1
|
Carvalho MI, Silva-Carvalho R, Prada J, Pinto C, Gregório H, Lobo L, Pires I, Queiroga FL. TGFβ in malignant canine mammary tumors: relation with angiogenesis, immunologic markers and prognostic role. Vet Q 2024; 44:1-12. [PMID: 39165025 PMCID: PMC11340227 DOI: 10.1080/01652176.2024.2390941] [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: 01/15/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/22/2024] Open
Abstract
Transforming growth factor-β (TGFβ) and FoxP3 regulatory T cells (Treg) are involved in human breast carcinogenesis. This topic is not well documented in canine mammary tumors (CMT). In this work, the tumoral TGFβ expression was assessed by immunohistochemistry in 67 malignant CMT and its correlation to previously determined FoxP3, VEGF, and CD31 markers and other clinicopathologic parameters was evaluated. The high levels of TGFβ were statistically significantly associated with skin ulceration, tumor necrosis, high histological grade of malignancy (HGM), presence of neoplastic intravascular emboli and presence of lymph node metastases. The observed levels of TGFβ were positively correlated with intratumoral FoxP3 (strong correlation), VEGF (weak correlation) and CD31 (moderate correlation). Tumors that presented a concurrent high expression of TGFβ/FoxP3, TGFβ/VEGF, and TGFβ/CD31 markers were statistically significantly associated with parameters of tumor malignancy (high HGM, presence of vascular emboli and nodal metastasis). Additionally, shorter overall survival (OS) time was statistically significantly associated with tumors with an abundant TGFβ expression and with concurrent high expression of TGFβ/FoxP3, TGFβ/VEGF, and TGFβ/CD31. The presence of lymph node metastasis increased 11 times the risk of disease-related death, arising as an independent predictor of poor prognosis in the multivariable analysis. In conclusion, TGFβ and Treg cells seem involved in tumor progression emerging as potential therapeutic targets for future immunotherapy studies.
Collapse
Affiliation(s)
- Maria Isabel Carvalho
- MVET Research in Veterinary Medicine. Faculty of Veterinary Medicine, Lusófona University – Lisbon Centre, Lisboa, Portugal
| | - Ricardo Silva-Carvalho
- CEB – Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS – Associate Laboratory, Braga, Guimarães, Portugal
| | - Justina Prada
- Veterinary and Animal Research Center (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Carla Pinto
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Hugo Gregório
- Anicura Centro Hospitalar Veterinário, Porto, Portugal
| | - Luis Lobo
- MVET Research in Veterinary Medicine. Faculty of Veterinary Medicine, Lusófona University – Lisbon Centre, Lisboa, Portugal
- Onevet Hospital Veterinário do Porto, Porto, Portugal
- Center for the Study of Animal Sciences, CECA-ICETA, University of Porto, Portugal
| | - Isabel Pires
- Veterinary and Animal Research Center (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Felisbina L. Queiroga
- Veterinary and Animal Research Center (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
- Center for the Study of Animal Sciences, CECA-ICETA, University of Porto, Portugal
| |
Collapse
|
2
|
Shao Y, Han S, Hou Z, Yang C, Zhao Y. Tumor-associated macrophages within the immunological milieu: An emerging focal point for therapeutic intervention. Heliyon 2024; 10:e36839. [PMID: 39281573 PMCID: PMC11401039 DOI: 10.1016/j.heliyon.2024.e36839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024] Open
Abstract
Tumor-associated macrophages play an important role in the tumor immune microenvironment, and regulating the function of tumor-associated macrophages has important therapeutic potential in tumor therapy. Mature macrophages could migrate to the tumor microenvironment, influencing multiple factors such as tumor cell proliferation, invasion, metastasis, extracellular matrix remodeling, immune suppression, and drug resistance. As a major component of the tumor microenvironment, tumor-associated macrophages crosstalk with other immune cells. Currently, tumor-associated macrophages have garnered considerable attention in tumor therapy, broadening the spectrum of drug selection to some extent, thereby aiding in mitigating the prevailing clinical drug resistance dilemma. This article summarizes the recent advances in tumor-associated macrophages concerning immunology, drug targeting mechanisms for tumor-associated macrophages treatment, new developments, and existing challenges, offering insights for future therapeutic approaches. In addition, this paper summarized the impact of tumor-associated macrophages on current clinical therapies, discussed the advantages and disadvantages of targeted tumor-associated macrophages therapy compared with existing tumor therapies, and predicted and discussed the future role of targeted tumor-associated macrophages therapy and the issues that need to be focused on.
Collapse
Affiliation(s)
- Yanchi Shao
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Song Han
- The First Hospital of Jilin University, Changchun, China
| | - Zhenxin Hou
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Chen Yang
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yanbin Zhao
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
3
|
Guo T, Xu J. Cancer-associated fibroblasts: a versatile mediator in tumor progression, metastasis, and targeted therapy. Cancer Metastasis Rev 2024; 43:1095-1116. [PMID: 38602594 PMCID: PMC11300527 DOI: 10.1007/s10555-024-10186-7] [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: 12/27/2023] [Accepted: 03/31/2024] [Indexed: 04/12/2024]
Abstract
Tumor microenvironment (TME) has been demonstrated to play a significant role in tumor initiation, progression, and metastasis. Cancer-associated fibroblasts (CAFs) are the major component of TME and exhibit heterogeneous properties in their communication with tumor cells. This heterogeneity of CAFs can be attributed to various origins, including quiescent fibroblasts, mesenchymal stem cells (MSCs), adipocytes, pericytes, endothelial cells, and mesothelial cells. Moreover, single-cell RNA sequencing has identified diverse phenotypes of CAFs, with myofibroblastic CAFs (myCAFs) and inflammatory CAFs (iCAFs) being the most acknowledged, alongside newly discovered subtypes like antigen-presenting CAFs (apCAFs). Due to these heterogeneities, CAFs exert multiple functions in tumorigenesis, cancer stemness, angiogenesis, immunosuppression, metabolism, and metastasis. As a result, targeted therapies aimed at the TME, particularly focusing on CAFs, are rapidly developing, fueling the promising future of advanced tumor-targeted therapy.
Collapse
Affiliation(s)
- Tianchen Guo
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Junfen Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
4
|
Lahane GP, Dhar A, Bhat A. Therapeutic approaches and novel antifibrotic agents in renal fibrosis: A comprehensive review. J Biochem Mol Toxicol 2024; 38:e23795. [PMID: 39132761 DOI: 10.1002/jbt.23795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 06/20/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024]
Abstract
Renal fibrosis (RF) is one of the underlying pathological conditions leading to progressive loss of renal function and end-stage renal disease (ESRD). Over the years, various therapeutic approaches have been explored to combat RF and prevent ESRD. Despite significant advances in understanding the underlying molecular mechanism(s), effective therapeutic interventions for RF are limited. Current therapeutic strategies primarily target these underlying mechanisms to halt or reverse fibrotic progression. Inhibition of transforming growth factor-β (TGF-β) signaling, a pivotal mediator of RF has emerged as a central strategy to manage RF. Small molecules, peptides, and monoclonal antibodies that target TGF-β receptors or downstream effectors have demonstrated potential in preclinical models. Modulating the renin-angiotensin system and targeting the endothelin system also provide established approaches for controlling fibrosis-related hemodynamic changes. Complementary to pharmacological strategies, lifestyle modifications, and dietary interventions contribute to holistic management. This comprehensive review aims to summarize the underlying mechanisms of RF and provide an overview of the therapeutic strategies and novel antifibrotic agents that hold promise in its treatment.
Collapse
Affiliation(s)
- Ganesh Panditrao Lahane
- Department of Pharmacy, Birla Institute of Technology and Sciences (BITS) Pilani, Hyderabad, Telangana, India
| | - Arti Dhar
- Department of Pharmacy, Birla Institute of Technology and Sciences (BITS) Pilani, Hyderabad, Telangana, India
| | - Audesh Bhat
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu and Kashmir, India
| |
Collapse
|
5
|
Xiang S, Li J, Zhang M. TGF-β1 inhibitor enhances the therapeutic effect of microwave ablation on hepatocellular carcinoma. Int J Hyperthermia 2024; 41:2359496. [PMID: 38909985 DOI: 10.1080/02656736.2024.2359496] [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: 09/18/2023] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Microwave ablation (MWA) is a widely adopted treatment technique for hepatocellular carcinoma (HCC). However, MWA alone is of limited use and has a high recurrence rate. Transforming growth factor-β1 (TGF-β1) is recognized as a potential therapeutic target for HCC patients. Therefore, this study was designed to investigate whether the TGF-β1 inhibitor could increase the efficacy of MWA therapy for HCC treatment. METHODS In vitro, HCC cells challenged with TGF-β1 inhibitor (SB-525334), or normal saline were then heated by microwave. Methyl tetrazolium assays were performed to detect cell survival rate and half-maximal drug inhibitory concentration (IC50). Cell viability and apoptosis were detected by cell counting kit-8 assays, flow cytometry and western blotting. In vivo, the mice injected with HepG2 cells received oral gavage of SB-525334 (20 mg/kg) or normal saline and MWA at a power of 15 W. Tumor volume was recorded. Expression of Ki67 and apoptosis-related proteins were detected by immunohistochemistry and western blotting. TUNEL assays were used to detect cell death ratio. Histopathological changes were examined by hematoxylin and eosin staining. The mechanisms associated with the function of MWA combined with TGF-β1 inhibitor in HCC development were explored by western blotting. RESULTS Combination of MWA and SB-525334 decreased the survival rate and promoted the apoptosis of HCC cells compared with MWA alone. SB-525334 enhanced the suppressive effect of MWA on tumor growth and amplified cell apoptosis. Mechanistically, MWA collaborated with SB-525334 inhibitor inactivated the TGF-β1/Smad2/Smad3 pathway. CONCLUSION TGF-β1 inhibitor enhances the therapeutic effect of MWA on HCC.
Collapse
Affiliation(s)
- Shufang Xiang
- Department of Ultrasound, The First Affiliated Hospital of Yangtze University, Jingzhou City, P. R.China
| | - Juan Li
- Department of Ultrasound, The First Affiliated Hospital of Yangtze University, Jingzhou City, P. R.China
| | - Mei Zhang
- Department of Ultrasound, The First Affiliated Hospital of Yangtze University, Jingzhou City, P. R.China
| |
Collapse
|
6
|
Ma R, Sun JH, Wang YY. The role of transforming growth factor-β (TGF-β) in the formation of exhausted CD8 + T cells. Clin Exp Med 2024; 24:128. [PMID: 38884843 PMCID: PMC11182817 DOI: 10.1007/s10238-024-01394-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024]
Abstract
CD8 + T cells exert a critical role in eliminating cancers and chronic infections, and can provide long-term protective immunity. However, under the exposure of persistent antigen, CD8 + T cells can differentiate into terminally exhausted CD8 + T cells and lose the ability of immune surveillance and disease clearance. New insights into the molecular mechanisms of T-cell exhaustion suggest that it is a potential way to improve the efficacy of immunotherapy by restoring the function of exhausted CD8 + T cells. Transforming growth factor-β (TGF-β) is an important executor of immune homeostasis and tolerance, inhibiting the expansion and function of many components of the immune system. Recent studies have shown that TGF-β is one of the drivers for the development of exhausted CD8 + T cells. In this review, we summarized the role and mechanisms of TGF-β in the formation of exhausted CD8 + T cells and discussed ways to target those to ultimately enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Rong Ma
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
- Cancer Institute, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Jin-Han Sun
- Graduate School, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yan-Yang Wang
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
- Cancer Institute, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
7
|
Arai M, Hanada M, Moriyama H, Ohmoto H, Miyake T, Naka K, Sawa M. Design and synthesis of novel thiazole-derivatives as potent ALK5 inhibitors. Bioorg Med Chem Lett 2024; 108:129797. [PMID: 38759932 DOI: 10.1016/j.bmcl.2024.129797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
TGF-β is an immunosuppressive cytokine and plays a key role in progression of cancer by inducing immunosuppression in tumor microenvironment. Therefore, inhibition of TGF-β signaling pathway may provide a potential therapeutic intervention in treating cancers. Herein, we report the discovery of a series of novel thiazole derivatives as potent inhibitors of ALK5, a serine-threonine kinase which is responsible for TGF-β signal transduction. Compound 29b was identified as a potent inhibitor of ALK5 with an IC50 value of 3.7 nM with an excellent kinase selectivity.
