cancer Predicts Response and Prolonged Survival with PD-(L)1 Inhibitor. BIOLOGY 2022; 11:biology11111575. [PMID: 36358276 PMCID: PMC9687176 DOI: 10.3390/biology11111575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 01/25/2023]
Abstract
PD-(L)1 inhibitor could improve the survival of locally advanced esophageal cancer (ESCA) patients, but we cannot tailor the treatment to common biomarkers. WNT signaling activation was associated with primary resistance to immunotherapy. In this study, we used our two clinical cohorts (BJCH n = 95, BJIM n = 21) and three public cohorts to evaluate and verify a new immunotherapeutic biomarker based on WNT signaling in ESCA patients. Our findings showed that WNT signaling-related genes stratified TCGA patients into Cluster 1, 2, and 3, among which, Cluster 3 had the worst prognosis. The most up- and down-regulated genes in Cluster 3 were IGFBP1 and WNT3A. Further analysis validated that IGFBP1hiWNT3Alo ESCA patients had significantly poor RFS and OS in the TCGA and BJCH cohorts. Interestingly, IGFBP1hiWNT3Alo patients had a good response and prognosis with immunotherapy in three independent cohorts, exhibiting better predictive value than PD-L1 expression (signature AUC = 0.750; PD-L1 AUC = 0.571). Moreover, IGFBP1hiWNT3Alo patients may benefit more from immunotherapy than standard treatment (p = 0.026). Immune cell infiltration analysis revealed a significant increase in DC infiltration in IGFBP1hiWNT3Alo patients post-immunotherapy (p = 0.022), which may enhance immune response. The IGFBP1hiWNT3Alo signature could predict patients who benefited from PD-(L)1 inhibitor treatment and may serve as a biomarker in ESCA.
Collapse
Affiliation(s)
- Meichen Liu
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing 100142, China
| | - Wanpu Yan
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing 100142, China
| | - Dongbo Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Disease, No.11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Jiancheng Luo
- Aiyi Technology Co., Ltd., Room 1004, Building 3, Greenland Qihang, Biomedical Industry Base, Daxing District, Beijing 102629, China
| | - Liang Dai
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing 100142, China
| | - Hongsong Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Disease, No.11 Xizhimen South Street, Xicheng District, Beijing 100044, China
- Correspondence: (H.C.); (K.-N.C.)
| | - Ke-Neng Chen
- Department of Thoracic Surgery I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing 100142, China
- Correspondence: (H.C.); (K.-N.C.)
| |
Collapse
|
14
|
Potential role of Marine Bioactive Compounds targeting signaling pathways in cancer: A review. Eur J Pharmacol 2022; 936:175330. [DOI: 10.1016/j.ejphar.2022.175330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/23/2022]
|
15
|
Shan J, Han D, Shen C, Lei Q, Zhang Y. Mechanism and strategies of immunotherapy resistance in colorectal cancer. Front Immunol 2022; 13:1016646. [PMID: 36238278 PMCID: PMC9550896 DOI: 10.3389/fimmu.2022.1016646] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/05/2022] [Indexed: 11/15/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world. Although there are standard treatment options for CRC, most patients respond poorly to these treatments. Immunotherapies have gradually emerged due to the increasing awareness and understanding of tumor immunity, exhibiting good therapeutic efficacy in various cancers. Immunotherapies include cytokines, immune checkpoint inhibitors (ICIs), and adoptive cell therapies. In particular, ICIs, which are antibodies against cytotoxic T lymphocyte-associated protein 4 (CTLA-4), programmed cell death 1 (PD-1), or its ligand PD-L1, have been successfully applied clinically for solid tumors, relieving the inhibitory effect of the tumor microenvironment on T cells. However, only a minority of patients with cancer achieve a durable clinical response during immunotherapy. Several factors restrict the efficacy of immunotherapy, leading to the development of drug resistance. In this review, we aimed to discuss the current status of immunotherapy for CRC and elaborate on the mechanisms that mediate resistance to immunotherapy and other potential therapeutic strategies.
Collapse
Affiliation(s)
- Jiqi Shan
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dong Han
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunyi Shen
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingyang Lei
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
- *Correspondence: Yi Zhang,
| |
Collapse
|
16
|
Schwarze JK, Tijtgat J, Awada G, Cras L, Vasaturo A, Bagnall C, Forsyth R, Dufait I, Tuyaerts S, Van Riet I, Neyns B. Intratumoral administration of CD1c (BDCA-1) + and CD141 (BDCA-3) + myeloid dendritic cells in combination with talimogene laherparepvec in immune checkpoint blockade refractory advanced melanoma patients: a phase I clinical trial. J Immunother Cancer 2022; 10:jitc-2022-005141. [PMID: 36113895 PMCID: PMC9486335 DOI: 10.1136/jitc-2022-005141] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Intratumoral (IT) myeloid dendritic cells (myDCs) play a pivotal role in initiating antitumor immune responses and relicensing of anti-tumor cytotoxic T lymphocytes within the tumor microenvironment. Talimogene laherparepvec (T-VEC) induces immunogenic cell death, thereby providing maturation signals and enhancing the release of tumor antigens that can be captured and processed by CD1c (BDCA-1)+ / CD141 (BDCA-3)+ myDCs, in order to reinvigorate the cancer-immunity cycle. METHODS In this phase I trial, patients with advanced melanoma who failed standard therapy were eligible for IT injections of ≥1 non-visceral metastases with T-VEC on day 1 followed by IT injection of CD1c (BDCA-1)+ myDCs +/- CD141 (BDCA-3)+ myDCs on day 2. T-VEC injections were repeated on day 21 and every 14 days thereafter. The number of IT administered CD1c (BDCA-1)+ myDCs was escalated from 0.5×106, to 1×106, to a maximum of 10×106 cells in three sequential cohorts. In cohort 4, all isolated CD1c (BDCA-1)+ / CD141 (BDCA-3)+ myDCs were used for IT injection. Primary objectives were safety and feasibility. Repetitive biopsies of treated lesions were performed. RESULTS In total, 13 patients were enrolled (cohort 1 n=2; cohort 2 n=2; cohort 3 n=3; cohort 4 n=6). Patients received a median of 6 (range 3-8) T-VEC injections. The treatment was safe with most frequent adverse events being fatigue (n=11 (85%)), fever (n=8 (62%)), and chills/influenza-like symptoms (n=6 (46%)). Nine (69%) and four patients (31%), respectively, experienced pain or redness at the injection-site. Clinical responses were documented in injected and non-injected lesions. Two patients (cohort 3) who previously progressed on anti-PD-1 therapy (and one patient also on anti-CTLA-4 therapy) developed a durable, pathologically confirmed complete response that is ongoing at 33 and 35 months following initiation of study treatment. One additional patient treated (cohort 4) had an unconfirmed partial response as best response; two additional patients had a mixed response (with durable complete responses of some injected and non-injected lesions). On-treatment biopsies revealed a strong infiltration by inflammatory cells in regressing lesions. CONCLUSIONS IT coinjection of autologous CD1c (BDCA-1)+ +/- CD141 (BDCA-3)+ myDCs with T-VEC is feasible, tolerable and resulted in encouraging early signs of antitumor activity in immune checkpoint inhibitor-refractory melanoma patients. TRIAL REGISTRATION NUMBER NCT03747744.
Collapse
Affiliation(s)
- Julia Katharina Schwarze
- Department of Medical Oncology/Laboratory of Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Jens Tijtgat
- Department of Medical Oncology/Laboratory of Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Gil Awada
- Department of Medical Oncology/Laboratory of Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Louise Cras
- Department of Anatomopathology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | | | | | - Ramses Forsyth
- Department of Anatomopathology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Inès Dufait
- Department of Radiotherapy, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Sandra Tuyaerts
- Department of Medical Oncology/Laboratory of Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Ivan Van Riet
- Stem Cell Laboratory, Department of Hematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Bart Neyns
- Department of Medical Oncology/Laboratory of Medical and Molecular Oncology (LMMO), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| |
Collapse
|
17
|
Lee CK, Chan SL, Chon HJ. Could We Predict the Response of Immune Checkpoint Inhibitor Treatment in Hepatocellular Carcinoma? Cancers (Basel) 2022; 14:3213. [PMID: 35804984 PMCID: PMC9264773 DOI: 10.3390/cancers14133213] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
The use of anti-programmed cell-death protein (ligand)-1 (PD-[L]1) is an important strategy for treating hepatocellular carcinoma (HCC). However, the treatment only benefits 10-20% of patients when used as a monotherapy. Therefore, the selection of patients for anti-PD-1/PD-L1 treatment is crucial for both patients and clinicians. This review aimed to explore the existing literature on tissue or circulating markers for the identification of responders or non-responders to anti-PD-1/PD-L1 in HCC. For the clinically available markers, both etiological factors (viral versus non-viral) and disease extent (intra-hepatic vs. extrahepatic) impact the responses to anti-PD-1/PD-L1, warranting further studies. Preliminary data suggested that inflammatory indices (e.g., neutrophil-lymphocyte ratio) may be associated with clinical outcomes of HCC during the anti-PD-1/PD-L1 treatment. Finally, although PD-L1 expression in tumor tissues is a predictive marker for multiple cancer types, its clinical application is less clear in HCC due to the lack of a clear-cut association with responders to anti-PD-1/PD-L1 treatment. Although all translational markers are not routinely measured in HCC, recent data suggest their potential roles in selecting patients for anti-PD-1/PD-L1 treatment. Such markers, including the immune classification of HCC, selected signaling pathways, tumor-infiltrating lymphocytes, and auto-antibodies, were discussed in this review.