Collapse
Affiliation(s)
- Mai Arai
- Carna Biosciences, Inc., 1-5-5 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Mitsuharu Hanada
- Carna Biosciences, Inc., 1-5-5 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hideki Moriyama
- Carna Biosciences, Inc., 1-5-5 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hiroshi Ohmoto
- Carna Biosciences, Inc., 1-5-5 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Takahiro Miyake
- Carna Biosciences, Inc., 1-5-5 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kazuhito Naka
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Masaaki Sawa
- Carna Biosciences, Inc., 1-5-5 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| |
Collapse
|
8
|
Tian Y, Gao X, Yang X, Chen S, Ren Y. Glioma-derived exosome Lncrna Agap2-As1 promotes glioma proliferation and metastasis by mediating Tgf-β1 secretion of myeloid-derived suppressor cells. Heliyon 2024; 10:e29949. [PMID: 38699039 PMCID: PMC11064146 DOI: 10.1016/j.heliyon.2024.e29949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 04/08/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
Background Glioma (GBM) is the most prevalent malignancy worldwide with high morbidity and mortality. Exosome-mediated transfer of long noncoding RNA (lncRNA) has been reported to be associated with human cancers, containing GBM. Meanwhile, myeloid-derived suppressor cells (MDSCs) play a vital role in mediating the immunosuppressive environments in GBM. Objectives This study is designed to explore the role and mechanism of exosomal (Exo) lncRNA AGAP2-AS1 on the MDSC pathway in GBM. Methods AGAP2-AS1, microRNA-486-3p (miR-486-3p), and Transforming growth factor beta-1 (TGF-β1) levels were detected by real-time quantitative polymerase chain reaction (RT-qPCR). Cell proliferation, apoptosis, migration, and invasion were detected by 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, and Transwell assays. E-cadherin, Vimentin, CD9, CD81, and TGF-β1 protein levels were examined using Western blot. Exosomes were detected by a transmission electron microscope (TEM). Binding between miR-486-3p and AGAP2-AS1 or TGF-β1 was predicted by LncBase or TargetScan and then verified using a dual-luciferase reporter assay. Results AGAP2-AS1 was highly expressed in GBM tissues and cells. Functionally, AGAP2-AS1 absence or TGF-β1 knockdown repressed tumor cell growth and metastasis. Furthermore, Exo-AGAP2-AS1 from GBM cells regulated TGF-β1 expression via sponging miR-486-3p in MDSCs. Exo-AGAP2-AS1 upregulation facilitated GBM cell growth and metastasis via the MDSC pathway. Conclusion Exo-AGAP2-AS1 boosted GBM cell development partly by regulating the MDSC pathway, hinting at a promising therapeutic target for GBM treatment.
Collapse
Affiliation(s)
- Yanlong Tian
- Department of Pathology, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, 712000, Shaanxi, China
| | - Xiao Gao
- Department of Pathology, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, 712000, Shaanxi, China
| | - Xuechao Yang
- Department of Pathology, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, 712000, Shaanxi, China
| | - Shangjun Chen
- Department of Neurosurgery, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, 712000, Shaanxi, China
| | - Yufeng Ren
- Department of Orthopaedics, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, 712000, Shaanxi, China
| |
Collapse
|
9
|
Rapoport BL, Anderson R. Strategies to optimize the promise of checkpoint-targeted anti-cancer therapy. Immunotherapy 2024; 16:565-568. [PMID: 38717385 PMCID: PMC11290365 DOI: 10.1080/1750743x.2024.2343271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/11/2024] [Indexed: 07/26/2024] Open
Affiliation(s)
- Bernardo L Rapoport
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, 0001, South Africa
- Medical Oncology Centre of Rosebank, Johannesburg, 2196, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, 0001, South Africa
| |
Collapse
|
10
|
Kgokolo MCM, Malinga NZ, Steel HC, Meyer PWA, Smit T, Anderson R, Rapoport BL. Transforming growth factor-β1 and soluble co-inhibitory immune checkpoints as putative drivers of immune suppression in patients with basal cell carcinoma. Transl Oncol 2024; 42:101867. [PMID: 38308919 PMCID: PMC10847768 DOI: 10.1016/j.tranon.2023.101867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/30/2023] [Accepted: 12/13/2023] [Indexed: 02/05/2024] Open
Abstract
The current study compared the levels and possible associations between systemic soluble immune checkpoints (sICPs, n = 17) and a group of humoral modulators of immune suppressor cells (n = 7) in a cohort of patients with basal cell carcinoma (BCC, n = 40) and a group of healthy control subjects (n = 20). The seven humoral modulators of immunosuppressor cells were represented by the enzymes, arginase 1 and fibroblast activation protein (FAP), the chemokine, RANTES (CCL5) and the cytokines, interleukin-10 and transforming growth factor-β1 (TGF-β1), as well as the M2-type macrophage markers, soluble CD163 (sCD163) and sCD206. The plasma levels of six co-inhibitory sICPs, sCTLA-4, sLAG-3, sPD-1, sPD-L1, sTIM-3 and sPD-L2 were significantly elevated in the cohort of BCC patients (p<0.001-p<0.00001), while that of sBTLA was significantly decreased (p<0.006). Of the co-stimulatory sICPs, sCD27 and sGITR were significantly increased (p<0.0002 and p<0.0538) in the cohort of BCC patients, while the others were essentially comparable with those of the control participants; of the dual active sICPs, sHVEM was significantly elevated (p<0.00001) and TLR2 comparable with the control group. A correlation heat map revealed selective, strong associations of TGF-β1 with seven co-stimulatory (z = 0.618468-0.768131) and four co-inhibitory (z = 0.674040-0.808365) sICPs, as well as with sTLR2 (z = 0.696431). Notwithstanding the association of BCC with selective elevations in the levels of a large group of co-inhibitory sICPs, our novel findings also imply the probable involvement of TGF-β1 in driving immunosuppression in this malignancy, possibly via activation of regulatory T cells. Notably, these abnormalities were present in patients with either newly diagnosed or recurrent disease.
Collapse
Affiliation(s)
- Mahlatse C M Kgokolo
- Department of Dermatology, School of Medicine, Faculty of Health Sciences, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa.
| | - Nonkululeko Z Malinga
- Department of Dermatology, School of Medicine, Faculty of Health Sciences, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
| | - Helen C Steel
- Department of Immunology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Pieter W A Meyer
- Department of Immunology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; Tshwane Academic Division of the National Health Laboratory Service, Pretoria, South Africa
| | - Teresa Smit
- The Medical Oncology Centre of Rosebank, Saxonwold, Johannesburg, South Africa
| | - Ronald Anderson
- Department of Immunology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Bernardo L Rapoport
- Department of Immunology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; The Medical Oncology Centre of Rosebank, Saxonwold, Johannesburg, South Africa.
| |
Collapse
|
11
|
Katsimperis S, Kapriniotis K, Manolitsis I, Bellos T, Angelopoulos P, Juliebø-Jones P, Somani B, Skolarikos A, Tzelves L. Early investigational agents for the treatment of benign prostatic hyperplasia'. Expert Opin Investig Drugs 2024; 33:359-370. [PMID: 38421373 DOI: 10.1080/13543784.2024.2326023] [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: 10/25/2023] [Accepted: 02/28/2024] [Indexed: 03/02/2024]
Abstract
INTRODUCTION Benign prostatic hyperplasia (BPH), as a clinical entity that affects many people, has always been in the forefront of interest among researchers, pharmaceutical companies, and physicians. Patients with BPH exhibit a diverse range of symptoms, while current treatment options can occasionally cause adverse events. All the aforementioned have led to an increased demand for more effective treatment options. AREAS COVERED This review summarizes the outcomes of new medications used in a pre-clinical and clinical setting for the management of male lower urinary tract symptoms (LUTS)/BPH and provides information about ongoing trials and future directions in the management of this condition. More specifically, sheds light upon drug categories, such as reductase‑adrenoceptor antagonists, drugs interfering with the nitric oxide (NO)/cyclic guanosine monophosphate (GMP) signaling pathway, onabotulinumtoxinA, vitamin D3 (calcitriol) analogues, selective cannabinoid (CB) receptor agonists, talaporfin sodium, inhibitor of transforming growth factor beta 1 (TGF-β1), drugs targeting the hormonal control of the prostate, phytotherapy, and many more. EXPERT OPINION Clinical trials are being conducted on a number of new medications that may emerge as effective therapeutic alternatives in the coming years.
Collapse
Affiliation(s)
- Stamatios Katsimperis
- 2nd University Department of Urology, Sismanoglio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Ioannis Manolitsis
- 2nd University Department of Urology, Sismanoglio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Themistoklis Bellos
- 2nd University Department of Urology, Sismanoglio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Angelopoulos
- 2nd University Department of Urology, Sismanoglio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Patrick Juliebø-Jones
- Department of Urology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Bhaskar Somani
- Department of Urology, University Hospital Southampton, Southampton, UK
| | - Andreas Skolarikos
- 2nd University Department of Urology, Sismanoglio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Lazaros Tzelves
- 2nd University Department of Urology, Sismanoglio Hospital, National and Kapodistrian University of Athens, Athens, Greece
- Department of Urology, University College of London Hospitals (UCLH), London, UK
| |
Collapse
|
12
|
Kim W, Ye Z, Simonenko V, Shahi A, Malikzay A, Long S, Xu JJ, Lu A, Horng JH, Wu CR, Chen PJ, Lu P, Evans DM. Codelivery of TGFβ and Cox2 siRNA inhibits HCC by promoting T-cell penetration into the tumor and improves response to Immune Checkpoint Inhibitors. NAR Cancer 2024; 6:zcad059. [PMID: 38204925 PMCID: PMC10776204 DOI: 10.1093/narcan/zcad059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/11/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Upregulation of TGFβ and Cox2 in the tumor microenvironment results in blockade of T-cell penetration into the tumor. Without access to tumor antigens, the T-cell response will not benefit from administration of the immune checkpoint antibodies. We created an intravenous polypeptide nanoparticle that can deliver two siRNAs (silencing TGFβ and Cox2). Systemic administration in mice, bearing a syngeneic orthotopic hepatocellular carcinoma (HCC), delivers the siRNAs to various cells in the liver, and significantly reduces the tumor. At 2 mg/kg (BIW) the nanoparticle demonstrated a single agent action and induced tumor growth inhibition to undetectable levels after five doses. Reducing the siRNAs to 1mg/kg BIW demonstrated greater inhibition in the presence of PD-L1 mAbs. After only three doses BIW, we could still recover a smaller tumor and, in tumor sections, showed an increase in penetration of CD4+ and CD8+ T-cells deeper into the remaining tumor that was not evident in animals treated with non-silencing siRNA. The combination of TGFβ and Cox2 siRNA co-administered in a polypeptide nanoparticle can act as a novel therapeutic alone against HCC and may augment the activity of the immune checkpoint antibodies. Silencing TGFβ and Cox2 converts an immune excluded (cold) tumor into a T-cell inflamed (hot) tumor.
Collapse
Affiliation(s)
- Wookhyun Kim
- Sirnaomics Inc., 20511 Seneca Meadows Parkway, Suite 200, Germantown, MD 20876, USA
| | - Zhou Ye
- Sirnaomics Inc., 20511 Seneca Meadows Parkway, Suite 200, Germantown, MD 20876, USA
| | - Vera Simonenko
- Sirnaomics Inc., 20511 Seneca Meadows Parkway, Suite 200, Germantown, MD 20876, USA
| | - Aashirwad Shahi
- Sirnaomics Inc., 20511 Seneca Meadows Parkway, Suite 200, Germantown, MD 20876, USA
| | - Asra Malikzay
- Sirnaomics Inc., 20511 Seneca Meadows Parkway, Suite 200, Germantown, MD 20876, USA
| | - Steven Z Long
- Sirnaomics Inc., 20511 Seneca Meadows Parkway, Suite 200, Germantown, MD 20876, USA
| | - John J Xu
- Suzhou Sirnaomics Pharmaceuticals, Ltd., Biobay, Suzhou, China
| | - Alan Lu
- Sirnaomics Inc., 20511 Seneca Meadows Parkway, Suite 200, Germantown, MD 20876, USA
| | - Jau-Hau Horng
- National Taiwan University College of Medicine, No. 1, Section 1, Ren’ai Rd, Zhongzheng District, Taipei City 100, Taiwan
| | - Chang-Ru Wu
- National Taiwan University College of Medicine, No. 1, Section 1, Ren’ai Rd, Zhongzheng District, Taipei City 100, Taiwan
| | - Pei-Jer Chen
- National Taiwan University College of Medicine, No. 1, Section 1, Ren’ai Rd, Zhongzheng District, Taipei City 100, Taiwan
| | - Patrick Y Lu
- Sirnaomics Inc., 20511 Seneca Meadows Parkway, Suite 200, Germantown, MD 20876, USA
| | - David M Evans
- Sirnaomics Inc., 20511 Seneca Meadows Parkway, Suite 200, Germantown, MD 20876, USA
| |
Collapse
|
13
|
Rodrigues DB, Reis RL, Pirraco RP. Modelling the complex nature of the tumor microenvironment: 3D tumor spheroids as an evolving tool. J Biomed Sci 2024; 31:13. [PMID: 38254117 PMCID: PMC10804490 DOI: 10.1186/s12929-024-00997-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer remains a serious burden in society and while the pace in the development of novel and more effective therapeutics is increasing, testing platforms that faithfully mimic the tumor microenvironment are lacking. With a clear shift from animal models to more complex in vitro 3D systems, spheroids emerge as strong options in this regard. Years of development have allowed spheroid-based models to better reproduce the biomechanical cues that are observed in the tumor-associated extracellular matrix (ECM) and cellular interactions that occur in both a cell-cell and cell-ECM manner. Here, we summarize some of the key cellular interactions that drive tumor development, progression and invasion, and how successfully are these interactions recapitulated in 3D spheroid models currently in use in the field. We finish by speculating on future advancements in the field and on how these can shape the relevance of spherical 3D models for tumor modelling.
Collapse
Affiliation(s)
- Daniel B Rodrigues
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, 4805-017, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, 4805-017, Guimarães, Portugal
| | - Rogério P Pirraco
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal.
- ICVS/3B's, PT Government Associate Laboratory, Braga, 4805-017, Guimarães, Portugal.
| |
Collapse
|
14
|
Richardson L, Wilcockson SG, Guglielmi L, Hill CS. Context-dependent TGFβ family signalling in cell fate regulation. Nat Rev Mol Cell Biol 2023; 24:876-894. [PMID: 37596501 DOI: 10.1038/s41580-023-00638-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2023] [Indexed: 08/20/2023]
Abstract
The transforming growth factor-β (TGFβ) family are a large group of evolutionarily conserved cytokines whose signalling modulates cell fate decision-making across varying cellular contexts at different stages of life. Here we discuss new findings in early embryos that reveal how, in contrast to our original understanding of morphogen interpretation, robust cell fate specification can originate from a noisy combination of signalling inputs and a broad range of signalling levels. We compare this evidence with novel findings on the roles of TGFβ family signalling in tissue maintenance and homeostasis during juvenile and adult life, spanning the skeletal, haemopoietic and immune systems. From these comparisons, it emerges that in contrast to robust developing systems, relatively small perturbations in TGFβ family signalling have detrimental effects at later stages in life, leading to aberrant cell fate specification and disease, for example in cancer or congenital disorders. Finally, we highlight novel strategies to target and amend dysfunction in signalling and discuss how gleaning knowledge from different fields of biology can help in the development of therapeutics for aberrant TGFβ family signalling in disease.
Collapse
Affiliation(s)
- Louise Richardson
- Developmental Signalling Laboratory, The Francis Crick Institute, London, UK
| | - Scott G Wilcockson
- Developmental Signalling Laboratory, The Francis Crick Institute, London, UK
| | - Luca Guglielmi
- Developmental Signalling Laboratory, The Francis Crick Institute, London, UK
- Division of Cell Biology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Caroline S Hill
- Developmental Signalling Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
15
|
Alzamami A. Implications of single-cell immune landscape of tumor microenvironment for the colorectal cancer diagnostics and therapy. Med Oncol 2023; 40:352. [PMID: 37950801 DOI: 10.1007/s12032-023-02226-z] [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: 09/11/2023] [Accepted: 10/18/2023] [Indexed: 11/13/2023]
Abstract
Colorectal cancer (CRC) originates from the polyps lining the colon and is among the most common types of cancer. With the increasing popularity of single-cell sequencing technologies, researchers have been able to better understand the immune landscape of colorectal cancer, by analyzing their expression and interactions in detail with the tumor microenvironment (TME) at single-cell level. Since the tumor-immune cell interactions play a critical part in the advancement as well as treatment response in colorectal cancer, the release of inhibitory factors such as T cells are important for recognizing and destroying cancer cells. Such information is vital to identify immunotherapeutic targets for cure and monitoring response to treatments. Therefore, a comprehensive single-cell studies-based overview of key immunogenic agents regulating the TME of CRC is provided in this review. Tumor-associated macrophages can promote tumor growth and resistance to treatment by releasing factors that inhibit the function of other immune cells. Additionally, colorectal cancer cells can express programmed cell death protein 1 and its ligand, which can also inhibit T-cell function. Researchers have found that certain types of immune cells, prominently T cells, natural killer, and dendritic cells, can have a positive impact on the prognosis of colorectal cancer patients. Treatments like immune checkpoint inhibitors and CAR-T therapies that help to release the inhibitory signals from the cancer cells allow the immune cells to function more effectively.
Collapse
Affiliation(s)
- Ahmad Alzamami
- Clinical Laboratory Science Department, College of Applied Medical Sciences, Shaqra University, 11961, Al-Quwayiyah, Saudi Arabia.
| |
Collapse
|
16
|
Ravassa S, López B, Treibel TA, San José G, Losada-Fuentenebro B, Tapia L, Bayés-Genís A, Díez J, González A. Cardiac Fibrosis in heart failure: Focus on non-invasive diagnosis and emerging therapeutic strategies. Mol Aspects Med 2023; 93:101194. [PMID: 37384998 DOI: 10.1016/j.mam.2023.101194] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 07/01/2023]
Abstract
Heart failure is a leading cause of mortality and hospitalization worldwide. Cardiac fibrosis, resulting from the excessive deposition of collagen fibers, is a common feature across the spectrum of conditions converging in heart failure. Eventually, either reparative or reactive in nature, in the long-term cardiac fibrosis contributes to heart failure development and progression and is associated with poor clinical outcomes. Despite this, specific cardiac antifibrotic therapies are lacking, making cardiac fibrosis an urgent unmet medical need. In this context, a better patient phenotyping is needed to characterize the heterogenous features of cardiac fibrosis to advance toward its personalized management. In this review, we will describe the different phenotypes associated with cardiac fibrosis in heart failure and we will focus on the potential usefulness of imaging techniques and circulating biomarkers for the non-invasive characterization and phenotyping of this condition and for tracking its clinical impact. We will also recapitulate the cardiac antifibrotic effects of existing heart failure and non-heart failure drugs and we will discuss potential strategies under preclinical development targeting the activation of cardiac fibroblasts at different levels, as well as targeting additional extracardiac processes.
Collapse
Affiliation(s)
- Susana Ravassa
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Begoña López
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Thomas A Treibel
- Institute of Cardiovascular Science, University College London, UK; Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - Gorka San José
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Blanca Losada-Fuentenebro
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Leire Tapia
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Antoni Bayés-Genís
- CIBERCV, Carlos III Institute of Health, Madrid, Spain; Servei de Cardiologia i Unitat d'Insuficiència Cardíaca, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain; ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Badalona, Spain
| | - Javier Díez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain.
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain.
| |
Collapse
|
17
|
Anderson R, Rapoport BL, Steel HC, Theron AJ. Pro-Tumorigenic and Thrombotic Activities of Platelets in Lung Cancer. Int J Mol Sci 2023; 24:11927. [PMID: 37569299 PMCID: PMC10418868 DOI: 10.3390/ijms241511927] [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: 06/20/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/13/2023] Open
Abstract
Aside from their key protective roles in hemostasis and innate immunity, platelets are now recognized as having multifaceted, adverse roles in the pathogenesis, progression and outcome of many types of human malignancy. The most consistent and compelling evidence in this context has been derived from the notable association of elevated circulating platelet counts with the onset and prognosis of various human malignancies, particularly lung cancer, which represents the primary focus of the current review. Key topics include an overview of the association of lung cancer with the circulating platelet count, as well as the mechanisms of platelet-mediated, pro-tumorigenic immunosuppression, particularly the role of transforming growth factor beta 1. These issues are followed by a discussion regarding the pro-tumorigenic role of platelet-derived microparticles (PMPs), the most abundant type of microparticles (MPs) in human blood. In this context, the presence of increased levels of PMPs in the blood of lung cancer patients has been associated with tumor growth, invasion, angiogenesis and metastasis, which correlate with disease progression and decreased survival times. The final section of the review addresses, firstly, the role of cancer-related platelet activation and thrombosis in the pathogenesis of secondary cardiovascular disorders and the associated mortality, particularly in lung cancer, which is second only to disease progression; secondly, the review addresses the potential role of antiplatelet agents in the adjunctive therapy of cancer.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (B.L.R.); (H.C.S.); (A.J.T.)
| | - Bernardo L. Rapoport
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (B.L.R.); (H.C.S.); (A.J.T.)
- The Medical Oncology Centre of Rosebank, Johannesburg 2196, South Africa
| | - Helen C. Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (B.L.R.); (H.C.S.); (A.J.T.)
| | - Annette J. Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (B.L.R.); (H.C.S.); (A.J.T.)
| |
Collapse
|
18
|
Aoki K, Nishito Y, Motoi N, Arai Y, Hiraoka N, Shibata T, Sonobe Y, Kayukawa Y, Hashimoto E, Takahashi M, Fujii E, Nishizawa T, Fukuda H, Ohashi K, Arai K, Mizoguchi Y, Yoshida Y, Watanabe SI, Yamashita M, Kitano S, Sakamoto H, Nagata Y, Mitsumori R, Ozaki K, Niida S, Kanai Y, Hirayama A, Soga T, Maruyama T, Tsukada K, Yabuki N, Shimada M, Kitazawa T, Natori O, Sawada N, Kato A, Yoshida T, Yasuda K, Mizuno H, Tsunoda H, Ochiai A. Tumor-infiltrating Leukocyte Profiling Defines Three Immune Subtypes of NSCLC with Distinct Signaling Pathways and Genetic Alterations. CANCER RESEARCH COMMUNICATIONS 2023; 3:1026-1040. [PMID: 37377611 PMCID: PMC10263066 DOI: 10.1158/2767-9764.crc-22-0415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/02/2023] [Accepted: 05/17/2023] [Indexed: 06/29/2023]
Abstract
Resistance to immune checkpoint blockade remains challenging in patients with non-small cell lung cancer (NSCLC). Tumor-infiltrating leukocyte (TIL) quantity, composition, and activation status profoundly influence responsiveness to cancer immunotherapy. This study examined the immune landscape in the NSCLC tumor microenvironment by analyzing TIL profiles of 281 fresh resected NSCLC tissues. Unsupervised clustering based on numbers and percentages of 30 TIL types classified adenocarcinoma (LUAD) and squamous cell carcinoma (LUSQ) into the cold, myeloid cell-dominant, and CD8+ T cell-dominant subtypes. These were significantly correlated with patient prognosis; the myeloid cell subtype had worse outcomes than the others. Integrated genomic and transcriptomic analyses, including RNA sequencing, whole-exome sequencing, T-cell receptor repertoire, and metabolomics of tumor tissue, revealed that immune reaction-related signaling pathways were inactivated, while the glycolysis and K-ras signaling pathways activated in LUAD and LUSQ myeloid cell subtypes. Cases with ALK and ROS1 fusion genes were enriched in the LUAD myeloid subtype, and the frequency of TERT copy-number variations was higher in LUSQ myeloid subtype than in the others. These classifications of NSCLC based on TIL status may be useful for developing personalized immune therapies for NSCLC. Significance The precise TIL profiling classified NSCLC into novel three immune subtypes that correlates with patient outcome, identifying subtype-specific molecular pathways and genomic alterations that should play important roles in constructing subtype-specific immune tumor microenvironments. These classifications of NSCLC based on TIL status are useful for developing personalized immune therapies for NSCLC.