Collapse
Affiliation(s)
- Choong-kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Stephen L. Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Hong Jae Chon
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea
| |
Collapse
|
18
|
Lou X, Gao H, Xu X, Ye Z, Zhang W, Wang F, Chen J, Zhang Y, Chen X, Qin Y, Yu X, Ji S. The Interplay of Four Main Pathways Recomposes Immune Landscape in Primary and Metastatic Gastroenteropancreatic Neuroendocrine Tumors. Front Oncol 2022; 12:808448. [PMID: 35664743 PMCID: PMC9158120 DOI: 10.3389/fonc.2022.808448] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/08/2022] [Indexed: 11/29/2022] Open
Abstract
Background The four major pathways in gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) including chromatin remodeling, DNA damage repair, activation of mTOR signaling, and telomere maintenance were mediated by some critical molecules and constituted critical processes of regulation in cancer-causing processes. However, the interplay and potential role of these pathway-related molecules in the tumor microenvironment of the primary and metastatic site remained unknown. Methods We systematically evaluated the mRNA expression of 34 molecules associated with the four pathways in 227 GEP−NEN samples from 5 datasets. We assigned the samples into two expression patterns of pathway-related molecules by an unsupervised clustering method. Subsequently, we explored the specific cell-related molecules, especially immune and stromal cells using the WGCNA method, based on differentially expressed genes (DEGs) responsible for the different patterns of pathway-related molecules, which provided a new method to qualify the pathway-related subtypes of individual tumors, then the PC_Score and PI_Score scoring systems were also constructed using obtained specific cell-related molecules. Furthermore, we performed the association of pathway-related subtypes with characteristics of immune landscape in primary and metastatic GEP-NENs. Results We demonstrated that the specific pathway-related molecules (SMARCA4, MLH1, TSC1, ATRX, and ATR) were associated with cytolytic activity. Then we identified the two distinct patterns of pathway-related molecules, which were characteristic with a significantly distinct immune landscape. Using WGCNA, we also identified the fibroblast-related molecules, including ASPN, COL10A1, COL3A1, EDNRA, MYL9, PRELP, RAB31, SPARC, and THBS2, and immune-related molecules including CASP1, CCL5, CTSS, CYBRD1, PMP22, and TFEC. Based on these specific markers, we identified four distinct pathway-related subtypes, characterized by immune and fibrotic enriched (I/FE), immune enriched (IE), fibrotic enriched (FE), and immune and fibrotic desert (I/FD), of which I/FE was characteristic with the highest PC_Score and PI_Score whereas I/FD presents the opposite trend. I/FE was positively correlated with immune landscape of T-cell activation and immunosuppression. Furthermore, the I/FE marked GEP-NENs with increased immune activation scores (T-cell costimulation, MHC I presentation, and APC costimulation). Importantly, the four distinct pathway-related subtypes were not conserved in different tumor sites, because I/FE was lacking in the liver metastatic site even though IE, FE, and I/FD also could be observed in the metastatic site. Conclusions This study was the first to perform a comprehensive analysis of the four major pathways in GEP-NENs. We demonstrated the potential function of these pathway-related molecules in immune landscapes. Our findings indicated that the primary and metastatic GEP-NENs had distinct antitumor phenotypes. This work highlighted the interplay and potential clinical utility of these pathway-related molecules in GEP-NENs.
Collapse
Affiliation(s)
- Xin Lou
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Heli Gao
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xiaowu Xu
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Zeng Ye
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wuhu Zhang
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Fei Wang
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jie Chen
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yue Zhang
- Department of Hepatopancreatobiliary Surgery, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xuemin Chen
- Department of Hepatopancreatobiliary Surgery, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yi Qin
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Correia C, Weiskittel TM, Ung CY, Villasboas Bisneto JC, Billadeau DD, Kaufmann SH, Li H. Uncovering Pharmacological Opportunities for Cancer Stem Cells-A Systems Biology View. Front Cell Dev Biol 2022; 10:752326. [PMID: 35359437 PMCID: PMC8962639 DOI: 10.3389/fcell.2022.752326] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/10/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer stem cells (CSCs) represent a small fraction of the total cancer cell population, yet they are thought to drive disease propagation, therapy resistance and relapse. Like healthy stem cells, CSCs possess the ability to self-renew and differentiate. These stemness phenotypes of CSCs rely on multiple molecular cues, including signaling pathways (for example, WNT, Notch and Hedgehog), cell surface molecules that interact with cellular niche components, and microenvironmental interactions with immune cells. Despite the importance of understanding CSC biology, our knowledge of how neighboring immune and tumor cell populations collectively shape CSC stemness is incomplete. Here, we provide a systems biology perspective on the crucial roles of cellular population identification and dissection of cell regulatory states. By reviewing state-of-the-art single-cell technologies, we show how innovative systems-based analysis enables a deeper understanding of the stemness of the tumor niche and the influence of intratumoral cancer cell and immune cell compositions. We also summarize strategies for refining CSC systems biology, and the potential role of this approach in the development of improved anticancer treatments. Because CSCs are amenable to cellular transitions, we envision how systems pharmacology can become a major engine for discovery of novel targets and drug candidates that can modulate state transitions for tumor cell reprogramming. Our aim is to provide deeper insights into cancer stemness from a systems perspective. We believe this approach has great potential to guide the development of more effective personalized cancer therapies that can prevent CSC-mediated relapse.
Collapse
Affiliation(s)
- Cristina Correia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Taylor M Weiskittel
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Choong Yong Ung
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | | | - Daniel D Billadeau
- Department of Immunology, Mayo Clinic, Rochester, MN, United States,Division of Oncology Research, Mayo Clinic, Rochester, MN, United States
| | - Scott H Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States,Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, United States,Division of Oncology Research, Mayo Clinic, Rochester, MN, United States
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States,*Correspondence: Hu Li,
| |
Collapse
|
20
|
Szeto C, Kurzrock R, Kato S, Goloubev A, Veerapaneni S, Preble A, Reddy S, Adashek J. Association of differential expression of immunoregulatory molecules and presence of targetable mutations may inform rational design of clinical trials. ESMO Open 2022; 7:100396. [PMID: 35158206 PMCID: PMC8850727 DOI: 10.1016/j.esmoop.2022.100396] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/07/2021] [Accepted: 01/03/2022] [Indexed: 12/31/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) and genomic biomarker-driven targeted therapies have revolutionized the modern oncologic treatment arsenal. The next step has been to combine targeted agents and ICIs. In doing so, some combination regimens may be more logical than others. Patients and methods Whole-exome and whole-transcriptome sequencing were performed on 2739 unselected later-stage clinical cases from 24 solid tumor subtypes in the NantHealth database, and data were also curated from 5746 similarly sequenced patients across 28 solid tumor subtypes in The Cancer Genome Atlas (TCGA). Significant differential expression of 10 immunoregulatory molecules [IRMs (genes)] was analyzed for association with mutant versus wild-type genes. Results Twenty-three significant associations between currently actionable variants and RNA-expressed checkpoint genes were identified in the TCGA cases; 10 were validated in the external cohort of 2739 clinical cases from NantHealth (P values were adjusted using Benjamini–Hochberg multiple hypothesis correction to reduce false-discovery rate). Within the same 5746 TCGA profiles, 2740 TCGA patients were identified as having one or more potentially oncogenic single-nucleotide variant (SNV) mutation within an established 50-gene hotspot panel. Of the 50 genes, SNVs within 15 were found to be significantly associated with differential expression of at least one IRM after adjusting for tissue enrichment; six were confirmed significant associations in an independent set of 2739 clinical cases from NantHealth. Conclusions Logically combining ICIs with targeted therapies may offer unique treatment strategies for patients with cancer. The presence of specific mutations impacts the expression of IRMs, an observation of potential importance for selecting combinations of gene- and immune-targeted therapeutics. Altered actionable genes correlated with specific checkpoint transcripts. Associations between IRMs and altered genes were validated in independent datasets. Combining immune- and gene-targeted drugs based on IRM/gene correlations merits study.
Collapse
|
21
|
Karagiannakos A, Adamaki M, Tsintarakis A, Vojtesek B, Fåhraeus R, Zoumpourlis V, Karakostis K. Targeting Oncogenic Pathways in the Era of Personalized Oncology: A Systemic Analysis Reveals Highly Mutated Signaling Pathways in Cancer Patients and Potential Therapeutic Targets. Cancers (Basel) 2022; 14:cancers14030664. [PMID: 35158934 PMCID: PMC8833388 DOI: 10.3390/cancers14030664] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is the second leading cause of death globally. One of the main hallmarks in cancer is the functional deregulation of crucial molecular pathways via driver genetic events that lead to abnormal gene expression, giving cells a selective growth advantage. Driver events are defined as mutations, fusions and copy number alterations that are causally implicated in oncogenesis. Molecular analysis on tissues that have originated from a wide range of anatomical areas has shown that mutations in different members of several pathways are implicated in different cancer types. In recent decades, significant efforts have been made to incorporate this knowledge into daily medical practice, providing substantial insight towards clinical diagnosis and personalized therapies. However, since there is still a strong need for more effective drug development, a deep understanding of the involved signaling mechanisms and the interconnections between these pathways is highly anticipated. Here, we perform a systemic analysis on cancer patients included in the Pan-Cancer Atlas project, with the aim to select the ten most highly mutated signaling pathways (p53, RTK-RAS, lipids metabolism, PI-3-Kinase/Akt, ubiquitination, b-catenin/Wnt, Notch, cell cycle, homology directed repair (HDR) and splicing) and to provide a detailed description of each pathway, along with the corresponding therapeutic applications currently being developed or applied. The ultimate scope is to review the current knowledge on highly mutated pathways and to address the attractive perspectives arising from ongoing experimental studies for the clinical implementation of personalized medicine.
Collapse
Affiliation(s)
- Alexandros Karagiannakos
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 48 Vassileos Constantinou Avenue, 11635 Athens, Greece; (A.K.); (M.A.); (A.T.)
| | - Maria Adamaki
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 48 Vassileos Constantinou Avenue, 11635 Athens, Greece; (A.K.); (M.A.); (A.T.)
| | - Antonis Tsintarakis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 48 Vassileos Constantinou Avenue, 11635 Athens, Greece; (A.K.); (M.A.); (A.T.)
| | - Borek Vojtesek
- Research Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, 65653 Brno, Czech Republic; (B.V.); (R.F.)
| | - Robin Fåhraeus
- Research Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, 65653 Brno, Czech Republic; (B.V.); (R.F.)