Collapse
Affiliation(s)
- Kazunori Aoki
- Department of Immune Medicine, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Yukari Nishito
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Noriko Motoi
- Department of Diagnostic Pathology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Yasuhito Arai
- Divison of Cancer Genomics, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Nobuyoshi Hiraoka
- Department of Analytical Pathology, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Tatsuhiro Shibata
- Divison of Cancer Genomics, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Yukiko Sonobe
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Yoko Kayukawa
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Eri Hashimoto
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Mina Takahashi
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Etsuko Fujii
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Takashi Nishizawa
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Hironori Fukuda
- Department of Immune Medicine, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Kana Ohashi
- Department of Immune Medicine, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Kosuke Arai
- Department of Immune Medicine, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Yukihiro Mizoguchi
- Department of Immune Medicine, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Yukihiro Yoshida
- Department of Thoracic Surgery, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Shun-ichi Watanabe
- Department of Thoracic Surgery, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Makiko Yamashita
- Advanced Medical Development Center, Cancer Research Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| | - Shigehisa Kitano
- Advanced Medical Development Center, Cancer Research Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| | - Hiromi Sakamoto
- Department of Clinical Genomics, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Yuki Nagata
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
- Bioresource Research Center, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Risa Mitsumori
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kouichi Ozaki
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Shumpei Niida
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Yae Kanai
- Department of Pathology, School of Medicine, Keio University, Sinjyuku-ku, Tokyo, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University Tsuruoka, Yamagata, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University Tsuruoka, Yamagata, Japan
| | - Toru Maruyama
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Keisuke Tsukada
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Nami Yabuki
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Mei Shimada
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Takehisa Kitazawa
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Osamu Natori
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Noriaki Sawada
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Atsuhiko Kato
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Teruhiko Yoshida
- Department of Genetic Medicine and Services, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Kazuki Yasuda
- National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hideaki Mizuno
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Hiroyuki Tsunoda
- Kamakura Research Laboratories, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Atsushi Ochiai
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| |
Collapse
|
19
|
Nixon BG, Gao S, Wang X, Li MO. TGFβ control of immune responses in cancer: a holistic immuno-oncology perspective. Nat Rev Immunol 2023; 23:346-362. [PMID: 36380023 PMCID: PMC10634249 DOI: 10.1038/s41577-022-00796-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2022] [Indexed: 11/16/2022]
Abstract
The immune system responds to cancer in two main ways. First, there are prewired responses involving myeloid cells, innate lymphocytes and innate-like adaptive lymphocytes that either reside in premalignant tissues or migrate directly to tumours, and second, there are antigen priming-dependent responses, in which adaptive lymphocytes are primed in secondary lymphoid organs before homing to tumours. Transforming growth factor-β (TGFβ) - one of the most potent and pleiotropic regulatory cytokines - controls almost every stage of the tumour-elicited immune response, from leukocyte development in primary lymphoid organs to their priming in secondary lymphoid organs and their effector functions in the tumour itself. The complexity of TGFβ-regulated immune cell circuitries, as well as the contextual roles of TGFβ signalling in cancer cells and tumour stromal cells, necessitates the use of rigorous experimental systems that closely recapitulate human cancer, such as autochthonous tumour models, to uncover the underlying immunobiology. The diverse functions of TGFβ in healthy tissues further complicate the search for effective and safe cancer therapeutics targeting the TGFβ pathway. Here we discuss the contextual complexity of TGFβ signalling in tumour-elicited immune responses and explain how understanding this may guide the development of mechanism-based cancer immunotherapy.
Collapse
Affiliation(s)
- Briana G Nixon
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School of Biomedical Sciences, Cornell University, New York, NY, USA
| | - Shengyu Gao
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xinxin Wang
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School of Biomedical Sciences, Cornell University, New York, NY, USA
| | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School of Biomedical Sciences, Cornell University, New York, NY, USA.
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
20
|
Chang J, Lo ZHY, Alenizi S, Kovacevic Z. Re-Shaping the Pancreatic Cancer Tumor Microenvironment: A New Role for the Metastasis Suppressor NDRG1. Cancers (Basel) 2023; 15:2779. [PMID: 37345116 DOI: 10.3390/cancers15102779] [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: 04/03/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 06/23/2023] Open
Abstract
Pancreatic cancer (PaC) is a highly aggressive disease, with poor response to current treatments and 5-year survival rates of 10-15%. PaC progression is facilitated by its interaction with the complex and multifaceted tumor microenvironment (TME). In the TME, cancer cells and surrounding stromal cells constantly communicate with each other via the secretion and uptake of factors including cytokines, chemokines, growth factors, metabolites, and extracellular vesicles (EVs), reshaping the landscape of PaC. Recent studies demonstrated that the metastasis suppressor N-myc downstream regulated 1 (NDRG1) not only inhibits oncogenic signaling pathways in PaC cells but also alters the communication between PaC cells and the surrounding stroma. In fact, NDRG1 was found to influence the secretome of PaC cells, alter cancer cell metabolism, and interfere with intracellular trafficking and intercellular communication between PaC cells and surrounding fibroblasts. This review will present recent advancements in understanding the role of NDRG1 in PaC progression, with a focus on how this molecule influences PaC-stroma communication and its potential for re-shaping the PaC TME.
Collapse
Affiliation(s)
- Jiawei Chang
- School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Sydney 2006, Australia
- Department of Physiology, School of Biomedical Sciences, Faculty of Medicine & Health, University of NSW, Sydney 2052, Australia
| | - Zoe H Y Lo
- School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Sydney 2006, Australia
| | - Shafi Alenizi
- School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Sydney 2006, Australia
| | - Zaklina Kovacevic
- School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Sydney 2006, Australia
- Department of Physiology, School of Biomedical Sciences, Faculty of Medicine & Health, University of NSW, Sydney 2052, Australia
| |
Collapse
|
21
|
Xiao H, Feng X, Liu M, Gong H, Zhou X. SnoRNA and lncSNHG: Advances of nucleolar small RNA host gene transcripts in anti-tumor immunity. Front Immunol 2023; 14:1143980. [PMID: 37006268 PMCID: PMC10050728 DOI: 10.3389/fimmu.2023.1143980] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
The small nucleolar RNA host genes (SNHGs) are a group of genes that can be transcript into long non-coding RNA SNHG (lncSNHG) and further processed into small nucleolar RNAs (snoRNAs). Although lncSNHGs and snoRNAs are well established to play pivotal roles in tumorigenesis, how lncSNHGs and snoRNAs regulate the immune cell behavior and function to mediate anti-tumor immunity remains further illustrated. Certain immune cell types carry out distinct roles to participate in each step of tumorigenesis. It is particularly important to understand how lncSNHGs and snoRNAs regulate the immune cell function to manipulate anti-tumor immunity. Here, we discuss the expression, mechanism of action, and potential clinical relevance of lncSNHGs and snoRNAs in regulating different types of immune cells that are closely related to anti-tumor immunity. By uncovering the changes and roles of lncSNHGs and snoRNAs in different immune cells, we aim to provide a better understanding of how the transcripts of SNHGs participate in tumorigenesis from an immune perspective.
Collapse
Affiliation(s)
- Hao Xiao
- Department of Clinical Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Clinical Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xin Feng
- Department of Clinical Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Clinical Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mengjun Liu
- Department of Clinical Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Clinical Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hanwen Gong
- Department of Clinical Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Clinical Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiao Zhou
- Department of Clinical Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Clinical Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- *Correspondence: Xiao Zhou,
| |
Collapse
|
22
|
Interactions between Platelets and Tumor Microenvironment Components in Ovarian Cancer and Their Implications for Treatment and Clinical Outcomes. Cancers (Basel) 2023; 15:cancers15041282. [PMID: 36831623 PMCID: PMC9953912 DOI: 10.3390/cancers15041282] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Platelets, the primary operatives of hemostasis that contribute to blood coagulation and wound healing after blood vessel injury, are also involved in pathological conditions, including cancer. Malignancy-associated thrombosis is common in ovarian cancer patients and is associated with poor clinical outcomes. Platelets extravasate into the tumor microenvironment in ovarian cancer and interact with cancer cells and non-cancerous elements. Ovarian cancer cells also activate platelets. The communication between activated platelets, cancer cells, and the tumor microenvironment is via various platelet membrane proteins or mediators released through degranulation or the secretion of microvesicles from platelets. These interactions trigger signaling cascades in tumors that promote ovarian cancer progression, metastasis, and neoangiogenesis. This review discusses how interactions between platelets, cancer cells, cancer stem cells, stromal cells, and the extracellular matrix in the tumor microenvironment influence ovarian cancer progression. It also presents novel potential therapeutic approaches toward this gynecological cancer.
Collapse
|
23
|
van Weverwijk A, de Visser KE. Mechanisms driving the immunoregulatory function of cancer cells. Nat Rev Cancer 2023; 23:193-215. [PMID: 36717668 DOI: 10.1038/s41568-022-00544-4] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/13/2022] [Indexed: 01/31/2023]
Abstract
Tumours display an astonishing variation in the spatial distribution, composition and activation state of immune cells, which impacts their progression and response to immunotherapy. Shedding light on the mechanisms that govern the diversity and function of immune cells in the tumour microenvironment will pave the way for the development of more tailored immunomodulatory strategies for the benefit of patients with cancer. Cancer cells, by virtue of their paracrine and juxtacrine communication mechanisms, are key contributors to intertumour heterogeneity in immune contextures. In this Review, we discuss how cancer cell-intrinsic features, including (epi)genetic aberrations, signalling pathway deregulation and altered metabolism, play a key role in orchestrating the composition and functional state of the immune landscape, and influence the therapeutic benefit of immunomodulatory strategies. Moreover, we highlight how targeting cancer cell-intrinsic parameters or their downstream immunoregulatory pathways is a viable strategy to manipulate the tumour immune milieu in favour of antitumour immunity.
Collapse
Affiliation(s)
- Antoinette van Weverwijk
- Division of Tumour Biology & Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Karin E de Visser
- Division of Tumour Biology & Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands.
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands.
| |
Collapse
|
24
|
Intratumoral pro-oxidants promote cancer immunotherapy by recruiting and reprogramming neutrophils to eliminate tumors. Cancer Immunol Immunother 2023; 72:527-542. [PMID: 36066649 PMCID: PMC9446783 DOI: 10.1007/s00262-022-03248-8] [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: 01/04/2022] [Accepted: 06/23/2022] [Indexed: 11/06/2022]
Abstract
Neutrophils have recently gained recognition for their potential in the fight against cancer. Neutrophil plasticity between the N1 anti-tumor and N2 pro-tumor subtypes is now apparent, as is the ability to polarize these individual subtypes by interventions such as intratumoral injection of various agents including bacterial products or pro-oxidants. Metabolic responses and the production of reactive oxygen species (ROS) such as hydrogen peroxide act as potent chemoattractants and activators of N1 neutrophils that facilitates their recruitment and ensuing activation of a toxic respiratory burst in tumors. Greater understanding of the precise mechanism of N1 neutrophil activation, recruitment and regulation is now needed to fully exploit their anti-tumor potential against cancers both locally and at distant sites. This systematic review critically analyzes these new developments in cancer immunotherapy.
Collapse
|
25
|
Roelands J, van der Ploeg M, Ijsselsteijn ME, Dang H, Boonstra JJ, Hardwick JCH, Hawinkels LJAC, Morreau H, de Miranda NFCC. Transcriptomic and immunophenotypic profiling reveals molecular and immunological hallmarks of colorectal cancer tumourigenesis. Gut 2022:gutjnl-2022-327608. [PMID: 36442992 DOI: 10.1136/gutjnl-2022-327608] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 11/12/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Biological insights into the stepwise development and progression of colorectal cancer (CRC) are imperative to develop tailored approaches for early detection and optimal clinical management of this disease. Here, we aimed to dissect the transcriptional and immunologic alterations that accompany malignant transformation in CRC and to identify clinically relevant biomarkers through spatial profiling of pT1 CRC samples. DESIGN We employed digital spatial profiling (GeoMx) on eight pT1 CRCs to study gene expression in the epithelial and stromal segments across regions of distinct histology, including normal mucosa, low-grade and high-grade dysplasia and cancer. Consecutive histology sections were profiled by imaging mass cytometry to reveal immune contextures. Finally, publicly available single-cell RNA-sequencing data was analysed to determine the cellular origin of relevant transcripts. RESULTS Comparison of gene expression between regions within pT1 CRC samples identified differentially expressed genes in the epithelium (n=1394 genes) and the stromal segments (n=1145 genes) across distinct histologies. Pathway analysis identified an early onset of inflammatory responses during malignant transformation, typified by upregulation of gene signatures such as innate immune sensing. We detected increased infiltration of myeloid cells and a shift in macrophage populations from pro-inflammatory HLA-DR+CD204- macrophages to HLA-DR-CD204+ immune-suppressive subsets from normal tissue through dysplasia to cancer, accompanied by the upregulation of the CD47/SIRPα 'don't eat me signal'. CONCLUSION Spatial profiling revealed the molecular and immunological landscape of CRC tumourigenesis at early disease stage. We identified biomarkers with strong association with disease progression as well as targetable immune processes that are exploitable in a clinical setting.
Collapse
Affiliation(s)
- Jessica Roelands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Manon van der Ploeg
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Hao Dang
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jurjen J Boonstra
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - James C H Hardwick
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lukas J A C Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
26
|
Shao X, Hua S, Feng T, Ocansey DKW, Yin L. Hypoxia-Regulated Tumor-Derived Exosomes and Tumor Progression: A Focus on Immune Evasion. Int J Mol Sci 2022; 23:ijms231911789. [PMID: 36233088 PMCID: PMC9570495 DOI: 10.3390/ijms231911789] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/24/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor cells express a high quantity of exosomes packaged with unique cargos under hypoxia, an important characteristic feature in solid tumors. These hypoxic tumor-derived exosomes are, crucially, involved in the interaction of cancer cells with their microenvironment, facilitating not only immune evasion, but increased cell growth and survival, enhanced angiogenesis, epithelial–mesenchymal transition (EMT), therapeutic resistance, autophagy, pre-metastasis, and metastasis. This paper explores the tumor microenvironment (TME) remodeling effects of hypoxic tumor-derived exosome towards facilitating the tumor progression process, particularly, the modulatory role of these factors on tumor cell immune evasion through suppression of immune cells, expression of surface recognition molecules, and secretion of antitumor soluble factor. Tumor-expressed exosomes educate immune effector cells, including macrophages, monocytes, T cells, natural killer (NK) cells, dendritic cells (DCs), γδ T lymphocytes, regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), mast cells, and B cells, within the hypoxic TME through the release of factors that regulate their recruitment, phenotype, and function. Thus, both hypoxia and tumor-derived exosomes modulate immune cells, growth factors, cytokines, receptor molecules, and other soluble factors, which, together, collaborate to form the immune-suppressive milieu of the tumor environment. Exploring the contribution of exosomal cargos, such as RNAs and proteins, as indispensable players in the cross-talk within the hypoxic tumor microenvironmental provides a potential target for antitumor immunity or subverting immune evasion and enhancing tumor therapies.
Collapse
|
27
|
Anti-TGF-β1 aptamer enhances therapeutic effect of tyrosine kinase inhibitor, gefitinib, on non-small cell lung cancer in xenograft model. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 29:969-978. [PMID: 36189081 PMCID: PMC9481871 DOI: 10.1016/j.omtn.2022.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 06/07/2022] [Indexed: 11/30/2022]
Abstract
Transforming growth factor β (TGF-β) is a multifunctional cytokine that plays crucial pathophysiological roles in various diseases, such as cancer and fibrosis. However, the disease modulation by targeting TGF-β1 isoform remains to be established, regardless of several studies employed with limited antibodies. Here, we developed an RNA aptamer to human active TGF-β1, named APT-β1, and characterized its properties in vitro and in vivo. APT-β1 bound to human and mouse active TGF-β1 proteins with high affinity and specificity and strongly inhibited TGF-β1-induced downstream signaling and cell morphology with 50% inhibition concentration (IC50) values at picomolar concentrations. In a xenograft mouse model of non-small cell lung cancer, APT-β1 alone showed no appreciable effect on tumor growth, while it greatly enhanced the anti-tumor effect of gefitinib, an approved tyrosine kinase inhibitor. These findings strongly suggest that the anti-TGF-β1 medication may be a promising cancer therapy to suppress repopulation of lung cancer in combination with certain anti-cancer drugs, such as gefitinib.
Collapse
|
28
|
Li L, Wen Q, Ding R. Therapeutic targeting of VEGF and/or TGF-β to enhance anti-PD-(L)1 therapy: The evidence from clinical trials. Front Oncol 2022; 12:905520. [PMID: 35957885 PMCID: PMC9360509 DOI: 10.3389/fonc.2022.905520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/30/2022] [Indexed: 11/19/2022] Open
Abstract
Normalizing the tumor microenvironment (TME) is a potential strategy to improve the effectiveness of immunotherapy. Vascular endothelial growth factor (VEGF) and transforming growth factor (TGF)-β pathways play an important role in the development and function of the TME, contributing to the immunosuppressive status of TME. To inhibit VEGF and/or TGF-β pathways can restore TME from immunosuppressive to immune-supportive status and enhance sensitivity to immunotherapy such as programmed death protein-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) inhibitors. In this review, we described the existing preclinical and clinical evidence supporting the use of anti-VEGF and/or anti-TGF-β therapies to enhance cancer immunotherapy. Encouragingly, adopting anti-VEGF and/or anti-TGF-β therapies as a combination treatment with anti-PD-(L)1 therapy have been demonstrated as effective and tolerable in several solid tumors in clinical trials. Although several questions need to be solved, the clinical value of this combination strategy is worthy to be studied further.
Collapse
Affiliation(s)
- Linwei Li
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qinglian Wen
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Qinglian Wen, ; Ruilin Ding,
| | - Ruilin Ding
- Institute of Drug Clinical Trial/GCP Center, Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Qinglian Wen, ; Ruilin Ding,
| |
Collapse
|
29
|
Marvin DL, Spaans VM, de Kroon CD, Slieker RC, Khelil M, Ten Dijke P, Ritsma L, Jordanova ES. Low Transforming Growth Factor-β Pathway Activity in Cervical Adenocarcinomas. Front Oncol 2022; 12:797453. [PMID: 35756604 PMCID: PMC9213724 DOI: 10.3389/fonc.2022.797453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 05/12/2022] [Indexed: 12/24/2022] Open
Abstract
Cervical cancer is the fourth most common cancer in women worldwide. Squamous cell carcinoma (SCC) and adenocarcinoma (AC) are the most common histological types, with AC patients having worse prognosis. Over the last two decades, incidence rates of AC have increased, highlighting the importance of further understanding AC tumorigenesis, and the need to investigate new treatment options. The cytokine TGF-β functions as a tumour suppressor in healthy tissue. However, in tumour cells this suppressive function can be overcome. Therefore there is an increasing interest in using TGF-β inhibitors in the treatment of cancer. Here, we hypothesize that TGF-β plays a different role in SCC and AC. Analysis of RNA-seq data from the TCGA, using a TGF-β response signature, resulted in separate clustering of the two subtypes. We further investigated the expression of TGF-β-signalling related proteins (TβR1/2, SMAD4, pSMAD2, PAI-1, αvβ6 and MMP2/9) in a cohort of 62 AC patients. Low TβR2 and SMAD4 expression was associated with worse survival in AC patients and interestingly, high PAI-1 and αvβ6 expression was also correlated with worse survival. Similar correlations of TβR2, PAI-1 and αvβ6 with clinical parameters were found in previously reported SCC analyses. However, when comparing expression levels between SCC and AC patient samples, pSMAD2, SMAD4, PAI-1 and αvβ6 showed lower expression in AC compared to SCC. Because of the low expression of core TβR1/2, (p-)SMAD2 and SMAD4 proteins and the correlation with worse prognosis, TGF-β pathway most likely leads to tumour inhibitory effects in AC and therefore the use of TGF-β inhibitors would not be recommended. However, given the correlation of PAI-1 and αvβ6 with poor prognosis, the use of TGF- β inhibitors might be of interest in SCC and in the subsets of AC patients with high expression of these TGF-β associated proteins.
Collapse
Affiliation(s)
- Dieuwke L Marvin
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Vivian M Spaans
- Department of Gynaecology and Obstetrics, Leiden University Medical Center, Leiden, Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Cor D de Kroon
- Department of Gynaecology and Obstetrics, Leiden University Medical Center, Leiden, Netherlands
| | - Roderick C Slieker
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.,Department of Epidemiology and Data Science, Amsterdam University Medical Center (UMC), location VU University Medical Center (VUmc), Amsterdam, Netherlands
| | - Maryam Khelil
- Department of Gynaecology and Obstetrics, Center Gynaecological Oncology Amsterdam, Amsterdam University Medical Center (UMC), location VU University Medical Center (VUmc), Amsterdam, Netherlands
| | - Peter Ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Laila Ritsma
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Ekaterina S Jordanova
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands.,Department of Gynaecology and Obstetrics, Center Gynaecological Oncology Amsterdam, Amsterdam University Medical Center (UMC), location VU University Medical Center (VUmc), Amsterdam, Netherlands.,Department of Urology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
30
|
Corvino D, Kumar A, Bald T. Plasticity of NK cells in Cancer. Front Immunol 2022; 13:888313. [PMID: 35619715 PMCID: PMC9127295 DOI: 10.3389/fimmu.2022.888313] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/12/2022] [Indexed: 12/19/2022] Open
Abstract
Natural killer (NK) cells are crucial to various facets of human immunity and function through direct cytotoxicity or via orchestration of the broader immune response. NK cells exist across a wide range of functional and phenotypic identities. Murine and human studies have revealed that NK cells possess substantial plasticity and can alter their function and phenotype in response to external signals. NK cells also play a critical role in tumor immunity and form the basis for many emerging immunotherapeutic approaches. NK cells can directly target and lyse malignant cells with their inherent cytotoxic capabilities. In addition to direct targeting of malignant cells, certain subsets of NK cells can mediate antibody-dependent cellular cytotoxicity (ADCC) which is integral to some forms of immune checkpoint-blockade immunotherapy. Another important feature of various NK cell subsets is to co-ordinate anti-tumor immune responses by recruiting adaptive and innate leukocytes. However, given the diverse range of NK cell identities it is unsurprising that both pro-tumoral and anti-tumoral NK cell subsets have been described. Here, NK cell subsets have been shown to promote angiogenesis, drive inflammation and immune evasion in the tumor microenvironment. To date, the signals that drive tumor-infiltrating NK cells towards the acquisition of a pro- or anti-tumoral function are poorly understood. The notion of tumor microenvironment-driven NK cell plasticity has substantial implications for the development of NK-based immunotherapeutics. This review will highlight the current knowledge of NK cell plasticity pertaining to the tumor microenvironment. Additionally, this review will pose critical and relevant questions that need to be addressed by the field in coming years.