- Inserm UMRS1131, Institut de Génétique Moléculaire, Université Paris 7, Hôpital St. Louis, F-75010 Paris, France
- Department of Medical Biosciences, Umeå University, 90185 Umeå, Sweden
- International Centre for Cancer Vaccine Science, University of Gdansk, 80-822 Gdansk, Poland
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 48 Vassileos Constantinou Avenue, 11635 Athens, Greece; (A.K.); (M.A.); (A.T.)
- Correspondence: (V.Z.); (K.K.)
| | - Konstantinos Karakostis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 48 Vassileos Constantinou Avenue, 11635 Athens, Greece; (A.K.); (M.A.); (A.T.)
- Inserm UMRS1131, Institut de Génétique Moléculaire, Université Paris 7, Hôpital St. Louis, F-75010 Paris, France
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Correspondence: (V.Z.); (K.K.)
| |
Collapse
|
22
|
Gao S, Jin Y, Zhang H. Pan-Cancer Analyses Reveal Oncogenic and Immunological Role of Dickkopf-1 (DKK1). Front Genet 2021; 12:757897. [PMID: 34899842 PMCID: PMC8654726 DOI: 10.3389/fgene.2021.757897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/05/2021] [Indexed: 12/15/2022] Open
Abstract
WNT signaling pathway inhibitor Dickkopf-1 (DKK1) is related to cancer progression; however, its diagnostic and prognostic potential have not been investigated in a pan-cancer perspective. In this study, multiple bioinformatic analyses were conducted to evaluate therapeutic value of DKK1 in human cancers. The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) project served as data resources. The Wilcoxon rank test was performed to evaluate the expression difference of DKK1 between cancer tissues and normal tissues. A Kaplan-Meier curve and Cox regression were used for prognosis evaluation. Single-sample gene set enrichment analysis (ssGSEA) was used to evaluate the association of DKK1 expression with the immune cell infiltration. The potential function of DKK1 was explored by STRING and clusterProfiler. We found that the expression level of DKK1 is significantly different in different cancer types. Importantly, we demonstrated that DKK1 is an independent risk factor in ESCA, LUAD, MESO, and STAD. Further analysis revealed that DKK1 had a large effect on the immune cell infiltration and markers of certain immune cells, such as Th1 and Th2 cells. PPI network analysis and further pathway enrichment analysis indicated that DKK1 was mainly involved in the WNT signaling pathway. Our findings suggested that DKK1 might serve as a marker of prognosis for certain cancers by affecting the WNT signaling pathway and tumor immune microenvironment.
Collapse
Affiliation(s)
- Shuang Gao
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Ye Jin
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Hongmei Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, China.,School of Clinical Medicine, North China University of Science and Technology, Tangshan, China.,Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
23
|
Newnes HV, Armitage JD, Audsley KM, Bosco A, Waithman J. Directing the Future Breakthroughs in Immunotherapy: The Importance of a Holistic Approach to the Tumour Microenvironment. Cancers (Basel) 2021; 13:cancers13235911. [PMID: 34885021 PMCID: PMC8656826 DOI: 10.3390/cancers13235911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Immunotherapies have changed the way we treat cancer and, while some patients have benefitted greatly, there are still those that do not respond to therapy. Understanding why some patients respond to therapy and others do not is critical in developing new immunotherapeutic strategies. The increasing awareness of the importance of investigating the tumour in its entirety, including the surrounding tissue and role of various immune cells is helping to differentiate responders and non-responders. In addition, the resolution gained by the development of sophisticated bioinformatic technologies allows for a deeper understanding of the complex roles of individual cells in the tumour. This advancement will be critical for the development of novel therapies to treat cancer. Abstract Immunotherapy has revolutionised the treatment of cancers by exploiting the immune system to eliminate tumour cells. Despite the impressive response in a proportion of patients, clinical benefit has been limited thus far. A significant focus to date has been the identification of specific markers associated with response to immunotherapy. Unfortunately, the heterogeneity between patients and cancer types means identifying markers of response to therapy is inherently complex. There is a growing appreciation for the role of the tumour microenvironment (TME) in directing response to immunotherapy. The TME is highly heterogeneous and contains immune, stromal, vascular and tumour cells that all communicate and interact with one another to form solid tumours. This review analyses major cell populations present within the TME with a focus on their diverse and often contradictory roles in cancer and how this informs our understanding of immunotherapy. Furthermore, we discuss the role of integrated omics in providing a comprehensive view of the TME and demonstrate the potential of leveraging multi-omics to decipher the underlying mechanisms of anti-tumour immunity for the development of novel immunotherapeutic strategies.
Collapse
|
24
|
Furman SA, Stern AM, Uttam S, Taylor DL, Pullara F, Chennubhotla SC. In situ functional cell phenotyping reveals microdomain networks in colorectal cancer recurrence. CELL REPORTS METHODS 2021; 1:100072. [PMID: 34888541 PMCID: PMC8653984 DOI: 10.1016/j.crmeth.2021.100072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/14/2021] [Accepted: 08/09/2021] [Indexed: 04/21/2023]
Abstract
Tumors are dynamic ecosystems comprising localized niches (microdomains), possessing distinct compositions and spatial configurations of cancer and non-cancer cell populations. Microdomain-specific network signaling coevolves with a continuum of cell states and functional plasticity associated with disease progression and therapeutic responses. We present LEAPH, an unsupervised machine learning algorithm for identifying cell phenotypes, which applies recursive steps of probabilistic clustering and spatial regularization to derive functional phenotypes (FPs) along a continuum. Combining LEAPH with pointwise mutual information and network biology analyses enables the discovery of outcome-associated microdomains visualized as distinct spatial configurations of heterogeneous FPs. Utilization of an immunofluorescence-based (51 biomarkers) image dataset of colorectal carcinoma primary tumors (n = 213) revealed microdomain-specific network dysregulation supporting cancer stem cell maintenance and immunosuppression that associated selectively with the recurrence phenotype. LEAPH enables an explainable artificial intelligence platform providing insights into pathophysiological mechanisms and novel drug targets to inform personalized therapeutic strategies.
Collapse
Affiliation(s)
- Samantha A. Furman
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Andrew M. Stern
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Shikhar Uttam
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - D. Lansing Taylor
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- SpIntellx, Inc., 2425 Sidney Street, Pittsburgh, PA 15203, USA
| | - Filippo Pullara
- SpIntellx, Inc., 2425 Sidney Street, Pittsburgh, PA 15203, USA
| | - S. Chakra Chennubhotla
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- SpIntellx, Inc., 2425 Sidney Street, Pittsburgh, PA 15203, USA
| |
Collapse
|
25
|
Hui J, Wang C, Miao Y, Liu R, Xu J. The pancancer landscape of Wnt family expression reveals potential biomarkers in urinary system tumors. Cancer Gene Ther 2021; 28:1035-1045. [PMID: 33311568 DOI: 10.1038/s41417-020-00273-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/03/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023]
Abstract
Immunotherapy and targeted therapy have been particularly effective in treating tumors of the urinary system; however, the mechanisms of the Wnt family of proteins in the tumorigenesis, development, and immune response of urinary system tumors are not fully understood. Here, we show that the Wnt family was extensively upregulated in and impacted the prognosis of patients with prostate adenocarcinoma (PRAD) and bladder urothelial carcinoma (BLCA). Moreover, the Wnt family correlated with the levels of infiltrating immune cells, including B cells, CD4 + T cells, CD8 + T cells, neutrophils, macrophages, and dendritic cells. The expression levels of Wnt family members were closely related to neoantigens, the mismatch repair system (MMRS) and DNA methyltransferases, and the mutation rate was generally low. Wnt family members are potential biomarkers for precision immunotherapy of urinary system tumors.
Collapse
Affiliation(s)
- Jialiang Hui
- Department of Organ Transplant, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Chengxinqiao Wang
- Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Yun Miao
- Department of Organ Transplant, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Ruiyu Liu
- Department of Urology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| | - Jian Xu
- Department of Organ Transplant, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| |
Collapse
|
26
|
Backman M, La Fleur L, Kurppa P, Djureinovic D, Elfving H, Brunnström H, Mattsson JSM, Lindberg A, Pontén V, Eltahir M, Mangsbo S, Gulyas M, Isaksson J, Jirström K, Kärre K, Leandersson K, Mezheyeuski A, Pontén F, Strell C, Lindskog C, Botling J, Micke P. Infiltration of NK and plasma cells is associated with a distinct immune subset in non-small cell lung cancer. J Pathol 2021; 255:243-256. [PMID: 34339045 DOI: 10.1002/path.5772] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/13/2021] [Accepted: 07/28/2021] [Indexed: 12/23/2022]
Abstract
Immune cells of the tumor microenvironment are central but erratic targets for immunotherapy. The aim of this study was to characterize novel patterns of immune cell infiltration in non-small cell lung cancer (NSCLC) in relation to its molecular and clinicopathologic characteristics. Lymphocytes (CD3+, CD4+, CD8+, CD20+, FOXP3+, CD45RO+), macrophages (CD163+), plasma cells (CD138+), NK cells (NKp46+), PD1+, and PD-L1+ were annotated on a tissue microarray including 357 NSCLC cases. Somatic mutations were analyzed by targeted sequencing for 82 genes and a tumor mutational load score was estimated. Transcriptomic immune patterns were established in 197 patients based on RNA sequencing data. The immune cell infiltration was variable and showed only poor association with specific mutations. The previously defined immune phenotypic patterns, desert, inflamed, and immune excluded, comprised 30, 13, and 57% of cases, respectively. Notably, mRNA immune activation and high estimated tumor mutational load were unique only for the inflamed pattern. However, in the unsupervised cluster analysis, including all immune cell markers, these conceptual patterns were only weakly reproduced. Instead, four immune classes were identified: (1) high immune cell infiltration, (2) high immune cell infiltration with abundance of CD20+ B cells, (3) low immune cell infiltration, and (4) a phenotype with an imprint of plasma cells and NK cells. This latter class was linked to better survival despite exhibiting low expression of immune response-related genes (e.g. CXCL9, GZMB, INFG, CTLA4). This compartment-specific immune cell analysis in the context of the molecular and clinical background of NSCLC reveals two previously unrecognized immune classes. A refined immune classification, including traits of the humoral and innate immune response, is important to define the immunogenic potency of NSCLC in the era of immunotherapy. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Max Backman
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Linnéa La Fleur
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Pinja Kurppa
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Dijana Djureinovic
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hedvig Elfving
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hans Brunnström
- Division of Pathology, Lund University, Skåne University Hospital, Lund, Sweden
| | | | - Amanda Lindberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Victor Pontén
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Mohamed Eltahir
- Department of Pharmaceutical Bioscience, Uppsala University, Uppsala, Sweden
| | - Sara Mangsbo
- Department of Pharmaceutical Bioscience, Uppsala University, Uppsala, Sweden
| | - Miklos Gulyas
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Johan Isaksson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Respiratory Medicine, Gävle Hospital, Gävle, Sweden
| | - Karin Jirström
- Division of Oncology and Therapeutic Pathology, Department of Clinical Sciences Lund, Lund, Sweden
| | - Klas Kärre
- Department of Microbiology, Cell and Tumor Biology, Karolinska Institutet, Stockholm, Sweden
| | - Karin Leandersson
- Cancer Immunology, Department of Translational Medicine, Lund University, Skånes University Hospital, Malmö, Sweden
| | - Artur Mezheyeuski
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Fredrik Pontén
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Carina Strell
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
27
|
Rodon J, Argilés G, Connolly RM, Vaishampayan U, de Jonge M, Garralda E, Giannakis M, Smith DC, Dobson JR, McLaughlin ME, Seroutou A, Ji Y, Morawiak J, Moody SE, Janku F. Phase 1 study of single-agent WNT974, a first-in-class Porcupine inhibitor, in patients with advanced solid tumours. Br J Cancer 2021; 125:28-37. [PMID: 33941878 PMCID: PMC8257624 DOI: 10.1038/s41416-021-01389-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND This Phase 1 study assessed the safety and efficacy of the Porcupine inhibitor, WNT974, in patients with advanced solid tumours. METHODS Patients (n = 94) received oral WNT974 at doses of 5-30 mg once-daily, plus additional dosing schedules. RESULTS The maximum tolerated dose was not established; the recommended dose for expansion was 10 mg once-daily. Dysgeusia was the most common adverse event (50% of patients), likely resulting from on-target Wnt pathway inhibition. No responses were seen by Response Evaluation Criteria in Solid Tumors (RECIST) v1.1; 16% of patients had stable disease (median duration 19.9 weeks). AXIN2 expression by RT-PCR was reduced in 94% of paired skin biopsies (n = 52) and 74% of paired tumour biopsies (n = 35), confirming inhibition of the Wnt pathway. In an exploratory analysis, an inverse association was observed between AXIN2 change and immune signature change in paired tumour samples (n = 8). CONCLUSIONS Single-agent WNT974 treatment was generally well tolerated. Biomarker analyses suggest that WNT974 may influence immune cell recruitment to tumours, and may enhance checkpoint inhibitor activity. CLINICAL TRIAL REGISTRATION NCT01351103.
Collapse
Affiliation(s)
- Jordi Rodon
- grid.411083.f0000 0001 0675 8654Vall d’Hebron University Hospital and Universitat Autònoma de Barcelona, Barcelona, Spain ,grid.240145.60000 0001 2291 4776Present Address: The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Guillem Argilés
- grid.411083.f0000 0001 0675 8654Vall d’Hebron University Hospital and Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Roisin M. Connolly
- grid.21107.350000 0001 2171 9311Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD USA ,grid.7872.a0000000123318773Present Address: CancerResearch@UCC, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Ulka Vaishampayan
- grid.477517.70000 0004 0396 4462Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI USA
| | - Maja de Jonge
- grid.5645.2000000040459992XDepartment of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Elena Garralda
- grid.488453.60000000417724902START Madrid, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - Marios Giannakis
- grid.38142.3c000000041936754XDana Farber Cancer Institute, Harvard Medical School, Boston, MA USA
| | - David C. Smith
- grid.214458.e0000000086837370University of Michigan, Ann Arbor, MI USA
| | - Jason R. Dobson
- grid.418424.f0000 0004 0439 2056Novartis Institutes for BioMedical Research, Cambridge, MA USA
| | - Margaret E. McLaughlin
- grid.418424.f0000 0004 0439 2056Novartis Institutes for BioMedical Research, Cambridge, MA USA
| | | | - Yan Ji
- grid.418424.f0000 0004 0439 2056Novartis Institutes for BioMedical Research, East Hanover, NJ USA
| | - Jennifer Morawiak
- grid.418424.f0000 0004 0439 2056Novartis Institutes for BioMedical Research, Cambridge, MA USA
| | - Susan E. Moody
- grid.418424.f0000 0004 0439 2056Novartis Institutes for BioMedical Research, Cambridge, MA USA
| | - Filip Janku
- grid.240145.60000 0001 2291 4776Department of Investigational Cancer Therapeutics (Phase 1 Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
28
|
Guo JN, Xia BR, Deng SH, Yang C, Pi YN, Cui BB, Jin WL. Deubiquitinating Enzymes Orchestrate the Cancer Stem Cell-Immunosuppressive Niche Dialogue: New Perspectives and Therapeutic Potential. Front Cell Dev Biol 2021; 9:680100. [PMID: 34179009 PMCID: PMC8220152 DOI: 10.3389/fcell.2021.680100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/17/2021] [Indexed: 11/13/2022] Open
Abstract
Cancer stem cells (CSCs) are sparks for igniting tumor recurrence and the instigators of low response to immunotherapy and drug resistance. As one of the important components of tumor microenvironment, the tumor associated immune microenvironment (TAIM) is driving force for the heterogeneity, plasticity and evolution of CSCs. CSCs create the inhibitory TAIM (ITAIM) mainly through four stemness-related signals (SRSs), including Notch-nuclear factor-κB axis, Hedgehog, Wnt and signal transducer and activator of transcription. Ubiquitination and deubiquitination in proteins related to the specific stemness of the CSCs have a profound impact on the regulation of ITAIM. In regulating the balance between ubiquitination and deubiquitination, it is crucial for deubiquitinating enzymes (DUBs) to cleave ubiquitin chains from substrates. Ubiquitin-specific peptidases (USPs) comprise the largest family of DUBs. Growing evidence suggests that they play novel functions in contribution of ITAIM, including regulating tumor immunogenicity, activating stem cell factors, upregulating the SRSs, stabilizing anti-inflammatory receptors, and regulating anti-inflammatory cytokines. These overactive or abnormal signaling may dampen antitumor immune responses. The inhibition of USPs could play a regulatory role in SRSs and reversing ITAIM, and also have great potential in improving immune killing ability against tumor cells, including CSCs. In this review, we focus on the USPs involved in CSCs signaling pathways and regulating ITAIM, which are promising therapeutic targets in antitumor therapy.
Collapse
Affiliation(s)
- Jun-Nan Guo
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Bai-Rong Xia
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Anhui Provincial Cancer Hospital, University of Science and Technology of China, Hefei, China
| | - Shen-Hui Deng
- Department of Anesthesiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chang Yang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ya-Nan Pi
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Bin-Bin Cui
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wei-Lin Jin
- Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Institute of Cancer Neuroscience, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| |
Collapse
|
29
|
Giacobino C, Canta M, Fornaguera C, Borrós S, Cauda V. Extracellular Vesicles and Their Current Role in Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13092280. [PMID: 34068657 PMCID: PMC8126043 DOI: 10.3390/cancers13092280] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In recent years, immunotherapy has shown great advancement, becoming a powerful tool to combat cancer. In this context, the use of biologically derived vesicles has also acquired importance for cancer immunotherapy. Extracellular vesicles are thus proposed to transport molecules able to trigger an immune response and thus fight cancer cells. As a particular immunotherapeutic approach, a new technique also consists in the exploitation of extracellular vesicles as new cancer vaccines. The present review provides basic notions on cancer immunotherapy and describes several clinical trials in which therapeutic anticancer vaccines are tested. In particular, the potential of extracellular vesicles-based therapeutic vaccines in the treatment of cancer patients is highlighted, even with advanced stage-cancer. A focus on the clinical studies, already completed or still in progress, is offered and a systematic collection and reorganization of the present literature on this topic is proposed to the reader. Abstract Extracellular vesicles (EVs) are natural particles formed by the lipid bilayer and released from almost all cell types to the extracellular environment both under physiological conditions and in presence of a disease. EVs are involved in many biological processes including intercellular communication, acting as natural carriers in the transfer of various biomolecules such as DNA, various RNA types, proteins and different phospholipids. Thanks to their transfer and targeting abilities, they can be employed in drug and gene delivery and have been proposed for the treatment of different diseases, including cancer. Recently, the use of EVs as biological carriers has also been extended to cancer immunotherapy. This new technique of cancer treatment involves the use of EVs to transport molecules capable of triggering an immune response to damage cancer cells. Several studies have analyzed the possibility of using EVs in new cancer vaccines, which represent a particular form of immunotherapy. In the literature there are only few publications that systematically group and collectively discuss these studies. Therefore, the purpose of this review is to illustrate and give a partial reorganization to what has been produced in the literature so far. We provide basic notions on cancer immunotherapy and describe some clinical trials in which therapeutic cancer vaccines are tested. We thus focus attention on the potential of EV-based therapeutic vaccines in the treatment of cancer patients, overviewing the clinically relevant trials, completed or still in progress, which open up new perspectives in the fight against cancer.