Collapse
Affiliation(s)
- Dillon Corvino
- Tumor-Immunobiology, Institute for Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Ananthi Kumar
- Tumor-Immunobiology, Institute for Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Tobias Bald
- Tumor-Immunobiology, Institute for Experimental Oncology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
31
|
Pu Y, Ji Q. Tumor-Associated Macrophages Regulate PD-1/PD-L1 Immunosuppression. Front Immunol 2022; 13:874589. [PMID: 35592338 PMCID: PMC9110638 DOI: 10.3389/fimmu.2022.874589] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/06/2022] [Indexed: 12/17/2022] Open
Abstract
Anti-programmed cell death 1 (PD-1) or anti-PD-ligand (L) 1 drugs, as classic immune checkpoint inhibitors, are considered promising treatment strategies for tumors. In clinical practice, some cancer patients experience drug resistance and disease progression in the process of anti-PD-1/PD-L1 immunotherapy. Tumor-associated macrophages (TAMs) play key roles in regulating PD-1/PD-L1 immunosuppression by inhibiting the recruitment and function of T cells through cytokines, superficial immune checkpoint ligands, and exosomes. There are several therapies available to recover the anticancer efficacy of PD-1/PD-L1 inhibitors by targeting TAMs, including the inhibition of TAM differentiation and re-education of TAM activation. In this review, we will summarize the roles and mechanisms of TAMs in PD-1/PD-L1 blocker resistance. Furthermore, we will discuss the therapies that were designed to deplete TAMs, re-educate TAMs, and intervene with chemokines secreted by TAMs and exosomes from M1 macrophages, providing more potential options to improve the efficacy of PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Yunzhou Pu
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qing Ji
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
32
|
Ogasawara M, Yamasaki-Yashiki S, Hamada M, Yamaguchi-Miyamoto T, Kawasuji T, Honda H, Yanagibashi T, Ikutani M, Watanabe Y, Fujimoto R, Matsunaga T, Nakajima N, Nagai Y, Takatsu K. Betulin Attenuates TGF-β1- and PGE 2-Mediated Inhibition of NK Cell Activity to Suppress Tumor Progression and Metastasis in Mice. Biol Pharm Bull 2022; 45:339-353. [PMID: 35228400 DOI: 10.1248/bpb.b21-00921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Transforming growth factor (TGF)-β1 and prostaglandin E2 (PGE2) are humoral factors critically involved in the induction of immunosuppression in the microenvironment of various types of tumors, including melanoma. In this study, we identified a natural compound that attenuated TGF-β1- and PGE2-induced immunosuppression and examined its effect on B16 melanoma growth in mice. By screening 502 natural compounds for attenuating activity against TGF-β1- or PGE2-induced suppression of cytolysis in poly(I:C)-stimulated murine splenocytes, we found that betulin was the most potent compound. Betulin also reduced TGF-β1- and PGE2-induced downregulation of perforin and granzyme B mRNA expression and cell surface expression of NKG2D and CD69 in natural killer (NK) cells. Cell depletion and coculture experiments showed that NK cells, dendritic cells, B cells, and T cells were necessary for the attenuating effects of betulin. Structure-activity relationship analysis revealed that two hydroxyl groups at positions C3 and C28 of betulin, their cis-configuration, and methyl group at C30 played crucial roles in its attenuating activity. In a subcutaneous implantation model of B16 melanoma in mice, intratumor administration of betulin and LY2157299, a TGF-β1 type I receptor kinase inhibitor, significantly retarded the growth of B16 melanoma. Notably, betulin increased significantly the number of CD69 positive NK cells in tumor sites at early stages of post-tumor cell injection. Our data suggest that betulin inhibits the growth of B16 melanoma by enhancing NK cell activity through attenuating the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Masaru Ogasawara
- Toyama Prefectural Institute for Pharmaceutical Research.,Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama
| | | | - Masahiro Hamada
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University
| | | | - Toru Kawasuji
- Toyama Prefectural Institute for Pharmaceutical Research
| | - Hiroe Honda
- Toyama Prefectural Institute for Pharmaceutical Research.,Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama
| | - Tsutomu Yanagibashi
- Toyama Prefectural Institute for Pharmaceutical Research.,Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama
| | - Masashi Ikutani
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama.,Graduate School of Integrated Sciences for Life, Hiroshima University.,Department of Immune Regulation, Research Institute, National Center for Global Health and Medicine
| | - Yasuharu Watanabe
- Toyama Prefectural Institute for Pharmaceutical Research.,Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama
| | - Ryota Fujimoto
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University
| | | | - Noriyuki Nakajima
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University
| | - Yoshinori Nagai
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama.,Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University
| | - Kiyoshi Takatsu
- Toyama Prefectural Institute for Pharmaceutical Research.,Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama
| |
Collapse
|
33
|
Tschernia NP, Gulley JL. Tumor in the Crossfire: Inhibiting TGF-β to Enhance Cancer Immunotherapy. BioDrugs 2022; 36:153-180. [PMID: 35353346 PMCID: PMC8986721 DOI: 10.1007/s40259-022-00521-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2022] [Indexed: 02/04/2023]
Abstract
Cancer immunotherapy using monoclonal antibodies targeting immune checkpoints has undoubtedly revolutionized the cancer treatment landscape in the last decade. Immune checkpoint inhibitors can elicit long-lasting, previously unheard-of responses in a number of tumor entities. Yet, even in such tumors as metastatic melanoma and non-small cell-lung cancer, in which immune checkpoint inhibition has become the first-line treatment of choice, only a minority of patients will benefit considerably from these treatments. This has been attributed to a number of factors, including an immune-suppressive tumor microenvironment (TME). Using different modalities to break these barriers is of utmost importance to expand the population of patients that benefit from immune checkpoint inhibition. The multifunctional cytokine transforming growth factor-β (TGF-β) has long been recognized as an immune-suppressive factor in the TME. A considerable number of drugs have been developed to target TGF-β, yet most of these have since been discontinued. The combination of anti-TGF-β agents with immune checkpoint inhibitors now has the potential to revive this target as a viable immunomodulatory therapeutic approach. Currently, a limited number of small molecular inhibitor and monoclonal antibody candidates that target TGF-β are in clinical development in combination with the following immune checkpoint inhibitors: SRK 181, an antibody inhibiting the activation of latent TGF-β1; NIS 793, a monoclonal antibody targeting TGF-β; and SHR 1701, a fusion protein consisting of an anti-PD-L1 monoclonal antibody fused with the extracellular domain of human TGF-β receptor II. Several small molecular inhibitors are also in development and are briefly reviewed: LY364947, a pyrazole-based small molecular inhibitor of the serine-threonine kinase activity of TGFβRI; SB-431542, an inhibitor targeting several TGF-β superfamily Type I activin receptor-like kinases as well as TGF-β1-induced nuclear Smad3 localization; and galunisertib, an oral small molecular inhibitor of the TGFβRI kinase. One of the most advanced agents in this area is bintrafusp alfa, a bifunctional fusion protein composed of the extracellular domain of TGF-β receptor II fused to a human IgG1 mAb blocking PD-L1. Bintrafusp alfa is currently in advanced clinical development and as an agent in this space with the most clinical experience, is a focused highlight of this review.
Collapse
Affiliation(s)
- Nicholas P Tschernia
- Genitourinary Malignancies Branch, Medical Oncology Service, National Cancer Institute, Building 10, Room 13N240, Bethesda, MD, 20892, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, Medical Oncology Service, National Cancer Institute, Building 10, Room 13N240, Bethesda, MD, 20892, USA.
| |
Collapse
|
34
|
Transforming growth factor-beta (TGF-β) in prostate cancer: A dual function mediator? Int J Biol Macromol 2022; 206:435-452. [PMID: 35202639 DOI: 10.1016/j.ijbiomac.2022.02.094] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 12/14/2022]
Abstract
Transforming growth factor-beta (TGF-β) is a member of a family of secreted cytokines with vital biological functions in cells. The abnormal expression of TGF-β signaling is a common finding in pathological conditions, particularly cancer. Prostate cancer (PCa) is one of the leading causes of death among men. Several genetic and epigenetic alterations can result in PCa development, and govern its progression. The present review attempts to shed some light on the role of TGF-β signaling in PCa. TGF-β signaling can either stimulate or inhibit proliferation and viability of PCa cells, depending on the context. The metastasis of PCa cells is increased by TGF-β signaling via induction of EMT and MMPs. Furthermore, TGF-β signaling can induce drug resistance of PCa cells, and can lead to immune evasion via reducing the anti-tumor activity of cytotoxic T cells and stimulating regulatory T cells. Upstream mediators such as microRNAs and lncRNAs, can regulate TGF-β signaling in PCa. Furthermore, some pharmacological compounds such as thymoquinone and valproic acid can suppress TGF-β signaling for PCa therapy. TGF-β over-expression is associated with poor prognosis in PCa patients. Furthermore, TGF-β up-regulation before prostatectomy is associated with recurrence of PCa. Overall, current review discusses role of TGF-β signaling in proliferation, metastasis and therapy response of PCa cells and in order to improve knowledge towards its regulation, upstream mediators of TGF-β such as non-coding RNAs are described. Finally, TGF-β regulation and its clinical application are discussed.
Collapse
|
35
|
Billmeier A, Khinvasara K, Lang F, Mohr J, Reidenbach D, Schork M, Yildiz I, Diken M. CIMT 2021: report on the 18th Annual Meeting of the Association for Cancer Immunotherapy. Hum Vaccin Immunother 2022; 18:2024416. [PMID: 35130105 PMCID: PMC8993083 DOI: 10.1080/21645515.2021.2024416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
After one year of absence, the 18th Annual Meeting of the Association for Cancer Immunotherapy (CIMT), Europe’s cancer immunotherapy meeting, took place virtually from 10 to 12 May 2021. Over 850 academic and clinical professionals from 30 countries met to discuss the recent advancements in cancer immunotherapy and the current progress in COVID19-related research. This meeting report summarizes the highlights of CIMT2021.
Collapse
Affiliation(s)
| | - Krutika Khinvasara
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Franziska Lang
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | | | - Daniel Reidenbach
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Maik Schork
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Ikra Yildiz
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - Mustafa Diken
- BioNTech SE, Mainz, Germany.,TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| |
Collapse
|
36
|
Arany PR. Photobiomodulation-Activated Latent Transforming Growth Factor-β1: A Critical Clinical Therapeutic Pathway and an Endogenous Optogenetic Tool for Discovery. Photobiomodul Photomed Laser Surg 2022; 40:136-147. [PMID: 34905400 DOI: 10.1089/photob.2021.0109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objective: The central role of the TGF-β pathway in embryonic development, immune responses, tissue healing, and malignancies is well established. Prior attempts with small molecules, peptides, and regulatory RNAs have failed mainly due to off-target effects in clinical studies. This review outlines the evidence for selectively activating the endogenous, latent transforming growth factor (TGF)-β1 with photobiomodulation (PBM) treatments. Background: Light treatments play a central role in current-directed energy therapeutics in medicine. Therapeutic use of low-dose light treatments has been noted since the 1960s. However, the breadth of treatments and inconsistencies with clinical outcomes have led to much skepticism. This can be primarily attributed to a lack of understanding of the fundamental light-tissue interactions and optimization of clinical treatment protocols. Methods: Recent advances in molecular mechanisms and improved biophotonic device technologies have led to a resurgence of interest in this field. Results: Over the past two decades, our work has focused on outlining a direct molecular mechanism involving PBM-generated redox-mediated activation of endogenous latent TGF-β1. Conclusions: Despite its critical roles in these processes, the complexity and cross talk in this potent growth factor signaling network have prevented the development of directed targeted therapeutics. PBM treatments offer a novel therapeutic and discovery tool in this aspect, especially with the growing evidence for its roles in cancer immunotherapy and stem cell biology.