Collapse
Affiliation(s)
- Carla Giacobino
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (C.G.); (M.C.)
| | - Marta Canta
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (C.G.); (M.C.)
| | - Cristina Fornaguera
- Grup d’Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Via Augusta 390, 08017 Barcelona, Spain; (C.F.); (S.B.)
| | - Salvador Borrós
- Grup d’Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Via Augusta 390, 08017 Barcelona, Spain; (C.F.); (S.B.)
| | - Valentina Cauda
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (C.G.); (M.C.)
- Correspondence:
| |
Collapse
|
30
|
Deyell M, Garris CS, Laughney AM. Cancer metastasis as a non-healing wound. Br J Cancer 2021; 124:1491-1502. [PMID: 33731858 PMCID: PMC8076293 DOI: 10.1038/s41416-021-01309-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/25/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023] Open
Abstract
Most cancer deaths are caused by metastasis: recurrence of disease by disseminated tumour cells at sites distant from the primary tumour. Large numbers of disseminated tumour cells are released from the primary tumour, even during the early stages of tumour growth. However, only a minority survive as potential seeds for future metastatic outgrowths. These cells must adapt to a relatively inhospitable microenvironment, evade immune surveillance and progress from the micro- to macro-metastatic stage to generate a secondary tumour. A pervasive driver of this transition is chronic inflammatory signalling emanating from tumour cells themselves. These signals can promote migration and engagement of stem and progenitor cell function, events that are also central to a wound healing response. In this review, we revisit the concept of cancer as a non-healing wound, first introduced by Virchow in the 19th century, with a new tumour cell-intrinsic perspective on inflammation and focus on metastasis. Cellular responses to inflammation in both wound healing and metastasis are tightly regulated by crosstalk with the surrounding microenvironment. Targeting or restoring canonical responses to inflammation could represent a novel strategy to prevent the lethal spread of cancer.
Collapse
Affiliation(s)
- Matthew Deyell
- grid.5386.8000000041936877XInstitute for Computational Biomedicine, Weill Cornell Medicine, New York, NY USA ,grid.5386.8000000041936877XDepartment of Physiology and Biophysics, Weill Cornell Medicine, New York, NY USA ,grid.5386.8000000041936877XSandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY USA ,grid.4444.00000 0001 2112 9282Chimie Biologie et Innovation, ESPCI Paris, Université PSL, CNRS, Paris, France
| | | | - Ashley M. Laughney
- grid.5386.8000000041936877XInstitute for Computational Biomedicine, Weill Cornell Medicine, New York, NY USA ,grid.5386.8000000041936877XDepartment of Physiology and Biophysics, Weill Cornell Medicine, New York, NY USA ,grid.5386.8000000041936877XSandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY USA
| |
Collapse
|
31
|
β-Catenin Activation in Hepatocellular Cancer: Implications in Biology and Therapy. Cancers (Basel) 2021; 13:cancers13081830. [PMID: 33921282 PMCID: PMC8069637 DOI: 10.3390/cancers13081830] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Liver cancer is a dreadful tumor which has gradually increased in incidence all around the world. One major driver of liver cancer is the Wnt–β-catenin pathway which is active in a subset of these tumors. While this pathway is normally important in liver development, regeneration and homeostasis, it’s excessive activation due to mutations, is detrimental and leads to tumor cell growth, making it an important therapeutic target. There are also some unique characteristics of this pathway activation in liver cancer. It makes the tumor addicted to specific amino acids and in turn to mTOR signaling, which can be treated by certain existing therapies. In addition, activation of the Wnt–β-catenin in liver cancer appears to alter the immune cell landscape making it less likely to respond to the new immuno-oncology treatments. Thus, Wnt–β-catenin active tumors may need to be treated differently than non-Wnt–β-catenin active tumors. Abstract Hepatocellular cancer (HCC), the most common primary liver tumor, has been gradually growing in incidence globally. The whole-genome and whole-exome sequencing of HCC has led to an improved understanding of the molecular drivers of this tumor type. Activation of the Wnt signaling pathway, mostly due to stabilizing missense mutations in its downstream effector β-catenin (encoded by CTNNB1) or loss-of-function mutations in AXIN1 (the gene which encodes for Axin-1, an essential protein for β-catenin degradation), are seen in a major subset of HCC. Because of the important role of β-catenin in liver pathobiology, its role in HCC has been extensively investigated. In fact, CTNNB1 mutations have been shown to have a trunk role. β-Catenin has been shown to play an important role in regulating tumor cell proliferation and survival and in tumor angiogenesis, due to a host of target genes regulated by the β-catenin transactivation of its transcriptional factor TCF. Proof-of-concept preclinical studies have shown β-catenin to be a highly relevant therapeutic target in CTNNB1-mutated HCCs. More recently, studies have revealed a unique role of β-catenin activation in regulating both tumor metabolism as well as the tumor immune microenvironment. Both these roles have notable implications for the development of novel therapies for HCC. Thus, β-catenin has a pertinent role in driving HCC development and maintenance of this tumor-type, and could be a highly relevant therapeutic target in a subset of HCC cases.
Collapse
|
32
|
BCL9/BCL9L promotes tumorigenicity through immune-dependent and independent mechanisms in triple negative breast cancer. Oncogene 2021; 40:2982-2997. [PMID: 33767438 DOI: 10.1038/s41388-021-01756-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 03/01/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023]
Abstract
Treatment of patients with triple-negative breast cancer (TNBC) has been challenging due to a lack of well-defined molecular targets. The Wnt/β-catenin pathway is known to be activated in many TNBC patients and BCL9 and BCL9L are important transcriptional co-activators of β-catenin, but whether inhibition of BCL9/BCL9L can suppress TNBC growth and the underlying mechanism are not fully understood. Here we demonstrate that the expression of BCL9 and BCL9L is directly correlated with malignancy in TNBC patient tumors and that BCL9 and BCL9L promote tumor cell growth, cell migration and metastasis in TNBC models. Mechanistically, we found that BCL9/BCL9L promotes tumorigenicity through both the Wnt and TGF-β pathways. Besides, BCL9/BCL9L expression inversely correlates with CD8+ T cell infiltration in TNBC and BCL9/BCL9L inhibits the infiltration of CD8+ T cells in the tumor microenvironment. hsBCL9CT-24, an inhibitor of BCL9/β-catenin peptides, promotes intratumoral infiltration of cytotoxic T cells, reducing regulatory T cells (Treg) and increasing dendritic cells (DCs). Inhibition of BCL9/BCL9L and TGF-β suppresses activity of Treg. TGF-β signaling increases tumor infiltration of cytotoxic CD8+ T cells. In accordance, genetic or pharmacological inhibition of BCL9/BCL9L synergizes with PD-1/L1 antibodies to inhibit tumor growth. In summary, these results suggest that targeting BCL9/BCL9L has a direct anti-tumor effect and also unleashes an anti-cancer immune response through inhibition of both Wnt and TGF-β signaling, suggesting a viable therapeutic approach for TNBC treatment.
Collapse
|
33
|
Kumar R, Gulia K. The convergence of nanotechnology‐stem cell, nanotopography‐mechanobiology, and biotic‐abiotic interfaces: Nanoscale tools for tackling the top killer, arteriosclerosis, strokes, and heart attacks. NANO SELECT 2021. [DOI: 10.1002/nano.202000192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Rajiv Kumar
- NIET National Institute of Medical Science Rajasthan India
| | - Kiran Gulia
- Materials and Manufacturing School of Engineering University of Wolverhampton Wolverhampton England, UK
| |
Collapse
|
34
|
Malvehy J, Samoylenko I, Schadendorf D, Gutzmer R, Grob JJ, Sacco JJ, Gorski KS, Anderson A, Pickett CA, Liu K, Gogas H. Talimogene laherparepvec upregulates immune-cell populations in non-injected lesions: findings from a phase II, multicenter, open-label study in patients with stage IIIB-IVM1c melanoma. J Immunother Cancer 2021; 9:jitc-2020-001621. [PMID: 33785610 PMCID: PMC8011715 DOI: 10.1136/jitc-2020-001621] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2021] [Indexed: 11/20/2022] Open
Abstract
Background Talimogene laherparepvec (T-VEC), an oncolytic virus, was designed to selectively replicate in and lyse tumor cells, releasing tumor-derived antigen to stimulate a tumor-specific immune response. Methods In this phase II study in patients with unresectable stage IIIB–IV melanoma, we evaluated non-injected lesions to establish whether baseline or change in intratumoral CD8+ T-cell density (determined using immunohistochemistry) correlated with T-VEC clinical response. Results Of 112 enrolled patients, 111 received ≥1 dose of T-VEC. After a median follow-up of 108.0 weeks, objective/complete response rates were 28%/14% in the overall population and 32%/18% in patients with stage IIIB–IVM1a disease. No unexpected toxicity occurred. Baseline and week 6 change from baseline CD8+ T-cell density results were available for 91 and 65 patients, respectively. Neither baseline nor change in CD8+ T-cell density correlated with objective response rate, changes in tumor burden, duration of response or durable response rate. However, a 2.4-fold median increase in CD8+ T-cell density in non-injected lesions from baseline to week 6 was observed. In exploratory analyses, multiparameter immunofluorescence showed that after treatment there was an increase in the proportion of infiltrating CD8+ T-cells expressing granzyme B and checkpoint markers (programmed death-1, programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte antigen-4) in non-injected lesions, together with an increase in helper T-cells. Consistent with T-cell infiltrate, we observed an increase in the adaptive resistance marker PD-L1 in non-injected lesions. Conclusions This study indicates that T-VEC induces systemic immune activity and alters the tumor microenvironment in a way that will likely enhance the effects of other immunotherapy agents in combination therapy. Trial registration number NCT02366195.
Collapse
Affiliation(s)
- Josep Malvehy
- Dermatology Department and IDIBAPS, Hospital Clinic of Barcelona, Barcelona, Catalunya, Spain
| | - Igor Samoylenko
- NN Blokhin Russian Cancer Research Center, Moscow, Russian Federation
| | - Dirk Schadendorf
- Department of Dermatology, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Ralf Gutzmer
- Department of Dermatology and Allergy, Skin Cancer Center Hannover, Hannover Medical School, Hannover, Germany
| | | | - Joseph J Sacco
- Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, Wirral, UK.,University of Liverpool, Liverpool, UK
| | | | | | | | - Kate Liu
- Amgen Inc, Thousand Oaks, California, USA
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
35
|
Li Z, Liu Y, Fang X, Shu Z. Nanomaterials Enhance the Immunomodulatory Effect of Molecular Targeted Therapy. Int J Nanomedicine 2021; 16:1631-1661. [PMID: 33688183 PMCID: PMC7935456 DOI: 10.2147/ijn.s290346] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/23/2021] [Indexed: 01/22/2023] Open
Abstract
Molecular targeted therapy, a tumor therapy strategy that inhibits specific oncogenic targets, has been shown to modulate the immune response. In addition to directly inhibiting the proliferation and metastasis of tumor cells, molecular targeted drugs can activate the immune system through a variety of mechanisms, including by promoting tumor antigen processing and presentation, increasing intratumoral T cell infiltration, enhancing T cell activation and function, and attenuating the immunosuppressive effect of the tumor microenvironment. However, poor water solubility, insufficient accumulation at the tumor site, and nonspecific targeting of immune cells limit their application. To this end, a variety of nanomaterials have been developed to overcome these obstacles and amplify the immunomodulatory effects of molecular targeted drugs. In this review, we summarize the impact of molecular targeted drugs on the antitumor immune response according to their mechanisms, highlight the advantages of nanomaterials in enhancing the immunomodulatory effect of molecular targeted therapy, and discuss the current challenges and future prospects.
Collapse
Affiliation(s)
- Zhongmin Li
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China
| | - Yilun Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China
| | - Zhenbo Shu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China
| |
Collapse
|
36
|
Xue D, Zheng Y, Wen J, Han J, Tuo H, Liu Y, Peng Y. Role of chemokines in hepatocellular carcinoma (Review). Oncol Rep 2021; 45:809-823. [PMID: 33650640 PMCID: PMC7859922 DOI: 10.3892/or.2020.7906] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 12/04/2020] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent malignant tumor worldwide, with an unsatisfactory prognosis, although treatments are improving. One of the main challenges for the treatment of HCC is the prevention or management of recurrence and metastasis of HCC. It has been found that chemokines and their receptors serve a pivotal role in HCC progression. In the present review, the literature on the multifactorial roles of exosomes in HCC from PubMed, Cochrane library and Embase were obtained, with a specific focus on the functions and mechanisms of chemokines in HCC. To date, >50 chemokines have been found, which can be divided into four families: CXC, CX3C, CC and XC, according to the different positions of the conserved N‑terminal cysteine residues. Chemokines are involved in the inflammatory response, tumor immune response, proliferation, invasion and metastasis via modulation of various signaling pathways. Thus, chemokines and their receptors directly or indirectly shape the tumor cell microenvironment, and regulate the biological behavior of the tumor. In addition, the potential application of chemokines in chemotaxis of exosomes as drug vehicles is discussed. Exosomes containing chemokines or expressing receptors for chemokines may improve chemotaxis to HCC and may thus be exploited for targeted drug delivery.
Collapse
Affiliation(s)
- Dongdong Xue
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, Hebei 050051, Shanghai 200065, P.R. China
| | - Ya Zheng
- Medical Center Laboratory, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Junye Wen
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, Hebei 050051, Shanghai 200065, P.R. China
| | - Jingzhao Han
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, Hebei 050051, Shanghai 200065, P.R. China
| | - Hongfang Tuo
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, Hebei 050051, Shanghai 200065, P.R. China
| | - Yifan Liu
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, Hebei 050051, Shanghai 200065, P.R. China
| | - Yanhui Peng
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, Hebei 050051, Shanghai 200065, P.R. China
| |
Collapse
|
37
|
Rückert M, Deloch L, Frey B, Schlücker E, Fietkau R, Gaipl US. Combinations of Radiotherapy with Vaccination and Immune Checkpoint Inhibition Differently Affect Primary and Abscopal Tumor Growth and the Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13040714. [PMID: 33572437 PMCID: PMC7916259 DOI: 10.3390/cancers13040714] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
Radiotherapy (RT) is known to have immune-modulatory properties. We hypothesized that RT and inactivated whole tumor cell vaccines generated with high hydrostatic pressure (HHP) synergize to retard the tumor growth which can be additionally improved with anti-PD-1 treatment. In abscopal tumor models, we injected mice with B16-F10 melanoma or TS/A mammary tumors. To evaluate the efficiency of RT in combination with HHP vaccines, we locally irradiated only one tumor with 2 × 8 Gy or 3 × 8 Gy. HHP vaccines further retarded the growth of locally irradiated (2 × 8 Gy) tumors. However, HHP vaccination combined with RT failed to induce abscopal anti-tumor immune responses, namely those to non-irradiated tumors, and even partly abrogated those which were induced with RT plus anti-PD-1. In the latter group, the abscopal effects were accompanied by an elevated infiltration of CD8+ T cells, monocytes/macrophages, and dendritic cells. 3 × 8 Gy failed to induce abscopal effects in association with increased expression of immunosuppressive checkpoint molecules compared to 2 × 8 Gy. We conclude that HHP vaccines induce anti-tumor effects, but only if the tumor microenvironment was previously modulated by hypofractionated RT with not too many fractions, but failed to improve RT plus anti-PD-induced abscopal responses that are characterized by distinct immune alterations.
Collapse
Affiliation(s)
- Michael Rückert
- Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.R.); (L.D.); (B.F.); (R.F.)
| | - Lisa Deloch
- Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.R.); (L.D.); (B.F.); (R.F.)
| | - Benjamin Frey
- Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.R.); (L.D.); (B.F.); (R.F.)
| | - Eberhard Schlücker
- Institute of Process Machinery and Systems Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany;
| | - Rainer Fietkau
- Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.R.); (L.D.); (B.F.); (R.F.)
| | - Udo S. Gaipl
- Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.R.); (L.D.); (B.F.); (R.F.)
- Correspondence: ; Tel.: +49-(0)9131-85-44258
| |
Collapse
|
38
|
Recent Advancements in the Mechanisms Underlying Resistance to PD-1/PD-L1 Blockade Immunotherapy. Cancers (Basel) 2021; 13:cancers13040663. [PMID: 33562324 PMCID: PMC7915065 DOI: 10.3390/cancers13040663] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/03/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Immune checkpoint blockade targeting PD-1/PD-L1 has a promising therapeutic efficacy in different tumors, but a significant percentage of patients cannot benefit from this therapy due to primary and acquired resistance during treatment. This review summarizes the recent findings of PD-L1 role in resistance to therapies through the PD-1/PD-L1 pathway and other correlating signaling pathways. A special focus will be given to the key mechanisms underlying resistance to the PD-1/PD-L1 blockade in cancer immunotherapy. Furthermore, we also discuss the promising combination of therapeutic strategies for patients resistant to the PD-1/PD-L1 blockade in order to enhance the efficacy of immune checkpoint inhibitors. Abstract Release of immunoreactive negative regulatory factors such as immune checkpoint limits antitumor responses. PD-L1 as a significant immunosuppressive factor has been involved in resistance to therapies such as chemotherapy and target therapy in various cancers. Via interacting with PD-1, PD-L1 can regulate other factors or lead to immune evasion of cancer cells. Besides, immune checkpoint blockade targeting PD-1/PD-L1 has promising therapeutic efficacy in the different tumors, but a significant percentage of patients cannot benefit from this therapy due to primary and acquired resistance during treatment. In this review, we described the utility of PD-L1 expression levels for predicting poor prognosis in some tumors and present evidence for a role of PD-L1 in resistance to therapies through PD-1/PD-L1 pathway and other correlating signaling pathways. Afterwards, we elaborate the key mechanisms underlying resistance to PD-1/PD-L1 blockade in cancer immunotherapy. Furthermore, promising combination of therapeutic strategies for patients resistant to PD-1/PD-L1 blockade therapy or other therapies associated with PD-L1 expression was also summarized.
Collapse
|
39
|
Cancer Stem Cells-Key Players in Tumor Relapse. Cancers (Basel) 2021; 13:cancers13030376. [PMID: 33498502 PMCID: PMC7864187 DOI: 10.3390/cancers13030376] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/10/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
Tumor relapse and treatment failure are unfortunately common events for cancer patients, thus often rendering cancer an uncurable disease. Cancer stem cells (CSCs) are a subset of cancer cells endowed with tumor-initiating and self-renewal capacity, as well as with high adaptive abilities. Altogether, these features contribute to CSC survival after one or multiple therapeutic approaches, thus leading to treatment failure and tumor progression/relapse. Thus, elucidating the molecular mechanisms associated with stemness-driven resistance is crucial for the development of more effective drugs and durable responses. This review will highlight the mechanisms exploited by CSCs to overcome different therapeutic strategies, from chemo- and radiotherapies to targeted therapies and immunotherapies, shedding light on their plasticity as an insidious trait responsible for their adaptation/escape. Finally, novel CSC-specific approaches will be described, providing evidence of their preclinical and clinical applications.