Collapse
Affiliation(s)
- Praveen R Arany
- Department of Oral Biology, Surgery and Biomedical Engineering, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
37
|
Mortezaee K, Majidpoor J. (Im)maturity in Tumor Ecosystem. Front Oncol 2022; 11:813897. [PMID: 35145911 PMCID: PMC8821092 DOI: 10.3389/fonc.2021.813897] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/27/2021] [Indexed: 01/10/2023] Open
Abstract
Tumors have special features that make them distinct from their normal counterparts. Immature cells in a tumor mass and their critical contributions to the tumorigenesis will open new windows toward cancer therapy. Incomplete cellular development brings versatile and unique functionality in the cellular tumor ecosystem, such as what is seen for highly potential embryonic cells. There is evidence that maturation of certain types of cells in this ecosystem can recover the sensitivity of the tumor. Therefore, understanding more about the mechanisms that contributed to this immaturity will render new therapeutic approaches in cancer therapy. Targeting such mechanisms can be exploited as a supplementary to the current immunotherapeutic treatment schedules, such as immune checkpoint inhibitor (ICI) therapy. The key focus of this review is to discuss the impact of (im)maturity in cellular tumor ecosystems on cancer progression, focusing mainly on immaturity in the immune cell compartment of the tumor, as well as on the stemness of tumor cells.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
38
|
Mirlekar B. Tumor promoting roles of IL-10, TGF-β, IL-4, and IL-35: Its implications in cancer immunotherapy. SAGE Open Med 2022; 10:20503121211069012. [PMID: 35096390 PMCID: PMC8793114 DOI: 10.1177/20503121211069012] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/07/2021] [Indexed: 12/23/2022] Open
Abstract
Cytokines play a critical role in regulating host immune response toward cancer and determining the overall fate of tumorigenesis. The tumor microenvironment is dominated mainly by immune-suppressive cytokines that control effector antitumor immunity and promote survival and the proliferation of cancer cells, which ultimately leads to enhanced tumor growth. In addition to tumor cells, the heterogeneous immune cells present within the tumor milieu are the significant source of immune-suppressive cytokines. These cytokines are classified into a broad range; however, in most tumor types, the interleukin-10, transforming growth factor-β, interleukin-4, and interleukin-35 are consistently reported as immune-suppressive cytokines that help tumor growth and metastasis. The most emerging concern in cancer treatment is hijacking and restraining the activity of antitumor immune cells in the tumor niche due to a highly immune-suppressive environment. This review summarizes the role and precise functions of interleukin-10, transforming growth factor-β, interleukin-4, and interleukin-35 in modulating tumor immune contexture and its implication in developing effective immune-therapeutic approaches. CONCISE CONCLUSION Recent effort geared toward developing novel immune-therapeutic approaches faces significant challenges due to sustained mutations in tumor cells and a highly immune-suppressive microenvironment present within the tumor milieu. The cytokines play a crucial role in developing an immune-suppressive environment that ultimately dictates the fate of tumorigenesis. This review critically covers the novel aspects of predominant immune-suppressive cytokines such as interleukin-10, transforming growth factor-β, interleukin-4, and interleukin-35 in dictating the fate of tumorigenesis and how targeting these cytokines can help the development of better immune-therapeutic drug regimens for the treatment of cancer.
Collapse
Affiliation(s)
- Bhalchandra Mirlekar
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
39
|
Abstract
Transforming growth factor-β (TGFβ) isoforms are upregulated and activated in myocardial diseases and have an important role in cardiac repair and remodelling, regulating the phenotype and function of cardiomyocytes, fibroblasts, immune cells and vascular cells. Cardiac injury triggers the generation of bioactive TGFβ from latent stores, through mechanisms involving proteases, integrins and specialized extracellular matrix (ECM) proteins. Activated TGFβ signals through the SMAD intracellular effectors or through non-SMAD cascades. In the infarcted heart, the anti-inflammatory and fibroblast-activating actions of TGFβ have an important role in repair; however, excessive or prolonged TGFβ signalling accentuates adverse remodelling, contributing to cardiac dysfunction. Cardiac pressure overload also activates TGFβ cascades, which initially can have a protective role, promoting an ECM-preserving phenotype in fibroblasts and preventing the generation of injurious, pro-inflammatory ECM fragments. However, prolonged and overactive TGFβ signalling in pressure-overloaded cardiomyocytes and fibroblasts can promote cardiac fibrosis and dysfunction. In the atria, TGFβ-mediated fibrosis can contribute to the pathogenic substrate for atrial fibrillation. Overactive or dysregulated TGFβ responses have also been implicated in cardiac ageing and in the pathogenesis of diabetic, genetic and inflammatory cardiomyopathies. This Review summarizes the current evidence on the role of TGFβ signalling in myocardial diseases, focusing on cellular targets and molecular mechanisms, and discussing challenges and opportunities for therapeutic translation.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
40
|
Lin H, Xia L, Lian J, Chen Y, Zhang Y, Zhuang Z, Cai H, You J, Guan G. Delineation of colorectal cancer ligand-receptor interactions and their roles in the tumor microenvironment and prognosis. J Transl Med 2021; 19:497. [PMID: 34876143 PMCID: PMC8650275 DOI: 10.1186/s12967-021-03162-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 11/22/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Immunotherapies targeting ligand-receptor interactions (LRIs) are advancing rapidly in the treatment of colorectal cancer (CRC), and LRIs also affect many aspects of CRC development. However, the pattern of LRIs in CRC and their effect on tumor microenvironment and clinical value are still unclear. METHODS We delineated the pattern of LRIs in 55,539 single-cell RNA sequencing (scRNA-seq) samples from 29 patients with CRC and three bulk RNA-seq datasets containing data from 1411 CRC patients. Then the influence of tumor microenvironment, immunotherapy and prognosis of CRC patients were comprehensively investigated. RESULTS We calculated the strength of 1893 ligand-receptor pairs between 25 cell types to reconstruct the spatial structure of CRC. We identified tumor subtypes based on LRIs, revealed the relationship between the subtypes and immunotherapy efficacy and explored the ligand-receptor pairs and specific targets affecting the abundance of tumor-infiltrating lymphocytes. Finally, a prognostic model based on ligand-receptor pairs was constructed and validated. CONCLUSION Overall, through the comprehensive and in-depth investigation of the existing ligand-receptor pairs, this study provides new ideas for CRC subtype classification, a new risk screening tool for CRC patients, and potential ligand-receptor pair targets and pathways for CRC therapy.
Collapse
Affiliation(s)
- Hexin Lin
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, 350001, Fujian, China
| | - Lu Xia
- Xiamen Cell Therapy Research Center, The First Affiliated Hospital of Xiamen University. School of Medicine, Xiamen University, Xiamen, China
| | - Jiabian Lian
- Department of Laboratory Medicine, Xiamen Key Laboratory of Genetic Testing, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yinan Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yiyi Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, 350001, Fujian, China
| | - Zhicheng Zhuang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, 350001, Fujian, China
| | - HuaJun Cai
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, 350001, Fujian, China
| | - Jun You
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China.,School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Guoxian Guan
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, 350001, Fujian, China.
| |
Collapse
|
41
|
Gulley JL, Schlom J, Barcellos-Hoff MH, Wang XJ, Seoane J, Audhuy F, Lan Y, Dussault I, Moustakas A. Dual inhibition of TGF-β and PD-L1: a novel approach to cancer treatment. Mol Oncol 2021; 16:2117-2134. [PMID: 34854206 PMCID: PMC9168966 DOI: 10.1002/1878-0261.13146] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/03/2021] [Accepted: 11/30/2021] [Indexed: 11/11/2022] Open
Abstract
Transforming growth factor β (TGF-β) and programmed death ligand 1 (PD-L1) initiate signaling pathways with complementary, nonredundant immunosuppressive functions in the tumor microenvironment (TME). In the TME, dysregulated TGF-β signaling suppresses antitumor immunity and promotes cancer fibrosis, epithelial-to-mesenchymal transition and angiogenesis. Meanwhile, PD-L1 expression inactivates cytotoxic T cells and restricts immunosurveillance in the TME. Anti-PD-L1 therapies have been approved for the treatment of various cancers, but TGF-β signaling in the TME is associated with resistance to these therapies. In this Review, we discuss the importance of the TGF-β and PD-L1 pathways in cancer, as well as clinical strategies using combination therapies that block these pathways separately or approaches with dual-targeting agents (bispecific and bifunctional immunotherapies) that may block them simultaneously. Currently, the furthest developed dual-targeting agent is bintrafusp alfa. This drug is a first-in-class bifunctional fusion protein that consists of the extracellular domain of the TGF-βRII receptor (a TGF-β "trap") fused to a human immunoglobulin G1 (IgG1) monoclonal antibody blocking PD-L1. Given the immunosuppressive effects of the TGF-β and PD-L1 pathways within the TME, colocalized and simultaneous inhibition of these pathways may potentially improve clinical activity and reduce toxicity.
Collapse
Affiliation(s)
- James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Xiao-Jing Wang
- Department of Pathology, University of Colorado, Aurora, CO, USA
| | - Joan Seoane
- ICREA, Vall D'Hebron Institute of Oncology, Universitat Autonoma de Barcelona, CIBERONC, Barcelona, Spain
| | | | - Yan Lan
- EMD Serono, Billerica, MA, USA
| | - Isabelle Dussault
- EMD Serono, Billerica, MA, USA.,Current affiliation: Fusion Pharmaceuticals, Boston, MA, USA
| | - Aristidis Moustakas
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
42
|
A four immune-related long noncoding RNAs signature as predictors for cervical cancer. Hum Cell 2021; 35:348-359. [PMID: 34846702 DOI: 10.1007/s13577-021-00654-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/22/2021] [Indexed: 10/19/2022]
Abstract
The progression, metastasis, and prognosis of cervical cancer (CC) is influenced by the tumor immune microenvironment. Studies proved that long non-coding RNAs (lncRNAs) to engage in cervical cancer development, especially immune-related lncRNAs, have emerged crucial in the tumor immune process. This study was set out to identify an immune-related lncRNA signature. In total, 13,838 lncRNA expression profiles and 328 immune genes were acquired from the clnical data of 306 CC tissues and 3 non-CC tissues. From the 433 identified immune-related lncRNAs, 4 candidate immune-related lncRNAs (SOX21-AS1, AC005332.4, NCK1-DT, LINC01871) were considered independent indicators of cervical cancer prognosis through the univariate and multivariate Cox regression analysis, and they were used to construct a prognostic and survival lncRNA signature model followed by the bootstrap method for further verification. Kaplan-Meier curves illustrated that cervical cancer patients could be divided into high-risk and low-risk groups with significant differences (P = 2.052e - 05), and the discrepancy of immune profiles between these two risk groups was illustrated by principal components analysis. Taken together, the novel survival predictive model created by the four immune-related lncRNAs showed promising clinical prediction value in cervical cancer.
Collapse
|
43
|
Trivedi T, Pagnotti GM, Guise TA, Mohammad KS. The Role of TGF-β in Bone Metastases. Biomolecules 2021; 11:1643. [PMID: 34827641 PMCID: PMC8615596 DOI: 10.3390/biom11111643] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
Complications associated with advanced cancer are a major clinical challenge and, if associated with bone metastases, worsen the prognosis and compromise the survival of the patients. Breast and prostate cancer cells exhibit a high propensity to metastasize to bone. The bone microenvironment is unique, providing fertile soil for cancer cell propagation, while mineralized bone matrices store potent growth factors and cytokines. Biologically active transforming growth factor β (TGF-β), one of the most abundant growth factors, is released following tumor-induced osteoclastic bone resorption. TGF-β promotes tumor cell secretion of factors that accelerate bone loss and fuel tumor cells to colonize. Thus, TGF-β is critical for driving the feed-forward vicious cycle of tumor growth in bone. Further, TGF-β promotes epithelial-mesenchymal transition (EMT), increasing cell invasiveness, angiogenesis, and metastatic progression. Emerging evidence shows TGF-β suppresses immune responses, enabling opportunistic cancer cells to escape immune checkpoints and promote bone metastases. Blocking TGF-β signaling pathways could disrupt the vicious cycle, revert EMT, and enhance immune response. However, TGF-β's dual role as both tumor suppressor and enhancer presents a significant challenge in developing therapeutics that target TGF-β signaling. This review presents TGF-β's role in cancer progression and bone metastases, while highlighting current perspectives on the therapeutic potential of targeting TGF-β pathways.
Collapse
Affiliation(s)
- Trupti Trivedi
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
| | - Gabriel M. Pagnotti
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
| | - Theresa A. Guise
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
| | - Khalid S. Mohammad
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
44
|
TIMP1 and TIMP2 Downregulate TGFβ Induced Decidual-like Phenotype in Natural Killer Cells. Cancers (Basel) 2021; 13:cancers13194955. [PMID: 34638439 PMCID: PMC8507839 DOI: 10.3390/cancers13194955] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Cancer patients are characterized by NK cells with altered surface markers, such as CD56 brightness, CD9, CD49a (pro-angiogenic) and PD-1, and TIM-3 (exhaustion), that favor immune escape. Transforming growth factor-beta (TGFβ) is a major tumor-derived cytokine that favors cancer growth and supports pro-angiogenic activities in NK cells by inducing pro-angiogenic molecules. TIMP-1 and TIMP-2 play a crucial role in extracellular matrix (ECM) regulation, wound healing, pregnancy and cancer, and there is increasing evidence that they are immune-modulatory. We found that recombinant TIMP-1 and -2 can partially contrast the induction of pro-tumor/pro-angiogenic decidual-like polarization of NK cells by TGFβ. Abstract Natural Killer (NK) cells have been found to be anergic, exhausted and pro-angiogenic in cancers. NK cell from healthy donors, exposed to TGFβ, acquire the CD56brightCD9+CD49a+ decidual-like-phenotype, together with decreased levels of NKG2D activation marker, increased levels of TIM-3 exhaustion marker, similar to cancer-associated NK cells. Tissue inhibitors of metalloproteases (TIMPs) exert dual roles in cancer. The role of TIMPs in modulating immune cells is a very novel concept, and the present is the first report studying their ability to contrast TGFβ action on NK cells. Here, we investigated the effects of TIMP1 and TIMP2 recombinant proteins in hindering decidual-like markers in NK cells, generated by polarizing cytolytic NK cells with TGFβ. The effects of TIMP1 or TIMP2 on NK cell surface antigens were determined by multicolor flow cytometry. We found that TIMP1 and TIMP2 were effective in interfering with TGFβ induced NK cell polarization towards a decidual-like-phenotype. TIMP1 and TIMP2 counteracted the effect of TGFβ in increasing the percentage of CD56bright, CD16−, CD9+ and CD49a+, and restoring normal levels for TIMP 1 and 2 also inhibited decrease levels of the activation marker NKG2D induced by TGFβ and decreased the TGFβ upregulated exhaustion marker TIM-3. NK cell degranulation capabilities against K562 cells were also decreased by TGFβ and not by TIMP1 or TIMP2. TIMP1 treatment could partially restore degranulation marker CD107a expression. Treatment with recombinant TIMP-1 or TIMP-2 showed a trend, although not statistically significant, to decrease CD49a+ and TIM-3+ expression and increase NKG2D in peripheral blood NK cells exposed to conditioned media from colon cancer cell lines. Our results suggest a potential role of TIMPs in controlling the tumor-associated cytokine TGFβ-induced NK cell polarization. Given the heterogeneity of released factors within the TME, it is clear that TGFβ stimulation represents a model to prove TIMP’s new properties, but it cannot be envisaged as a soloist NK cell polarizing agent. Therefore, further studies from the scientific community will help defining TIMPs immunomodulatory activities of NK cells in cancer, and their possible future diagnostic–therapeutic roles.
Collapse
|
45
|
Tuleta I, Frangogiannis NG. Fibrosis of the diabetic heart: Clinical significance, molecular mechanisms, and therapeutic opportunities. Adv Drug Deliv Rev 2021; 176:113904. [PMID: 34331987 PMCID: PMC8444077 DOI: 10.1016/j.addr.2021.113904] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 01/02/2023]
Abstract
In patients with diabetes, myocardial fibrosis may contribute to the pathogenesis of heart failure and arrhythmogenesis, increasing ventricular stiffness and delaying conduction. Diabetic myocardial fibrosis involves effects of hyperglycemia, lipotoxicity and insulin resistance on cardiac fibroblasts, directly resulting in increased matrix secretion, and activation of paracrine signaling in cardiomyocytes, immune and vascular cells, that release fibroblast-activating mediators. Neurohumoral pathways, cytokines, growth factors, oxidative stress, advanced glycation end-products (AGEs), and matricellular proteins have been implicated in diabetic fibrosis; however, the molecular links between the metabolic perturbations and activation of a fibrogenic program remain poorly understood. Although existing therapies using glucose- and lipid-lowering agents and neurohumoral inhibition may act in part by attenuating myocardial collagen deposition, specific therapies targeting the fibrotic response are lacking. This review manuscript discusses the clinical significance, molecular mechanisms and cell biology of diabetic cardiac fibrosis and proposes therapeutic targets that may attenuate the fibrotic response, preventing heart failure progression.
Collapse
Affiliation(s)
- Izabela Tuleta
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY, USA
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY, USA.
| |
Collapse
|
46
|
Kondratyeva L, Chernov I, Kopantzev E, Didych D, Kuzmich A, Alekseenko I, Kostrov S, Sverdlov E. Pancreatic Lineage Specifier PDX1 Increases Adhesion and Decreases Motility of Cancer Cells. Cancers (Basel) 2021; 13:cancers13174390. [PMID: 34503200 PMCID: PMC8430990 DOI: 10.3390/cancers13174390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 12/16/2022] Open
Abstract
Intercellular interactions involving adhesion factors are key operators in cancer progression. In particular, these factors are responsible for facilitating cell migration and metastasis. Strengthening of adhesion between tumor cells and surrounding cells or extracellular matrix (ECM), may provide a way to inhibit tumor cell migration. Recently, we demonstrated that PDX1 ectopic expression results in the reduction of pancreatic cancer line PANC-1 cell motility in vitro and in vivo, and we now provide experimental data confirming the hypothesis that suppression of migration may be related to the effect of PDX1 on cell adhesion. Cell migration analyses demonstrated decreased motility of pancreatic Colo357 and PANC-1 cell lines expressing PDX1. We observed decreased expression levels of genes associated with promoting cell migration and increased expression of genes negatively affecting cell motility. Expression of the EMT regulator genes was only mildly induced in cells expressing PDX1 during the simulation of the epithelial-mesenchymal transition (EMT) by the addition of TGFβ1 to the medium. PDX1-expressing cancer cell lines showed increased cell adhesion to collagen type I, fibronectin, and poly-lysine. We conclude that ectopic expression of PDX1 reduces the migration potential of cancer cells, by increasing the adhesive properties of cells and reducing the sensitivity to TGFβ1-induced EMT.
Collapse
Affiliation(s)
- Liya Kondratyeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Ulitsa Miklukho-Maklaya, 117997 Moscow, Russia; (I.C.); (E.K.); (D.D.); (A.K.); (I.A.)
- Correspondence: (L.K.); (E.S.)
| | - Igor Chernov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Ulitsa Miklukho-Maklaya, 117997 Moscow, Russia; (I.C.); (E.K.); (D.D.); (A.K.); (I.A.)
| | - Eugene Kopantzev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Ulitsa Miklukho-Maklaya, 117997 Moscow, Russia; (I.C.); (E.K.); (D.D.); (A.K.); (I.A.)
| | - Dmitry Didych
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Ulitsa Miklukho-Maklaya, 117997 Moscow, Russia; (I.C.); (E.K.); (D.D.); (A.K.); (I.A.)
| | - Alexey Kuzmich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Ulitsa Miklukho-Maklaya, 117997 Moscow, Russia; (I.C.); (E.K.); (D.D.); (A.K.); (I.A.)
- Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Ploshchad’ Akademika Kurchatova, 123182 Moscow, Russia;
| | - Irina Alekseenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Ulitsa Miklukho-Maklaya, 117997 Moscow, Russia; (I.C.); (E.K.); (D.D.); (A.K.); (I.A.)
- Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Ploshchad’ Akademika Kurchatova, 123182 Moscow, Russia;
| | - Sergey Kostrov
- Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Ploshchad’ Akademika Kurchatova, 123182 Moscow, Russia;
| | - Eugene Sverdlov
- Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Ploshchad’ Akademika Kurchatova, 123182 Moscow, Russia;
- Correspondence: (L.K.); (E.S.)
| |
Collapse
|
47
|
van Oost S, Meijer DM, Kuijjer ML, Bovée JVMG, de Miranda NFCC. Linking Immunity with Genomics in Sarcomas: Is Genomic Complexity an Immunogenic Trigger? Biomedicines 2021; 9:1048. [PMID: 34440251 PMCID: PMC8391750 DOI: 10.3390/biomedicines9081048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022] Open
Abstract
Sarcomas comprise a collection of highly heterogeneous malignancies that can be grossly grouped in the categories of sarcomas with simple or complex genomes. Since the outcome for most sarcoma patients has barely improved in the last decades, there is an urgent need for improved therapies. Immunotherapy, and especially T cell checkpoint blockade, has recently been a game-changer in cancer therapy as it produced significant and durable treatment responses in several cancer types. Currently, only a small fraction of sarcoma patients benefit from immunotherapy, supposedly due to a general lack of somatically mutated antigens (neoantigens) and spontaneous T cell immunity in most cancers. However, genomic events resulting from chromosomal instability are frequent in sarcomas with complex genomes and could drive immunity in those tumors. Improving our understanding of the mechanisms that shape the immune landscape of sarcomas will be crucial to overcoming the current challenges of sarcoma immunotherapy. This review focuses on what is currently known about the tumor microenvironment in sarcomas and how this relates to their genomic features. Moreover, we discuss novel therapeutic strategies that leverage the tumor microenvironment to increase the clinical efficacy of immunotherapy, and which could provide new avenues for the treatment of sarcomas.
Collapse
Affiliation(s)
- Siddh van Oost
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (S.v.O.); (D.M.M.); (M.L.K.); (N.F.C.C.d.M.)
| | - Debora M. Meijer
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (S.v.O.); (D.M.M.); (M.L.K.); (N.F.C.C.d.M.)
| | - Marieke L. Kuijjer
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (S.v.O.); (D.M.M.); (M.L.K.); (N.F.C.C.d.M.)
- Centre for Molecular Medicine Norway (NCMM), Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
| | - Judith V. M. G. Bovée
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (S.v.O.); (D.M.M.); (M.L.K.); (N.F.C.C.d.M.)
| | - Noel F. C. C. de Miranda
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (S.v.O.); (D.M.M.); (M.L.K.); (N.F.C.C.d.M.)
| |
Collapse
|
48
|
Two Complementarity Immunotherapeutics in Non-Small-Cell Lung Cancer Patients-Mechanism of Action and Future Concepts. Cancers (Basel) 2021; 13:cancers13112836. [PMID: 34200219 PMCID: PMC8201041 DOI: 10.3390/cancers13112836] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/19/2021] [Accepted: 05/31/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Here, we focused on the most important mechanisms of action of combined immunotherapy with modern anticancer approaches in patients with non-small-cell lung cancer. This knowledge is extremely important for lung cancer clinicians. First, it facilitates proper involvement of the patient in the treatment and monitoring its effectiveness. More importantly, the knowledge of the immunotherapy mechanisms will certainly allow quick recognition of the side effects of such a therapy, which are totally different of those observed after chemotherapy. Side effects of combination therapies can occur at any stage of treatment, and even after completion thereof. This review article could particularly explain the mechanism of action of combined immunotherapy, which have different targets in patients. Abstract Due to the limited effectiveness of immunotherapy used as first-line monotherapy in patients with non-small-cell lung cancer (NSCLC), the concepts of combining classical immunotherapy based on immune checkpoint antibodies with other treatment methods have been developed. Pembrolizumab and atezolizumab were registered in combination with chemotherapy for the treatment of metastatic NSCLC, while durvalumab found its application in consolidation therapy after successful chemoradiotherapy in patients with locally advanced NSCLC. Exceptionally attractive, due to their relatively low toxicity and high effectiveness, are treatment approaches in which a combination of two different immunotherapy methods is applied. This method is based on observations from clinical trials in which nivolumab and ipilimumab were used as first-line therapy for advanced NSCLC. It turned out that the dual blockade of immune checkpoints activated T lymphocytes in different compartments of the immune response, at the same time affecting the downregulation of immune suppressor cells (regulatory T cells). These experiments not only resulted in the registration of combination therapy with nivolumab and ipilimumab, but also initiated other clinical trials using immune checkpoint inhibitors (ICIs) in combination with other ICIs or activators of costimulatory molecules found on immune cells. There are also studies in which ICIs are associated with molecules that modify the tumour environment. This paper describes the mechanism of the synergistic effect of a combination of different immunotherapy methods in NSCLC patients.
Collapse
|
49
|
Immunosuppressive Effects of Myeloid-Derived Suppressor Cells in Cancer and Immunotherapy. Cells 2021; 10:cells10051170. [PMID: 34065010 PMCID: PMC8150533 DOI: 10.3390/cells10051170] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/01/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
The primary function of myeloid cells is to protect the host from infections. However, during cancer progression or states of chronic inflammation, these cells develop into myeloid-derived suppressor cells (MDSCs) that play a prominent role in suppressing anti-tumor immunity. Overcoming the suppressive effects of MDSCs is a major hurdle in cancer immunotherapy. Therefore, understanding the mechanisms by which MDSCs promote tumor growth is essential for improving current immunotherapies and developing new ones. This review explores mechanisms by which MDSCs suppress T-cell immunity and how this impacts the efficacy of commonly used immunotherapies.
Collapse
|