Collapse
|
40
|
Kwee SA, Tiirikainen M. Beta-catenin activation and immunotherapy resistance in hepatocellular carcinoma: mechanisms and biomarkers. ACTA ACUST UNITED AC 2021; 7. [PMID: 33553649 PMCID: PMC7861492 DOI: 10.20517/2394-5079.2020.124] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mutations involving CTNNB1, the gene encoding beta-catenin, and other molecular alterations that affect the Wnt/beta-catenin signaling pathway are exceptionally common in hepatocellular carcinoma. Several of these alterations have also been associated with scarcity of immune cells in the tumor microenvironment and poor clinical response to immune checkpoint inhibitor therapy. In light of these associations, tumor biomarkers of beta-catenin status could have the potential to serve as clinical predictors of immunotherapy outcome. This editorial review article summarizes recent pre-clinical and clinical research pertaining to associations between beta-catenin activation and diminished anti-tumor immunity. Potential non-invasive biomarkers that may provide a window into this oncogenic mechanism of immune evasion are also presented and discussed.
Collapse
Affiliation(s)
- Sandi A Kwee
- Cancer Biology Program (SAK) and Population Sciences in the Pacific Program (MT), University of Hawaii Cancer Center, University of Hawaii, Honolulu, Hawaii 96813, USA.,Hamamatsu/Queen's PET Imaging Center, The Queen's Medical Center, Honolulu, Hawaii 96813, USA
| | - Maarit Tiirikainen
- Cancer Biology Program (SAK) and Population Sciences in the Pacific Program (MT), University of Hawaii Cancer Center, University of Hawaii, Honolulu, Hawaii 96813, USA
| |
Collapse
|
41
|
Molecularly targeted photothermal ablation improves tumor specificity and immune modulation in a rat model of hepatocellular carcinoma. Commun Biol 2020; 3:783. [PMID: 33335270 PMCID: PMC7746712 DOI: 10.1038/s42003-020-01522-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Thermal ablation is a standard therapy for patients with hepatocellular carcinoma (HCC). Contemporary ablation devices are imperfect, as they lack tumor specificity. An ideal ablation modality would generate thermal energy only within tumoral tissue. Furthermore, as hyperthermia is known to influence tumor immunity, such a tumor-specific ablation modality may have the ability to favorably modulate the tumor immune landscape. Here we show a clinically relevant thermal ablation modality that generates tumor-specific hyperthermia, termed molecularly targeted photothermal ablation (MTPA), that is based upon the excellent localization of indocyanine green to HCC. In a syngeneic rat model, we demonstrate the tumor-specific hyperthermia generated by MTPA. We also show through spatial and transcriptomic profiling techniques that MTPA favorably modulates the intratumoral myeloid population towards tumor immunogenicity and diminishes the systemic release of oncogenic cytokines relative to conventional ablation modalities. Nina Muñoz et al. present a novel thermal ablation technique, termed molecularly targeted photothermal ablation (MTPA) that generates hyperthermia specifically within tumor tissue in a clinically-relevant rat model of liver cancer. They further show that MTPA modulates the intratumoral immune profile toward stronger tumor immunogenicity while reducing the release of oncogenic cytokines relative to conventional ablation techniques.
Collapse
|
42
|
Behl T, Sharma A, Sharma L, Sehgal A, Zengin G, Brata R, Fratila O, Bungau S. Exploring the Multifaceted Therapeutic Potential of Withaferin A and Its Derivatives. Biomedicines 2020; 8:E571. [PMID: 33291236 PMCID: PMC7762146 DOI: 10.3390/biomedicines8120571] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Withaferin A (WA), a manifold studied, C28-steroidal lactone withanolide found in Withania somnifera. Given its unique beneficial effects, it has gathered attention in the era of modern science. Cancer, being considered a "hopeless case and the leading cause of death worldwide, and the available conventional therapies have many lacunae in the form of side effects. The poly pharmaceutical natural compound, WA treatment, displayed attenuation of various cancer hallmarks by altering oxidative stress, promoting apoptosis, and autophagy, inhibiting cell proliferation, reducing angiogenesis, and metastasis progression. The cellular proteins associated with antitumor pathways were also discussed. WA structural modifications attack multiple signal transduction pathways and enhance the therapeutic outcomes in various diseases. Moreover, it has shown validated pharmacological effects against multiple neurodegenerative diseases by inhibiting acetylcholesterinases and butyrylcholinesterases enzyme activity, antidiabetic activity by upregulating adiponectin and preventing the phosphorylation of peroxisome proliferator-activated receptors (PPARγ), cardioprotective activity by AMP-activated protein kinase (AMPK) activation and suppressing mitochondrial apoptosis. The current review is an extensive survey of various WA associated disease targets, its pharmacokinetics, synergistic combination, modifications, and biological activities.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Aditi Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh 173229, India; (A.S.); (L.S.)
| | - Lalit Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh 173229, India; (A.S.); (L.S.)
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, Konya 42250, Turkey;
| | - Roxana Brata
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (R.B.); (O.F.)
| | - Ovidiu Fratila
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (R.B.); (O.F.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| |
Collapse
|
43
|
Inflammation and immunity in ovarian cancer. EJC Suppl 2020; 15:56-66. [PMID: 33240443 PMCID: PMC7569134 DOI: 10.1016/j.ejcsup.2019.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/15/2019] [Accepted: 12/27/2019] [Indexed: 12/30/2022] Open
Abstract
The standard first-line therapy for ovarian cancer is a combination of surgery and carboplatin/paclitaxel-based chemotherapy. Patients with longer survival and improved response to chemotherapy usually present T-cell inflamed tumours. The presence of tumour-infiltrating T cells (TILs) notably varies among the different subtypes of ovarian tumours, being highest in high-grade serous ovarian carcinoma, intermediate in endometrioid tumours, and lowest in low-grade serous, mucinous and clear cell tumours. Interestingly, the presence of TILs is often accompanied by a strong immunosuppressive tumour environment. A better understanding of the immune response against ovarian cancer and the tumour immune evasion mechanisms will enable improved prognostication, response prediction and immunotherapy of this disease. This article provides an overview of some ovarian cancer cell features relevant for antitumour response, such as tumour-associated antigens, including neoantigens, expression of inhibitory molecules, and other mechanisms of immune evasion. Moreover, we describe relevant immune cell types found in epithelial ovarian tumours, including T and B lymphocytes, regulatory T cells, natural killer cells, tumour-associated macrophages, myeloid-derived suppressor cells and neutrophils. We focus on how these components influence the burden of the tumour and the clinical outcome. The presence of spontaneous tumour-specific T lymphocytes and the existence of multiple immune evasion mechanisms in epithelial ovarian cancer (EOC) support the immunogenicity of this tumour. Tumour-infiltrating T lymphocytes (TILs) have been associated with disease outcome in EOC, indicating their clinical significance. The subtypes of EOC, mutations in TP53 and breast and ovarian cancer susceptibility protein 1/2 and the immune expression signature are factors associated to TIL density in EOC. The tumour microenvironment in EOC consists of a dynamic and complex network of soluble factors, inhibitory receptors and immunosuppressive cells.
Collapse
|
44
|
Schwarze JK, Awada G, Cras L, Tijtgat J, Forsyth R, Dufait I, Tuyaerts S, Van Riet I, Neyns B. Intratumoral Combinatorial Administration of CD1c (BDCA-1) + Myeloid Dendritic Cells Plus Ipilimumab and Avelumab in Combination with Intravenous Low-Dose Nivolumab in Patients with Advanced Solid Tumors: A Phase IB Clinical Trial. Vaccines (Basel) 2020; 8:vaccines8040670. [PMID: 33182610 PMCID: PMC7712037 DOI: 10.3390/vaccines8040670] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/31/2022] Open
Abstract
Intratumoral (IT) myeloid dendritic cells (myDCs) play a pivotal role in re-licensing antitumor cytotoxic T lymphocytes. IT injection of the IgG1 monoclonal antibodies ipilimumab and avelumab may induce antibody-dependent cellular cytotoxicity, thereby enhancing the release of tumor antigens that can be captured and processed by CD1c (BDCA-1)+ myDCs. Patients with advanced solid tumors after standard care were eligible for IT injections of ≥1 lesion with ipilimumab (10 mg) and avelumab (40 mg) and intravenous (IV) nivolumab (10 mg) on day 1, followed by IT injection of autologous CD1c (BDCA-1)+ myDCs on day 2. IT/IV administration of ipilimumab, avelumab, and nivolumab was repeated bi-weekly. Primary objectives were safety and feasibility. Nine patients were treated with a median of 21 × 106 CD1c (BDCA-1)+ myDCs, and a median of 4 IT/IV administrations of ipilimumab, avelumab, and nivolumab. The treatment was safe with mainly injection-site reactions, but also immune-related pneumonitis (n = 2), colitis (n = 1), and bullous pemphigoid (n = 1). The best response was a durable partial response in a patient with stage IV melanoma who previously progressed on checkpoint inhibitors. Our combinatorial therapeutic approach, including IT injection of CD1c (BDCA-1)+ myDCs, is feasible and safe, and it resulted in encouraging signs of antitumor activity in patients with advanced solid tumors.
Collapse
Affiliation(s)
- Julia Katharina Schwarze
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (J.T.); (I.D.); (S.T.); (B.N.)
- Correspondence: ; Tel.: +32-2-477-64-15
| | - Gil Awada
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (J.T.); (I.D.); (S.T.); (B.N.)
| | - Louise Cras
- Department of Anatomopathology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (L.C.); (R.F.)
| | - Jens Tijtgat
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (J.T.); (I.D.); (S.T.); (B.N.)
| | - Ramses Forsyth
- Department of Anatomopathology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (L.C.); (R.F.)
| | - Inès Dufait
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (J.T.); (I.D.); (S.T.); (B.N.)
| | - Sandra Tuyaerts
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (J.T.); (I.D.); (S.T.); (B.N.)
| | - Ivan Van Riet
- Stem Cell Laboratory, Department of Hematology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium;
| | - Bart Neyns
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (J.T.); (I.D.); (S.T.); (B.N.)
| |
Collapse
|
45
|
Stefanski CD, Prosperi JR. Wnt-Independent and Wnt-Dependent Effects of APC Loss on the Chemotherapeutic Response. Int J Mol Sci 2020; 21:E7844. [PMID: 33105836 PMCID: PMC7660076 DOI: 10.3390/ijms21217844] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/20/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Resistance to chemotherapy occurs through mechanisms within the epithelial tumor cells or through interactions with components of the tumor microenvironment (TME). Chemoresistance and the development of recurrent tumors are two of the leading factors of cancer-related deaths. The Adenomatous Polyposis Coli (APC) tumor suppressor is lost in many different cancers, including colorectal, breast, and prostate cancer, and its loss correlates with a decreased overall survival in cancer patients. While APC is commonly known for its role as a negative regulator of the WNT pathway, APC has numerous binding partners and functional roles. Through APC's interactions with DNA repair proteins, DNA replication proteins, tubulin, and other components, recent evidence has shown that APC regulates the chemotherapy response in cancer cells. In this review article, we provide an overview of some of the cellular processes in which APC participates and how they impact chemoresistance through both epithelial- and TME-derived mechanisms.
Collapse
Affiliation(s)
- Casey D. Stefanski
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46617, USA;
- Mike and Josie Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Jenifer R. Prosperi
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46617, USA;
- Mike and Josie Harper Cancer Research Institute, South Bend, IN 46617, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine-South Bend, South Bend, IN 46617, USA
| |
Collapse
|
46
|
de Bono JS, Guo C, Gurel B, De Marzo AM, Sfanos KS, Mani RS, Gil J, Drake CG, Alimonti A. Prostate carcinogenesis: inflammatory storms. Nat Rev Cancer 2020; 20:455-469. [PMID: 32546840 DOI: 10.1038/s41568-020-0267-9] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/24/2020] [Indexed: 02/06/2023]
Abstract
Prostate cancer is a major cause of cancer morbidity and mortality. Intra-prostatic inflammation is a risk factor for prostate carcinogenesis, with diet, chemical injury and an altered microbiome being causally implicated. Intra-prostatic inflammatory cell recruitment and expansion can ultimately promote DNA double-strand breaks and androgen receptor activation in prostate epithelial cells. The activation of the senescence-associated secretory phenotype fuels further 'inflammatory storms', with free radicals leading to further DNA damage. This drives the overexpression of DNA repair and tumour suppressor genes, rendering these genes susceptible to mutagenic insults, with carcinogenesis accelerated by germline DNA repair gene defects. We provide updates on recent advances in elucidating prostate carcinogenesis and explore novel therapeutic and prevention strategies harnessing these discoveries.
Collapse
Affiliation(s)
- Johann S de Bono
- The Institute of Cancer Research, London, UK.
- The Royal Marsden NHS Foundation Trust, Sutton, UK.
| | - Christina Guo
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Bora Gurel
- The Institute of Cancer Research, London, UK
| | | | - Karen S Sfanos
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ram S Mani
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jesús Gil
- MRC London Institute of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | | | - Andrea Alimonti
- Institute of Oncology Research, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
47
|
Pérez-Ruiz E, Melero I, Kopecka J, Sarmento-Ribeiro AB, García-Aranda M, De Las Rivas J. Cancer immunotherapy resistance based on immune checkpoints inhibitors: Targets, biomarkers, and remedies. Drug Resist Updat 2020; 53:100718. [PMID: 32736034 DOI: 10.1016/j.drup.2020.100718] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/09/2020] [Accepted: 07/13/2020] [Indexed: 01/05/2023]
Abstract
Cancer is one of the main public health problems in the world. Systemic therapies such as chemotherapy and more recently target therapies as well as immunotherapy have improved the prognosis of this large group of complex malignant diseases. However, the frequent emergence of multidrug resistance (MDR) mechanisms is one of the major impediments towards curative treatment of cancer. While several mechanisms of drug chemoresistance are well defined, resistance to immunotherapy is still insufficiently unclear due to the complexity of the immune response and its dependence on the host. Expression and regulation of immune checkpoint molecules (such as PD-1, CD279; PD-L1, CD274; and CTLA-4, CD152) play a key role in the response to immunotherapy. In this regard, immunotherapy based on immune checkpoints inhibitors (ICIs) is a common clinical approach for treatment of patients with poor prognosis when other first-line therapies have failed. Unfortunately, about 70 % of patients are classified as non-responders, or they progress after initial response to these ICIs. Multiple factors can be related to immunotherapy resistance: characteristics of the tumor microenvironment (TME); presence of tumor infiltrating lymphocytes (TILs), such as CD8 + T cells associated with treatment-response; presence of tumor associated macrophages (TAMs); activation of certain regulators (like PIK3γ or PAX4) found present in non-responders; a low percentage of PD-L1 expressing cells; tumor mutational burden (TMB); gain or loss of antigen-presenting molecules; genetic and epigenetic alterations correlated with resistance. This review provides an update on the current state of immunotherapy resistance presenting targets, biomarkers and remedies to overcome such resistance.
Collapse
Affiliation(s)
- Elisabeth Pérez-Ruiz
- Medical Oncology Department, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Costa del Sol, 29603 Marbella, Malaga, Spain.
| | - Ignacio Melero
- Immunology and Immunotherapy Department, Clinica Universidad de Navarra (CUN), Center for Applied Medical Research (CIMA), Universidad de Navarra (UNAV), 31008 Pamplona, Spain.
| | - Joanna Kopecka
- Department of Oncology, Turin School of Medicine, University of Turin, 10126 Turin, Italy.
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology and University Clinic of Hematology and Coimbra Institute for Clinical and Biomedical Research - Group of Environment Genetics and Oncobiology (iCBR/CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), and Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.
| | - Marilina García-Aranda
- Research Unit, Hospital Costa del Sol, Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Instituto de Investigación Biomédica de Málaga (IBIMA), 29603 Marbella, Malaga, Spain.
| | - Javier De Las Rivas
- Cancer Research Center (CiC-IBMCC, CSIC/USAL), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), and Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain.
| |
Collapse
|
48
|
Gajos-Michniewicz A, Czyz M. WNT Signaling in Melanoma. Int J Mol Sci 2020; 21:E4852. [PMID: 32659938 PMCID: PMC7402324 DOI: 10.3390/ijms21144852] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
WNT-signaling controls important cellular processes throughout embryonic development and adult life, so any deregulation of this signaling can result in a wide range of pathologies, including cancer. WNT-signaling is classified into two categories: β-catenin-dependent signaling (canonical pathway) and β-catenin-independent signaling (non-canonical pathway), the latter can be further divided into WNT/planar cell polarity (PCP) and calcium pathways. WNT ligands are considered as unique directional growth factors that contribute to both cell proliferation and polarity. Origin of cancer can be diverse and therefore tissue-specific differences can be found in WNT-signaling between cancers, including specific mutations contributing to cancer development. This review focuses on the role of the WNT-signaling pathway in melanoma. The current view on the role of WNT-signaling in cancer immunity as well as a short summary of WNT pathway-related drugs under investigation are also provided.
Collapse
Affiliation(s)
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92–215 Lodz, Poland;
| |
Collapse
|
49
|
Timperi E, Barnaba V. Viral Hepatitides, Inflammation and Tumour Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1263:25-43. [PMID: 32588321 DOI: 10.1007/978-3-030-44518-8_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this chapter, we discuss the role of hepatitis B virus (HBV) and hepatitis C virus (HCV) infections in the establishment of hepatocellular carcinoma (HCC), highlighting the key role of the multiple, non-mutually exclusive, pathways involved in the modulation of immune responses and in the orchestration of a chronic low-level inflammation state favouring HCC development. In particular, we discuss (i) HCC as a classical paradigm of inflammation-linked cancer; (ii) the role of the most relevant inflammatory cytokines involved (i.e. IL-6, TNF-α, IL-18, IL-1β, TGF-β IL-10); (iii) the role of T cell exhaustion by immune checkpoints; (iv) the role of the Wnt3a/β-catenin signalling pathway and (v) the role of different subsets of suppressor cells.
Collapse
Affiliation(s)
- Eleonora Timperi
- INSERM U932, Institut Curie, PSL Research University, Paris, France.,Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Vincenzo Barnaba
- INSERM U932, Institut Curie, PSL Research University, Paris, France. .,Istituto Pasteur, Fondazione Cenci Bolognetti, Rome, Italy. .,Center for Life Nano Science, Istituto Italiano di Tecnologia, Rome, Italy.
| |
Collapse
|
50
|
Chang RB, Beatty GL. The interplay between innate and adaptive immunity in cancer shapes the productivity of cancer immunosurveillance. J Leukoc Biol 2020; 108:363-376. [PMID: 32272502 DOI: 10.1002/jlb.3mir0320-475r] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022] Open
Abstract
The immune system is a vital determinant of cancer and shapes its trajectory. Notably, the immune reaction to cancer harbors dual potential for suppressing or promoting cancer development and progression. This polarity of the immune response is determined, in part, by the character of the interplay between innate and adaptive immunity. On the one hand, the innate immune compartment is a necessary proponent of cancer immunity by supporting an immunostimulatory state that enables T cell immunosurveillance. However, in the setting of cancer, innate immune cells are commonly polarized with immune-suppressive properties and as a result, orchestrate a tolerogenic niche that interferes with the cytotoxic potential of tumor antigen-specific T cells. Here, we discuss the role of innate immunity as a positive and negative regulator of adaptive immunosurveillance; moreover, we highlight how tumor cells may skew leukocytes toward an immunosuppressive state and, as such, subvert the phenotypic plasticity of the immune compartment to advance disease progression. These observations establish the precedent for novel therapeutic strategies that aim to restore the tumor microenvironment to an immunoreactive state and, in doing so, condition and maintain the immunogenicity of tumors to yield deep and durable responses to immunotherapy.
Collapse
Affiliation(s)
- Renee B Chang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gregory L Beatty
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|